WO2002085922A2 - Proteins and nucleic acids encoding same - Google Patents

Proteins and nucleic acids encoding same Download PDF

Info

Publication number
WO2002085922A2
WO2002085922A2 PCT/US2002/011634 US0211634W WO02085922A2 WO 2002085922 A2 WO2002085922 A2 WO 2002085922A2 US 0211634 W US0211634 W US 0211634W WO 02085922 A2 WO02085922 A2 WO 02085922A2
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
amino acid
protein
nucleic acid
seq
Prior art date
Application number
PCT/US2002/011634
Other languages
French (fr)
Other versions
WO2002085922A3 (en
Inventor
Carol E. A. Pena
Xiaojia Guo
Richard A. Shimkets
Muralidhara Padigaru
Ramesh Kekuda
Kimberly A. Spytek
Fuad Mehraban
James N. Topper
Uriel M. Malyankar
Scott Wasserman
R. Shlomit Edinger
Glennda Smithson
Erik Gunther
Laszlo Komuves
Original Assignee
Curagen Corporation
Millennium Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Curagen Corporation, Millennium Pharmaceuticals, Inc. filed Critical Curagen Corporation
Priority to EP02723842A priority Critical patent/EP1383533A4/en
Priority to CA002443770A priority patent/CA2443770A1/en
Publication of WO2002085922A2 publication Critical patent/WO2002085922A2/en
Publication of WO2002085922A3 publication Critical patent/WO2002085922A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals

Definitions

  • the invention relates to polynucleotides and the polypeptides encoded by such polynucleotides, as well as vectors, host cells, antibodies and recombinant methods for producing the polypeptides and polynucleotides, and methods for using the same.
  • the invention generally relates to nucleic acids and polypeptides encoded therefrom. More specifically, the invention relates to nucleic acids encoding cytoplasmic, nuclear, membrane bound, and secreted polypeptides, as well as vectors, host cells, antibodies, and recombinant methods for producing these nucleic acids and polypeptides.
  • Heart disease is the primary cause of death in most western societies. Death from heart disease is often induced by platelet-dependent ischemic syndromes which are initiated by atherosclerosis and arteriosclerosis and include, but are not limited to, acute myocardial infarction, clironic unstable angina, transient ischemic attacks and strokes, peripheral vascular disease, arterial thrombosis, preeclampsia, embolism, restenosis and/or thrombosis following angioplasty, carotid endarterectomy, anastomosis of vascular grafts, and chronic cardiovascular devices (e.g., in-dwelling catheters or shunts "extracorporeal circulating devices").
  • platelet-dependent ischemic syndromes which are initiated by atherosclerosis and arteriosclerosis and include, but are not limited to, acute myocardial infarction, clironic unstable angina, transient ischemic attacks and strokes, peripheral vascular disease, arterial thrombosis, preeclampsi
  • Thrombospondin-l-like proteins associate with the extracellular matrix and inhibits angiogenesis in vivo.
  • Thrombospondin-like proteins block capillary-like tube formation and endothelial cell proliferation.
  • the antiangiogenic activity is mediated by a region that contains 3 type 1 (properdin or thrombospondin) repeats.
  • Selectin-like proteins such as P-selectin, also called GMP-140, CD62, or selectin P, is a 140-kD adhesion molecule, expressed at the surface of activated cells, that mediates the interaction of activated endothelial cells or platelets with leukocytes.
  • the protein In endothelial cells, the protein is localized to the membranes of Weibel-Palade bodies, the intracellular storage granules for von Willebrand factor.
  • Many disease states are characterized by uncontrolled cell proliferation. These diseases involve a variety of cell types and include disorders such as cancer, psoriasis, pulmonary fibrosis, glomeralonepl ritis, atherosclerosis and restenosis following angioplasty.
  • Vital cellular functions such as cell proliferation and signal transduction are regulated in part by the balance between the activities of protein-tyrosine kinases (PTK) and protein-tyrosine phosphatases (PTPase). Oncogenesis can result from an imbalance.
  • PTK protein-tyrosine kinases
  • PPPase protein-tyrosine phosphatases
  • the invention is based in part upon the discovery of nucleic acid sequences encoding novel polypeptides.
  • novel nucleic acids and polypeptides are referred to herein as NONX, or ⁇ ON1, ⁇ ON2, ⁇ ON3, ⁇ ON4, ⁇ ON5, ⁇ ON6, ⁇ ON7, ⁇ ON8, ⁇ ON9, ⁇ ONlOa, ⁇ ONlOb, ⁇ ON11, ⁇ ON12, ⁇ ON13, ⁇ ON14, ⁇ ON15, and ⁇ ON16 nucleic acids and polypeptides.
  • nucleic acids and polypeptides as well as variants, derivatives, homologs, analogs and fragments thereof, will hereinafter be collectively designated as " ⁇ ONX” nucleic acid or polypeptide sequences.
  • the invention provides an isolated ⁇ ONX nucleic acid molecule encoding a ⁇ ONX polypeptide that includes a nucleic acid sequence that has identity to the nucleic acids disclosed in SEQ ID ⁇ OS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33.
  • the NOVX nucleic acid molecule will hybridize under stringent conditions to a nucleic acid sequence complementary to a nucleic acid molecule that includes a protein-coding sequence of a NONX nucleic acid sequence.
  • the invention also includes an isolated nucleic acid that encodes a ⁇ ONX polypeptide, or a fragment, homolog, analog or derivative thereof.
  • the nucleic acid can encode a polypeptide at least 80% identical to a polypeptide comprising the amino acid sequences of SEQ ID ⁇ OS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34.
  • the nucleic acid can be, for example, a genomic DNA fragment or a cDNA molecule that includes the nucleic acid sequence of any of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33.
  • an oligonucleotide e.g., an oligonucleotide which includes at least 6 contiguous nucleotides of a NONX nucleic acid (e.g., SEQ ID ⁇ OS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33) or a complement of said oligonucleotide.
  • a NONX nucleic acid e.g., SEQ ID ⁇ OS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33
  • NONX polypeptides SEQ ID ⁇ OS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34.
  • the NONX polypeptides include an amino acid sequence that is substantially identical to the amino acid sequence of a human ⁇ ONX polypeptide.
  • the invention also features antibodies that immunoselectively bind to ⁇ ONX polypeptides, or fragments, homologs, analogs or derivatives thereof.
  • the invention includes pharmaceutical compositions that include therapeutically- or prophylactically-effective amounts of a therapeutic and a pharmaceutically- acceptable carrier.
  • the therapeutic can be, e.g., a ⁇ ONX nucleic acid, a ⁇ ONX polypeptide, or an antibody specific for a ⁇ OVX polypeptide.
  • the invention includes, in one or more containers, a therapeutically- or prophylactically-effective amount of this pharmaceutical composition.
  • the invention includes a method of producing a polypeptide by culturing a cell that includes a ⁇ ONX nucleic acid, under conditions allowing for expression of the ⁇ ONX polypeptide encoded by the D ⁇ A. If desired, the ⁇ ONX polypeptide can then be recovered.
  • the invention includes a method of detecting the presence of a
  • ⁇ ONX polypeptide in a sample in a sample.
  • a sample is contacted with a compound that selectively binds to the polypeptide under conditions allowing for formation of a complex between the polypeptide and the compound.
  • the complex is detected, if present, thereby identifying the ⁇ ONX polypeptide within the sample.
  • the invention also includes methods to identify specific cell or tissue types based on their expression of a ⁇ ONX.
  • Also included in the invention is a method of detecting the presence of a ⁇ ONX nucleic acid molecule in a sample by contacting the sample with a ⁇ ONX nucleic acid probe or primer, and detecting whether the nucleic acid probe or primer bound to a ⁇ ONX nucleic acid molecule in the sample.
  • the invention provides a method for modulating the activity of a ⁇ ONX polypeptide by contacting a cell sample that includes the ⁇ ONX polypeptide with a compound that binds to the ⁇ ONX polypeptide in an amount sufficient to modulate the activity of said polypeptide.
  • the compound can be, e.g., a small molecule, such as a nucleic acid, peptide, polypeptide, peptidomimetic, carbohydrate, lipid or other organic (carbon containing) or inorganic molecule, as further described herein.
  • a therapeutic in the manufacture of a medicament for treating or preventing disorders or syndromes including, e.g., those described for the individual NONX nucleotides and polypeptides herein, and/or other pathologies and disorders of the like.
  • the therapeutic can be, e.g., a ⁇ ONX nucleic acid, a ⁇ ONX polypeptide, or a ⁇ ONX- specific antibody, or biologically-active derivatives or fragments thereof.
  • the compositions of the present invention will have efficacy for treatment of patients suffering from the diseases and disorders disclosed below and/or other pathologies and disorders of the like.
  • the polypeptides can be used as immunogens to produce antibodies specific for the invention, and as vaccines. They can also be used to screen for potential agonist and antagonist compounds.
  • a cD ⁇ A encoding ⁇ ONX may be useful in gene therapy, and ⁇ ONX may be useful when administered to a subject in need thereof.
  • the compositions of the present invention will have efficacy for treatment of patients suffering from the diseases and disorders disclosed above and/or other pathologies and disorders of the like.
  • the invention further includes a method for screening for a modulator of disorders or syndromes including, e.g., the diseases and disorders disclosed above and/or other pathologies and disorders of the like.
  • the method includes contacting a test compound with a ⁇ ONX polypeptide and determining if the test compound binds to said ⁇ ONX polypeptide. Binding of the test compound to the ⁇ ONX polypeptide indicates the test compound is a modulator of activity, or of latency or predisposition to the aforementioned disorders or syndromes.
  • Also within the scope of the invention is a method for screening for a modulator of activity, or of latency or predisposition to an disorders or syndromes including, e.g., the diseases and disorders disclosed above and/or other pathologies and disorders of the like by administering a test compound to a test animal at increased risk for the aforementioned disorders or syndromes.
  • the test animal expresses a recombinant polypeptide encoded by a ⁇ ONX nucleic acid. Expression or activity of ⁇ ONX polypeptide is then measured in the test animal, as is expression or activity of the protein in a control animal which recombinantly- expresses ⁇ ONX polypeptide and is not at increased risk for the disorder or syndrome.
  • the expression of NOVX polypeptide in both the test animal and the control animal is compared. A change in the activity of NONX polypeptide in the test animal relative to the control animal indicates the test compound is a modulator of latency of the disorder or syndrome.
  • the mvention includes a method for determining the presence of or predisposition to a disease associated with altered levels of a ⁇ ONX polypeptide, a ⁇ ONX nucleic acid, or both, in a subject (e.g., a human subject).
  • the method includes measuring the amount of the NONX polypeptide in a test sample from the subject and comparing the amount of the polypeptide in the test sample to the amount of the ⁇ ONX polypeptide present in a control sample.
  • An alteration in the level of the ⁇ ONX polypeptide in the test sample as compared to the control sample indicates the presence of or predisposition to a disease in the subject.
  • the predisposition includes, e.g., the diseases and disorders disclosed above and/or other pathologies and disorders of the like.
  • the expression levels of the new polypeptides of the invention can be used in a method to screen for various cancers as well as to determine the stage of cancers.
  • the invention includes a method of treating or preventing a pathological condition associated with a disorder in a mammal by administering to the subject a ⁇ ONX polypeptide, a ⁇ ONX nucleic acid, or a ⁇ ONX-specific antibody to a subject (e.g., a human subject), in an amount sufficient to alleviate or prevent the pathological condition.
  • the disorder includes, e.g., the diseases and disorders disclosed above and/or other pathologies and disorders of the like.
  • the invention can be used in a method to identity the cellular receptors and downstream effectors of the invention by any one of a number of techniques commonly employed in the art.
  • the present invention provides novel nucleotides and polypeptides encoded thereby.
  • nucleic acid sequences and their polypeptides.
  • the sequences are collectively referred to as “NONX nucleic acids” or “ ⁇ ONX polynucleotides” and the corresponding encoded polypeptides are referred to as “ ⁇ ONX polypeptides” or “NONX proteins.” Unless indicated otherwise, " ⁇ ONX” is meant to refer to any of the novel sequences disclosed herein. Table A provides a summary of the ⁇ ONX nucleic acids and their encoded polypeptides.
  • NOVX nucleic acids and their encoded polypeptides are useful in a variety of applications and contexts.
  • the various NOVX nucleic acids and polypeptides according to the invention are useful as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. Additionally, NOVX nucleic acids and polypeptides can also be used to identify proteins that are members of the family to which the NONX polypeptides belong.
  • the ⁇ ONX genes and their corresponding encoded proteins are useful for preventing, treating or ameliorating medical conditions, e.g., by protein or gene therapy.
  • Pathological conditions can be diagnosed by determining the amount of the new protein in a sample or by determining the presence of mutations in the new genes. Specific uses are described for each of the sixteen genes, based on the tissues in which they are most highly expressed. Uses include developing products for the diagnosis or treatment of a variety of diseases and disorders.
  • the ⁇ ONX nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance ⁇ ONX activity or function.
  • the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cell growth, cell metabolism, cell differentiation, cell proliferation, and/or cell signaling.
  • NONX or a fragment or derivative thereof may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of ⁇ ONX.
  • disorders include, but are not limited to, cancers such as adenocarcinoma, leukemia, lymphoma, melanoma, myeloma, sarcoma, teratocarcinoma, and, in particular, cancers of the adrenal gland, bladder, bone, bone marrow, brain, breast, cervix, gall bladder, ganglia, gastrointestinal tract, heart, kidney, liver, lung, muscle, ovary, pancreas, parathyroid, penis, prostate, salivary glands, skin, spleen, testis, thymus, thyroid, and uterus; neurological disorders such as epilepsy, ischemic cerebrovascular disease, stroke, cerebral neoplasms, Alzheimer's disease, Pick's disease, Huntington's disease, dementia, Parkinson's disease and other extrapyr
  • the ⁇ ONX nucleic acids and proteins of the invention are useful in potential diagnostic and therapeutic applications and as a research tool. These include serving as a specific or selective nucleic acid or protein diagnostic and or prognostic marker, wherein the presence or amount of the nucleic acid or the protein are to be assessed. These also include potential therapeutic applications such as the following: (i) a protein therapeutic, (ii) a small molecule drug target, (iii) an antibody target (therapeutic, diagnostic, drug targeting/cytotoxic antibody), (iv) a nucleic acid useful in gene therapy (gene delivery/gene ablation), (v) an agent promoting tissue regeneration in vitro and in vivo, and (vi) a biological defense weapon. Additional utilities for the ⁇ ONX nucleic acids and polypeptides according to the invention are disclosed herein.
  • ⁇ OV1 A ⁇ OV1 polypeptide has been identified as aPaladin-like protein (also referred to as CG93221-01).
  • the disclosed novel ⁇ OV1 nucleic acid (SEQ ID ⁇ O:l) of2600 nucleotides is shown in Table 1A.
  • the novel NOV1 nucleic acid sequences maps to the chromosome 10.
  • An ORF begins with an ATG initiation codon at nucleotides 15-17 and ends with a TAG codon at nucleotides 2583-2585.
  • a putative untranslated region and/or downstream from the termination codon is underlined in Table 1A, and the start and stop codons are in bold letters.
  • a variant sequence can include a single nucleotide polymorphism (SNP).
  • SNP can, in some instances, be referred to as a "cSNP" to denote that the nucleotide sequence containing the SNP originates as a cDNA.
  • the NOV1 protein (SEQ ID NO:2) encoded by SEQ ID NO.T is 856 amino acid residues in length and is presented using the one-letter amino acid code in Table IB. Psort analysis predicts the NO VI protein of the invention to be localized in the cytoplasm with a certainty of 0.4500.
  • the "E- value” or “Expect” value is a numeric indication of the probability that the aligned sequences could have achieved their similarity to the BLAST query sequence by chance alone, within the database that was searched.
  • the Expect value (E) is a parameter that describes the number of hits one can "expect” to see just by chance when searching a database of a particular size. It decreases exponentially with the Score (S) that is assigned to a match between two sequences. Essentially, the E value describes the random background noise that exists for matches between sequences.
  • the Expect value is used to create a significance threshold for reporting results.
  • the default value used for blasting is typically set to 0.0001, with the filter to remove low complexity sequence turned off.
  • the Expect value is also used instead of the P value (probability) to report the significance of matches.
  • P value probability
  • an E value of one assigned to a hit can be interpreted as meaning that in a database of the current size one might expect to see one match with a similar score simply by chance.
  • An E value of zero means that one would not expect to see any matches with a similar score simply by chance. See, e.g., http://www.ncbi.nlm.nih.gov/Education/BLASTinfo/.
  • nucleic acid sequence of this invention has 2063 of 2508 bases (82%) identical to a gb:GENBANK-ID:MMPAL
  • the full amino acid sequence of the protein of the invention was found to have 695 of 859 amino acid residues (80%) identical to, and 754 of 859 amino acid residues (87%) similar to, the 859 amino acid residue ptnr:SPTREMBL-ACC:P70261 protein ⁇ xom Mus musculus (PALADIN GENE).
  • NOV1 also has homology to the proteins shown in the BLASTP data in Table ID. Table ID. BLAST results for NOVl
  • Table IE A multiple sequence alignment is given in Table IE, with the NOVl protein being shown on line 1 in Table IE in a ClustalW analysis, and comparing the NOVl protein with the related protein sequences shown in Table ID.
  • This BLASTP data is displayed graphically in the ClustalW in Table IE.
  • the NOVl Clustal W alignment shown in Table IE was modified to begin at amino residue 1050.
  • the data in Table IE includes all of the regions overlapping with the NOVl protein sequences.
  • NOVl has, for example, multiple Paladin gene signature sequences and homology to other members of the Paladin-like Protein Family.
  • NOV 1 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts.
  • NOVl nucleic acids and polypeptides can be used to identify proteins that are members of the Paladin like family of proteins.
  • the NOVl nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOVl activity or function.
  • nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular metabolism and signal transduction.
  • These molecules can be used to treat, e.g., cardiomyopathy, atherosclerosis, hypertension, congenital heart defects, aortic stenosis, atrial septal defect (ASD),atrioventricular (A-V) canal defect, ductus arteriosus, pulmonary stenosis, subaortic stenosis, ventricular septal defect (VSD), valve diseases, tuberous sclerosis, scleroderma, obesity, transplantation, adrenoleukodystrophy, congenital adrenal hyperplasia, diabetes, Von Hippel-Lindau (NHL) syndrome, pancreatitis, obesity, hyperthyroidism and hypothyroidism, hypercalceimia, ulcers, cirrhosis, transplantation, inflammatory bowel disease, divertic
  • NOVl nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins.
  • the NOVl nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the Paladin-like protein family.
  • Paladin proteins are a family of protein-tyrosine phosphatases.
  • the protein phosphatases can be divided into 2 large families: the serine/threonine phosphatases, which are metalloproteins, and the protein-tyrosine phosphatases, which proceed via a thiol-phosphate enzyme intermediate.
  • the protein-tyrosine phosphatase family includes the VHl-like dual- specificity phosphatases. These phosphatases dephosphorylate phosphotyrosine- as well as phosphoserine- and phosphothreonine-containing substrates. Members of the dual-specificity phosphatase protein family inactivate mitogen-activated protein (MAP) kinase through dephosphorylation of critical threonine and tyrosine residues. Members of the MAP kinase family play a pivotal role in cellular signal transduction. Using a subtractive screen of mouse gastrulation, Pearce et al. (1996) identified a novel mouse gene, paladin, with similarity to the dual specificity protein phosphatase family.
  • MAP mitogen-activated protein
  • the NOVl nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of cardiac and endocrine physiology.
  • the NOVl nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat muscle and nervous system disorders, e.g., cardiomyopathy, atherosclerosis, hypertension, congenital heart defects, aortic stenosis, atrial septal defect (ASD),atrioventricular (A-V) canal defect, ductus arteriosus, pulmonary stenosis, subaortic stenosis, ventricular septal defect (VSD), valve diseases, tuberous sclerosis, scleroderma, obesity, transplantation, adrenoleukodystrophy, congenital adrenal hyperplasia, diabetes, Von Hippel-Lindau (VHL) syndrome, pancreatitis, obesity, hyperthyroidism and hypothyroidism, hypercalceimia, ulcer
  • NOVl nucleic acids and polypeptides are useful for detecting specific cell types. For example, expression analysis has demonstrated that a NOVl nucleic acid is expressed in brown adipose, heart, aorta, vein, umbilical vein, adrenal gland/suprarenal gland, pancreas, thyroid, salivary glands, parotid salivary glands, stomach, liver, gall bladder, small intestine, colon, bone marrow, lymphoid tissue, spleen, lymph node, tonsils, thymus, cartilage, muscle, brain, thalamus, hypothalamus, pituitary gland, amygdala, substantia nigra, hippocampus, spinal chord, cervix, mammary gland/breast, ovary, placenta, uterus, vulva, prostate, testis, lung, lung pleura, kidney, retina, dermis. Additional utilities for NOVl nucleic acids and polypeptides according to
  • a NOV2 polypeptide has been identified as a Plasma Membrane Ring Finger-like protein (also referred to as CG93210-01).
  • the disclosed novel NOV2 nucleic acid (SEQ ID NO:3) of 1205 nucleotides is shown in Table 2A.
  • the novel NOV2 nucleic acid sequences maps to the chromosome 22.
  • An ORF begins with an ATG initiation codon at nucleotides 17-19 and ends with a ATT codon at nucleotides 1149-1151.
  • a putative untranslated region and/or downstream from the termination codon is underlined in Table 2A, and the start and stop codons are in bold letters.
  • the NOV2 protein (SEQ ID NO:4) encoded by SEQ ID NO:3 is 378 amino acid residues in length and is presented using the one-letter amino acid code in Table 2B. Psort analysis predicts the NOV2 protein of the invention to be localized at the plasma membrane with a certainty of 0.6400.
  • nucleic acid sequence of this invention has 287 of 489 bases (58%) identical to a gb:GENBANK-ID:SSI132828]acc:AJ132828.1 mRNA from Spermatozopsis similis (mRNA for p210 protein, partial).
  • the full amino acid sequence of the protein of the invention was found to have 341 of 379 amino acid residues (89%>) identical to, and 355 of 379 amino acid residues (93%) similar to, the 379 amino acid residue ptnr:SPTREMBL-ACC:Q9DCWl protein from Mus musculus (0610009 J22RTK PROTEIN).
  • NOV2 also has homology to the proteins shown in the BLASTP data in Table 2D.
  • NOV2 has, for example, a Ring Finger signature sequence and homology to other members of the Plasma Membrane Ring Finger-like Protein Family.
  • NOV 2 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts.
  • NOV2 nucleic acids and polypeptides can be used to identify proteins that are members of the Plasma Membrane Ring Finger-like Protein Family.
  • the NOV2 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOVl activity or function.
  • nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular metabolism and signal transduction.
  • small molecules that modulate or inhibit, e.g., cellular activation, cellular metabolism and signal transduction.
  • These molecules can be used to treat, e.g., anemia, ataxia-telangiectasia, autoimmume disease, immunodeficiencies, diabetes, autoimmune disease, renal artery stenosis, interstitial nephritis, glomerulonephritis, polycystic kidney disease, systemic lupus erythematosus, renal tubular acidosis, IgA nephropathy, hypercalceimia, Lesch-Nyhan syndrome, cancer, trauma, regeneration (in vitro and in vivo), viral/bacterial/parasitic infections, as well as other diseases, disorders and conditions.
  • anemia e.g.,
  • NOV2 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins.
  • the NOV2 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the Plasma Membrane Ring Finger-like Protein Family .
  • the NOV2 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of immune and renal physiology.
  • the NOV2 nucleic acids and polypeptides, antibodies and related compounds according to the mvention may be used to treat muscle and nervous system disorders, e.g., anemia, ataxia-telangiectasia, autoimmume disease, immunodeficiencies, diabetes, autoimmune disease, renal artery stenosis, interstitial nephritis, glomerulonephritis, polycystic kidney disease, systemic lupus erythematosus, renal tubular acidosis, IgA nephropathy, hypercalceimia, Lesch-Nyhan syndrome, cancer, trauma, regeneration (in vitro and in vivo), viral/bacterial/parasitic infections, as well as other diseases, disorders and conditions.
  • muscle and nervous system disorders e.g., anemia, ataxia-
  • the NOV2 nucleic acids and polypeptides are useful for detecting specific cell types.
  • expression analysis has demonstrated that a NOV2 nucleic acid is expressed in peripheral blood, and a pool of various mammalian tissues.
  • Expression information was derived from the tissue sources of the sequences that were included in the derivation of the sequence of CuraGen Aec. No. CG93210-01.
  • the sequence is predicted to be expressed in the following tissues because of the expression pattern of (GENBANK-ID: gb:GENBANK- ID:SSI132828
  • NOV2 nucleic acids and polypeptides according to the invention are disclosed herein.
  • NOV3 A NOV3 polypeptide has been identified as a Thrombospondin type 1 (tsp_l) domain containing protein (also referred to as CG93275-01).
  • the disclosed novel NOV3 nucleic acid (SEQ ID NO:5) of 799 nucleotides is shown in Table 3 A.
  • the novel NOV3 nucleic acid sequences maps to the chromosome 16.
  • An ORF begins with an ATG initiation codon at nucleotides 51-53 and ends with a TGA codon at nucleotides 744-746.
  • a putative untranslated region and/or downstream from the termination codon is underlined in Table 3 A, and the start and stop codons are in bold letters.
  • the NOV3 protein (SEQ ID NO:6) encoded by SEQ TD NO:5 is 231 amino acid residues in length and is presented using the one-letter amino acid code in Table 3B. Psort analysis predicts the NOV3 protein of the invention to be localized in the cytoplasm with a certainty of 0.4500.
  • nucleic acid sequence of this invention has 392 of 396 bases (98%) identical to an EST AA057409 mRNA from human).
  • the full amino acid sequence of the protein of the invention was found to have 74 of 216 amino acid residues (34%) identical to, and 107 of 216 amino acid residues (49%) similar to, the 237 amino acid residue ptnr:SPTREMBL-ACC:Q9HBS6 protein from Homo sapiens (HYPOTHETICAL 25.7 KDA PROTEIN).
  • NOV3 also has homology to the proteins shown in the BLASTP data in Table 3D.
  • ADAMTS-10 precursor A disintegrin and metalloproteinase with thrombospondin motifs 10) (ADAM-TS 10) (ADAM-TSIO) (Fragment); SEQ ID NO:49
  • the NOV3 Clustal W alignment shown in Table 3E was modified to begin at amino residue 1321.
  • the data in Table 3E includes all of the regions overlapping with the NOV3 protein sequences.
  • the presence of identifiable domains in the protein disclosed herein was determined by searches using algorithms such as PROSITE, Blocks, Pfam, ProDomain, Prints and then determining the Interpro number by crossing the domain match (or numbers) using the Interpro website (http:www.ebi.ac.uk/interpro/).
  • Table 3F lists the domain description from DOMAIN analysis results against NOV3.
  • NOV3 has, for example, tliree tsp_l domain signature sequences and homology to other members of the tsp_l Domain-containing Protein Family.
  • NOV 3 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts.
  • NOV3 nucleic acids and polypeptides can be used to identify proteins that are members of the tsp_l Domain-containing Protein Family.
  • the NOV3 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOV3 activity or function.
  • nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular metabolism, and signal transduction. These molecules can be used to treat, e.g., Von Hippel-Lindau (VHL) syndrome, diabetes, tuberous sclerosis, fertility, hypogonadism, as well as other diseases, disorders and conditions.
  • VHL Von Hippel-Lindau
  • various NOV3 nucleic acids and polypeptides according to the invention are useful, ter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins.
  • the NOV3 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the tsp_l Domain-containing Protein Family.
  • Thrombospondin type 1 domain (TSP1, IPR000884) is a repeat found in the thrombospondin protein where it is repeated 3 times.
  • the tsp_l domain is repeated three times in the NOV3 polypeptide.
  • proteins involved in the complement pathway properdin, C6, C7, C8A, C8B, C9 (Patthy,L., J. Mol. Biol.
  • the NOV3 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of urogenital physiology.
  • the NOV3 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat reproductive and metabolic disorders, e.g., Von Hippel-Lindau (VHL) syndrome, diabetes, tuberous sclerosis, fertility, hypogonadism, as well as other diseases, disorders and conditions.
  • VHL Von Hippel-Lindau
  • the NOV3 nucleic acids and polypeptides are useful for detecting specific cell types.
  • NOV3 nucleic acid is expressed in eye and testis.
  • a NOV4 polypeptide has been identified as a Protocadherin Alpha C2 Short Form-like protein (also referred to as CG93187-01).
  • the disclosed novel NOV4 nucleic acid (SEQ ID NO: 7) of 600 nucleotides is shown in Table 4A.
  • the novel NOV4 nucleic acid sequences maps to the chromosome 11.
  • An ORF begins with an ATG initiation codon at nucleotides 41-43 and ends with a
  • TAG codon at nucleotides 2546-2548.
  • a putative untranslated region and/or downstream from the termination codon is underlined in Table 4A, and the start and stop codons are in bold letters.
  • the NOV4 protein (SEQ LD NO:8) encoded by SEQ ID NO:7 is 835 amino acid residues in length and is presented using the one-letter amino acid code in Table 4B. Psort analysis predicts the NOV4 protein of the invention to be localized at the plasma membrane with a certainty of 0.7900.
  • nucleic acid sequence of this invention has 273 of 415 bases (65%) identical to a gb:GENBANK-ID:AF061573
  • the full amino acid sequence of the protein of the invention was found to have 273 of 415 amino acid residues (65%) identical to, and 273 of 415 amino acid residues (65%) similar to, the 4076 amino acid residue gb:GENBANK- ID:AF061573jacc:AF061573.2 protein from Homo sapiens (protocadherin (PCDH8) mRNA, complete eds).
  • PCDH8 protocadherin
  • NOV4 also has homology to the proteins shown in the BLASTP data in Table 4D.
  • EGF LAG seven-pass G-type receptor 2 >gi
  • N0V4 492 FE DESSBTLRL ⁇ HAL 542 gi 1 17461472 I 936 TTjVDSYTgEIRVgRSP 987 gi
  • the NOV4 Clustal W alignment shown in Table 4E was modified to begin at amino residue 1201 and end at amino acid residue 2760.
  • the data in Table IE includes all of the regions overlapping with the NOV4 protein sequences.
  • NOV4 has, for example, seven Cadherin domain signature sequences and homology to other members of the Protocadherin Alpha C2 Short Form
  • NOV4 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts.
  • NOV4 nucleic acids and polypeptides can be used to identify proteins that are members of the Protocadherin Alpha C2 Short Form Protein-like Protein Family.
  • the NOV4 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOV4 activity or function.
  • the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular metabolism, and signal transduction.
  • VHL Von Hippel-Lindau
  • Alzheimer's disease stroke, tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, cerebral palsy, epilepsy, Lesch- Nyhan syndrome, multiple sclerosis, ataxia-telangiectasia, leukodystrophies, behavioral disorders, addiction, anxiety, pain, neurodegeneration, systemic lupus erythematosus, autoimmune disease, asthma, emphysema, scleroderma, allergy, ARDS, fertility, endometriosis, hypogonadism, hemophilia, hypercoagulation, idiopathic thrombocytopenic purpura, autoimmune disease, allergies, immunodeficiencies, transplantation, graft versus host disease (GVHD), lymphaedema, as well as other diseases, disorders and conditions.
  • VHL Von Hippel-Lindau
  • NOV4 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins.
  • the NOV4 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the Protocadherin Alpha C2 Short Form Protein-like Protein Family. Cadherins (Takeichi, Annu. Rev. Biochem. 59: 237-252 (1990); Takeichi Trends
  • cadherin expression causes morphological changes involving the positional segregation of cells into layers, suggesting they may play an important role in the sorting of different cell types during morphogenesis, histogenesis and regeneration. They may also be involved in the regulation of tight and gap junctions, and in the control of intercellular spacing.
  • Cadherins are evolutionary related to the desmogleins which are component of intercellular desmosome junctions involved in the interaction of plaque proteins.
  • cadherins comprise a number of domains: these include a signal sequence; a propeptide of around 130 residues; an extracellular domain of around 600 residues; a single transmembrane domain; and a well-conserved C-terminal cytoplasmic domain of about 150 residues.
  • the extracellular domain can be subdivided into 5 parts, 4 of which are repeats of about 110 residues, and the fifth contains 4 conserved cysteines.
  • the calcium-binding region of cadherins is thought to be located in the extracellular domain. This indicates that the sequence of the invention has properties similar to those of other proteins known to contain this/these domain(s) and similar to the properties of these domains.
  • Maniatis et al. has identified 52 novel human cadherin-like genes organized into three closely linked clusters (Wu and Maniatis, Cell 97(6):779-90 (1999).) Comparison of the genomic DNA sequences with those of representative cDNAs reveals a striking genomic organization similar to that of immunoglobulin and T cell receptor gene clusters.
  • the N- terminal extracellular and transmembrane domains of each cadherin protein are encoded by a distinct and unusually large exon. These exons are organized in a tandem array.
  • the C-terminal cytoplasmic domain of each protein is identical and is encoded by three small exons located downstream from the cluster of N-terminal exons. This unusual organization has interesting implications regarding the molecular code required to establish complex networks of neuronal connections in the brain and the mechanisms of cell-specific cadherin- like gene expression.
  • the NOV4 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of urogenital, nerve, and endocrine physiology.
  • the NOV4 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat reproductive and nervous system disorders, e.g., Von Hippel-Lindau (VHL) syndrome, Alzheimer's disease, stroke, tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, cerebral palsy, epilepsy, Lesch-Nyhan syndrome, multiple sclerosis, ataxia-telangiectasia, leukodystrophies, behavioral disorders, addiction, anxiety, pain, neurodegeneration, systemic lupus erythematosus, autoimmune disease, asthma, emphysema, scleroderma, allergy, ARDS, fertility, endometriosis, hypogonadism, hemophilia, hypercoagulation
  • NOV4 nucleic acids and polypeptides are useful for detecting specific cell types.
  • expression analysis has demonstrated that a NOV4 nucleic acid is expressed in Heart, Aorta, Umbilical Vein, Thyroid, Colon, Peripheral Blood, Spleen, Lymph node, Bone, Cartilage, Brain, Left cerebellum, Right Cerebellum, Parietal Lobe, Temporal Lobe, Cerebral Medulla/Cerebral white matter, Hippocampus, Cervix, Mammary glandBreast, Ovary, Placenta, Uterus, Testis, Lung, and Retina.
  • NOV5 nucleic acids and polypeptides according to the invention are disclosed herein.
  • a NOV5 polypeptide has been identified as a Nuclear protein-like protein (also referred to as CG95083-01).
  • the disclosed novel NOV5 nucleic acid (SEQ ID NO:9) of 2322 nucleotides is shown in Table 5 A.
  • An ORF begins with an ATG initiation codon at nucleotides 70-72 and ends with a TAA codon at nucleotides 2320-2322.
  • a putative untranslated region and/or downstream from the termination codon is underlined in Table 5 A, and the start and stop codons are in bold letters.
  • a variant sequence can include a single nucleotide polymorphism (SNP).
  • SNP can, in some instances, be referred to as a "cSNP” to denote that the nucleotide sequence containing the SNP originates as a cDNA.
  • the NOV5 protein (SEQ ID NO: 10) encoded by SEQ ID NO:9 is 750 amino acid residues in length and is presented using the one-letter amino acid code in Table 5B. Psort analysis predicts the NOV5 protein of the invention to be localized in the nucleus with a certainty of 0.3000.
  • nucleic acid sequence of this invention has 443 of 754 bases (58%) identical to a gb:GENBANK-ID:AB037783
  • NOV5 also has homology to the proteins shown in the BLASTP data in Table 5D. Table 5D. BLAST results for NOV5
  • NOV5 has, for example, an RhoGEF signature sequence and a FYVE Zinc Finger signature sequence, aw well as homology to other members of the Nuclear Protein-like Protein Family.
  • NOV5 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts.
  • NOV5 nucleic acids and polypeptides can be used to identify proteins that are members of the Nuclear Protein-like Protein Family.
  • the NOV5 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOV5 activity or function.
  • nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular replication, and signal transduction.
  • These molecules can be used to treat, e.g., Cardiovascular diseases, Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus , Pulmonary stenosis, Subaortic stenosis, Ventricular septal defect (VSD), valve diseases, Tuberous sclerosis, Scleroderma, Obesity, Transplantation, DiabetesNon Hippel-Lindau (VHL) syndrome , Pancreatitis, Obesity, Von Hippel-Lindau (VHL) syndrome , Alzheimer's disease, Stroke, Tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's
  • NOV5 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins.
  • the NOV5 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the Nuclear Protein-like Protein Family.
  • NOV5 nucleic acids and polypeptides, antibodies and related compounds according to the mvention will be useful in therapeutic and diagnostic applications in the mediation of cardiac and nerve physiology.
  • the NOV5 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat cardiovascular and nervous system disorders, e.g., Cardiovascular diseases,
  • Cardiomyopathy Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus , Pulmonary stenosis, Subaortic stenosis, Ventricular septal defect (VSD), valve diseases, Tuberous sclerosis, Scleroderma, Obesity, Transplantation, DiabetesNon Hippel-Lindau (VHL) syndrome , Pancreatitis, Obesity, Von Hippel-Lindau (VHL) syndrome , Alzheimer's disease, Stroke, Tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, Cerebral palsy, Epilepsy, Lesch-Nyhan syndrome, Multiple sclerosis, Ataxia-telangiectasia, Leukodystrophies, Behavioral disorders, Addiction, Anxiety, Pain, Neuroprotection as well as other diseases, disorders and conditions.
  • the NOV5 nucleic acids and polypeptides are useful for detecting specific cell types. For example, expression analysis has demonstrated that a NOV5 nucleic acid is expressed in Brown adipose, Vein, Umbilical Vein, Adrenal Gland/Suprarenal gland, Gall Bladder, Small Intestine, Colon, Lymphoid tissue, Spleen, Lymph node, Thymus, Brain, Temporal Lobe, Basal Ganglia Cerebral nuclei, Substantia Nigra, Spinal Chord, Cervix, Ovary, Uterus, Testis, Lung, Lung Pleura, Larynx, Urinary Bladder, Kidney.
  • a NOV6 polypeptide has been identified as a Secretory Protein-like protein (also referred to as CG94989-01).
  • the disclosed novel NOV6 nucleic acid (SEQ ID NO:ll) of 2372 nucleotides is shown in Table 6A.
  • the novel NOV6 nucleic acid sequences maps to the chromosome 17.
  • An ORF begins with an ATG initiation codon at nucleotides 99-101 and ends with a
  • TAA codon at nucleotides 1710-1712.
  • a putative untranslated region and/or downstream from the termination codon is underlined in Table 6A, and the start and stop codons are in bold letters.
  • the NOV6 protein (SEQ ID NO:12) encodedby SEQ ID NO:11 is 537 amino acid residues in length and is presented using the one-letter amino acid code in Table 6B. Psort analysis predicts the NOV6 protein ofthe invention to be localized outside the cell with a certainty of0.6997.
  • the full amino acid sequence of the protein of the invention was found to have 395 of 395 amino acid residues (100%) identical to, and 395 of 395 amino acid residues (100%) similar to, the 468 amino acid residue ptnr:SPTREMBL-ACC:O60276 protein from Homo sapiens (KIAA0523 PROTEIN)(Fig. 3B).
  • NOV6 also has homology to the proteins shown in the BLASTP data in Table 6D.
  • the NOV6 Clustal W alignment shown in Table 6E was modified to begin at amino residue 841 and end at amino acid residue 1860.
  • the data in Table 6E includes all of the regions overlapping with the NOV6 protein sequences.
  • the presence of identifiable domains in the protein disclosed herein was determined by searches using algorithms such as PROSITE, Blocks, Pfam, ProDomain, Prints and then determining the Interpro number by crossing the domain match (or numbers) using the
  • NOV6 has, for example, has homology to other members of the Secretory Protein-like Protein Family.
  • NOV6 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts.
  • NOV6 nucleic acids and polypeptides can be used to identify proteins that are members of the Secretory Protein-like Protein Family.
  • the NOV6 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOV6 activity or function.
  • the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation and signal transduction.
  • NOV6 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins.
  • the NOV6 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the Secretory Protein-like Protein Family.
  • the NOV6 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of cardiac physiology.
  • the NOV6 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat cardiac and vascular system disorders, e.g., Cardiovascular diseases, Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus as well as other diseases, disorders and conditions
  • NOV6 nucleic acids and polypeptides are useful for detecting specific cell types. For example, expression analysis has demonstrated that a NOV6 nucleic acid is expressed in Aorta.
  • NOV7 A NOV7 polypeptide has been identified as a Transmission Blocking Target Antigen
  • S230 Precursor-like protein also referred to as CG94978-01.
  • the disclosed novel NOV7 nucleic acid (SEQ ID NO:13) of 1629 nucleotides is shown in Table 7A.
  • the novel NOV7 nucleic acid sequences maps to the chromosome 1.
  • An ORF begins with an ATG initiation codon at nucleotides 1-3 and ends with a TGA codon at nucleotides 1627- 1629.
  • a putative untranslated region and/or downstream from the termination codon is underlined in Table 7A, and the start and stop codons are in bold letters.
  • the NOV7 protein (SEQ ID NO: 14) encoded by SEQ ID NO: 13 is 542 amino acid residues in length and is presented using the one-letter amino acid code in Table 7B. Psort analysis predicts the NOV7 protein of the invention to be localized in the cytoplasm with a certainty of 0.4500.
  • nucleic acid sequence of this invention has 874 of 876 bases (99%) identical to a gb:GENBANK-LD:AK022517
  • the full amino acid sequence of the protein of the invention was found to have 290 of 292 amino acid residues (99%) identical to, and 290 of 292 amino acid residues (99%) similar to, the 525 amino acid residue ptnr:SPTREMBL- ACC.Q9H9Z3 protein from Homo sapiens (CDNA FLJ12455 FIS, CLONE NT2RM1000563, WEAKLY SIMILAR TO TRANSMISSION- BLOCKING TARGET ANTIGEN S230 PRECURSOR).
  • NOV7 also has homology to the proteins shown in the BLASTP data in Table 7D.
  • Table 7E A multiple sequence alignment is given in Table 7E, with the NOV7 protein being shown on line 1 in Table 7E in a ClustalW analysis, and comparing the NOV7 protein with the select related protein sequences shown in Table 7D.
  • This BLASTP data is displayed graphically in the ClustalW in Table 7E.
  • N0V7 420 EARJSLLIVESRRRLVSILESRVLQLHREFL ⁇ A'LRL.
  • the presence of identifiable domains in the protein disclosed herein was determined by searches using algorithms such as PROSITE, Blocks, Pfam, ProDomain, Prints and then determining the Interpro number by crossing the domain match (or numbers) using the
  • NOV7 has, for example, three Blocking NT2RM1000563 Transmission-FIS Antigen Weakly Precursor Peptidase A2 signature sequences and homology to other members of the Transmission Blocking Target Antigen S230 Precursor-like Protein Family.
  • NOV7 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts. For example, NOV7 nucleic acids and polypeptides can be used to identify proteins that are members of the Transmission Blocking Target Antigen S230 Precursor-like Protein Family.
  • the NOV7 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOV7 activity or function.
  • the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation and signal transduction.
  • Cardiovascular diseases can be used to treat, e.g., Cardiovascular diseases, Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus , Pulmonary stenosis, Subaortic stenosis, Ventricular septal defect (VSD), valve diseases, Tuberous sclerosis, Scleroderma, Obesity, Transplantation, DiabetesNon Hippel-Lindau (VHL) syndrome , Pancreatitis, Obesity, Von Hippel-Lindau (VHL) syndrome , Alzheimer's disease, Stroke, Tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, Cerebral palsy, Epilepsy, Lesch- ⁇ yhan syndrome, Multiple sclerosis, Ataxia-telangiectasia, Leukodystrophies, Behavioral disorders, Addiction,
  • NOV7 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins.
  • the NOV7 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the Transmission Blocking Target Antigen S230 Precursor-like Protein Family.
  • the NOV7 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of cardiac and nerve physiology.
  • the NOV7 nucleic acids and polypeptides, antibodies and related compounds according to the mvention may be used to treat cardiovascular and nervous system disorders, e.g., Cardiovascular diseases,
  • Cardiomyopathy Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus , Pulmonary stenosis, Subaortic stenosis, Ventricular septal defect (VSD), valve diseases, Tuberous sclerosis, Scleroderma, Obesity, Transplantation, DiabetesNon Hippel-Lindau (VHL) syndrome, Pancreatitis, Obesity, Von Hippel-Lindau (VHL) syndrome, Alzheimer's disease, Stroke, Tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, Cerebral palsy, Epilepsy, Lesch- ⁇ yhan syndrome, Multiple sclerosis, Ataxia-telangiectasia, Leukodystrophies, Behavioral disorders, Addiction, Anxiety, Pain, Neuroprotection
  • NOV7 nucleic acids and polypeptides are useful for detecting specific cell types.
  • expression analysis has demonstrated that a NOV7 nucleic acid is expressed in Adipose, Heart, Aorta, Coronary Artery, Umbilical Vein, Pancreas, Liver, Gall Bladder, Colon, Bone Marrow, Thymus, Bone, Cartilage, Synovium/Synovial membrane, Skeletal Muscle, Brain, Left cerebellum, Right Cerebellum, Thalamus, Hypothalamus, Pituitary Gland, Frontal Lobe, Parietal Lobe, Cerebral Medulla/Cerebral white matter, Basal Ganglia/Cerebral nuclei, Substantia Nigra, Hippocampus, Cervix, Mammary gland Breast, Uterus,
  • Oviduct/Uterine Tube/Fallopian tube Prostate, Testis, Lung, Bronchus, Larynx, Kidney, Retina, Skin, Epidermis.
  • a NOV8 polypeptide has been identified as a Nuclear Protein-like protein (also referred to as CG94713-01).
  • the disclosed novel NOV8 nucleic acid (SEQ ID NO:15) of 3807 nucleotides is shown in Table 8A.
  • the novel NOV8 nucleic acid sequences maps to the chromosome 1.
  • An ORF begins with an ATG initiation codon at nucleotides 16-18 and ends with a TGA codon at nucleotides 3793-3795.
  • a putative untranslated region and/or downstream from the termination codon is underlined in Table 8 A, and the start and stop codons are in bold letters.
  • a variant sequence can include a single nucleotide polymorphism (SNP).
  • SNP can, in some instances, be referred to as a "cSNP" to denote that the nucleotide sequence containing the SNP originates as a cDNA.
  • the NOV8 protein (SEQ ID NO: 16) encoded by SEQ ID NO: 15 is 1259 amino acid residues in length and is presented using the one-letter amino acid code in Table 8B. Psort analysis predicts the NOV8 protein of the invention to be localized in the nucleus with a certainty of 0.7600.
  • nucleic acid sequence of a human ATIP isoform +1 2954 1.5e-307 In a BLAST search of public sequence databases, it was found, for example, that the nucleic acid sequence of this invention has 3751 of 3751 bases (100%) identical to a gb:GENBANK-ID:AB033114
  • the full amino acid sequence of the protein of the invention was found to have 1245 of 1245 amino acid residues (100%) identical to, and 1245 of 1245 amino acid residues (100%) similar to, the 1245 amino acid residue ptnr:SPTREMBL- ACC:Q9ULD2 protein from Homo sapiens (KIAA1288 PROTEIN).
  • NOV8 also has homology to the proteins shown in the BLASTP data in Table 8D.
  • Table 8E A multiple sequence alignment is given in Table 8E, with the NOV8 protein being shown on line 1 in Table 8E in a ClustalW analysis, and comparing the NOV8 protein with the related protein sequences shown in Table 8D.
  • This BLASTP data is displayed graphically in the ClustalW in Table 8E.
  • NOV8 has, for example, an RNA polymerase omega subunit signature sequence and homology to other members of the Nuclear Protein-like Protein Family.
  • NOV8 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts.
  • NOV8 nucleic acids and polypeptides can be used to identify proteins that are members of the Nuclear Protein-like Protein Family.
  • the NOV8 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOV8 activity or function.
  • nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular replication, and signal transduction.
  • These molecules can be used to treat, e.g., Cardiovascular diseases, Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus , Pulmonary stenosis, Subaortic stenosis, Ventricular septal defect (VSD), valve diseases, Tuberous sclerosis, Scleroderma, Obesity, Transplantation, DiabetesNon Hippel-Lindau (VHL) syndrome , Pancreatitis, Obesity, Hyperparathyroidism, Hypoparathyroidism as well as other diseases, disorders and conditions.
  • Cardiovascular diseases e.g., Cardiomyopathy, Atherosclerosis, Hypertension,
  • NOV8 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins.
  • the NOV8 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the Nuclear Protein-like Protein Family.
  • NOV8 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of cardiac or endocrine physiology.
  • the NOV8 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat infection, cardiovascular system, immune system, and nervous system disorders, e.g., Cardiovascular diseases, Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect,
  • Ductus arteriosus Pulmonary stenosis, Subaortic stenosis, Ventricular septal defect (VSD), valve diseases, Tuberous sclerosis, Scleroderma, Obesity, Transplantation, DiabetesNon Hippel-Lindau (VHL) syndrome , Pancreatitis, Obesity, Hyperparathyroidism, Hypoparathyroidism as well as other diseases, disorders and conditions.
  • VSD Ventricular septal defect
  • VHL Ventricular septal defect
  • the NOV8 nucleic acids and polypeptides are useful for detecting specific cell types.
  • NOV8 nucleic acid is expressed in Heart, Aorta, Coronary Artery, Vein, Umbilical Vein, Adrenal Gland/Suprarenal gland, Pancreas, Islets of Langerhans, Parathyroid Gland, Thyroid, Pineal Gland, Tongue, Salivary Glands, Stomach, Liver, Small Intestine, Colon, Ascending Colon, Lymphoid tissue, Spleen, Brain, Thalamus, Hypothalamus, Temporal Lobe, Amygdala, Cerebral Medulla/Cerebral white matter, Basal Ganglia/Cerebral nuclei, Substantia Nigra, Hippocampus, Spinal Chord, Cervix, Mammary gland/Breast, Ovary, Placenta, Uterus, Prostate, Testis, Lung, Nasoepithelium, Larynx, Urinary Bladder, Kidney, Kidney Cortex, Re
  • ANOV9 polypeptide has been identified as a Hemicentin precursor-like protein (also referred to as CG94702-01).
  • the disclosed novel NOV9 nucleic acid (SEQ ID NO:17) of 11796 nucleotides is shown in Table 9A.
  • the novel NOV9 nucleic acid sequences maps to the chromosome 9.
  • An ORF begins with an ATG initiation codon at nucleotides 1-3 and ends with a TAA codon at nucleotides 11794-11796.
  • a putative untranslated region and/or downstream from the termination codon is underlined in Table 9A, and the start and stop codons are inbold letters.
  • a variant sequence can include a single nucleotide polymorphism (SNP).
  • SNP can, in some instances, be referred to as a "cSNP" to denote that the nucleotide sequence containing the SNP originates as a cDNA.
  • the NOV9 protein (SEQ ID NO: 18) encoded by SEQ ID NO: 17 is 3931 amino acid residues in length and is presented using the one-letter amino acid code in Table 9B. Psort analysis predicts the NOV9 protein of the invention to be localized at the plasma membrane with a certainty of 0.7300.
  • nucleic acid sequence of this invention has 625 of 1067 bases (58%) identical to a gb:GENBANK-ID:HSLTGFBP4
  • the full amino acid sequence of the protein of the invention was found to have 502 of 1665 amino acid residues (30%) identical to, and 767 of 1665 amino acid residues (46%) similar to, the 5198 amino acid residue ⁇ tnr:SPTREMBL-ACC:O76518 protein from Caenorhabditis elegans (HEMICENTIN PRECURSOR).
  • NOV9 also has homology to the proteins shown in the BLASTP data in Table 9D.
  • the NOV9 Clustal W alignment shown in Table 9E was modified to begin at amino residue 4080.
  • the data in Table 9E includes all of the regions overlapping with the NOV9 protein sequences.
  • NON9 has, for example, thirty-three immunoglobulin (ig) signature sequences and four epidermal growth factor (EGF) signature sequences, as well as homology to other members of the Hemicentin Precursor-like Protein Family.
  • ⁇ ON9 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts.
  • ⁇ ON9 nucleic acids and polypeptides can be used to identify proteins that are members of the Hemicentin Precursor-like Protein Family.
  • the ⁇ ON9 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance ⁇ ON9 activity or function.
  • nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular differentiation, and signal transduction.
  • small molecules that modulate or inhibit, e.g., cellular activation, cellular differentiation, and signal transduction.
  • These molecules can be used to treat, e.g., Cardiovascular diseases, Hyperparathyroidism, Hypoparathyroidism, Lymphedema, Allergies as well as other diseases, disorders and conditions..
  • ⁇ ON9 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins.
  • the ⁇ ON9 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the Hemicentin Precursor-like Protein Family.
  • Hemicentrin is an extracellular matrix protein with a modular sturcture. Like ⁇ ON9, the hemicentrin structure includes many immunoglobulin domains flanked by EGF domains. The protein is likely involved in cellular differentiation of epithelial tissue.
  • the NON9 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of cardiac, immune and endocrine physiology. As such, the ⁇ ON9 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat cardiovascular, immune, and endocrine disorders, e.g., Cardiovascular diseases,
  • Hyperparathyroidism Hypoparathyroidism, Lymphedema, Allergies as well as other diseases, disorders and conditions.
  • the ⁇ ON9 nucleic acids and polypeptides are useful for detecting specific cell types. For example, expression analysis has demonstrated that a ⁇ ON9 nucleic acid is expressed in Adipose, Thyroid, Colon, Lymph node, Bone, Myometrium, Prostate, Testis, Aorta, Nein.
  • ⁇ OV10 A ⁇ ON10 polypeptide has been identified as a Selectin-like protein.
  • the novel ⁇ ON10 nucleic acid sequences maps to the chromosome 9.
  • Two alternative novel ⁇ OV10, ⁇ ONlOa and ⁇ ONlOb, nucleic acids and encoded polypeptides are provided.
  • a ⁇ ON10 variant is ⁇ ONlOa (alternatively referred to herein as CG94661-01), which includes the 1268 nucleotide sequence (SEQ D ⁇ O:19) shown in Table 10A.
  • a NOVlOa ORF begins with a ATG initiation codon at nucleotides 145-147 and ends with a TGA codon at nucleotides 871-873. Putative untranslated regions upstream from the initiation codon and downstream from the termination codon are underlined in Table 10A, and the start and stop codons are in bold letters.
  • the NOVlOa polypeptide (SEQ LD ⁇ O:20) encoded by SEQ ID NO:19 is 242 amino acid residues in length and is presented using the one-letter amino acid code in Table 10B.
  • the Psort profile for the NOVlOa predicts that this peptide is likely to be localized at the plasma membrane with a certainty of 0.7000.
  • NOV10 variant is the novel NOV 10b (alternatively referred to herein as CG94661-02), which includes the 887 nucleotide sequence (SEQ ID NO:21) shown in Table IOC.
  • NOV 10b was created by polymerase chain reaction (PCR) using the primers detailed in Example 1, Table 17. Primers were designed based on in silico predictions of the full length or some portion (one or more exons) of the cDNA/protein sequence of the invention. The PCR product derived by exon linking, covering the entire open reading frame, was cloned into the pCR2.1 vector from Invitrogen to provide clone 143260::COR100348691_extn.698976.C20.
  • the NOVl 0b ORF begins with a Kozak consensus ATG initiation codon at nucleotides 72-74 and ends with a TGA codon at nucleotides 1958-1960. Putative untranslated regions upstream from the initiation codon and downstream from the termination codon are underlined in Table 10C, and the start and stop codons are in bold letters.
  • a variant sequence can include a single nucleotide polymorphism (SNP).
  • SNP can, in some instances, be referred to as a "cSNP" to denote that the nucleotide sequence containing the SNP originates as a cDNA.
  • the NOVlOb protein (SEQ ID NO:22) encoded by SEQ ID NO:21 is 242 amino acid residues in length and is presented using the one-letter code in Table 10D.
  • the Psort profile for NOVl Ob predicts that this sequence is likely to be localized at the plasma membrane with a certainty of 0.7000.
  • NOVlOa differs from NOVlOb at amino acid position 18 (T>M) and amino acid position 50 (S>P) as shown in Tables 10B and 10D.
  • NOVlOa nucleic acid sequence of this invention has 438 of 447 bases (97%) identical to a gb:GENBANK-ID:HSM802384
  • the full amino acid sequence of the protein of the invention was found to have 110 of 139 amino acid residues (79%) identical to, and 123 of 139 amino acid residues (88%) similar to, the 269 amino acid residue ptm:SPTREMBL- ACC:Q9D176 protein from Mus musculus (170001711 IRK PROTEIN).
  • the NOV 10b nucleic acid sequence of this invention has 438 of 447 bases (97%) identical to a gb:GENBANK-
  • the NOVIO Clustal W alignment shown in Table 10F was modified to begin at amino residue 1600 and end at amino acid residue 2000.
  • the data in Table 10F includes all of the regions overlapping with the NOVIO protein sequences.
  • the NOVIO Clustal W alignment shown in Table 10G was modified to begin at amino residue 1601.
  • the data in Table 10G includes all of the regions overlapping with the NOVIO protein sequences.
  • NOV10 has, for example, a Sushi domain (SCR repeat) signature sequences and homology to other members of the Selectin-like Protein Family.
  • SCR repeat Sushi domain
  • NOV10 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts.
  • NOV10 nucleic acids and polypeptides can be used to identify proteins that are members of the Selectin-like Protein Family.
  • the NOV10 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOV10 activity or function.
  • nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular adhesion and signal transduction.
  • These molecules can be used to treat, e.g., Cardiovascular diseases, Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus , Pulmonary stenosis, Subaortic stenosis, Ventricular septal defect (VSD), valve diseases, Tuberous sclerosis, Scleroderma, Obesity, Transplantation, Von Hippel-Lindau (VHL) syndrome,
  • Cardiovascular diseases Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus , Pul
  • Cirrhosis Transplantation, Diabetes, Autoimmune disease, Renal artery stenosis, Interstitial nephritis, Glomerulonephritis, Polycystic kidney disease, Systemic lupus erythematosus, Renal tubular acidosis, IgA nephropathy, Hypercalceimia, Lesch-Nyhan syndrome, Systemic lupus erythematosus, Autoimmune disease, Asthma, Emphysema, Scleroderma, allergy as well as other diseases, disorders and conditions.
  • NOV10 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins.
  • the NOV10 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of protems belonging to the Selectin-like Protein Family.
  • the NOV10 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of cardiac and immune physiology.
  • the NOV10 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat cardiovascular and immune disorders, e.g., Cardiovascular diseases, Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus , Pulmonary stenosis, Subaortic stenosis, Ventricular septal defect (VSD), valve diseases, Tuberous sclerosis, Scleroderma, Obesity, Transplantation, Von Hippel-Lindau (VHL) syndrome, Cirrhosis, Transplantation, Diabetes, Autoimmune disease, Renal artery stenosis, Interstitial nephritis, Glomerulonephriti
  • NOVIO nucleic acids and polypeptides are useful for detecting specific cell types.
  • expression analysis has demonstrated that a NOVIO nucleic acid is expressed in Heart, Thyroid, Parotid Salivary glands, Liver, Colon, Ascending Colon, Bone Marrow, Peripheral Blood, Lymphoid tissue, Spleen, Lymph node, Tonsils, Thymus, Cerebellum, Spinal Chord, Cervix, Mammary gland/Breast, Ovary, Placenta, Uterus, Oviduct/Uterine Tube/Fallopian tube, Vulva, Prostate, Testis, Lung, Kidney, Kidney Cortex, Retina, Skin.
  • a NOVl 1 polypeptide has been identified as a Nuclear Protein-like protein (also referred to as CG94325-01).
  • the disclosed novel NOV11 nucleic acid (SEQ ID NO:23) of 8670 nucleotides is shown in Table 11 A.
  • the novel NOVl 1 nucleic acid sequences maps to the chromosome 15.
  • An ORF begins with an ATG initiation codon at nucleotides 204-206 and ends with a TAA codon at nucleotides 7152-7154.
  • a putative untranslated region and/or downstream from the termination codon is underlined in Table 11 A, and the start and stop codons are in bold letters.
  • a variant sequence can include a single nucleotide polymorphism (SNP).
  • SNP can, in some instances, be referred to as a "cSNP" to denote that the nucleotide sequence containing the SNP originates as a cDNA.
  • the NOVl 1 protein (SEQ ID NO:24) encoded by SEQ ID NO:23 is 2316 amino acid residues in length and is presented using the one-letter amino acid code in Table 1 IB. Psort analysis predicts the NOVl 1 protein of the invention to be localized at the nucleus with a certainty of 0.8800.
  • nucleic acid sequence of this invention has 5584 of 5584 bases (100%) identical to a gb:GENBANK-ID:AB046790
  • the full amino acid sequence of the protein of the invention was found to have 1790 of 1793 amino acid residues (99%) identical to, and 1792 of 1793 amino acid residues (99%) similar to, the 1833 amino acid residue ptnr:SPTREMBL- ACC:Q9NR92 protein from Homo sapiens (AF15Q14 PROTEIN).
  • NOVl 1 also has homology to the proteins shown in the BLASTP data in Table 1 ID.
  • Table 1 IE A multiple sequence alignment is given in Table 1 IE, with the NOVl 1 protein being shown on line 1 in Table 1 IE in a ClustalW analysis, and comparing the NOVl 1 protein with the related protein sequences shown in Table 1 ID.
  • This BLASTP data is displayed graphically in the ClustalW in Table 1 IE.
  • NOVll nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts.
  • NOVl 1 nucleic acids and polypeptides can be used to identify proteins that are members of the Nuclear Protein-like Protein Family.
  • the NOVl 1 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOVl 1 activity or function.
  • the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular replication, and signal transduction. These molecules can be used to treat, e.g.
  • VHL Von Hippel-Lindau
  • VHL Cirrhosis, Transplantation, Hemophilia, hypercoagulation, Idiopathic thrombocytopenic purpura, autoimmume disease, allergies, immunodeficiencies, transplantation, Graft vesus host, Cardiovascular diseases, Von Hippel-Lindau (VHL) syndrome , Alzheimer's disease, Stroke, Tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, Cerebral palsy, Epilepsy, Lesch-Nyhan syndrome, Multiple sclerosis, Ataxia-telangiectasia, Leukodystrophies, Behavioral disorders, Addiction, Anxiety, Pain, Neuroprotection, Systemic lupus erythematosus , Autoimmune disease, Asthma, Emphysema, Scleroderma, allergy, as well as other diseases, disorders and conditions.
  • NOVl 1 nucleic acids and polypeptides according to the invention are useful, ter alia, as novel members of the protein families according to the presence of sequence relatedness to previously described proteins.
  • the NOVll nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of cardiac, immune, and nerve physiology.
  • the NOVl 1 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat cardiovascular, immune, and nervous system disorders, e.g., Von Hippel-Lindau (VHL) syndrome, Cirrhosis,
  • VHL Von Hippel-Lindau
  • Leukodystrophies Leukodystrophies, Behavioral disorders, Addiction, Anxiety, Pain, Neuroprotection, Systemic lupus erythematosus , Autoimmune disease, Asthma, Emphysema, Scleroderma, allergy, as well as other diseases, disorders and conditions.
  • the NOVl 1 nucleic acids and polypeptides are useful for detecting specific cell types.
  • expression analysis has demonstrated that a NOVl 1 nucleic acid is expressed in Adipose, Aorta, Artery, Coronary Artery, Umbilical Vein, Thyroid, Liver, Small Intestine, Duodenum, Colon, Ascending Colon, Bone Marrow, Lymph node, Tonsils, Thymus, Cartilage, Muscle, Brain, Cervix, Uterus, Vulva, Prostate, Testis, Lung, Bronchus, Urinary Bladder, Kidney, Skin, Epidermis, Dermis.
  • a NOV 12 polypeptide has been identified as a Plasma Membrane Protein-like protein (also referred to as CG94282-01).
  • the disclosed novel NOV12 nucleic acid (SEQ ID NO:25) of 8811 nucleotides is shown in Table 12 A.
  • the novel NOV 12 nucleic acid sequences maps to the chromosome 12.
  • An ORF begins with an ATG initiation codon at nucleotides 1-3 and ends with a TAG codon at nucleotides 4378-4380.
  • a putative untranslated region and/or downstream from the termination codon is underlined in Table 12 A, and the start and stop codons are in bold letters.
  • a variant sequence can include a single nucleotide polymorphism (SNP).
  • SNP can, in some instances, be referred to as a "cSNP" to denote that the nucleotide sequence containing the SNP originates as a cDNA.
  • the NOV12 protein (SEQ ID NO:26) encoded by SEQ ID NO:25 is 1459 amino acid residues in length and is presented using the one-letter amino acid code in Table 12B. Psort analysis predicts the NOV12 protein of the invention to be localized at the plasma membrane with a certainty of 0.6500.
  • nucleic acid sequence of this invention has 6444 of 6447 bases (99%) identical to a gb:GENBANK-ID:AB033034
  • KIAA1208 protein partial eds.
  • the full amino acid sequence of the protein of the invention was found to have 663 of 663 amino acid residues (100%) identical to, and 663 of 663 amino acid residues (100%) similar to, the 663 amino acid residue ptnr:SPTREMBL-ACC:Q9ULL2 protein from Homo sapiens (KIAA1208 PROTEIN).
  • NOV 12 also has homology to the proteins shown in the BLASTP data in Table 12D.
  • Table 12E A multiple sequence alignment is given in Table 12E, with the NOV12 protein being shown on line 1 in Table 12E in a ClustalW analysis, and comparing the NOV 12 protein with the related protein sequences shown in Table 12D.
  • This BLASTP data is displayed graphically in the ClustalW in Table 12E.
  • the NOV 12 Clustal W alignment shown in Table 12E was modified to end at amino residue 1200.
  • the data in Table IE includes all of the regions overlapping with the NOV12 protein sequences.
  • NOV12 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts.
  • NOV12 nucleic acids and polypeptides can be used to identify proteins that are members of the Plasma Membrane Protein-like Protein Family.
  • the NOVl 2 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOVl 2 activity or function.
  • the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation and signal transduction.
  • VHL DiabetesNon Hippel-Lindau
  • Pancreatitis Obesity
  • Cardiomyopathy Atherosclerosis
  • Hypertension Congenital heart defects
  • Aortic stenosis Atrial septal defect (ASD)
  • Atrioventricular (A-V) canal defect Ductus arteriosus
  • Pulmonary stenosis Subaortic stenosis
  • Ventricular septal defect VSD
  • valve diseases Tuberous sclerosis, Scleroderma, Obesity, Transplantation, Von Hippel-Lindau (NHL) syndrome, Cirrhosis, Transplantation, Non Hippel-Lindau (NHL) syndrome , Alzheimer's disease, Stroke, Tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, Cerebral palsy, Epilepsy, Lesch- ⁇ yhan syndrome, Multiple sclerosis, Ataxia-telangiec
  • NOV 12 nucleic acids and polypeptides according to the invention are useful, ter alia, as novel members of the protein families according to the sequence relatedness to previously described proteins.
  • the NON12 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of cardiac, nerve, and immune physiology.
  • the ⁇ ON12 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat cardiovascular, immune, and nervous system disorders, e.g., DiabetesNon Hippel-Lindau (VHL) syndrome, Pancreatitis, Obesity, Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus , Pulmonary stenosis, Subaortic stenosis, Ventricular septal defect (VSD), valve diseases, Tuberous sclerosis, Scleroderma, Obesity, Transplantation, Von Hippel-Lindau (VHL) syndrome, Cirrhosis, Transplantation, Von Hippel-Lindau (VHL) syndrome , Alzheimer's disease, Stroke, Tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, Cere
  • NOV12 nucleic acids and polypeptides are useful for detecting specific cell types.
  • expression analysis has demonstrated that a NOV 12 nucleic acid is expressed in Pancreas, Uterus, Epidermis, Heart, Coronary Artery, Adrenal Gland/Suprarenal gland, Pancreas, Parathyroid Gland, Salivary Glands, Liver, Small Intestine, Bone Marrow, Peripheral Blood, Lymphoid tissue, Lymph node, Cartilage, Brain, Hypothalamus, Spinal Chord, Mammary gland/Breast, Uterus, Prostate, Testis, Lung, Kidney, Epidermis, Hair Follicle.
  • a NOVl 3 polypeptide has been identified as a BHLH Factor MATH6-like protein (also referred to as CG94399-01).
  • the disclosed novel NOV13 nucleic acid (SEQ ID NO:27) of 2244 nucleotides is shown in Table 13 A.
  • the novel NOV 13 nucleic acid sequences maps to the chromosome 2.
  • An ORF begins with an ATG initiation codon at nucleotides 105-107 and ends with a TGA codon at nucleotides 1062-1064.
  • a putative untranslated region and/or downstream from the termination codon is underlined in Table 13 A, and the start and stop codons are in bold letters.
  • a variant sequence can include a single nucleotide polymorphism (SNP).
  • SNP can, in some instances, be referred to as a "cSNP" to denote that the nucleotide sequence containing the SNP originates as a cDNA.
  • the NOV13 protein (SEQ D NO:28) encoded by SEQ ID NO:27 is 319 amino acid residues in length and is presented using the one-letter amino acid code in Table 13B. Psort analysis predicts the NOVl 3 protein of the invention to be localized at the nucleus with a certainty of 0.7000.
  • nucleic acid sequence of this invention has 372 of 657 bases (56%) identical to a gb:GENBANK-ID:HSBBICP4A
  • Bovine herpesvirus 1 Bovine herpesvirus type 1 early-intermediate transcription control protein (BICP4) gene, complete eds).
  • the full amino acid sequence of the protein of the invention was found to have 238 of 322 amino acid residues (73%) identical to, and 244 of 322 amino acid residues (75%) similar to, the 322 amino acid residue ⁇ tnr:TREMBLNEW-ACC:BAB39468 protein from Mus musculus (BHLH FACTOR MATH6).
  • NOV 13 also has homology to the proteins shown in the BLASTP data in Table 13D.
  • NOV 13 has, for example, a Helix-loop-helix domain and a Helix-loop-helix DNA binding domain (HLH) signature sequence as well as homology to other members of the BHLH Factor MATH6-like Protein Family.
  • HHL Helix-loop-helix DNA binding domain
  • NOV 13 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts.
  • NOV 13 nucleic acids and polypeptides can be used to identify proteins that are members of the BHLH Factor MATH6-like Protein Family.
  • the NOV13 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOVl 3 activity or function.
  • the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular replication, and signal transduction.
  • small molecules that modulate or inhibit, e.g., cellular activation, cellular replication, and signal transduction.
  • VHL DiabetesNon Hippel-Lindau
  • Pancreatitis Obesity
  • Inflammatory bowel disease Diverticular disease
  • Non Hippel-Lindau (NHL) syndrome Non Hippel-Lindau
  • Alzheimer's disease, Stroke Tuberous sclerosis
  • hypercalceimia Parkinson's disease
  • Huntington's disease Cerebral palsy
  • Epilepsy Lesch- ⁇ yhan syndrome
  • Multiple sclerosis Ataxia-telangiectasia, Leukodystrophies, Behavioral disorders, Addiction, Anxiety, Pain, Neuroprotection, Systemic lupus erythematosus , Autoimmune
  • NOV13 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins.
  • the NOVl 3 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of protems belonging to the BHLH Factor MATH6-like Protein Family.
  • bHLH basic helix-loop-helix proteins
  • class A ubiquitous
  • class B tissue-specific
  • the HLH proteins lacking the basic domain (Emc, Id) function as negative regulators since they form heterodimers, but fail to bind DNA.
  • the hairy-related proteins hairy, E(spl), deadpan
  • the proteins of this subfamily act together with co-repressor proteins, like groucho, through their C-terminal motif WRPW.
  • MATH6 (Inoue, et al, Genes to Cells 6: 977-86 (2001)) is a distant homolog of Drosophila proneuronal gene Atonal Murine expression is higest in developing nervous system (ventricular zone and mantle layer, spinal cord, dorsal root ganglia). MATH6 is expressed by neuronal precursor cells and designated neurons, e.g., cerebellar Purkinje cells. The closest mammalian homolog to MATH6 is NeuroD. NeuroD point mutations and
  • NeuroD gene knockout animals have severe diabetes and die perinatally.
  • the NeuroD knockout animals lack beta-Islet cells and could not be rescued with insulin administration.
  • the NeuroD knockout animals are deaf due to a loss of inner ear sensory neurons.
  • the NON 13 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of metabolism as well as nerve and immune physiology.
  • the ⁇ ON13 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat metabolic, hearing, nervous system, immune disorders, e.g., DiabetesNon Hippel-Lindau (NHL) syndrome , Pancreatitis, Obesity, Inflammatory bowel disease, Diverticular disease, Non Hippel-Lindau (NHL) syndrome , Alzheimer's disease, Stroke, Tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, Cerebral palsy, Epilepsy, Lesch- ⁇ yhan syndrome, Multiple sclerosis, Ataxia-telangiectasia, Leukodystrophies, Behavioral disorders, Addiction, Anxiety, Pain, Neuroprotection, Systemic lupus erythematosus , Autoimmune disease, Asthma, Emphysema, Scleroderma, allergy as well as other diseases, disorders and conditions.
  • NDL DiabetesNon Hippel-Lind
  • the NON13 nucleic acids and polypeptides are useful for detecting specific cell types. For example, expression analysis has demonstrated that a ⁇ ON13 nucleic acid is expressed in Pancreas, Umbilical Nein, Small Intestine, Cartilage, Synovium Synovial membrane, Brain, Placenta, Oviduct/Uterine Tube/Fallopian tube, Lung, Brain, Uterus.
  • a NOV14 polypeptide has been identified as a Putative Protein-Tyrosine Phosphatase- like protein (also referred to as CG94366-01).
  • the disclosed novel NOV14 nucleic acid (SEQ ID NO:29) of nucleotides is shown in Table 14A.
  • the novel NOV14 nucleic acid sequences maps to the chromosome 22.
  • An ORF begins with an ATG initiation codon at nucleotides 248-250 and ends with a TAA codon at nucleotides 1679-1681.
  • a putative untranslated region and/or downstream from the termination codon is underlined in Table 14 A, and the start and stop codons are in bold letters.
  • the NOV14 protein (SEQ ID NO:30) encoded by SEQ D NO:29 is 477 amino acid residues in length and is presented using the one-letter amino acid code in Table 14B. Psort analysis predicts the NOV14 protein ofthe invention to be localized at the plasma membrane with a certainty of0.6000.
  • nucleic acid sequence of this invention has 1105 of 1427 bases (77%) identical to a gb:GENBANK-ID:AF007118
  • the full amino acid sequence of the protein of the invention was found to have 369 of 462 amino acid residues (79%) identical to, and 402 of 462 amino acid residues (87%) similar to, the 551 amino acid residue ptn ⁇ SWISSNEW- ACC:P56180 protein from Homo sapiens (PUTATIVE PROTETN-TYROSINE PHOSPHATASE TPTE (EC 3.1.3.48)).
  • NOVl 4 also has homology to the proteins shown in the BLASTP data in Table 14D.
  • Table 14E A multiple sequence alignment is given in Table 14E, with the NOV 14 protein being shown on line 1 in Table 14E in a ClustalW analysis, and comparing the NOV14 protein with the related protein sequences shown in Table 14D.
  • This BLASTP data is displayed graphically in the ClustalW in Table 14E.
  • NOVl 4 has, for example, a dual specificity protein phosphatase signature sequence and homology to other members of the Putative Protein-Tyrosine Phosphatase-like Protein Family.
  • NOV14 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts.
  • NOV14 nucleic acids and polypeptides can be used to identify proteins that are members of the Putative Protein-Tyrosine Phosphatase-like Protein Family.
  • the NOV 14 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOV14 activity or function.
  • nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular replication, and signal transduction. These molecules can be used to treat, e.g., Cardiovascular diseases, cystitis, incontinence as well as other diseases, disorders and conditions hi addition, various NOV 14 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins.
  • the NOV14 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the Putative Protein-Tyrosine Phosphatase-like Protein Family.
  • PTPs protein tyrosine phosphatases
  • the protein tyrosine phosphatases which hydrolyze the phosphate monoesters of tyrosine residues, all share a common active site motif and are classified into 3 groups. These include the receptor-like PTPs, the intracellular PTPs, and the dual-specificity PTPs, which can dephosphorylate at serine and threonine residues as well as at tyrosines.
  • Diamond et al (1994) described a PTP from regenerating rat liver that is a member of a fourth class. The gene, which they designated Prll, was one of many immediate-early genes.
  • PTP(CAAXl) and PTP(CAAX2) represent a novel class of isoprenylated, oncogenic PTPs.
  • Peng et al. (1998) reported that the human PTP(CAAXl) gene, or PRL1, is composed of 6 exons and contains 2 promoters. The predicted mouse, rat, and human PRL1 proteins are identical. Zeng et al (1998) determined that the human PRL1 and PRL2 proteins share 87% amino acid sequence identity.
  • the NOV14 nucleic acids and polypeptides, antibodies and related compounds according to the mvention will be useful in therapeutic and diagnostic applications in the mediation of cardiac and renal physiology.
  • the NOVl 4 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat cardiovascular and urogenital system disorders, e.g., Cardiovascular diseases, cystitis, incontinence as well as other diseases, disorders and conditions.
  • the NOV 14 nucleic acids and polypeptides are useful for detecting specific cell types. For example, expression analysis has demonstrated that a NOVl 4 nucleic acid is expressed in Urinary bladder.
  • NOV14 nucleic acids and polypeptides according to the invention are disclosed herein.
  • a NOVl 5 polypeptide has been identified as a Leucine Rich Repeat (LRR)-like protein (also referred to as CG95387-02).
  • LRR Leucine Rich Repeat
  • SEQ ID NO:31 The disclosed novel NOVl 5 nucleic acid (SEQ ID NO:31) of 3136 nucleotides is shown in Table 15A.
  • the novel NOV15 nucleic acid sequences maps to the chromosome 19.
  • An ORF begins with an ATG initiation codon at nucleotides 330-332 and ends with a TAA codon at nucleotides 2331-2333.
  • a putative untranslated region and/or downstream from the termination codon is underlined in Table 15 A, and the start and stop codons are in bold letters.
  • a variant sequence can include a single nucleotide polymo ⁇ hism (SNP).
  • SNP can, in some instances, be referred to as a "cSNP" to denote that the nucleotide sequence containing the SNP originates as a cDNA.
  • the NOVl 5 protein (SEQ D NO:32) encoded by SEQ ID NO:31 is 667 amino acid residues in length and is presented using the one-letter amino acid code in Table 15B. Psort analysis predicts the NOVl 5 protein of the invention to be localized at the plasma membrane with a certainty of 0.7900.
  • nucleic acid sequence of this invention has 2227 of 2228 bases (99%) identical to a gb:GENBANK-ID:AK027073
  • the full amino acid sequence of the protein of the invention was found to have 444 of 444 amino acid residues (100%) identical to, and 444 of 444 amino acid residues (100%) similar to, the 444 amino acid residue ptnr:SPTREMBL-ACC:Q9H5H8 protein from Homo sapiens (CDNA: FLJ23420 FIS, CLONE HEP22352).
  • NOV 15 also has homology to the proteins shown in the BLASTP data in Table 15D.
  • the NOVl 5 Clustal W alignment shown in Table 15E was modified to begin at amino residue 121.
  • the data in Table 15E includes all of the regions overlapping with the NOVl 5 protein sequences.
  • ⁇ OV15 has, for example, eight Leucine Rich Repeat (LRR) signature sequences and homology to other members of the LRR-like Protein Family.
  • LRR Leucine Rich Repeat
  • NOVl 5 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts.
  • NOVl 5 nucleic acids and polypeptides can be used to identify proteins that are members of the LRR-like Protein Family.
  • the NOV 15 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOVl 5 activity or function.
  • nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular replication, and signal transduction.
  • These molecules can be used to treat, e.g., cancer, trauma, regeneration (in vitro and in vivo), viral/bacterial/parasitic infections, Alzheimer's disease, stroke, tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, cerebral palsy, epilepsy, Lesch-Nyhan syndrome, multiple sclerosis, ataxia-telangiectasia, leukodystrophies, behavioral disorders, addiction, anxiety, pain, neurodegeneration, Von Hippel-Lindau (VHL) syndrome, cirrhosis, transplantation, Hirschsprung's disease , Crohn's Disease, appendicitis, osteoporosis, hypercalceimia, arthritis, ankylosing spondylitis, scoliosis, systemic lupus
  • NOVl 5 nucleic acids and polypeptides according to the invention are useful, ter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins.
  • the NOVl 5 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the LRR-like Protein Family.
  • LRR Proteins are a family of proteins characterized by a structural motif rich in leucine residues. They are either transmembrane or secreted proteins and are involved in protein- protein interactions. Members of this family have been implicated in extracellular matrix assembly and cellular growth.
  • LRRs are relatively short motifs (22-28 residues in length) found in a variety of cytoplasmic, membrane and extracellular proteins. Although these proteins are associated with widely different functions, a common property involves protein-protein interaction. Little is known about' the 3D structure of LRRs, although it is believed that they can form amphipathic structures with hydrophobic surfaces capable of interacting with membranes. In vitro studies of a synthetic LRR from Drosophila Toll protein have indicated that the peptides form gels by adopting beta-sheet structures that form extended filaments (Packman et al. FEBS Lett.1991; 291 : 87-91). These results are consistent with the idea that LRRs mediate protein-protein interactions and cellular adhesion. Other functions of LRR-containing proteins include, for example, binding to enzymes and vascular repair.
  • the NOVl 5 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of cardiac, immune, and nerve physiology.
  • the NOV 15 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat cardiovascular, nervous, and immune system disorders, e.g., cancer, trauma, regeneration (in vitro and in vivo), viral/bacterial/parasitic infections, Alzheimer's disease, stroke, tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, cerebral palsy, epilepsy, Lesch-Nyhan syndrome, multiple sclerosis, ataxia-telangiectasia, leukodystrophies, behavioral disorders, addiction, anxiety, pain, neurodegeneration, Von Hippel-Lindau (VHL) syndrome, cirrhosis, transplantation, Hirschsprung's disease, Crohn's Disease, appendicitis, osteoporosis, hypercalceimia, arthritis, anky
  • NOV 15 nucleic acid is expressed in Coronary Artery, Parotid Salivary glands, Liver, Colon, Bone, Synovium/Synovial membrane, and Brain.
  • NOV 15 nucleic acids and polypeptides according to the invention are disclosed herein.
  • NOV16 is disclosed herein.
  • a NOVl 6 polypeptide has been identified as a RhoGEF-like protein (also referred to as CG95419-02).
  • the disclosed novel NOV16 nucleic acid (SEQ ID NO:33) of 5372 nucleotides is shown in Table 16A.
  • the novel NOVl 6 nucleic acid sequences maps to the chromosome 5.
  • Ail ORF begins with an ATG initiation codon at nucleotides 61-63 and ends with a TAA codon at nucleotides 5179-5181.
  • a putative untranslated region and/or downstream from the termination codon is underlined in Table 16 A, and the start and stop codons are in bold letters.
  • the NOV16 protein (SEQ ID NO:34) encoded by SEQ ID NO:33 is 1706 amino acid residues in length and is presented using the one-letter amino acid code in Table 16B. Psort analysis predicts the NOVl 6 protein of the invention to be localized in the cytoplasm with a certainty of 0.4500.
  • nucleic acid sequence of this invention has 4339 of 5274 bases (82%) identical to a gb:GENBANK-rD:MMU73199
  • the full amino acid sequence of the protein of the mvention was found to have 1350 of 1670 amino acid residues (80%) identical to, and 1460 of 1670 amino acid residues (87%) similar to, the 1693 amino acid residue ptnr:SWISSPROT-ACC:P97433 protein from as musculus (RHO-GUANINE NUCLEOTIDE EXCHANGE FACTOR (RHOGEF) (RIP2)).
  • NOVl 6 also has homology to the proteins shown in the BLASTP data in Table 16D.
  • Table 16D BLAST results for NOV16
  • Table 16E A multiple sequence alignment is given in Table 16E, with the NOV16 protein being shown on line 1 in Table 16E in a ClustalW analysis, and comparing the NOV16 protein with the related protein sequences shown in Table 16D.
  • This BLASTP data is displayed graphically in the ClustalW in Table 16E.
  • Rho interacting protein 2 > gi]7106395]/ Rho interacting protein 2; Rho specific exchange factor [Mus musculus]; SEQ ID NO: 1;
  • Rho interacting protein 2 > gi
  • NOV 16 has, for example, a RhoGEF signature sequence and homology to other members of the RhoGEF- like Protein Family.
  • NOVl 6 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts.
  • NOVl 6 nucleic acids and polypeptides can be used to identify proteins that are members of the RhoGEF-like Protein Family.
  • the NOVl 6 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOVl 6 activity or function.
  • nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular replication, and signal transduction. These molecules can be used to treat, e.g.
  • VHL Von Hippel-Lindau
  • graft versus host disease Alzheimer's disease, stroke, tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, cerebral palsy, epilepsy, Lesch-Nyhan syndrome, multiple sclerosis, ataxia-telangiectasia, leukodystrophies, behavioral disorders, addiction, anxiety, pain, neurodegeneration, cirrhosis, transplantation, adrenoleukodystrophy , congenital adrenal hyperplasia, hemophilia, hypercoagulation, idiopathic thrombocytopenic purpura, autoimmune disease, allergies, immunodeficiencies
  • NOV 16 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins.
  • the NOVl 6 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the RhoGEF-like Protein Family.
  • GEF Guide nucleotide exchange factor
  • Rho/Rac/Cdc42-like GTPases is also called Dbl-homologous (DH) domain.
  • DH Dbl-homologous domain.
  • PH pleckstrin homology domains invariably occur C-terminal to RhoGEF/DH domains.
  • binding to the beta/gamma subunit of heterotrimeric G proteins binding to lipids, e.g. phosphatidylinositol-4,5-bisphosphate, binding to phosphorylated Ser/Thr residues, attachment to membranes by an unknown mechanism.
  • the DAG_PE-binding domain binds two zinc ions; the ligands of these metal ions are probably the six cysteines and two histidines that are conserved in this domain and can regulate signal transduction by the PKC family of kinases.
  • NOVl 6 belongs to the guanine nucleotide exchange factor family of proteins which play a significant role in signal transduction.
  • the guanine nucleotide exchange factor (GEF) domain that regulates GTP binding protein signaling.
  • the GEF domain regulates positively the signaling cascades that utilize GTP-binding proteins (such as those of the ras superfamily) that function as molecular switches in fundamental events such as signal transduction, cytoskeleton dynamics and intracellular trafficking.
  • FGD1 farnesoid dyplasia protein
  • Rho family GTPase Cdc42Hs Rho family GTPase Cdc42Hs
  • the GEF domain of FGD1 has also been shown to activate 2 kinases involved in cell proliferation; the Jun NH2- terminal kinase and the p70 S6 kinase (Zheng et al; J. Biol. Chem 1996 Dec 27;271(52):33169-72).
  • NOV16 polypeptide may play an important role in normal development as well as disease.
  • This class of molecules (GEFs) is also being considered as a good drug target as the guanine nucleotide exchange factor RasGRP is a high -affinity target for diacylglycerol and phorbol esters and is bound by bryostatin 1, a compound currently in clinical trials (Lorenzo et al; Mol. Pharmacol 2000 May; 57(5):840-6).
  • the homolog of RhoGEF, DRhoGEF2 fail to gastrulate due to a defect in cell shape changes required for tissue invagination and the mRNA is found throughout oogenesis and embryogenesis (Barrett et al.
  • RhoGEF also interacts with c-Jun amino-terminal kinase (JNK) interacting protein- 1 (JIP-1).
  • JEP-1 might function as a scaffold protein by complexing specific components of the JNK signaling pathway, namely JNK, mitogen-activated protein kinase kinase 7, and mixed lineage kinase 3 (Meyer et al; J Biol Chem 1999; 274(49):35113-8).

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Toxicology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

Disclosed are polypeptides and nucleic acids encoding same. Also disclosed are vectors, host cells, antibodies and recombinant methods for producing the polypeptides and polynucleotides, as well as methods for using same.

Description

PROTEINS AND NUCLEIC ACIDS ENCODING SAME
FIELD OF THE INVENTION
The invention relates to polynucleotides and the polypeptides encoded by such polynucleotides, as well as vectors, host cells, antibodies and recombinant methods for producing the polypeptides and polynucleotides, and methods for using the same.
BACKGROUND OF THE INVENTION
The invention generally relates to nucleic acids and polypeptides encoded therefrom. More specifically, the invention relates to nucleic acids encoding cytoplasmic, nuclear, membrane bound, and secreted polypeptides, as well as vectors, host cells, antibodies, and recombinant methods for producing these nucleic acids and polypeptides.
Heart disease is the primary cause of death in most western societies. Death from heart disease is often induced by platelet-dependent ischemic syndromes which are initiated by atherosclerosis and arteriosclerosis and include, but are not limited to, acute myocardial infarction, clironic unstable angina, transient ischemic attacks and strokes, peripheral vascular disease, arterial thrombosis, preeclampsia, embolism, restenosis and/or thrombosis following angioplasty, carotid endarterectomy, anastomosis of vascular grafts, and chronic cardiovascular devices (e.g., in-dwelling catheters or shunts "extracorporeal circulating devices"). These syndromes represent a variety of stenotic and occlusive vascular disorders thought to be initiated by platelet activation either on vessel walls or within the lumen by blood-borne mediators but are manifested by platelet aggregates which form thrombi that restrict blood flow.
For example, Thrombospondin-l-like proteins associate with the extracellular matrix and inhibits angiogenesis in vivo. In. vitro, Thrombospondin-like proteins block capillary-like tube formation and endothelial cell proliferation. The antiangiogenic activity is mediated by a region that contains 3 type 1 (properdin or thrombospondin) repeats.
In addition, Selectin-like proteins such as P-selectin, also called GMP-140, CD62, or selectin P, is a 140-kD adhesion molecule, expressed at the surface of activated cells, that mediates the interaction of activated endothelial cells or platelets with leukocytes. In endothelial cells, the protein is localized to the membranes of Weibel-Palade bodies, the intracellular storage granules for von Willebrand factor. Many disease states are characterized by uncontrolled cell proliferation. These diseases involve a variety of cell types and include disorders such as cancer, psoriasis, pulmonary fibrosis, glomeralonepl ritis, atherosclerosis and restenosis following angioplasty. Vital cellular functions such as cell proliferation and signal transduction are regulated in part by the balance between the activities of protein-tyrosine kinases (PTK) and protein-tyrosine phosphatases (PTPase). Oncogenesis can result from an imbalance.
SUMMARY OF THE INVENTION
The invention is based in part upon the discovery of nucleic acid sequences encoding novel polypeptides. The novel nucleic acids and polypeptides are referred to herein as NONX, or ΝON1, ΝON2, ΝON3, ΝON4, ΝON5, ΝON6, ΝON7, ΝON8, ΝON9, ΝONlOa, ΝONlOb, ΝON11, ΝON12, ΝON13, ΝON14, ΝON15, and ΝON16 nucleic acids and polypeptides. These nucleic acids and polypeptides, as well as variants, derivatives, homologs, analogs and fragments thereof, will hereinafter be collectively designated as "ΝONX" nucleic acid or polypeptide sequences. hi one aspect, the invention provides an isolated ΝONX nucleic acid molecule encoding a ΝONX polypeptide that includes a nucleic acid sequence that has identity to the nucleic acids disclosed in SEQ ID ΝOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33. In some embodiments, the NOVX nucleic acid molecule will hybridize under stringent conditions to a nucleic acid sequence complementary to a nucleic acid molecule that includes a protein-coding sequence of a NONX nucleic acid sequence. The invention also includes an isolated nucleic acid that encodes a ΝONX polypeptide, or a fragment, homolog, analog or derivative thereof. For example, the nucleic acid can encode a polypeptide at least 80% identical to a polypeptide comprising the amino acid sequences of SEQ ID ΝOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34. The nucleic acid can be, for example, a genomic DNA fragment or a cDNA molecule that includes the nucleic acid sequence of any of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33.
Also included in the invention is an oligonucleotide, e.g., an oligonucleotide which includes at least 6 contiguous nucleotides of a NONX nucleic acid (e.g., SEQ ID ΝOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33) or a complement of said oligonucleotide.
Also included in the invention are substantially purified NONX polypeptides (SEQ ID ΝOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34). In certain embodiments, the NONX polypeptides include an amino acid sequence that is substantially identical to the amino acid sequence of a human ΝONX polypeptide.
The invention also features antibodies that immunoselectively bind to ΝONX polypeptides, or fragments, homologs, analogs or derivatives thereof. In another aspect, the invention includes pharmaceutical compositions that include therapeutically- or prophylactically-effective amounts of a therapeutic and a pharmaceutically- acceptable carrier. The therapeutic can be, e.g., a ΝONX nucleic acid, a ΝONX polypeptide, or an antibody specific for a ΝOVX polypeptide. In a further aspect, the invention includes, in one or more containers, a therapeutically- or prophylactically-effective amount of this pharmaceutical composition.
In a further aspect, the invention includes a method of producing a polypeptide by culturing a cell that includes a ΝONX nucleic acid, under conditions allowing for expression of the ΝONX polypeptide encoded by the DΝA. If desired, the ΝONX polypeptide can then be recovered. In another aspect, the invention includes a method of detecting the presence of a
ΝONX polypeptide in a sample. In the method, a sample is contacted with a compound that selectively binds to the polypeptide under conditions allowing for formation of a complex between the polypeptide and the compound. The complex is detected, if present, thereby identifying the ΝONX polypeptide within the sample. The invention also includes methods to identify specific cell or tissue types based on their expression of a ΝONX.
Also included in the invention is a method of detecting the presence of a ΝONX nucleic acid molecule in a sample by contacting the sample with a ΝONX nucleic acid probe or primer, and detecting whether the nucleic acid probe or primer bound to a ΝONX nucleic acid molecule in the sample.
In a further aspect, the invention provides a method for modulating the activity of a ΝONX polypeptide by contacting a cell sample that includes the ΝONX polypeptide with a compound that binds to the ΝONX polypeptide in an amount sufficient to modulate the activity of said polypeptide. The compound can be, e.g., a small molecule, such as a nucleic acid, peptide, polypeptide, peptidomimetic, carbohydrate, lipid or other organic (carbon containing) or inorganic molecule, as further described herein.
Also within the scope of the invention is the use of a therapeutic in the manufacture of a medicament for treating or preventing disorders or syndromes including, e.g., those described for the individual NONX nucleotides and polypeptides herein, and/or other pathologies and disorders of the like.
The therapeutic can be, e.g., a ΝONX nucleic acid, a ΝONX polypeptide, or a ΝONX- specific antibody, or biologically-active derivatives or fragments thereof. For example, the compositions of the present invention will have efficacy for treatment of patients suffering from the diseases and disorders disclosed below and/or other pathologies and disorders of the like. The polypeptides can be used as immunogens to produce antibodies specific for the invention, and as vaccines. They can also be used to screen for potential agonist and antagonist compounds. For example, a cDΝA encoding ΝONX may be useful in gene therapy, and ΝONX may be useful when administered to a subject in need thereof. By way of non-limiting example, the compositions of the present invention will have efficacy for treatment of patients suffering from the diseases and disorders disclosed above and/or other pathologies and disorders of the like.
The invention further includes a method for screening for a modulator of disorders or syndromes including, e.g., the diseases and disorders disclosed above and/or other pathologies and disorders of the like. The method includes contacting a test compound with a ΝONX polypeptide and determining if the test compound binds to said ΝONX polypeptide. Binding of the test compound to the ΝONX polypeptide indicates the test compound is a modulator of activity, or of latency or predisposition to the aforementioned disorders or syndromes. Also within the scope of the invention is a method for screening for a modulator of activity, or of latency or predisposition to an disorders or syndromes including, e.g., the diseases and disorders disclosed above and/or other pathologies and disorders of the like by administering a test compound to a test animal at increased risk for the aforementioned disorders or syndromes. The test animal expresses a recombinant polypeptide encoded by a ΝONX nucleic acid. Expression or activity of ΝONX polypeptide is then measured in the test animal, as is expression or activity of the protein in a control animal which recombinantly- expresses ΝONX polypeptide and is not at increased risk for the disorder or syndrome. Next, the expression of NOVX polypeptide in both the test animal and the control animal is compared. A change in the activity of NONX polypeptide in the test animal relative to the control animal indicates the test compound is a modulator of latency of the disorder or syndrome.
In yet another aspect, the mvention includes a method for determining the presence of or predisposition to a disease associated with altered levels of a ΝONX polypeptide, a ΝONX nucleic acid, or both, in a subject (e.g., a human subject). The method includes measuring the amount of the NONX polypeptide in a test sample from the subject and comparing the amount of the polypeptide in the test sample to the amount of the ΝONX polypeptide present in a control sample. An alteration in the level of the ΝONX polypeptide in the test sample as compared to the control sample indicates the presence of or predisposition to a disease in the subject. Preferably, the predisposition includes, e.g., the diseases and disorders disclosed above and/or other pathologies and disorders of the like. Also, the expression levels of the new polypeptides of the invention can be used in a method to screen for various cancers as well as to determine the stage of cancers.
In a further aspect, the invention includes a method of treating or preventing a pathological condition associated with a disorder in a mammal by administering to the subject a ΝONX polypeptide, a ΝONX nucleic acid, or a ΝONX-specific antibody to a subject (e.g., a human subject), in an amount sufficient to alleviate or prevent the pathological condition. In preferred embodiments, the disorder, includes, e.g., the diseases and disorders disclosed above and/or other pathologies and disorders of the like. h yet another aspect, the invention can be used in a method to identity the cellular receptors and downstream effectors of the invention by any one of a number of techniques commonly employed in the art. These include but are not limited to the two-hybrid system, affinity purification, co-precipitation with antibodies or other specific-interacting molecules. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In the case of conflict, the present specification, including definitions, will control, h addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
Other features and advantages of the invention will be apparent from the following detailed description and claims.
DETAILED DESCRIPTION OF THE INVENTION The present invention provides novel nucleotides and polypeptides encoded thereby.
Included in the invention are the novel nucleic acid sequences and their polypeptides. The sequences are collectively referred to as "NONX nucleic acids" or "ΝONX polynucleotides" and the corresponding encoded polypeptides are referred to as "ΝONX polypeptides" or "NONX proteins." Unless indicated otherwise, "ΝONX" is meant to refer to any of the novel sequences disclosed herein. Table A provides a summary of the ΝONX nucleic acids and their encoded polypeptides.
TABLE A. Sequences and Corresponding SEQ ID Numbers
Figure imgf000007_0001
NOVX nucleic acids and their encoded polypeptides are useful in a variety of applications and contexts. The various NOVX nucleic acids and polypeptides according to the invention are useful as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. Additionally, NOVX nucleic acids and polypeptides can also be used to identify proteins that are members of the family to which the NONX polypeptides belong.
The ΝONX genes and their corresponding encoded proteins are useful for preventing, treating or ameliorating medical conditions, e.g., by protein or gene therapy. Pathological conditions can be diagnosed by determining the amount of the new protein in a sample or by determining the presence of mutations in the new genes. Specific uses are described for each of the sixteen genes, based on the tissues in which they are most highly expressed. Uses include developing products for the diagnosis or treatment of a variety of diseases and disorders.
The ΝONX nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance ΝONX activity or function. Specifically, the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cell growth, cell metabolism, cell differentiation, cell proliferation, and/or cell signaling.
In one embodiment of the present invention, NONX or a fragment or derivative thereof may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of ΝONX. Examples of such disorders include, but are not limited to, cancers such as adenocarcinoma, leukemia, lymphoma, melanoma, myeloma, sarcoma, teratocarcinoma, and, in particular, cancers of the adrenal gland, bladder, bone, bone marrow, brain, breast, cervix, gall bladder, ganglia, gastrointestinal tract, heart, kidney, liver, lung, muscle, ovary, pancreas, parathyroid, penis, prostate, salivary glands, skin, spleen, testis, thymus, thyroid, and uterus; neurological disorders such as epilepsy, ischemic cerebrovascular disease, stroke, cerebral neoplasms, Alzheimer's disease, Pick's disease, Huntington's disease, dementia, Parkinson's disease and other extrapyramidal disorders, amyotrophic lateral sclerosis and other motor neuron disorders, progressive neural muscular atrophy, retinitis pigmentosa, hereditary ataxias, multiple sclerosis and other demyelinating diseases, bacterial and viral meningitis, brain abscess, subdural empyema, epidural abscess, suppurative intracranial thrombophlebitis, myelitis and radiculitis, viral central nervous system disease, prion diseases including kuru, Creutzfeldt- Jakob disease, and Gerstmann-Straussler-Scheinker syndrome, fatal familial insomnia, nutritional and metabolic diseases of the nervous system, neurofibromatosis, tuberous sclerosis, cerebelloretinal hemangioblastomatosis, encephalotrigeminal syndrome, mental retardation and other developmental disorders of the central nervous system, cerebral palsy, neuroskeletal disorders, autonomic nervous system disorders, cranial nerve disorders, spinal cord diseases, muscular dystrophy and other neuromuscular disorders, peripheral nervous system disorders, dermatomyositis and polymyositis, inherited, metabolic, endocrine, and toxic myopathies, myasthenia gravis, periodic paralysis, mental disorders including mood, anxiety, and schizophrenic disorders, akathesia, amnesia, catatonia, diabetic neuropathy, tardive dyskinesia, dystonias, paranoid psychoses, postherpetic neuralgia, and Tourette's disorder; and disorders of vesicular transport such as cystic fibrosis, glucose-galactose malabsorption syndrome, hypercholesterolemia, diabetes mellitus, diabetes insipidus, hyper- and hypoglycemia, Grave's disease, goiter, Cushing's disease, Addison's disease, gastrointestinal disorders including ulcerative colitis, gastric and duodenal ulcers, other conditions associated with abnormal vesicle trafficking including acquired immunodeficiency syndrome (AIDS), allergic reactions, autoimmune hemolytic anemia, proliferative glomerulonephritis, inflammatory bowel disease, multiple sclerosis, myasthenia gravis, rheumatoid arthritis, osteoarthritis, scleroderma, Chediak-Higashi syndrome, Sjogren's syndrome, systemic lupus erythiematosus, toxic shock syndrome, traumatic tissue damage, and viral, bacterial, fungal, helminthic, and protozoal infections, as well as additional indications listed for the individual NONX clones.
The ΝONX nucleic acids and proteins of the invention are useful in potential diagnostic and therapeutic applications and as a research tool. These include serving as a specific or selective nucleic acid or protein diagnostic and or prognostic marker, wherein the presence or amount of the nucleic acid or the protein are to be assessed. These also include potential therapeutic applications such as the following: (i) a protein therapeutic, (ii) a small molecule drug target, (iii) an antibody target (therapeutic, diagnostic, drug targeting/cytotoxic antibody), (iv) a nucleic acid useful in gene therapy (gene delivery/gene ablation), (v) an agent promoting tissue regeneration in vitro and in vivo, and (vi) a biological defense weapon. Additional utilities for the ΝONX nucleic acids and polypeptides according to the invention are disclosed herein.
ΝOV1 AΝOV1 polypeptide has been identified as aPaladin-like protein (also referred to as CG93221-01). The disclosed novel ΝOV1 nucleic acid (SEQ ID ΝO:l) of2600 nucleotides is shown in Table 1A. The novel NOV1 nucleic acid sequences maps to the chromosome 10. An ORF begins with an ATG initiation codon at nucleotides 15-17 and ends with a TAG codon at nucleotides 2583-2585. A putative untranslated region and/or downstream from the termination codon is underlined in Table 1A, and the start and stop codons are in bold letters.
Table IA. NOV1 Nucleotide Sequence (SEQ TD NO:l)
GCTGCTGGCAGACTATGGGTACAΆCGGCCAGCACAGCCCAGCAGACGGTCTCGGCAGGCACCCCATT TGAGGGCCTACAGGGCAGTGGCACGATGGACAGTCGGCACTCCGTCAGCATCCACTCCTTCCAGAGC ACTAGCTTGCATAACAGCAAGGCCAAGTCCATCATCCCCAACAAGGTGGCCCCTGTTGTGATCACGT ACAACTGCAAGGAGGAGTTCCAGATCCATGATGAGCTGCTCAAGGCTCATTACACGTTGGGCCGGCT CTCGGACAACACCCCTGAGCACTACCTGGTGCAAGGCCGCTACTTCCTGGTGCGGGATGTCACTGAG AAGATGGATGTGCTGGGCACCGTGGGAAGCTGTGGGGCCCCCAACTTCCGGCAGGTGCAGGGTGGGC TCACTGTGTTCGGCATGGGACAGCCCAGCCTCTTAGGGTTCAGGCGGGTCCTCCAGAAACTCCAGAA GGACGGACATAGGGAGTGTGTCATCTTCTGTGTGCGGGAGGAACCTGTGCTTTTCCTGCGTGCAGAT GAGGACTTTGTGTCCTACACACCTCGAGACAAGCAGAACCTTCATGAGAACCTCCAGGGCCTTGGAC CCGGGGTCCGGGTGGAGAGCCTGGAGCTGGCCATCCGGAAAGAGATCCACGACTTTGCCCAGCTGAG CGAGAACACATACCATGTGTACCATAACACCGAGGACCTGTGGGGGGAGCCCCATGCTGTGGCCATC CATGGTGAGGACGACTTGCATGTGACGGAGGAGGTGTACAAGCGGCCCCTCTTCCTGCAGCCCACCT ACAGGTACCACCGCCTGCCCCTGCCCGAGCAAGGGAGTCCCCTGGAGGCCCAGTTGGACGCCTTTGT CAGTGTTCTCCGGGAGACCCCCAGCCTGCTGCAGCTCCGTGATGCCCACGGGCCTCCCCCAGCCCTC GTCTTCAGCTGCCAGATGGGCGTGGGCAGGACCAACCTGGGCATGGTCCTGGGCACCCTCATCCTGC TTCACCGCAGTGGGACCACCTCCCAGCCAGAGGCTGCCCCCACGCAGGCCAAGCCCCTGCCTATGGA GCAGTTCCAGGTGATCCAGAGCTTTCTCCGCATGGTGCCCCAGGGAAGGAGGATGGTGGAAGAGGTG GACAGAGCCATCACTGCCTGTGCCGAGTTGCATGACCTGAAAGAAGTGGTCTTGGAAAACCAGAAGA AGTTAGAAGGTATGCGACCGGAGAGCCCAGCCCAGGGAAGCGGCAGCCGACACAGCGTCTGGCAGAG GGCGCTGTGGAGCCTGGAGCGATACTTCTACCTGATCCTGTTTAACTACTACCTTCATGAGCAGTAC CCGCTGGCCTTTGCCCTCAGTTTCAGCCGCTGGCTGTGTGCCCACCCTGAGCTGTACCGCCTGCCCG TGACGCTGAGCTCAGCAGGCCCTGTGGCTCCGAGGGACCTCATCGCCAGGGGCTCCCTACGGGAGGA CGATCTGGTCTCCCCGGACGCGCTCAGCACTGTCAGAGAGATGGATGTGGCCAACTTCCGGCGGGTG CCCCGCATGCCCATCTACGGCACGGCCCAGCCCAGCGCCAAGGCCCTGGGGAGCATCCTGGCCTACC TGACGGACGCCAAGAGGAGGCTGCGGAAGGTTGTCTGGGTGAGCCTTCGGGAGGAGGCCGTGTTGGA GTGTGACGGGCACACCTACAGCCTGCGGTGGCCTGGGCCCCCTGTGGCTCCTGACCAGCTGGAGACC CTGGAGGCCCAGCTGAAGGCCCATCTAAGCGAGCCTCCCCCAGGCAAGGAGGGCCCCCTGACCTACA GGTTCCAGACCTGCCTTACCATGCAGGAGGTCTTCAGCCAGCACCGCAGGGCCTGTCCTGGCCTCAC CTACCACCGCATCCCCATGCCGGACTTCTGTGCCCCCCGAGAGGAGGACTTTGACCAGCTGCTGGAG GCCCTGCGGGCCGCCCTCTCCAAGGACCCAGGCACTGGCTTCGTGTTCAGCTGCCTCAGCGGCCAGG GCCGTACCACAACTGCGATGGTGGTGGCTGTCCTGGCCTTCTGGCACATCCAAGGCTTCCCCGAGGT GGGTGAGGAGGAGCTCGTGAGTGTGCCTGATGCCAAGTTCACTAAGGGTGAATTTCAGGTAGTAATG AAGGTGGTGCAGCTGCTACCCGATGGGCACCGTGTGAAGAAGGAGGTGGACGCAGCGCTGGACACTG TCAGCGAGACCATGACGCCCATGCACTACCACCTGCGGGAGATCATCATCTGCACCTACCGCCAGGC GAAGGCAGCGAAAGAGGCGCAGGAAATGCGGAGGCTGCAGCTGCGGAGCCTGCAGTACTTGGAGCGC TATGTCTGCCTGATTCTCTTCAACGCGTACCTCCACCTGGAGAAGGCCGACTCCTGGCAGAGGCCCT TCAGCACCTGGATGCAGGAGGTGGCATCGAAGGCTGGCATCTACGAGATCCTTAACGAGCTGGGCTT CCCCGAGCTGGAGAGCGGGGAGGACCAGCCCTTCTCCAGGCTGCGCTACCGGTGGCAGGAGCAGAGC TGCAGCCTCGAGCCCTCTGCCCCCGAGGACTTGCTGTAGGGGGCCTTACTCCCT
Variant sequences of NOV1 are included in Example 3, Table 18. A variant sequence can include a single nucleotide polymorphism (SNP). A SNP can, in some instances, be referred to as a "cSNP" to denote that the nucleotide sequence containing the SNP originates as a cDNA.
The NOV1 protein (SEQ ID NO:2) encoded by SEQ ID NO.T is 856 amino acid residues in length and is presented using the one-letter amino acid code in Table IB. Psort analysis predicts the NO VI protein of the invention to be localized in the cytoplasm with a certainty of 0.4500.
Table IB. Encoded NO VI protein sequence (SEQ ID NO:2)
MGTTASTAQQTVSAGTPFEG QGSGTMDSRHSVSIHSFQSTSLHNSKAKSIIPNKVAPWITYNC KEEFQIHDEL KAHYTLGRLSDNTPEHYLVQGRYFLVRDVTE MDVLGTVGSCGAPNFRQVQGGL TVFGMGQPS LGFRRVLQKLQKDGHRECVIFCVREEPVLFLRADEDFVSYTPRDKQN HEN QGL GPGVRVES ELAIRKEIHDFAQ SENTYHVYHNTEDL GEPHAVAIHGEDDLHVTEEVYKRP F QPTYRYHRLPLPEQGSP EAQLDAFVSVLRETPSLLQLRDAHGPPPALVFSCQMGVGRTNLGMVL GTLILLHRSGTTSQPEAAPTQAKPLPMEQFQVIQSFLRMVPQGRRMVEEVDRAITACAELHDLKE W ENQK LEGIRPESPAQGSGSRHSV QRAL SLERYFY I FNYYLHEQYPLAFALSFSR LC AHPELYRLPVT SSAGPVAPRD IARGS REDDLVSPDA STVREMDVANFRRVPRMPIYGTAQP SAKALGSILAYLTDAKRR RKWWVSLREEAVLECDGHTYSLR PGPPVAPDQLET EAQLKAHL SEPPPGKEGP TYRFQTC TMQEVFSQHRRACPGLTYHRIPMPDFCAPREEDFDQL EALRAALS DPGTGFVFSCLSGQGRTTTAMWAVLAF HIQGFPEVGEEELVSVPDAKFTKGEFQWMKWQL LPDGHRVKKEVDAALDTVSETMTPMHYH REIIICTYRQAKAAKEAQEMRRLQ RSLQYLERYVC LILFNAYLH EKADS QRPFST MQEVASKAGIYEI NELGFPELESGEDQPFSRLRYR QEQSC SLEPSAPED
In all BLAST alignments described herein, the "E- value" or "Expect" value is a numeric indication of the probability that the aligned sequences could have achieved their similarity to the BLAST query sequence by chance alone, within the database that was searched. The Expect value (E) is a parameter that describes the number of hits one can "expect" to see just by chance when searching a database of a particular size. It decreases exponentially with the Score (S) that is assigned to a match between two sequences. Essentially, the E value describes the random background noise that exists for matches between sequences.
The Expect value is used to create a significance threshold for reporting results. The default value used for blasting is typically set to 0.0001, with the filter to remove low complexity sequence turned off. In BLAST 2.0, the Expect value is also used instead of the P value (probability) to report the significance of matches. For example, an E value of one assigned to a hit can be interpreted as meaning that in a database of the current size one might expect to see one match with a similar score simply by chance. An E value of zero means that one would not expect to see any matches with a similar score simply by chance. See, e.g., http://www.ncbi.nlm.nih.gov/Education/BLASTinfo/. Occasionally, a string of X's or N's will result from a BLAST search. This is a result of automatic filtering of the query for low- complexity sequence that is performed to prevent artifactual hits The filter substitutes any low-complexity sequence that it finds with the letter "N" in nucleotide sequence (e.g., "NNNNNNNN") or the letter "X" in protein sequences (e.g., "XXX"). Low-complexity regions can result in high scores that reflect compositional bias rather than significant position- by-position alignment. Wootton and Federhen. Methods Enzymol 266:554-571, (1996).
A search against the Patp database, a proprietary database that contains sequences published in patents and patent publications, yielded several homologous proteins shown in Table IC.
Table IC. Patp results for NOV1
Smallest Sum
Reading High Prob Sequences producing High-scoring Segment Pairs: Frame Score P(N)
>patp:AAB41108 Human ORFX 0RF872 polypeptide +1 4187 0.0
>patp:AAB35276 Murine dual specificity phosphatase DSP-11 +1 120 5.2e- -06
>patp.-A&B73211 Murine phosphatase AA023073 m +1 120 5.2e- -06
>patp:AAB73231 Human phosphatase BAA91172 h +1 115 1.8e- -05
>patp:AAG67455 Amino acid sequence of a human polypeptide 4-1 115 1.8e- -05
In a BLAST search of public sequence databases, it was found, for example, that the nucleic acid sequence of this invention has 2063 of 2508 bases (82%) identical to a gb:GENBANK-ID:MMPAL|acc:X99384.1 mRNA from Mus musculus (Paladin gene). The full amino acid sequence of the protein of the invention was found to have 695 of 859 amino acid residues (80%) identical to, and 754 of 859 amino acid residues (87%) similar to, the 859 amino acid residue ptnr:SPTREMBL-ACC:P70261 protein ϊxom Mus musculus (PALADIN GENE). NOV1 also has homology to the proteins shown in the BLASTP data in Table ID. Table ID. BLAST results for NOVl
Gene Index/ Protein/ Organism Length Identity Positives Expect Identifier (aa) (%) (%) gi|6331287|dbj |BAA8 KIAA1274 protein 752 752/752 752/752 0.0 6588.1] (AB033100) [Homo sapiens] (100%) (100%) gi 114738662 I ref |XP_ KIAA protein 748 747/748 747/748 0.0 046314. l| (similar to mouse (99%) (99%) (XM 046314) paladin) [Homo sapiens] gi I 7305365 | ref |NP_0 paladin [Mus 859 673/841 730/841 0.0 38781. l| musculus] (80%) (86%) (NM 013753) gi 115228672 I ref |NP_ putative protein 1232 207/821 340/821 7e-45 191760. l| [Arabidopsis (25%) (41%) (NM 116066) thaliana] gi 112836455 I dbj |BAB data source : SPTR, 144 24/60 33/60 2e-04 23663.ll (AK004912) source (40%) (55%) key:Q9NX48, evidence : ISS-homo log to CDNA
FLJ20442 FIS,
CLONE
KAT04828~putative
[Mus musculus]
A multiple sequence alignment is given in Table IE, with the NOVl protein being shown on line 1 in Table IE in a ClustalW analysis, and comparing the NOVl protein with the related protein sequences shown in Table ID. This BLASTP data is displayed graphically in the ClustalW in Table IE.
Table IE. ClustalW Analysis of NOVl l) > NOVl; SEQ ID NO:2
2) > gi|6331287/ KIAA1274 protein [Homo sapiens]; SEQ ID NO:35
3) > gi]1473866/ KIAA protein (similar to paladin) [Homo sapiens]; SEQ ID NO:36
4) > gi|7305365/ paladin [Mus musculus]; SEQ ID NO:37
5) > gi|1522867/ putative protein [Arabidopsis thaliana]; SEQ ID NO:38
6) > gi|1283645/ data source: SPTR, source key: Q9NX48, evidence: ISS-homolog to cDNA FLJ20442
FIS clone KAT04828 putative [Mus musculus]; SEQ ID NO:39
10 20 30 40 50
NOVl MGTTASTAQQTVSAGTPFEGLQGSGT- -MDSRHSVS - IHSFQSTSLHNSK gi I 6331287 gi j 1473866 gi j 7305365 MGTTASTAQQTVSAGTSLEGLQGGSSSSMDSQHSLGGVQSFRATSLHNSK gi 1 1522867 gi 1 1283645
60 70 80 90 100
NOVl A SIIPNKVAPWITYFCKEEFQIHDELL AHYTLGRLSDNTPEHYLVQG
AKSIIPNKVAPWITYNCKEEFQIHDELLKAHYR GRLSDATPEHY VQG MSIPKEPEQVMKMRDGSVLGKK
Figure imgf000012_0001
Figure imgf000013_0001
Figure imgf000014_0001
560 570 580 590 600
NOVl VREMDVANFRRVPRMPIYGTAQPSAKALGSILAYLHDAKI gi|6331287 VREMDVANFRRVPRMPIYGTAQPSAKALGSILAYLHDAKI gijl473866 VREMDVANFRRVPRMPIYGTAQPSAKALGSILAYLHDAKI giJ7305365 VRE DVAWFRRVPRMPIYGTAQPSAKALGiSlLAYLBDAiα gi j 1522867 SPGCQILN PEJ3 EGAPJJg3EggGF|j|v^ g @TIDGIR§ IER GSSR gij 1283645
Figure imgf000014_0002
Figure imgf000015_0001
1010 1020 1030 1040 1050
NOVl PlsJSRLRYRWQEQS CSLB 9SSPER
PiaSRLRYRWQEQS CSLS asjaPEg
CSLB SgPEl
RDPiaJaCDVGiBl _ QINGAPHVYKVDRYPVYSMgτPTISGAKKMLAYLGTKLKEEGGGSTERIV
Figure imgf000015_0002
The NOVl Clustal W alignment shown in Table IE was modified to begin at amino residue 1050. The data in Table IE includes all of the regions overlapping with the NOVl protein sequences.
The presence of identifiable domains in the protein disclosed herein was determined by searches using algorithms such as PROSITE, Blocks, Pfam, ProDomain, Prints and then determining the Interpro number by crossing the domain match (or numbers) using the Interpro website (http:www.ebi.ac.uk/interpro/). Table IF lists the domain description from DOMAIN analysis results against NOVl.
Figure imgf000016_0001
Consistent with other known members of the Paladin-like family of proteins, NOVl has, for example, multiple Paladin gene signature sequences and homology to other members of the Paladin-like Protein Family. NOV 1 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts. For example, NOVl nucleic acids and polypeptides can be used to identify proteins that are members of the Paladin like family of proteins. The NOVl nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOVl activity or function. Specifically, the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular metabolism and signal transduction. These molecules can be used to treat, e.g., cardiomyopathy, atherosclerosis, hypertension, congenital heart defects, aortic stenosis, atrial septal defect (ASD),atrioventricular (A-V) canal defect, ductus arteriosus, pulmonary stenosis, subaortic stenosis, ventricular septal defect (VSD), valve diseases, tuberous sclerosis, scleroderma, obesity, transplantation, adrenoleukodystrophy, congenital adrenal hyperplasia, diabetes, Von Hippel-Lindau (NHL) syndrome, pancreatitis, obesity, hyperthyroidism and hypothyroidism, hypercalceimia, ulcers, cirrhosis, transplantation, inflammatory bowel disease, diverticular disease, hemophilia, hypercoagulation, idiopathic thrombocytopenic purpura, autoimmume disease, allergies, immunodeficiencies, transplantation, graft vesus host, hemophilia, hypercoagulation, idiopathic thrombocytopenic purpura, autoimmume disease, allergies, immunodeficiencies, transplantation, graft versus host disease (GVHD), lymphedema, Alzheimer's disease, stroke, tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, cerebral palsy, epilepsy, Lesch-Nyhan syndrome, multiple sclerosis, ataxia-telangiectasia, leukodystrophies, behavioral disorders, addiction, anxiety, pain, neuroprotection, cancer, trauma, regeneration (in vitro and in vivo), viral/bacterial/parasitic infections, as well as other diseases, disorders and conditions. In addition, various NOVl nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. For example, the NOVl nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the Paladin-like protein family. Paladin proteins are a family of protein-tyrosine phosphatases. The protein phosphatases can be divided into 2 large families: the serine/threonine phosphatases, which are metalloproteins, and the protein-tyrosine phosphatases, which proceed via a thiol-phosphate enzyme intermediate. The protein-tyrosine phosphatase family includes the VHl-like dual- specificity phosphatases. These phosphatases dephosphorylate phosphotyrosine- as well as phosphoserine- and phosphothreonine-containing substrates. Members of the dual-specificity phosphatase protein family inactivate mitogen-activated protein (MAP) kinase through dephosphorylation of critical threonine and tyrosine residues. Members of the MAP kinase family play a pivotal role in cellular signal transduction. Using a subtractive screen of mouse gastrulation, Pearce et al. (1996) identified a novel mouse gene, paladin, with similarity to the dual specificity protein phosphatase family.
The NOVl nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of cardiac and endocrine physiology. As such, the NOVl nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat muscle and nervous system disorders, e.g., cardiomyopathy, atherosclerosis, hypertension, congenital heart defects, aortic stenosis, atrial septal defect (ASD),atrioventricular (A-V) canal defect, ductus arteriosus, pulmonary stenosis, subaortic stenosis, ventricular septal defect (VSD), valve diseases, tuberous sclerosis, scleroderma, obesity, transplantation, adrenoleukodystrophy, congenital adrenal hyperplasia, diabetes, Von Hippel-Lindau (VHL) syndrome, pancreatitis, obesity, hyperthyroidism and hypothyroidism, hypercalceimia, ulcers, cirrhosis, transplantation, inflammatory bowel disease, diverticular disease, hemophilia, hypercoagulation, idiopathic thrombocytopenic purpura, autoimmume disease, allergies, immunodeficiencies, transplantation, graft vesus host, hemophilia, hypercoagulation, idiopathic thrombocytopenic purpura, autoimmume disease, allergies, immunodeficiencies, transplantation, graft versus host disease (GVHD), lymphedema, Alzheimer's disease, stroke, tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, cerebral palsy, epilepsy, Lesch-Nyhan syndrome, multiple sclerosis, ataxia-telangiectasia, leukodystrophies, behavioral disorders, addiction, anxiety, pain, neuroprotection, cancer, trauma, regeneration (in vitro and in vivo), viral/bacterial/parasitic infections, as well as other diseases, disorders and conditions.
The NOVl nucleic acids and polypeptides are useful for detecting specific cell types. For example, expression analysis has demonstrated that a NOVl nucleic acid is expressed in brown adipose, heart, aorta, vein, umbilical vein, adrenal gland/suprarenal gland, pancreas, thyroid, salivary glands, parotid salivary glands, stomach, liver, gall bladder, small intestine, colon, bone marrow, lymphoid tissue, spleen, lymph node, tonsils, thymus, cartilage, muscle, brain, thalamus, hypothalamus, pituitary gland, amygdala, substantia nigra, hippocampus, spinal chord, cervix, mammary gland/breast, ovary, placenta, uterus, vulva, prostate, testis, lung, lung pleura, kidney, retina, dermis. Additional utilities for NOVl nucleic acids and polypeptides according to the invention are disclosed herein.
NOV2
A NOV2 polypeptide has been identified as a Plasma Membrane Ring Finger-like protein (also referred to as CG93210-01). The disclosed novel NOV2 nucleic acid (SEQ ID NO:3) of 1205 nucleotides is shown in Table 2A. The novel NOV2 nucleic acid sequences maps to the chromosome 22.
An ORF begins with an ATG initiation codon at nucleotides 17-19 and ends with a ATT codon at nucleotides 1149-1151. A putative untranslated region and/or downstream from the termination codon is underlined in Table 2A, and the start and stop codons are in bold letters.
Table 2A. NOV2 Nucleotide Sequence (SEQ ID NO:3)
CτCGCCGGGTCCGGCCATGGGCCCCGCCGCTCGCCCCGCGCTGAGATCGCCGCCGCGGCCTCCGCCG CCGCCTCCGTCTCCGCTGCTGCTGCTGCTGCCCCTGCTGCCGCTGTGGCTGGGCCTGGCGGGGCCCG GGGCCGCGGCGGACGGCAGCGAGCCGGCGGCCGGGGCGGGGCGGGGCGGAGCCCGCGCCGTGCGGGT GGACGTGAGACTGCCGCGCCAGGACGCTCTGGTCCTGGAGGGCGTCAGGATCGGCTCCGAAGCCGAC CCGGCGCCCCTGCTGGGCGGTCGTCTGCTGCTGATGGACATCGTGGATGCCGAGCAGGAGGCACCAG TGGAAGGCTGGATTGCAGTGGCATACGTGGGCAAGGAGCAGGCGGCCCAGTTCCACCAGGAGAATAA GGGCAGTGGCCCGCAGGCCTATCCCAAGGCCCTGGTCCAGCAGATGCGGCGGGCCCTCTTCCTGGGT GCCTCTGCCCTGCTTCTTCTCATCCTGAACCACAACGTGGTCCGAGAGCTGGACATATCCCAGCTTC TGCTCAGGCCAGTGATCGTCCTCCATTATTCCTCCAATGTCACCAAGCTGTTGGATGCATTGCTGCA GAGGACCCAGGCCACGGCTGAGATCACCAGCGGAGAGTCCCTGTCTGCCAATATCGAGTGGAAGTTG ACCTTGTGGACCACCTGTGGCCTCTCCAAGGATGGCTATGGAGGATGGCAGGACTTGGTCTGCCTTG GAGGCAGTCGTGCCCAGGAGCAGAAACCCCTGCAGCAGCTGTGGAACGCCATCCTGCTGGTGGCCAT GCTCCTGTGCACAGGCCTCGTGGTCCAGGCCCAGCGGCAGGCGTCGCGGCAGAGCCAGCGGGAGCTC GGAGGCCAGGTGGACCTGTTTAAGCGCCGCGTGGTGCGGAGACTGGCATCCCTCAAGACACGGCGCT GCCGGCTGAGCAGGGCAGCGCAGGGCCTCCCAGATCCGGGTGCTGAGACCTGTGCGGTGTGCCTGGA CTACTTCTGCAACAAACAGTGGCTCCGGGTGCTGCCCTGTAAGCACGAGTTTCACCGAGACTGTGTG GACCCCTGGCTGATGCTCCAGCAGACCTGCCCACTGTGCAAATTCAACGTCCTGGGTGAGCACCGCT ACTCCGATGATTAGCTGCCCAGCTGGACTCTGCACATGGGGATGGACCCCTCCTGCCTGCACCCCG
The NOV2 protein (SEQ ID NO:4) encoded by SEQ ID NO:3 is 378 amino acid residues in length and is presented using the one-letter amino acid code in Table 2B. Psort analysis predicts the NOV2 protein of the invention to be localized at the plasma membrane with a certainty of 0.6400.
Table 2B. Encoded NOV2 protein sequence (SEQ ID NO:4)
MGPAARPALRSPPPPPPPPPSPLLLLLPLLPL LGLAGPGAAADGSEPAAGAGRGGARAVRVDVR LPRQDALVLEGVRIGSEADPAPLLGGRLLLMDIVDAEQEAPVEG IAVAYVGKEQAAQFHQENKG SGPQAYPKALVQQMRRALFLGASALLLLILNHNWRELDISQLLLRPVIVLHYSSNVTKLLDALD QRTQATAEITSGΞSLSANIEWKLTLWTTCGLSKDGYGGWQDLVCLGGSRAQEQKPLQQLWNAILL VAMLLCTGLWQAQRQASRQSQRELGGQVDLFKRRWRRLASLKTRRCRLSRAAQGLPDPGAETC AVCLDYFCNKQWLRVLPCKHEFHRDCVDPWLMLQQTCPLCKFNVLGEHRYSDD
A search against the Patp database, a proprietary database that contains sequences published in patents and patent publications, yielded several homologous proteins shown in Table 2C.
Table 2C. Patp results for NOV2
Smallest Sum
Reading High Prob
Sequences producing High-scoring Segment Pairs Frame Score P(N)
>patp.-AAB42695 Human ORFX ORF2459 polypeptide +1 1715 3.0e-176
>patp:AAM79288 Human protein SEQ ID NO 1950 +1 612 2.3e-59
>patp:AAM80272 Human protein SEQ ID NO 3918 +1 534 4.2e-51
>patp-.AAU28202 Novel human secretory protein +1 201 5.1e-13
>patp:ABB50251 Human transcription factor TRFX- 102 +1 148 5.5e-13
In a BLAST search of public sequence databases, it was found, for example, that the nucleic acid sequence of this invention has 287 of 489 bases (58%) identical to a gb:GENBANK-ID:SSI132828]acc:AJ132828.1 mRNA from Spermatozopsis similis (mRNA for p210 protein, partial). The full amino acid sequence of the protein of the invention was found to have 341 of 379 amino acid residues (89%>) identical to, and 355 of 379 amino acid residues (93%) similar to, the 379 amino acid residue ptnr:SPTREMBL-ACC:Q9DCWl protein from Mus musculus (0610009 J22RTK PROTEIN).
NOV2 also has homology to the proteins shown in the BLASTP data in Table 2D.
Figure imgf000020_0001
A multiple sequence alignment is given in Table 2E, with the NOV2 protein being shown on line 1 in Table 2E in a ClustalW analysis, and comparing the NOV2 protein with the related protein sequences shown in Table 2D. This BLASTP data is displayed graphically in the ClustalW in Table 2E. Table 2E. ClustalW Analysis of NOV2
1) > NOV2; SEQ ID NO:4
2) >gi|12832380|/ data source:SPTR, source key:Q9Y6U7, evidence:ISS~homolog to WUGSC:H_DJ130H16.6 PROTEIN(FRAGMENT)~putative [Mus musculus]; SEQ ID NO:40
3) >gi|5441942|/ supported by mouse EST AA538043 (NID:g2284036) [Homo sapiens]; SEQ ID NO:41
4) >gi|17485136|/ similar to data source:SPTR, source key:Q9Y6U7 evidence. SS-homolog to WUGSC:H_DJ130H16.6 PROTEIN (FRAGMENT)~putative [Homo sapiens]; SEQ ID NO:42
5) >gi| 17861674|/ GH20973p [Drosophila melanogaster]; SEQ ID NO:43
6) >gi|18485962|/ similar to goliath (H. sapiens) [Drosophila melanogaster]; SEQ ID NO:44
Figure imgf000021_0001
87 89 87 118
Figure imgf000021_0002
118
Figure imgf000021_0003
Figure imgf000022_0001
The presence of identifiable domains in the protein disclosed herein was determined by searches using algorithms such as PROSITE, Blocks, Pfam, ProDomam, Prints and then determining the Interpro number by crossing the domain match (or numbers) using the Interpro website (http:www.ebi.ac.uk/interpro/). Table 2F lists the domain description from DOMAIN analysis results against NON2.
Figure imgf000022_0002
Consistent with other known members of the Membrane Ring Finger-like family of proteins, NOV2 has, for example, a Ring Finger signature sequence and homology to other members of the Plasma Membrane Ring Finger-like Protein Family. NOV 2 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts. For example, NOV2 nucleic acids and polypeptides can be used to identify proteins that are members of the Plasma Membrane Ring Finger-like Protein Family. The NOV2 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOVl activity or function. Specifically, the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular metabolism and signal transduction. These molecules can be used to treat, e.g., anemia, ataxia-telangiectasia, autoimmume disease, immunodeficiencies, diabetes, autoimmune disease, renal artery stenosis, interstitial nephritis, glomerulonephritis, polycystic kidney disease, systemic lupus erythematosus, renal tubular acidosis, IgA nephropathy, hypercalceimia, Lesch-Nyhan syndrome, cancer, trauma, regeneration (in vitro and in vivo), viral/bacterial/parasitic infections, as well as other diseases, disorders and conditions.
In addition, various NOV2 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. For example, the NOV2 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the Plasma Membrane Ring Finger-like Protein Family .
The NOV2 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of immune and renal physiology. As such, the NOV2 nucleic acids and polypeptides, antibodies and related compounds according to the mvention may be used to treat muscle and nervous system disorders, e.g., anemia, ataxia-telangiectasia, autoimmume disease, immunodeficiencies, diabetes, autoimmune disease, renal artery stenosis, interstitial nephritis, glomerulonephritis, polycystic kidney disease, systemic lupus erythematosus, renal tubular acidosis, IgA nephropathy, hypercalceimia, Lesch-Nyhan syndrome, cancer, trauma, regeneration (in vitro and in vivo), viral/bacterial/parasitic infections, as well as other diseases, disorders and conditions.
The NOV2 nucleic acids and polypeptides are useful for detecting specific cell types. For example, expression analysis has demonstrated that a NOV2 nucleic acid is expressed in peripheral blood, and a pool of various mammalian tissues. Expression information was derived from the tissue sources of the sequences that were included in the derivation of the sequence of CuraGen Aec. No. CG93210-01. The sequence is predicted to be expressed in the following tissues because of the expression pattern of (GENBANK-ID: gb:GENBANK- ID:SSI132828|acc:AJ132828.1) a closely related Spermatozopsis similis mRNA for ρ210 protein, partial homolog in species Spermatozopsis similis :kidney.
Additional utilities for NOV2 nucleic acids and polypeptides according to the invention are disclosed herein.
NOV3 A NOV3 polypeptide has been identified as a Thrombospondin type 1 (tsp_l) domain containing protein (also referred to as CG93275-01). The disclosed novel NOV3 nucleic acid (SEQ ID NO:5) of 799 nucleotides is shown in Table 3 A. The novel NOV3 nucleic acid sequences maps to the chromosome 16. An ORF begins with an ATG initiation codon at nucleotides 51-53 and ends with a TGA codon at nucleotides 744-746. A putative untranslated region and/or downstream from the termination codon is underlined in Table 3 A, and the start and stop codons are in bold letters.
Table 3A. NOV3 Nucleotide Sequence (SEQ ID NO:5)
GAATATATTTAGTGTGTTGTTTTTTTTTTTAATGTGGCTACTGAAACCTAATGGGAATGCAAATAGA ACTTTTTTGTCTTCTCAAGTGTTCCAAGACCTGTGGACGAGGGGTGAGGAAGCGTGAACTCCTCTGC AAGGGCTCTGCCGCAGAAACCCTCCCCGAGAGCCAGTGTACCAGTCTCCCCAGACGTGAGCTGCAGG AGGGCTGTGTGCTTGGACGATGCCCCAAGAACAGCCGGCTACAGTGGGTCGCTTCTTCGTGGAGCGA GTGTTCTGCAACCTGTGGTTTGGGTGTGAGGAAGAGGGAGATGAAGTGCAGCGAGAAGGGCTTCCAG GGAAAGCTGATAACTTTCCCAGAGCGAAGATGCCGTAATATTAAGAAACCAAATCTGGACTTGGAAG AGACCTGCAACCGACGGGCTTGCCCAGCCCATCCAGTGTACAACATGGTAGCTGGATGGTATTCATT GCCGTGGCAGCAGTGCACAGTCACCTGTGGGGGAGGGGTCCAGACCCGGTCAGTCCACTGTGTTCAG CAAGGCCGGCCTTCCTCAAGTTGTCTGCTCCATCAGAAACCTCCGGTGCTACGAGCCTGTAATACAA ACTTCTGTCCAGCTCCTGAAAAGAGAGAGGATCCATCCTGCGTAGATTTCTTCAACTGGTGTCACCT AGTTCCTCAGCATGGTGTCTGCAACCACAAGTTTTACGGAAAACAATGCTGCAAGTCATGCACAAGG AAGATCTGATCTTGGTGTCCTCCCCAGCCTTAGGGCCAGGGGCTTACCTTTCAACCTCTAGA
The NOV3 protein (SEQ ID NO:6) encoded by SEQ TD NO:5 is 231 amino acid residues in length and is presented using the one-letter amino acid code in Table 3B. Psort analysis predicts the NOV3 protein of the invention to be localized in the cytoplasm with a certainty of 0.4500.
Table 3B. Encoded NOV3 protein sequence (SEQ ID NO: 6)
MG QIELFCLLKCSKTCGRGVRKRELLCKGSAAETLPESQCTSLPRPELQEGCVLGRCPKNSRLQ WVASSWSECSATCGLG KREMKCSEKGFQGKLITFPERRCRNIKKPNLDLEETCNRRACPAHPV Y MVAG YSLP QQCTVTCGGGVQTRSVHCVQQGRPSSSCLLHQKPPVLRACNTNFCPAPEKRED PSCVDFFN CHLVPQHGVCNHKFYGKQCCKSCTR I
A search against the Patp database, a proprietary database that contains sequences published in patents and patent publications, yielded several homologous proteins shown in Table 3C.
Table 3C. Patp results for NOV3
Smallest Sum
Reading High Prob
Sequences producing High-scoring Segment Pairs: Frame Score P(N)
>patp:AAE09696 Human gene 7 encoding protein HE8CY61 +1 1248 9.2e-127
>patp:AAE09699 Human gene 10 encoding protein HUVHR16 +1 1245 1.9e-126
>patp:AAU72893 Human metalloprotease partial sequence #5 +1 1204 4.2e-122
>patp:AAU72891 Human metalloprotease partial sequence #3 +1 693 3.5e-70
>patp:AAB21253 Human metalloproteinase KIAA0605 +1 327 5.5e-28 In a BLAST search of public sequence databases, it was found, for example, that the nucleic acid sequence of this invention has 392 of 396 bases (98%) identical to an EST AA057409 mRNA from human). The full amino acid sequence of the protein of the invention was found to have 74 of 216 amino acid residues (34%) identical to, and 107 of 216 amino acid residues (49%) similar to, the 237 amino acid residue ptnr:SPTREMBL-ACC:Q9HBS6 protein from Homo sapiens (HYPOTHETICAL 25.7 KDA PROTEIN).
NOV3 also has homology to the proteins shown in the BLASTP data in Table 3D.
Table 3D. BLAST results for NOV3
Gene Index/ Protein/ Organism Length Identity Positives Expect Identifier (aa) (%) (%) gi 118598706 I ref |XP_ hypothetical 1123 181/183 183/183 e-100 091253.11 (XM_091253 protein XP_091253 (98%) (99%) ) [Homo sapiens] gi|19171150]emb|CAC ADAMTS18 protein 1081 61/62 62/62 4e-27 83612. ll (AJ311903) [Homo sapiens] (98%) (99%) gi I 7662202 I ref |NP_0 KIAA0605 gene 951 79/216 99/216 9e-23 55509. ll (NM 014694) product (36%) (45%) [Homo sapiens] gi 118561227 I ref |XP_ hypothetical 1365 51/112 74/112 4e-21 094442. ll (XM 094442 protein XP_094442 (45%) (65%) [Homo sapiens] gi 117432918 I sp|Q9H3 HUMAN ADAMTS-10 223 74/223 104/223 5e-20 24 AT10 precursor (A (33% (46%) disintegrin and metalloproteinase with thrombospondin motifs 10) (ADAM- TS 10) (ADAM- TS10) (Fragment)
A multiple sequence alignment is given in Table 3E, with the NOV3 protein being shown on line 1 in Table 3E in a ClustalW analysis, and comparing the NOV3 protein with the related protein sequences shown in Table 3D. This BLASTP data is displayed graphically in the ClustalW in Table 3E. Table 3E. ClustalW Analysis of NO V3
1) >N0V3; SEQ ID N0:6
2) >gi| 18598706|/ hypothetical protein XP_091253 [Homo sapiens]; SEQ ID NO:45
3) >gijl917115θj/ ADAMTS18 protein [Homo sapiens]; SEQ ID NO:46
4) >gi|7662202|/ KIAA0605 gene product [Homo sapiens]; SEQ ID NO:47 5) >gij 18561227|/ hypothetical protein XP_094442 [Homo sapiens]; SEQ ID NO:48
6) >gi|17432918|/ AT10JHUMAN ADAMTS-10 precursor (A disintegrin and metalloproteinase with thrombospondin motifs 10) (ADAM-TS 10) (ADAM-TSIO) (Fragment); SEQ ID NO:49
Figure imgf000025_0001
Figure imgf000026_0001
gij
Figure imgf000026_0002
1390 1400 1410 1420 1430 1440
Figure imgf000026_0003
1450 1460 1470 1480 1490 1500
Figure imgf000026_0004
1510 1520 1530 1540 1550 1560
Figure imgf000026_0005
1570 1580 1590 1600 1610 1620
Figure imgf000026_0006
1630 1640 1650 1660 1670 1680
Figure imgf000026_0007
The NOV3 Clustal W alignment shown in Table 3E was modified to begin at amino residue 1321. The data in Table 3E includes all of the regions overlapping with the NOV3 protein sequences. The presence of identifiable domains in the protein disclosed herein was determined by searches using algorithms such as PROSITE, Blocks, Pfam, ProDomain, Prints and then determining the Interpro number by crossing the domain match (or numbers) using the Interpro website (http:www.ebi.ac.uk/interpro/). Table 3F lists the domain description from DOMAIN analysis results against NOV3.
Figure imgf000027_0001
Consistent with other known members of the Thrombospondin type 1 (tsp_l) family of proteins, NOV3 has, for example, tliree tsp_l domain signature sequences and homology to other members of the tsp_l Domain-containing Protein Family. NOV 3 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts. For example, NOV3 nucleic acids and polypeptides can be used to identify proteins that are members of the tsp_l Domain-containing Protein Family. The NOV3 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOV3 activity or function. Specifically, the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular metabolism, and signal transduction. These molecules can be used to treat, e.g., Von Hippel-Lindau (VHL) syndrome, diabetes, tuberous sclerosis, fertility, hypogonadism, as well as other diseases, disorders and conditions. hi addition, various NOV3 nucleic acids and polypeptides according to the invention are useful, ter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. For example, the NOV3 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the tsp_l Domain-containing Protein Family. Thrombospondin type 1 domain (TSP1, IPR000884) is a repeat found in the thrombospondin protein where it is repeated 3 times. Likewise, the tsp_l domain is repeated three times in the NOV3 polypeptide. Now a number of proteins involved in the complement pathway (properdin, C6, C7, C8A, C8B, C9) (Patthy,L., J. Mol. Biol. 202: 689-696 (1988)) as well as extracellular matrix protein like mindin, F-spondin (Okamoto, et al, Development 126: 3637-3648 (1999)), SCO-spondin and even the circumsporozoite surface protein 2 and TRAP proteins of Plasmodium (Wengelnik, et al, EMBO J. 18: 5195-5204 (1999); Rogers, et al, Mol Biochem. Parasitol. 53: 45-51 (1992)) contain one or more instance of this repeat. It has been involved in cell-cell interraction, inhibition of angiogenesis (Krutzsch, βt al, Circulation 100: 1423-1431 (1999)), apoptosis [Krutzsch, et al, Cancer Res. 57: 1735-1742 (1997)).
The NOV3 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of urogenital physiology. As such, the NOV3 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat reproductive and metabolic disorders, e.g., Von Hippel-Lindau (VHL) syndrome, diabetes, tuberous sclerosis, fertility, hypogonadism, as well as other diseases, disorders and conditions. The NOV3 nucleic acids and polypeptides are useful for detecting specific cell types.
For example, expression analysis has demonstrated that a NOV3 nucleic acid is expressed in eye and testis.
Additional utilities for NOV3 nucleic acids and polypeptides according to the invention are disclosed herein.
NOV4
A NOV4 polypeptide has been identified as a Protocadherin Alpha C2 Short Form-like protein (also referred to as CG93187-01). The disclosed novel NOV4 nucleic acid (SEQ ID NO: 7) of 600 nucleotides is shown in Table 4A. The novel NOV4 nucleic acid sequences maps to the chromosome 11. An ORF begins with an ATG initiation codon at nucleotides 41-43 and ends with a
TAG codon at nucleotides 2546-2548. A putative untranslated region and/or downstream from the termination codon is underlined in Table 4A, and the start and stop codons are in bold letters.
Table 4A. NOV4 Nucleotide Sequence (SEQ ID NO:7)
CACCATAAAAGCTCAGAAAATAGACTTTTCCTCTGCCTCTATGGAGGGGCAGGCCAGATCTGGGGAA GGGATGGGACAGCCTGGCATGAAGAGCCCCAGGCCCCACCTCCTGCTACCATTGCTGCTGCTGCTGC TGCTGCTGCTGTCTTCGCCTCGCCGTGCACGCGTGCGCCTCCCAGAGGACCAGCCGCCTGGGCCCGC GGCTGGCACGCTCCTAGCCCGCGACCCGCATCTGGGCGAGGCTGCACGCGTGTCCTATCGGCTGGCA TCTGGCGGGGACGGCCACTTCCGGCTGCACTCAAGCACTGGAGCGCTGTCCGTGGTGCGGCCGTTGG ACCGCGAACAACGAGCTGAGCACGTACTGACAGTGGTGGCCTCAGACCGAGCTCCCCGCCCGCGCTC GGCCACGCAGGTCCTGACCGTCAGTGTCGCTGACGTCAACGACGAGGCGCCTACTTTCCAGCAGCAG GAGTACAGCGTCCTCTTGCGTGAGAACAACCCTCCTGGCACATCTCTGCTCACCCTGCGAGCAACCG ACCCCGACGTGGGGGCCAACGGGCAAGTGACTTATGGAGGCGTCTCTAGCGAAAGCTTTTCTCTGGA TCCTGACACTGGTGTTCTCACGACTCTTCGGGCCCTGGATCGAGAGGAACAGGAGGAGATCAACCTG ACAGTGTATGCCCAGGACAGGGGCTCACCTCCTCAGTTAACGCATGTCACTGTTCGAGTGGCTGTGG AGGATGAGAATGACCATGCACCAACCTTTGGGAGTGCCCATCTCTCTCTGGAGGTGCCTGAGGGCCA GGACCCCCAGACCCTTACCATGCTTCGGGCCTCTGATCCAGATGTGGGAGCCAATGGGCAGTTGCAG TACCGCATCCTAGATGGGGACCCATCAGGAGCCTTTGTCCTAGACCTTGCTTCTGGAGAGTTTGGCA CCATGCGGCCACTAGACAGAGAAGTGGAGCCAGCTTTCCAGCTGAGGATAGAGGCCCGGGATGGAGG CCAGCCAGCTCTCAGTGCCACGCTGCTTTTGACAGTGACAGTGCTGGATGCCAATGACCATGCTCCA GCCTTTCCTGTGCCTGCCTACTCGGTGGAGGTGCCGGAGGATGTGCCTGCAGGGACCCTGCTGCTGC AGCTACAGGCTCATGACCCTGATGCTGGAGCTAATGGCCATGTGACCTACTACCTGGGCGCCGGTAC AGCAGGAGCCTTCCTGCTGGAGCCCAGCTCTGGAGAACTGGTGTTGCTTGAACCTCTAGACTTTGAA AGCCTGACACAGTACAATCTAACAGTGGCTGCAGCTGACCGTGGGCAGCCACCCCAAAGCTCAGTCG TGCCAGTCACTGTCACTGTACTAGATGTCAATGACAACCCACCTGTCTTTACCCGAGCATCCTACCG TGTGACAGTACCTGAGGACACACCTGTTGGAGCTGAGCTGCTGCATGTAGAGGCCTCTGACGCTGAC CCTGCCCTCATGGCCTCCTCAGGCGACCCATCAGGGCTCTTTGAGCTGGATGAGAGCTCAGGCACCT TGCGACTGGCCCATGCCCTGGACTGTGAGACCCAGGCTCGACATCAGCTTGTAGTACAGGCTGCTGA CCCTGCTGGTGCACACTTTGCTTTGGCACCAGTGACAATTGAGGTCCAGGATGTGAATGATCATGGC CCAGCCTTCCCACTGAACTTACTCAGCACCAGCGTGGCCGAGAATCAGCCTCCAGGCACTCTCGTGA CCACTCTGCATGCAATCGACGGGGATGCTGGGGCTTTTGGGAGGCTCCGTTACAGCCTGTTGGAGGC TGGGCCAGGACCTGAGGGCCGTGAGGCATTTGCACTGAACAGCTCAACAGGGGAGTTGCGTGCGCGA GTGCCCTTTGACTATGAGCACACAGAAAGCTTCCGGCTGCTGGTGGGTGCTGCTGATGCTGGGAATC TCTCAGCCTCTGTCACTGTGTCGGTGCTAGTGACTGGAGAGGATGAGTATGACCCTGTATTTCTGGC ACCAGCTTTCCACTTCCAAGTGCCCGAAGGTGCCCGGCGTGGCCACAGCTTGGGTCACGTGCAGGCC ACAGATGAGGATGGGGGTGCCGATGGCCTGGTTCTGTATTCCCTTGCCACCTCTTCCCCCTATTTTG GTATTAACCAGACTACAGGAGCCCTGTACCTGCGGGTGGACAGTCGGGCACCAGGCAGCGGAACAGC CACCTCTGGGGGTGGGGGCCGGACCCGGCGGGAAGCACCACGGGAGCTGGGGCTCCACCTGGACTCT TACCAGAGTCACTCCAAGTCCTGTCTCAGGCAGAATACTCAGATCTATTCCAAGCACCTTCCCTGGG ATCTCAGGCGCATACTGAGAACCAGTGGGACAGGGTTGAGAGAGAGAGCCAACCGAGAATCTCAAAT GAACCAAACTGAGAAAGATGCCCCTCAGTGGGGCTACAGACCGACACCCCACCATGGGGCAACAGAA AAACCAAGACCCCCTCCCCAAAGGAATCAAACCAATCGGGAAAAGGAAGGAGGCGTTGGCCGTGCCT AGGATAT
The NOV4 protein (SEQ LD NO:8) encoded by SEQ ID NO:7 is 835 amino acid residues in length and is presented using the one-letter amino acid code in Table 4B. Psort analysis predicts the NOV4 protein of the invention to be localized at the plasma membrane with a certainty of 0.7900.
Table 4B. Encoded NOV4 protein sequence (SEQ ID NO:8) EGQARSGEGMGQPGMKSPRPHLLLPLLLLLLLLLSSPRRARVRLPEDQPPGPAAGTLLARDPHL GEAARVSYRLASGGDGHFRLHSSTGALSWRPLDREQRAEHVLTWASDRAPRPRSATQVLTVSV AD NDEAPTFQQQEYSVLLRE NPPGTSLLTLRATDPDVGANGQVTYGGVSSESFSLDPDTGVLT TLRALDREEQEEINLTVYAQDRGSPPQLTHVTVRVAVEDENDHAPTFGSAHLSLEVPEGQDPQTL TMLRASDPDVGANGQLQYRILDGDPSGAFVLDLASGEFGTMRPLDREVEPAFQLRIEARDGGQPA LSATLLLTVTVLDA DHAPAFPVPAYSVEVPEDVPAGTLLLQLQAHDPDAGANGHVTYYLGAGTA GAFLLEPSSGELVLLEPLDFΞSLTQY LTVAAADRGQPPQSSWPVTVTVLDVNDNPPVFTRASY RVTVPEDTPVGAELLHVEASDADPALMASSGDPSGLFELDESSGTLRLAHALDCETQARHQLWQ AADPAGAHFALAPVTIEVQDV DHGPAFPLNLLSTSVAΞNQPPGTLVTTLHAIDGDAGAFGRLRY SLLEAGPGPEGREAFALNSSTGELRARVPFDYEHTESFRLLVGAADAGNLSASVTVSVLVTGEDE YDPVFLAPAFHFQVPΞGARRGHSLGHVQATDEDGGADGLVLYSLATSSPYFGINQTTGALYLRVD SRAPGSGTATSGGGGRTRREAPRELGLHLDSYQSHSKSCLRQNTQIYSKHLP DLRRILRTSGTG LRERANRESQMNQTEKDAPQWGYRPTPHHGATEKPRPPPQRNQTNREKEGGVGRA
A search against the Patp database, a proprietary database that contains sequences published in patents and patent publications, yielded several homologous protems shown in Table 4C. Table 4C. Patp results for NOV4
Smallest Sum
Reading High Prob
Sequences producing High-scoring Segment Pairs: Frame Score P(N)
>patp:AAU07054 Human Flamingo protein +1 968 1.8e-98
>patp:AAU07053 Human Flamingo polypeptide +1 968 2.0e-98
>patp:ABG21921 Novel human diagnostic protein #21912 +1 642 3.1e-64
>patp-.ABG21921 Novel human diagnostic protein #21912 +1 642 3.1e-64
In a BLAST search of public sequence databases, it was found, for example, that the nucleic acid sequence of this invention has 273 of 415 bases (65%) identical to a gb:GENBANK-ID:AF061573|acc:AF061573.2 mRNA from Homo sapiens (protocadherin (PCDH8) mRNA, complete eds). The full amino acid sequence of the protein of the invention was found to have 273 of 415 amino acid residues (65%) identical to, and 273 of 415 amino acid residues (65%) similar to, the 4076 amino acid residue gb:GENBANK- ID:AF061573jacc:AF061573.2 protein from Homo sapiens (protocadherin (PCDH8) mRNA, complete eds).
NOV4 also has homology to the proteins shown in the BLASTP data in Table 4D.
Figure imgf000030_0001
Figure imgf000031_0002
A multiple sequence alignment is given in Table 4E, with the NOV4 protein being shown on line 1 in Table 4E in a ClustalW analysis, and comparing the NOV4 protein with the related protein sequences shown in Table 4D. This BLASTP data is displayed graphically in the ClustalW in Table 4E.
Table 4E. ClustalW Analysis of NOV4 l) > NOV4; SEQ ID NO: 8
2) >gi|17461472|/ similar to protocadherin 16 (H. sapiens) [Homo sapiens]; SEQ ID NO:50 3) >gi|16933557|/ protocadherin 16 precursor; fibroblast cadherin FIBl; cadherin 19; fibroblast cadherin 1; dachsous homologue [Homo sapiens] ; SEQ ID NO: 51
4) >gi|6753408|/ cadherin EGF LAG seven-pass G-type receptor [Mus musculus] ; SEQ ID NO:52
5) >gi|13325064|/ cadherin EGF LAG seven-pass G-type receptor 2; EGF-like-domain, multiple 2; epidermal growth factor-like 2; multiple epidermal growth factor-like domains 3; cadherin, EGF LAG seven-pass G-type receptor 2, flamingo (Drosophila) homolog; ; SEQ ID NO: 53
6) >gi|10727655|gb|AAF58763.2|(AE003828) stan gene product [Drosophila melanogaster] ; SEQ ID NO:54
1210 1220 1230 1240 1250 1260
Figure imgf000031_0001
1270 1280 1290 1300 1310 1320
Figure imgf000032_0001
1330 1340 1350 1360 1370 1380
Figure imgf000032_0002
1450 1460 1470 1480 1490 1500
Figure imgf000032_0003
1570 1580 1590 1600 1610 1620
Figure imgf000032_0004
1630 1640 1650 1660 1670 1680
Figure imgf000032_0005
Figure imgf000033_0001
0
NOV4 152 211 gi 117461472 586 645 gij 16933557 1661 1720 giJ6753408| 1460 1518 gijl3325064 1370 1428 gi 110727655 1559
Figure imgf000033_0002
1617
1810 1820 1830 1840 1850 1860
Figure imgf000033_0003
1870 1880 1890 1900 1910 1920
Figure imgf000033_0004
1930 1940 1950 1960 1970 1980
Figure imgf000033_0005
1990 2000 2010 2020 2030 2040
Figure imgf000033_0006
2050 2060 2070 2080 2090 2100
Figure imgf000033_0007
2110 2120 2130 2140 2150 2160
Figure imgf000033_0008
2170 2180 2190 2200 2210 2220
N0V4 492 FE DESSBTLRLΘHAL 542 gi 1 17461472 I 936 TTjVDSYTgEIRVgRSP
Figure imgf000033_0009
987 gi|l6933557 2011 TψBSYTSJEIRVgRSP LGPRDRVjJftV Tg e RPARS ATG 11 VGLQGEA- - 206 gij 6753408 I 1877 LKVRVKDgCDVEDPCASSPCPPHRPCRDT DSΥSCICΪ S YFGKKCVDACLLNPCKHVAA 1936 gijl3325064 1783 ESlNVEQgCSLPDPCDSNPCPANβYCSNDWDS^SCaCJ YYGDNCTNVCDLNPCEHQSV 1842 gi 10727655 1955 IRENVEDSJCESRgQCP-DHCPNH^SCQSSWDlJSTCEcj JJYVGTDCAPIJCTVRPCASG-V 2012
2230 2240 2250 2260 2270 2280
Figure imgf000034_0001
2290 2300 2310 2320 2330 2340
N0V4 598 EAFALNSSHGEL&AR VPFDfEHTESFgLLVGAADAGNLSASfflTVSVLVTGEjJEp? 651 gi 117461472 1038 GjfFSIQPSHGAITVRSA EGLDFEVSPRL|LVLQAESGGAFAFT|LTLTLQDANDN- 1092 gijl6933557 2113 GTFSIQPSJJGAITVRSA EGLDfEVSPRLgLyLQAESGGAFAFTfl LTLQ ,DA DN 2167 gij 6753408 I 1996 TNGQCQCKENYYKPPAQDACLPCDCΪPHGSHSgACDMDTGQCACKP' JGRQCNRcfaSPF 2055 gijl3325064 1902 T|SGECHCKENHYHPPGSPTCLLCDCS;PTGSLSgVCDPEDGQCPCKP GRQCDRCDNPS" 1961 gij 10727655 2073 TpGQCYCKgNHYQPPNETACLSCDCySIGSFSGACNPLTGQCECREGEl'GRRCDSCSsKlP f 2132
2350 2360 2370 2380 2390 2400
Figure imgf000034_0002
2470 2480 2490 2500 2510 2520
NOV4 725 SGGG-GRTRREAPR 738
Figure imgf000034_0003
2530 2540 2550 2560 2570 2580
Figure imgf000034_0004
2650 2660 2670 2680 2690 2700
Figure imgf000035_0001
The NOV4 Clustal W alignment shown in Table 4E was modified to begin at amino residue 1201 and end at amino acid residue 2760. The data in Table IE includes all of the regions overlapping with the NOV4 protein sequences.
The presence of identifiable domains in the protein disclosed herein was determined by searches using algorithms such as PROSITE, Blocks, Pfam, ProDomain, Prints and then determining the Interpro number by crossing the domain match (or numbers) using the Interpro website (http:www.ebi.ac.uk/interpro/). Table 4F lists the domain description from DOMAIN analysis results against NOV4.
Figure imgf000035_0002
Consistent with other known members of the Protocadherin Alpha C2 Short Form Protein-like family of proteins, NOV4 has, for example, seven Cadherin domain signature sequences and homology to other members of the Protocadherin Alpha C2 Short Form
Protein-like Protein Family. NOV4 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts. For example, NOV4 nucleic acids and polypeptides can be used to identify proteins that are members of the Protocadherin Alpha C2 Short Form Protein-like Protein Family. The NOV4 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOV4 activity or function. Specifically, the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular metabolism, and signal transduction. These molecules can be used to treat, e.g., Von Hippel-Lindau (VHL) syndrome, Alzheimer's disease, stroke, tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, cerebral palsy, epilepsy, Lesch- Nyhan syndrome, multiple sclerosis, ataxia-telangiectasia, leukodystrophies, behavioral disorders, addiction, anxiety, pain, neurodegeneration, systemic lupus erythematosus, autoimmune disease, asthma, emphysema, scleroderma, allergy, ARDS, fertility, endometriosis, hypogonadism, hemophilia, hypercoagulation, idiopathic thrombocytopenic purpura, autoimmune disease, allergies, immunodeficiencies, transplantation, graft versus host disease (GVHD), lymphaedema, as well as other diseases, disorders and conditions. hi addition, various NOV4 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. For example, the NOV4 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the Protocadherin Alpha C2 Short Form Protein-like Protein Family. Cadherins (Takeichi, Annu. Rev. Biochem. 59: 237-252 (1990); Takeichi Trends
Genet. 3: 213-217 (1987)), first discovered in mouse teratocarcinoma cells (Liaw, EMBOJ. 9: 2701-2708 (1990)), are a family of animal glycoproteins responsible for calcium-dependent cell-cell adhesion. Cadherins preferentially interact with themselves in a homophilic manner in connecting cells; thus acting as both receptor and ligand. There are a number of different isoforms distributed in a tissue-specific manner in a wide variety of organisms. Cells containing different cadherins tend to segregate in vitro, while those that contain the same cadherins tend to preferentially aggregate together. This observation is linked to the finding that cadherin expression causes morphological changes involving the positional segregation of cells into layers, suggesting they may play an important role in the sorting of different cell types during morphogenesis, histogenesis and regeneration. They may also be involved in the regulation of tight and gap junctions, and in the control of intercellular spacing. Cadherins are evolutionary related to the desmogleins which are component of intercellular desmosome junctions involved in the interaction of plaque proteins.
Structurally, cadherins comprise a number of domains: these include a signal sequence; a propeptide of around 130 residues; an extracellular domain of around 600 residues; a single transmembrane domain; and a well-conserved C-terminal cytoplasmic domain of about 150 residues. The extracellular domain can be subdivided into 5 parts, 4 of which are repeats of about 110 residues, and the fifth contains 4 conserved cysteines. The calcium-binding region of cadherins is thought to be located in the extracellular domain. This indicates that the sequence of the invention has properties similar to those of other proteins known to contain this/these domain(s) and similar to the properties of these domains.
Maniatis et al. has identified 52 novel human cadherin-like genes organized into three closely linked clusters (Wu and Maniatis, Cell 97(6):779-90 (1999).) Comparison of the genomic DNA sequences with those of representative cDNAs reveals a striking genomic organization similar to that of immunoglobulin and T cell receptor gene clusters. The N- terminal extracellular and transmembrane domains of each cadherin protein are encoded by a distinct and unusually large exon. These exons are organized in a tandem array. By contrast, the C-terminal cytoplasmic domain of each protein is identical and is encoded by three small exons located downstream from the cluster of N-terminal exons. This unusual organization has interesting implications regarding the molecular code required to establish complex networks of neuronal connections in the brain and the mechanisms of cell-specific cadherin- like gene expression.
The NOV4 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of urogenital, nerve, and endocrine physiology. As such, the NOV4 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat reproductive and nervous system disorders, e.g., Von Hippel-Lindau (VHL) syndrome, Alzheimer's disease, stroke, tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, cerebral palsy, epilepsy, Lesch-Nyhan syndrome, multiple sclerosis, ataxia-telangiectasia, leukodystrophies, behavioral disorders, addiction, anxiety, pain, neurodegeneration, systemic lupus erythematosus, autoimmune disease, asthma, emphysema, scleroderma, allergy, ARDS, fertility, endometriosis, hypogonadism, hemophilia, hypercoagulation, idiopathic thrombocytopenic purpura, autoimmune disease, allergies, immunodeficiencies, transplantation, graft versus host disease (GVHD), lymphaedema, as well as other diseases, disorders and conditions.
The NOV4 nucleic acids and polypeptides are useful for detecting specific cell types. For example, expression analysis has demonstrated that a NOV4 nucleic acid is expressed in Heart, Aorta, Umbilical Vein, Thyroid, Colon, Peripheral Blood, Spleen, Lymph node, Bone, Cartilage, Brain, Left cerebellum, Right Cerebellum, Parietal Lobe, Temporal Lobe, Cerebral Medulla/Cerebral white matter, Hippocampus, Cervix, Mammary glandBreast, Ovary, Placenta, Uterus, Testis, Lung, and Retina.
Additional utilities for NOV4 nucleic acids and polypeptides according to the invention are disclosed herein. NOV5
A NOV5 polypeptide has been identified as a Nuclear protein-like protein (also referred to as CG95083-01). The disclosed novel NOV5 nucleic acid (SEQ ID NO:9) of 2322 nucleotides is shown in Table 5 A.
An ORF begins with an ATG initiation codon at nucleotides 70-72 and ends with a TAA codon at nucleotides 2320-2322. A putative untranslated region and/or downstream from the termination codon is underlined in Table 5 A, and the start and stop codons are in bold letters.
Table 5A. NOV5 Nucleotide Sequence (SEQ ID NO:9)
GTGGAAGGACAGTCCAGAGCCCTTGTCATCGCACAGGAACTGCTATCTTCAGAGAAAGCATACGTGG AGATGCTCCAGCACTTAAATCTGTTCCTGGCAGAGCAGGCTATCAGCAGGAGAGGCCAGGGCTCCAA AGCCCCAGGGGAAATCTGCCAAGGAGGACTTGTGCTCAGTCCTATCAACCTGTGGGTAACAGACCTT TTGGTGTTTCAGGATTTCCATGGAGCTGTCATGAGGGCCTTGGATGACATGGACCATGAAGGCAGAG ACACATTGGCCCGGGAGGAGCTGAGGCAGGGCCTGAGTGAACTCCCAGCCATCCACGACCTTCATCA AGGCATCCTGGAGGAGCTGGAGGAAAGGCTGTCAAATTGGGAGAGCCAGCAGAAGGTAGCTGACGTC TTCCTTGCCCGGGAGCAGGGGTTTGATCACCACGCCACTCACATCCTGCAGTTCGACAGGTACCTAG GTCTGCTCAGTGAGAATTGCCTCCACTCTCCCCGGCTGGCAGCTGCTGTCCGTGAATTTGAGCAGAG TGTACAAGGAGGCAGCCAGACTGCGAAGCATCGGCTGCTGCGGGTGGTTCAACGCCTCTTCCAGTAC CAAGTGCTCCTCACAGACTATTTAAACAACCTTTGTCCGGACTCCGCCGAGTACGACAACACACAGG GTGCACTGAGCCTCATCTCCAAAGTCACAGACCGTGCCAACGACAGCATGGAGCAAGGGGAAAACCT GCAGAAGCTGGTCCACATTGAGCACAGCGTCCGGGGCCAAGGGGATCTCCTCCAGCCAGGAAGGGAG TTTCTGAAGGAAGGGACGCTGATGAAAGTAACAGGGAAAAACAGACGGCCCCGGCACCTATTTCTGA TGAACGATGTGCTCCTGTACACCTATCCCCAGAAGGATGGGAAGTACCGGCTGAAGAACACATTGGC TGTGGCCAACATGAAGGCTCTTTACCATGGGGAAGGGGAAGGAGGAAGCACCTTTCTCAGCATGGAG GTTTGTTCCCTTTTGGAACCAAAGGCTCCACCGAGGAGCCTGTTAGAAAAAGGCATGGGAGACGTGG TCACTGGCAGGTACTTGTCCAACATGACAGTGCACCTGGGGTTGCCCGGGCTGGGCCCTGAGCATGA CGCTCTGCAGCCTTCCCAGCGGTGGGTCAGCCGCCCTGTGATGGAGAAAGTGCCCTACGCTCTAAAG ATTGAGACTTCCGAGTCCTGCCTGATGCTGTCTGCGAGGCTGCAGGTCAGGAAGTCCAAGGTCAAGG CACTGACTGATTCGGTGTCTGCAGCCCTGGGAGTTAGGGGAATATCATTATTCCAGTGTAAGAAGAA ACAGACCCAAGGACAGCTAATGGACCAGTGGTCTGCTCGTAAACCTAGTCTGGCAGGTGATCTCTTC TTTGCTGGTGGTTCTGGGCAGTGTGAGAGGTGCAGGCTCAAGGGGCATCTGAGTGAGAACCTCATCC ATGCCGAGATGGAGGCCCATGCCCGCAGCTCCTGTGCAGAGAGGGACGAGTGGTATGGCTGTCTGAG CAGAGCCCTCCCTGAGGACTACAAGGCCCAGGCGCTGGCTGCATTCCACCATAGCGTGGAGATACGA GAGAGGCTGGGGGTTAGCCTTGGGGAGAGGCCCCCCACCCTGGTGCCTGTCACACACGTCATGATGT GCATGAACTGCGGCTGCGACTTCTCCCTCACCCTGCGGCGTCATCACTGTCACGCCTGTGGCAAGCA GATCGTGTGCCGGAACTGTTCGCGGAACAAGTACCCGCTGAAGTACCTGAAGGACAGGATGGCCAAG GTCTGCGACGGCTGCTTCGGGGAGCTGAAGAAGCGGGGCAGGGCTGTCCCGGGCCTGATGAGAGTTA CAGAGCGGCCTGTGAGCATGAGCTTCCCGCTGTCTTCACCCCGCTTCTCGGGCAGTGCCTTTTCATC CGTCTTCCAGAGCATTAACCCCTCGACCTTCAAGAAGCAGAAGAAAGTCCCTTCAGCCCTGACAGAG GTAGCTGCCTCTGGAGAGGGCTCTGCCATCAGTGGCTATCTCAGCCGGTGTAAGAGGGGCAAGCGGC ACTGGAAGAAGCTCTGGTTTGTCATCAAAGGCAAAGTTCTCTACACCTACATGGCCAGTGAGGACAA AGTGGCCTTGGAGAGTATGCCTCTGCTAGGCTTCACCATTGCTCCAGAAAAGGAAGAGGGCAGCAGT GAAGTAGGACCTATTTTTCACCTTTACCACAAGAAAACCCTATTTTATAGCTTCAAAGCAGAAGATA CCAATTCATGGATCGAGGCCATGGAAGATGCGAGTGTGTTATAG
Variant sequences of NOV5 are included in Example 3, Table 19. A variant sequence can include a single nucleotide polymorphism (SNP). A SNP can, in some instances, be referred to as a "cSNP" to denote that the nucleotide sequence containing the SNP originates as a cDNA. The NOV5 protein (SEQ ID NO: 10) encoded by SEQ ID NO:9 is 750 amino acid residues in length and is presented using the one-letter amino acid code in Table 5B. Psort analysis predicts the NOV5 protein of the invention to be localized in the nucleus with a certainty of 0.3000.
Table 5B. Encoded NOV5 protein sequence (SEQ ID NO:10)
MLQHLNLFLAEQAISRRGQGSKAPGEICQGGLVLSPINL VTDLLVFQDFHGAVMRALDDMDHΞG RDTLAREELRQGLSELPAIHDLHQGILEELEΞRLSNESQQKVADVFLAREQGFDHHATHILQFD RYLGLLSΞNCLHSPRLAAAVREFEQSVQGGSQTAKHRLLRWQRLFQYQVLLTDYLNLCPDSAE YDNTQGALSLISKVTDRANBSMEQGENLQ LVHIEHSVRGQGDLLQPGREFLKEGTL KVTGKMR RPRHLFLMNDVLLYTYPQKDGKYRLKNTLAVAMMKALYHGEGEGGSTFLSMEVCSLLEPKAPPRS LLEKGMGDWTGRYLSNMTVHLGLPGLGPEHDALQPSQRWVSRPVMEKVPYALKIETSESCLMLS ARLQVRKSKVALTDSVSAALGVRGISLFQCKKKQTQGQL DQWSARKPSLAGDLFFAGGSGQCE RCRLKGHLSENLIHAEMEAHARSSCAΞRDE YGCLSRALPEDYKAQALAAFHHSVΞIRERLGVSL GERPPTLVPVTHVMMCMNCGCDFSLTLRRHHCHACGKQIVCRNCSRNKYPLKYL DRMAKVCDGC FGELKKRGRAVPGLMRVTERPVSMSFPLSSPRFSGSAFSSVFQSINPSTFKKQKKVPSALTEVAA SGEGSAISGYLSRCKRGKRH KKL FVIKGKVLYTYMASEDKVALESMPLLGFTIAPEKEEGSSE VGPIFHLYHKKTLFYSFKAEDTNSWIEAMEDASVL
A search against the Patp database, a proprietary database that contains sequences published in patents and patent publications, yielded several homologous proteins shown in Table 5C.
Table 5C. Patp results for NOV5
Smallest Sum
Reading High Prob Sequences producing High-scoring Segment Pairs: Frame Score P(N)
>patp:AAB93568 Human protein sequence SEQ ID NO: 12972 +1 577 1.7e-95
>patp:AAY51248 Rat actin-binding protein frabin +1 312 1.9e-41
>patp:AAU21630 Novel human neoplastic disease polypeptide +1 256 1.6e-38
>patp:AAU27818 Human full-length polypeptide #143 +1 300 2.6e-29
>patp:ABG00573 Novel human diagnostic protein #564 +1 261 1.8e-26
In a BLAST search of public sequence databases, it was found, for example, that the nucleic acid sequence of this invention has 443 of 754 bases (58%) identical to a gb:GENBANK-ID:AB037783|acc:AB037783.1 mRNA from Homo sapiens (mRNA for
KIAA1362 protein, partial eds). The full amino acid sequence of the protein of the invention was found to have 114 of 263 amino acid residues (43%) identical to, and 173 of 263 amino acid residues (65%) similar to, the 699 amino acid residue ρtnr:SPTREMBL-ACC:Q9P2I5 protein from Homo sapiens (KIAA1362 PROTEIN). NOV5 also has homology to the proteins shown in the BLASTP data in Table 5D. Table 5D. BLAST results for NOV5
Gene Index/ Protein/ Organism Length Identity Positives Expect Identifier (aa) (%) (%) gi I 89229211 ref |NP_0 hypothetical 432 135/284 169/284 5e-57 60821. ll (NM 018351) protein FLJ11183 (47%) (58%) [Homo sapiens] gi I 16716345 I ref |NP_ ethanol decreased 431 131/284 171/284 2e-55 444302. ll (NM 053072 4 [Λfus musculus] (46%) (60%) gi|7243105|dbj |BAA9 KIAA1362 protein 699 111/251 166/251 2e-54 2600. ll (AB037783) [Homo sapiens] (44%) (65%) gi 113648298 ref XP hypothetical 204 115/222 141/222 le-49
012133.2) protein FLJ11183 (51%) (62%) ,
(XM_012133) [Homo sapiens] gi| 15426438 |gb|AAHl Similar to 376 103/221 129/221 4e-40 3319.11 AH13319 hypothetical (46%) (57%) (BC013319) protein FLJ11183 [Homo sapiens]
A multiple sequence alignment is given in Table 5E, with the NOV5 protein being shown on line 1 in Table 5E in a ClustalW analysis, and comparing the NOV5 protein with the related protein sequences shown in Table 5D. This BLASTP data is displayed graphically in the ClustalW in Table 5E.
Table 5E. ClustalW Analysis of NOV5
1) > NOV5; SEQ ID NO:10
2) >gi|8922921|/ hypothetical protein FLJ11183 [Homo sapiens]; SEQ ID NO:55
3) >gi| 167163451/ ethanol decreased 4 [Mus musculus]; SEQ ID NO:56
4) >giJ7243105|/ KIAA1362 protein [Homo sapiens]; SEQ ID NO:57
5) >gi|13648298|/ hypothetical protein FLJ11183 [Homo sapiens]; SEQ ID NO:58
6) >gijl 5426438|/ Similar to hypothetical protein FLJ11183 [Homo sapiens]; SEQ ID NO:59
10 20 30 40 50 60
0
1
20
6
80
Figure imgf000040_0001
190 200 210 220 230 240
N0V5 36 INL VTDLLVFQDFHGAVMRAL DDMDHE 6
40
24
00
Figure imgf000041_0001
Figure imgf000041_0002
gi 15426438] 167 iTiaaB 174
Figure imgf000042_0001
790 800 810 820 830 840
N0V5 601 RVTERPySMSFPLSE 660
8922921| 287 HQHSPRIGSPGNHKS .- - -ιa-ewBft.tflBSM3aa- ιa«waa«gjyaBaa«BtaiiιiMa-BBiaMtt 341 gi 16716345| 286 ιπaτ,EraagS33asiπgai 340 gi 7243105 I 617 HQHSPRIGSPGNHKSP jKEVSANTEDSSMSG 671 gi 13648298 I 59 HQHSPRIGSPGNHKSP SRKQKKIPAALKEVSANTEDSSMSG. 113 gi 15426438 287 HQHSPRIGSPGNHKSP 341
Figure imgf000042_0002
The presence of identifiable domains in the protein disclosed herein was determined by searches using algorithms such as PROSITE, Blocks, Pfam, ProDomain, Prints and then determining the Interpro number by crossing the domain match (or numbers) using the Interpro website (http:www.ebi.ac.uk/interpro/). Table 5F lists the domain description from DOMAIN analysis results against NOV5.
Figure imgf000042_0003
Consistent with other known members of the Nuclear Protein-like family of proteins, NOV5 has, for example, an RhoGEF signature sequence and a FYVE Zinc Finger signature sequence, aw well as homology to other members of the Nuclear Protein-like Protein Family. NOV5 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts. For example, NOV5 nucleic acids and polypeptides can be used to identify proteins that are members of the Nuclear Protein-like Protein Family. The NOV5 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOV5 activity or function. Specifically, the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular replication, and signal transduction. These molecules can be used to treat, e.g., Cardiovascular diseases, Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus , Pulmonary stenosis, Subaortic stenosis, Ventricular septal defect (VSD), valve diseases, Tuberous sclerosis, Scleroderma, Obesity, Transplantation, DiabetesNon Hippel-Lindau (VHL) syndrome , Pancreatitis, Obesity, Von Hippel-Lindau (VHL) syndrome , Alzheimer's disease, Stroke, Tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, Cerebral palsy, Epilepsy, Lesch-Nyhan syndrome, Multiple sclerosis, Ataxia-telangiectasia, Leukodystrophies, Behavioral disorders, Addiction, Anxiety, Pain, Neuroprotection as well as other diseases, disorders and conditions. hi addition, various NOV5 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. For example, the NOV5 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the Nuclear Protein-like Protein Family.
The NOV5 nucleic acids and polypeptides, antibodies and related compounds according to the mvention will be useful in therapeutic and diagnostic applications in the mediation of cardiac and nerve physiology. As such, the NOV5 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat cardiovascular and nervous system disorders, e.g., Cardiovascular diseases,
Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus , Pulmonary stenosis, Subaortic stenosis, Ventricular septal defect (VSD), valve diseases, Tuberous sclerosis, Scleroderma, Obesity, Transplantation, DiabetesNon Hippel-Lindau (VHL) syndrome , Pancreatitis, Obesity, Von Hippel-Lindau (VHL) syndrome , Alzheimer's disease, Stroke, Tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, Cerebral palsy, Epilepsy, Lesch-Nyhan syndrome, Multiple sclerosis, Ataxia-telangiectasia, Leukodystrophies, Behavioral disorders, Addiction, Anxiety, Pain, Neuroprotection as well as other diseases, disorders and conditions.
The NOV5 nucleic acids and polypeptides are useful for detecting specific cell types. For example, expression analysis has demonstrated that a NOV5 nucleic acid is expressed in Brown adipose, Vein, Umbilical Vein, Adrenal Gland/Suprarenal gland, Gall Bladder, Small Intestine, Colon, Lymphoid tissue, Spleen, Lymph node, Thymus, Brain, Temporal Lobe, Basal Ganglia Cerebral nuclei, Substantia Nigra, Spinal Chord, Cervix, Ovary, Uterus, Testis, Lung, Lung Pleura, Larynx, Urinary Bladder, Kidney.
Additional utilities for NOV5 nucleic acids and polypeptides according to the invention are disclosed herein.
NOV6
A NOV6 polypeptide has been identified as a Secretory Protein-like protein (also referred to as CG94989-01). The disclosed novel NOV6 nucleic acid (SEQ ID NO:ll) of 2372 nucleotides is shown in Table 6A. The novel NOV6 nucleic acid sequences maps to the chromosome 17. An ORF begins with an ATG initiation codon at nucleotides 99-101 and ends with a
TAA codon at nucleotides 1710-1712. A putative untranslated region and/or downstream from the termination codon is underlined in Table 6A, and the start and stop codons are in bold letters.
Table 6A. NOV6 Nucleotide Sequence (SEQ ID NO:ll)
CCGGCAAGGATGACGCCTCCGGAGGCCCTGGCCTCACTCCCACCTGGGCGCTAGGAGCCATCCCGGG GCTCCAGCCAGGAGCCCTGCTGCCCAGGGGCATGGCCAAACCTTTCTTCCGACTCCAGAAGTTTCTC CGCCGAACACAGTTCCTGCTGTTCTTCCTCACGGCTGCCTACCTGATGACCGGCAGCCTGCTGCTGC TGCAGCGGGTCCGCGTGGCTCTCCCACAGGGCCCCCGGGCACCCGGCCCCCTGCAGACCTTGCCAGT GGCCGCCGTGGCGCTGGGCGTGGGCTTGCTGGACAGCAGAGCCCTGCACGACCCTCGAGTCAGCCCA GAGCTGCTGCTGGGTGTGGACATGCTGCAGAGCCCCCTGACCCGGCCCCGGCCCGGCCCCCGCTGGC TCCGGAGCCGCAACTCGGAGCTGCGTCAGTTGCGTCGCCGCTGGTTCCACCACTTCATGAGTNGACT CCCAGGGACCGCCCGCCCTGGGCCCCGAGGCTGCCAGGCCCGCCATCCACAGCCGAGGTCCTATGTC TACGCCGGCTTGGAGGCCGGGGCGGAGTGTTACTGCGGGAACCGGCTGCCAGCGGTGAGCGTGGGGC TGGAAGAGTGTAACCATGAGTGCAAAGGCGAGAAGGGCTCTGTGTGCGGGGCTGTGGACCGGCTCTC CGTGTACCGTGTGGACGAGCTGCAGCCGGGCTCCAGGAAGCGGCGGACCGCCACCTACCGCGGATGC TTCCGACTGCCAGAGAACATCACACATGCCTTCCCCAGCTCCCTGATACAGGCCAATGTGACCGTGG GGACTTGCTCGGGCTTTTGTTCCCAGAAAGAGTTCCCCTTGGCCATTCTCAGGGGCTGGGAATGCTA CTGTGCTTACCCTACCCCCCGGTTCAACCTGCGGGATGCCATGGACAGCTCAGTATGTGGCCAGGAC CCTGAGGCACAGAGGCTGGCAGAATACTGTGAGGTCTACCAGACACCTGTGCAAGACACTCGTTGTA CAGACAGGAGGTTCCTGCCTAACAAATCCAAAGTGTTTGTGGCTTTGTCAAGCTTCCCAGGAGCCGG GAACACGTGGGCACGGCACCTCATTGAGCATGCCACTGGCTTCTATACAGGGAGCTACTACTTTGAT GGAACCCTCTACAACAAAGGGTTCAAGGGCGAAAAGGACCACTGGCGGAGCCGACGCACCATCTGTG TCAAAACCCACGAGAGTGGCAGGAGGGAGATTGAGATGTTTGATTCAGCCATCCTGCTAATCCGGAA CCCATACAGGTCCCTGGTGGCAGAATTCAACAGAAAATGTGCCGGGCACCTGGGATATGCAGCTGAC CGCAACTGGAAGAGCAAAGAGTGGCCGGACTTTGTCAACAGCTACGCCTCGTGGTGGTCCTCGCACG TCCTGGACTGGCTCAAGTACGGGAAGCGGCTGCTGGTGGTGCACTACGAGGAGCTGCGGCGCAGCCT GGTGCCCACGTTACGGGAGATGGTGGCCTTCCTCAACGTGTCTGTGAGCGAGGAGCGGCTGCTCTGC GTGGAGAACAACAAGGAGGGCAGCTTCCGGCGGCGCGGCCGGCGCTCCCACGACCCTGAGCCCTTCA CCCCGGAGATGAAAGACTTGATCAATGGCTACATCCGGACGGTGGACCAAGCCCTGCGTGACCACAA CTGGACGGGGCTGCCCAGGGAGTATGTGCCCAGATGATAGGCCTGGCCCACGCCGCCGCCCCCGCTG AGTGACGCAATCGCACCACGGGGCTGCGCTCCCCACTCTGATGCTCAGGCCCGTGGCCTCACTGGGA CGAACGGTGGGTGGGGGGCTCACCCTGGTGCTGCCTCCCGCACAAGGAGACCTGGACACAACAGACA CACATCACAAGGCGAACACAAATGGACACACATACCTGGCCACGAACCCACACCTCCTCAGACACTC AGACACCACTCCAGGCTCATAGCCCCGTCTTGATGCAGAGAAGCCACCCACGTGGGGTGTGCCAGGC ACCCCCAGCTACAAATGCAGCCACGCACAGACGTAACACACAGGTGCCAGGCCGTGTGCTCCTGGAG GCTGGCTGGCTGTCTCTCTCACACAGATACACGTGCGCTCCCTGGGATCCGGGAGGCCCTGGGCTTC CTGTGTGTAGCCCTGGCATAGACTTGCTCGTCAGGGTGTTTGACTCTGGGATGCTGGGCCGGGCAGA CATTTATGCTCTGAGCAGCAAGGACCATTGGGATGGAGGTGGGCACAAAGACTGCTGCTTCCAGGGT GTGCGGCCCTGGCCGTGTGTCTGACATCCCATAAATGTGTGTGTGGTGTGACTACGGGCACCACAAA CTCCGCAAAAAAAAAAAAAAAAAAAAA
The NOV6 protein (SEQ ID NO:12) encodedby SEQ ID NO:11 is 537 amino acid residues in length and is presented using the one-letter amino acid code in Table 6B. Psort analysis predicts the NOV6 protein ofthe invention to be localized outside the cell with a certainty of0.6997.
Table 6B. Encoded NOV6 protein sequence (SEQ ID NO:12)
MAKPFFRLQKFLRRTQFLLFFLTAAYLMTGSLLLLQRVRVALPQGPRAPGPLQTLPVAAVALGVG LLDSRALHDPRVSPELLLGVDMLQSPLTRPRPGPR LRSRNSELRQLRRR FHHFMSXLPGTARP GPRGCQARHPQPRSYVYAGLEAGAECYCGNRLPAVSVGLEECNHECKGEKGSVCGAVDRLSVYRV DELQPGSRKRRTATYRGCFRLPΞNITHAFPSSLIQANVTVGTCSGFCSQKEFPLAILRGWECYCA YPTPRFNLRDAMDSSVCGQDPEAQRLAEYCEVYQTPVQDTRCTDRRFLPNKSKVFVALSSFPGAG NT ARHLIEHATGFYTGSYYFDGTLY KGFKGEKDHWRSRRTICVKTHESGRREIEMFDSAILLI PJΓPYRSLVAEFNRKCAGHLGYAADRIWKSKΈ PDFVNSYASW SSHVLD LKYGKRLLVVHYEEL RRSLVPTLREMVAFLNVSVSEERLLCVENNKEGSFRRRGRRSHDPEPFTPEMKDLINGYIRTVDQ
ALRDHN TGLPREYVPR
A search against the Patp database, a proprietary database that contains sequences published in patents and patent publications, yielded several homologous proteins shown in Table 6C.
Table 6C. Patp results for NOV6
Smallest
Sum
Reading High Prob
Sequences producing High- scoring Segment Pairs : Frame Score P(N)
>patp .-ABB15485 Human nervous system related polypeptide +1 92 0 . 0036
>patp:AAU50001 Propionibacteπum acnes im unogenic protein +1 82 0 . 042
>patp :AAU50001 Propionibacterium acnes immunogenic protein +1 82 0.042
>patp:AAU18674 Renal and cardiovascular-associated protein +1 79 0.085
>patp:AAB95341 Human protein sequence SEQ ID NO:17621 +1 99 0.17 In a BLAST search of public sequence databases, it was found, for example, that the nucleic acid sequence of this invention has 2188 of 2189 bases (99%) identical to a gb:GENBANK-rD:AK000243]acc:AK000243.1 mRNA from Homo sapiens (cDNA FLJ20236 fis, clone COLF5810, highly similar to ABOl 1095 Homo sapiens mRNA for KIAA0523 protein). The full amino acid sequence of the protein of the invention was found to have 395 of 395 amino acid residues (100%) identical to, and 395 of 395 amino acid residues (100%) similar to, the 468 amino acid residue ptnr:SPTREMBL-ACC:O60276 protein from Homo sapiens (KIAA0523 PROTEIN)(Fig. 3B).
NOV6 also has homology to the proteins shown in the BLASTP data in Table 6D.
Figure imgf000046_0001
A multiple sequence alignment is given in Table 6E, with the NOV6 protein being shown on line 1 in Table 6E in a ClustalW analysis, and comparing the NOV6 protein with the related protein sequences shown in Table 6D. This BLASTP data is displayed graphically in the ClustalW in Table 6E.
Table 6E. ClustalW Analysis of NOV6
1) >N0V6; SEQ ID N0:12
2) >gi| 14602977|/ Similar to KIAA0789 gene product [Homo sapiens]; SEQ ID NO:60
3) >gi|3043570|/ KIAA0523 protein [Homo sapiens]; SEQ ID NO:61
4) >gi| 18489296|/ CG9164 [Drosophila melanogaster]; SEQ ID NO:62
5) >gij 16944644]/ hypothetical protein [Neurospora crassa]; SEQ ID NO:63
6) >gi|l 1359357|/ beta-1,3 exoglucanase (EC 3.2.1.-) precursor - fungus [Trichoderma harzianum]; SEQ ID NO:64
850 860 870 880 890 900 2 2
00
Figure imgf000047_0001
74
910 920 930 940 950 960
N0V6 33 LLLQRVRVAfflPQGPRAPGPLQTLPVAAVALGfflGIiLDSR-ALHDPRVSPEfflLLGVDMLQSP 91 gi 1 14602977 I 33 LLLQRVRVAaPQGPRAPGPLQTfjPVAAVALGgGgLDSR-ALHDPRVSPESLLGVDMLQSP 91 gi J 3043570 | X 1 gi j l8489296 | X x gi j l6944644 J 901 LFFTPGAAP^ADPIVYSGAWTpWNDLTFGfflHVVNFIGTLRATKVGPTfflPDGSARFKYL 960 gi j 11359357 j 275 RAISINNCG3GIDMTAAESITLDSSISGTP8GJKTSFRRNQSPATSNS3IVENLSLNNV 334
Figure imgf000047_0002
1030 1040 1050 1060 1070 1080
Figure imgf000047_0003
1090 1100 1110 1120 1130 1140
Figure imgf000047_0004
1150 1160 1170 1180 1190 1200
Figure imgf000047_0005
1210 1220 1230 1240 1250 1260
Figure imgf000047_0006
1270 1280 1290 1300 1310 1320
Figure imgf000047_0007
0
Figure imgf000048_0001
1390 1400 1410 1420 1430 1440
Figure imgf000048_0002
1510 1520 1530 1540 1550 1560
0
Figure imgf000048_0003
1570 1580 1590 1600 1610 1620
Figure imgf000048_0004
1690 1700 1710 1720 1730 1740
N0V6 494 454 gi| 14602977 ] 532 532 gi|3043570| 425 425 gi|l8489296| 284 284 gi| 16944644 | 1 166880 GSNTGVAPTNSASVTPTNSASVATTISVSVAPTASDAPTTSITLSVAPGSSSSTTAPAW 1739 gi| 11359357 | 766 766
1750 1760 1770 1780 1790 1800
Figure imgf000048_0005
Figure imgf000049_0001
The NOV6 Clustal W alignment shown in Table 6E was modified to begin at amino residue 841 and end at amino acid residue 1860. The data in Table 6E includes all of the regions overlapping with the NOV6 protein sequences.
The presence of identifiable domains in the protein disclosed herein was determined by searches using algorithms such as PROSITE, Blocks, Pfam, ProDomain, Prints and then determining the Interpro number by crossing the domain match (or numbers) using the
Interpro website (http:www.ebi.ac.uk/interpro/). Table 6F lists the domain description from
DOMAIN analysis results against NOV6.
Figure imgf000049_0002
Consistent with other known members of the Secretory Protein-like family of proteins,
NOV6 has, for example, has homology to other members of the Secretory Protein-like Protein Family. NOV6 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts. For example, NOV6 nucleic acids and polypeptides can be used to identify proteins that are members of the Secretory Protein-like Protein Family. The NOV6 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOV6 activity or function. Specifically, the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation and signal transduction. These molecules can be used to treat, e.g., Cardiovascular diseases, Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus as well as other diseases, disorders and conditions. In addition, various NOV6 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. For example, the NOV6 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the Secretory Protein-like Protein Family.
The NOV6 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of cardiac physiology. As such, the NOV6 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat cardiac and vascular system disorders, e.g., Cardiovascular diseases, Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus as well as other diseases, disorders and conditions
The NOV6 nucleic acids and polypeptides are useful for detecting specific cell types. For example, expression analysis has demonstrated that a NOV6 nucleic acid is expressed in Aorta.
Additional utilities for NOV6 nucleic acids and polypeptides according to the mvention are disclosed herein.
NOV7 A NOV7 polypeptide has been identified as a Transmission Blocking Target Antigen
S230 Precursor-like protein (also referred to as CG94978-01). The disclosed novel NOV7 nucleic acid (SEQ ID NO:13) of 1629 nucleotides is shown in Table 7A. The novel NOV7 nucleic acid sequences maps to the chromosome 1.
An ORF begins with an ATG initiation codon at nucleotides 1-3 and ends with a TGA codon at nucleotides 1627- 1629. A putative untranslated region and/or downstream from the termination codon is underlined in Table 7A, and the start and stop codons are in bold letters.
Table 7A. NOV7 Nucleotide Sequence (SEQ ID NO:13)
ATGGCGGTGCCCGGCGAGGCGGAGGAGGAGGCGACAGTTTACCTGGTAGTGAGCGGTATCCCCTCCG TGTTGCGCTCGGCCCATTTACGGAGCTATTTTAGCCAGTTCCGAGAAGAGCGCGGCGGTGGCTTCCT CTGTTTCCACTACCGGCATCGGCCTGAGCGGGCCCCTCCGCAGGCCGCTCCTAACTCTGCCCTAATT CCTACCGACCCAGCCGCTGAGGGCCAGCTTCTCTCTCAGACTTCGGCCACCGATGTCCGGCCTCTCT CCACTCGAGACTCTACTCCAATCCAGACCCGCACCTGCTGCTGCGTCATCTCGGTAAGGGGGTTGGC TCAAGCTCAGAGGCTTATTCGCATGTACTCGGGCCGCCGGTGGCTGGATTCTCACGGGACTTGGCTA CCGGGTCGCTGTCTCATCCGCAGACTTCGGCTACCTACGGAGGCATCAGGTCTGGGCTCCTTTCCCT TCAAGACCCGGAAGGAACTGCAGAGTTGGAAGGCAGAGAATGAAGCCTTCACCCTGGCTGACCTGAA GCAACTGCCGGAGCTGAACCCACCAGTGCTGATGCCCAGAGGGAATGTGGGGACTCCCCTGCGGGTC TTTTTGGAGTTGATCCGGGCCTGCCGCCTACCCCCTCGGATCATCACCCAGCTGCAGCTCCAGTTCC CCAAGACAGGTTCCTCGCGGCGCTACGGCAATGTGCCTTTTGAGTATGAGGACTCAGAGACTGTGGA GCAGGAAGAGCTTGTGTATACAGCAGAGGGTGAAGAAATACCCCAAGGAACCTACCTGGCAGATATA CCAGCCAGCCCCTGTGGAGAGCCTGAGGAAGAAGTGGGGAAGGAAGAGGAAGAAGAGTCTCACTCAG ATGAGCTGTTCGGGTGTGCTGTGGTCATCCTCCCTGCGCACCTACAGCCGCAGACCGCCGGTGGGGG GCGGGGGATGCCGGGCTGCCGCATCAGCGCCTGCGGCCCGGGGGCCCAGGAGGGGACGGCAGAGCAG AGGTCGCCGCCGCCGCCCTGGGATCCCATGCCGTCCTCTCAGCCCCCGCCCCCAACTCCGACCTTGA CTCCTACCCCGACCCCGGGTCAGTCCCCGCCGCTGCCGGACGCAGCTGGGGCTTCAGCAGGCGCGGC CGAGGACCAGGAGCTGCAGCGCTGGCGCCAGGGCGCTAGCGGGATCGCGGGGCTCGCCGGCCCCGGA GGGGGCTCTGGCGCGGCTGCGGGGGCGGGGGGCCGCGCGCTGGAGCTGGCCGAAGCACGGCGGCGGC TGCTGGAGGTGGAGGGCCGCCGGCGCCTGGTGTCGGAGCTGGAGAGCCGCGTGCTGCAGCTGCACCG CGTTTTCTTGGCGGCCGAGCTGCGCCTGGCGCACCGCGCGGAGAGCCTGAGCCGCCTGAGCGGCGGC GTGGCGCAGGCCGAGCTCTACCTGGCGGCTCACGGGTCGCGCCTCAAGAAGGGCCCGCGCCGCGGCC GCCGCGGCCGACCCCCCGCGCTGCTGGCCTCGGCGCTGGGCCTGGGCGGCTGCGTGCCCTGGGGTGC CGGGCGACTGCGGCGCGGCCACGGCCCCGAGCCCGACTCGCCCTTCCGCCGCAGCCCGCCCCGCGGC CCCGCCTCCCCGCAGCGCTGA
The NOV7 protein (SEQ ID NO: 14) encoded by SEQ ID NO: 13 is 542 amino acid residues in length and is presented using the one-letter amino acid code in Table 7B. Psort analysis predicts the NOV7 protein of the invention to be localized in the cytoplasm with a certainty of 0.4500.
Table 7B. Encoded NOV7 protein sequence (SEQ ID NO:14)
MAVPGEAEEEATVYL SGIPSVLRSAHLRSYFSQFREERGGGFLCFHYRHRPERAPPQAAPNSA LIPTDPAAEGQLLSQTSATDVRPLSTRDSTPIQTRTCCCVISVRGLAQAQRLIRMYSGRR LDSH GTWLPGRCLIRRLRLPTEASGLGSFPFKTRKELQSWKAENEAFTLADLKQLPELNPPVLMPRG V GTPLRVFLELIRAGRLPPRIITQLQLQFPKTGSSRRYGNVPFEYEDSETVEQEELVYTAEGEEIP QGTYLADIPASPCGEPEEΞVGKEEEEESHSDELFGCAWILPAHLQPQTAGGGRGMPGCRISACG PGAQEGTAEQRSPPPPWDPMPSSQPPPPTPTLTPTPTPGQSPPLPDAAGASAGAAEDQELQRWRQ GASGIAGLAGPGGGSGAAAGAGGRALELAEARRRLLEVEGRRRLVSELESRVLQLHRVFLAAELR LAHRAESLSRLSGGVAQAELYLAAHGSRLKKGPRRGRRGRPPALLASALGLGGCVPWGAGRLRRG HGPEPDSPFRRSPPRGPASPQR
A search against the Patp database, a proprietary database that contains sequences published in patents and patent publications, yielded several homologous proteins shown in Table 7C.
Table 7C. Patp results for NOV7
Smallest Sum
Reading High Prob Sequences producing High-scoring Segment Pairs: Frame Score P(N)
>patp:AAU33166 Novel human secreted protein #3657 +1 1533 5.8e-157
>patp:AAE04880 Human protease protein-7 (PRTS-7) +1 1533 5.8e-157
>patp:AAB94023 Human protein sequence SEQ ID NO: 14157 +1 1519 1.8e-155
>patp:AAU33124 Novel human secreted protein #3615 +1 390 7.7e-36
>patp:AAG02700 Human secreted protein, SEQ ID NO: 6781 +1 268 3.5e-22
In a BLAST search of public sequence databases, it was found, for example, that the nucleic acid sequence of this invention has 874 of 876 bases (99%) identical to a gb:GENBANK-LD:AK022517|acc:AK022517.1 mRNA from Homo sapiens (cDNA FLJ12455 fis, clone NT2RM1000563, weakly similar to TRANSMISSION-BLOCKING TARGET ANTIGEN S230 PRECURSOR). The full amino acid sequence of the protein of the invention was found to have 290 of 292 amino acid residues (99%) identical to, and 290 of 292 amino acid residues (99%) similar to, the 525 amino acid residue ptnr:SPTREMBL- ACC.Q9H9Z3 protein from Homo sapiens (CDNA FLJ12455 FIS, CLONE NT2RM1000563, WEAKLY SIMILAR TO TRANSMISSION- BLOCKING TARGET ANTIGEN S230 PRECURSOR).
NOV7 also has homology to the proteins shown in the BLASTP data in Table 7D.
Table 7D. BLAST results for NOV7
Gene Index/ Protein/ Organism Length Identity Positives Expect Identif ier (aa) ( % ) ( %) gi 1 1854515 | ref | XP_ hypothetical 525 274/274 274/274 e- 147 084046 . l l (XM 084046 protein FLJ12455 ( 100% ) (100%) [Homo sapiens] gi 1 11545793 | ref | NP_ hypothetical 525 272/274 272/274 e- 145 071361 . l l (NM 02207E protein FLJ12455 ( 99% ) ( 99%) [Homo sapiens] gi 1 18545156 I ref | XP_ similar to 107 83/84 84/84 6e- 37 086159 . l l (XM 08615S hypothetical ( 98% ) (99%) protein FLJ12455 [Homo sapiens] gi 1 18545158 I ref | XP_ hypothetical 141 135/137 136/137 2e-33 097448 . 1 ] (XM 097448 protein XP_09744ϊ ( 98%) ( 98%) [Homo sapiens] gi 1 17562286 I ref | NP_ K07B1 . 7b . p 487 76/237 119/237 3e-30 505420 . l | (NM 073019 [ Caenorhabdi tis ( 32%) ( 50%) elegans]
A multiple sequence alignment is given in Table 7E, with the NOV7 protein being shown on line 1 in Table 7E in a ClustalW analysis, and comparing the NOV7 protein with the select related protein sequences shown in Table 7D. This BLASTP data is displayed graphically in the ClustalW in Table 7E.
Table 7E. ClustalW Analysis of NOV7
1) > N0V7; SEQ ID N0:14
2) > gi|18545154|/ hypothetical protein FLJ12455 [Homo sapiens]; SEQ ID NO:65
3) > gi|l 1545793)/ hypothetical protein FLJ12455 [Homo sapiens]; SEQ ID NO:66
10 20 30 40 50 60
NOV7 1 YAVPGEAEEEATVYLWSGIPSVLRSAΗLRSYFSQFREERGGGFLCFΗYRΗRPERAPPQ gi 118545154 I 1 viAVPGEAEEEATVYLWSGIPSVLRSAΗLRSYFSQFREERGGGFLCFΗYRΗRPERAPPQi gij 11545793 j 1 VIAVPGEAEEEATVYLWSGIPSVLRSAΗLRSYFSQFREERGGGFLCFΗYRΗRPERAPPQ
70 80 90 100 110 120
NOV7 61 SALIPTDPAAEGOLLSOTSATDVRPLSTRDSTPlOTRTCCCVISVRGLAOAORLIRiy 120 gi 118545154 61 iPNSALIPTDPAAEGQLLSQTSATDVRPLSTRDSTPIQTRTCCCVISVRGLAQAQRLIf 120 gij 11545793 61 iPNSALIPTDPAAEGQLLSQTSATDVRPLSTRDSTPIQTRTCCCVISVRGLAQAQRLIl 120
130 140 150 160 170 180
N0V7 121 IHGTWLPGRCLIRRLRLPTEASGLGSF gi 118545154 121 rSGRR LDSHGT LPGRCLIRRLRLPTEASGLGSFPFKTRKELQS KAENEAFTLADLKζ gi 111545793 121 fSGRR LDSHGTWLPGRCLIRRLRLPTEASGLGUFPFKTRKELQS KAENEAFTLADLKC
190 200 210 220 230 240
N0V7 181 PELNPPVLMPRGNVGTPLRVFLELIRACRLPPRIITQLQLQFPKTGSSRRYG VPFEYE gi 118545154 181 JPELNPPVLMPRGNVGTPLRVFLELIRACRLPPRIITQLQLQFPKTGSSRRYGNVPFEYE gij 11545793 181 JPELNPPVLMPRGNVGTPLRVFLELIRACRLPPRIITQLQLQFPKTGSSRRYGNVPFEYE
250 260 270 280 290 300
Figure imgf000053_0001
430 440 450 460 470 480
N0V7 420 EARJSLLIVESRRRLVSILESRVLQLHREFLΞA'LRL. RAESLSJgLSGgVAQAELYJgAΛ 479 gi 118545154 412 (IGRKVMERQG WAEGQGLGCRCSGVPEALDSDGQI 462 gij 11545793 412 3IGRKVMER0G WAEGOGLGCRCSGVPEALDSDGOI 462
Figure imgf000053_0002
The presence of identifiable domains in the protein disclosed herein was determined by searches using algorithms such as PROSITE, Blocks, Pfam, ProDomain, Prints and then determining the Interpro number by crossing the domain match (or numbers) using the
Interpro website (http:www.ebi.ac.uk/interpro/). Table 7F lists the domain description from DOMAIN analysis results against NOV7.
Figure imgf000053_0003
Figure imgf000054_0001
Consistent with other known members of the Transmission Blocking Target Antigen S230 Precursor-like family of proteins, NOV7 has, for example, three Blocking NT2RM1000563 Transmission-FIS Antigen Weakly Precursor Peptidase A2 signature sequences and homology to other members of the Transmission Blocking Target Antigen S230 Precursor-like Protein Family. NOV7 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts. For example, NOV7 nucleic acids and polypeptides can be used to identify proteins that are members of the Transmission Blocking Target Antigen S230 Precursor-like Protein Family. The NOV7 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOV7 activity or function. Specifically, the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation and signal transduction. These molecules can be used to treat, e.g., Cardiovascular diseases, Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus , Pulmonary stenosis, Subaortic stenosis, Ventricular septal defect (VSD), valve diseases, Tuberous sclerosis, Scleroderma, Obesity, Transplantation, DiabetesNon Hippel-Lindau (VHL) syndrome , Pancreatitis, Obesity, Von Hippel-Lindau (VHL) syndrome , Alzheimer's disease, Stroke, Tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, Cerebral palsy, Epilepsy, Lesch-Νyhan syndrome, Multiple sclerosis, Ataxia-telangiectasia, Leukodystrophies, Behavioral disorders, Addiction, Anxiety, Pain, Neuroprotection
In addition, various NOV7 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. For example, the NOV7 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the Transmission Blocking Target Antigen S230 Precursor-like Protein Family. The NOV7 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of cardiac and nerve physiology. As such, the NOV7 nucleic acids and polypeptides, antibodies and related compounds according to the mvention may be used to treat cardiovascular and nervous system disorders, e.g., Cardiovascular diseases,
Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus , Pulmonary stenosis, Subaortic stenosis, Ventricular septal defect (VSD), valve diseases, Tuberous sclerosis, Scleroderma, Obesity, Transplantation, DiabetesNon Hippel-Lindau (VHL) syndrome, Pancreatitis, Obesity, Von Hippel-Lindau (VHL) syndrome, Alzheimer's disease, Stroke, Tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, Cerebral palsy, Epilepsy, Lesch-Νyhan syndrome, Multiple sclerosis, Ataxia-telangiectasia, Leukodystrophies, Behavioral disorders, Addiction, Anxiety, Pain, Neuroprotection
The NOV7 nucleic acids and polypeptides are useful for detecting specific cell types. For example, expression analysis has demonstrated that a NOV7 nucleic acid is expressed in Adipose, Heart, Aorta, Coronary Artery, Umbilical Vein, Pancreas, Liver, Gall Bladder, Colon, Bone Marrow, Thymus, Bone, Cartilage, Synovium/Synovial membrane, Skeletal Muscle, Brain, Left cerebellum, Right Cerebellum, Thalamus, Hypothalamus, Pituitary Gland, Frontal Lobe, Parietal Lobe, Cerebral Medulla/Cerebral white matter, Basal Ganglia/Cerebral nuclei, Substantia Nigra, Hippocampus, Cervix, Mammary gland Breast, Uterus,
Oviduct/Uterine Tube/Fallopian tube, Prostate, Testis, Lung, Bronchus, Larynx, Kidney, Retina, Skin, Epidermis.
Additional utilities for NOV7 nucleic acids and polypeptides according to the invention are disclosed herein.
NOV8
A NOV8 polypeptide has been identified as a Nuclear Protein-like protein (also referred to as CG94713-01). The disclosed novel NOV8 nucleic acid (SEQ ID NO:15) of 3807 nucleotides is shown in Table 8A. The novel NOV8 nucleic acid sequences maps to the chromosome 1.
An ORF begins with an ATG initiation codon at nucleotides 16-18 and ends with a TGA codon at nucleotides 3793-3795. A putative untranslated region and/or downstream from the termination codon is underlined in Table 8 A, and the start and stop codons are in bold letters.
Table 8A. NOV8 Nucleotide Sequence (SEQ ID NO:15)
ATGATAAAATAGAAGATGAATTGCAAACCTTCTTTACCAGTGATAAAGATGGAAATTACACATGCAT ACAACCCGAAATCACCACCTACACAAAACTCTTCAGCCAGCAGTGTGAACTGGAATTCTGCCAACCC AGATGACATGGTGGTTGATTATGAAACTGACCCTGCTGTAGTTACTGGTGAAAATATTTCTTTAAGC CTTCAGGGTGTTGAAGTATTTGGTCATGAAAAGTCTTCTAGTGATTTCATTAGTAAGCAGGTGTTAG ATATGCATAAAGATTCTATTTGTCAGTGTCCTGCACTTGTAGGTACTGAGAAGCCCAAATATCTGCA ACACAGTTGTCATTCCCTAGAAGCAGTTGAGGGCCAGAGTGTTGAGCCATCTTTGCCTTTTGTGTGG AAGCCTAATGACAATTTGAACTGTGCAGGCTACTGTGATGCCTTGGAGCTGAACCAAACATTTGACA TGACAGTGGATAAAGTTAACTGCACCTTTATATCACATCATGCCATCGGAAAGAGTCAGTCCTTCCA TACTGCTGGAAGCCTGCCACCAACTGGTAGGAGAAGTGGAAGTACATCTTCTTTATCCTATTCCACT TGGACATCTTCCCATTCTGATAAGACGCATGCAAGAGAAACTACTTATGATAGAGAAAGCTTTGAAA ACCCTCAAGTCACACCATCAGAAGCCCAAGACATGACTTACACAGCATTTTCTGATGTGGTGATGCA AAGTGAGGTTTTTGTTTCAGATATTGGAAATCAGTGTGCATOTTCTTCAGGAAAGGTCACCAGTGAG TACACAGATGGATCACAACAAAGACTAGTTGGAGAAAAAGAGACACAAGCACTAACACCAGTTTCTG ATGGCATGGAAGTCCCCAATGATTCTGCATTACAAGAGTTCTTTTGTTTATCCCATGATGAATCCAA TAGCGAACCACATTCACAGAGCTCATACAGGCACAAGGAAATGGGCCAAAATCTGAGAGAGACAGTG TCCTATTGTCTTATTGATGATGAATGCCCTTTAATGGTGCCAGCTTTTGATAAGAGCGAAGCTCAAG TGCTGAACCCAGAGCATAAAGTCACTGAGACTGAAGACACACAAATGGTCTCCAAAGGAAAGGATTT GGGAACCCAAAATCATACCTCAGAATTGATTCTAAGTAGCCCGCCAGGACAAAAGGTGGGCTCGTCA TTTGGACTGACTTGGGATGCAAATGATATGGTCATTAGCACAGACAAAACGATGTGCATGTCAACAC CAGTCCTAGAACCCACAAAAGTAACCTTTTCTGTTTCACCGATTGAAGCGACGGAGAAATGTAAGAA AGTGGAGAAGGGTAATCGAGGGCTTAAAAACATACCAGACTCGAAGGAGGCACCTGTGAACCTGTGT AAACCCAGTTTAGGAAAATCAACAATCAAAACGAATACCCCAATAGGCTGCAAAGTTAGAAAAACTG AAATTATAAGTTACCCAAGACCAAACTTCAAGAATGTCAAAGCAAAAGTTATGTCTAGAGCAGTGTT GCAGCCCAAAGATGCTGCTTTATCAAAGGTCACGCCCAGACCTCAGCAGACCAGTGCCTCATCACCC TCATCAGTGAATTCAAGACAACAAACAGTCTTGAGCAGAACACCGAGATCTGACTTGAATGCAGACA AAAAAGCAGAAATTCTAATTAACAAGACACATAAGCAGCAGTTTAATAAACTCATTACTAGCCAGGC TGTGCATGTTACAACTCATTCTAAAAATGCTTCACACAGGGTTCCAAGAACAACATCTGCCGTGAAA TCGAATCAGGAAGATGTTGACAAAGCCAGTTCTTCTAACTCAGCATGCGAGACCGGGTCCGTTTCTG CGTTGTTTCAGAAGATCAAAGGCATACTCCCTGTTAAAATGGAAAGTGCAGAATGTTTGGAAATGAC CTATGTTCCCAACATTGATAGGATTAGCCCTGAAAAGAAGGGTGAAAAAGAAAATGGGACATCTATG GAAAAACAAGAGCTGAAACAAGAGATTATGAATGAGACTTTTGAATATGGTTCTCTGTTTTTGGGCT CTGCTTCAAAAACAACGACCACCTCAGGTAGGAATATATCCAAGCCTGACTCCTGCGGTTTGAGGCA AATAGCTGCTCCAAAAGCCAAAGTGGGGCCCCCTGTTTCCTGTTTGAGGCGGAACAGTGACAATAGA AATCCCAGTGCTGATCGAGCCGTATCTCCTCAGAGGATCAGGCGTGTGTCCAGTTCTGGAAAGCCTA CATCCTTGAAAACTGCACAGTCGTCATGGGTGAATTTGCCTAGACCACTTCCTAAATCCAAAGCATC TTTGAAAAGTCCTGCGCTGCGGAGGACAGGAAGCACCCCCTCAATAGCCAGCACCCACAGTGAGCTG AGCACTTACAGCAACAATTCTGGTAATGCCGCTGTCATCAAATATGAGGAGAAACCTCCAAAACCAG CATTTCAGAATGGTTCCTCAGGATCCTTTTATTTGAAGCCTTTGGTATCCAGGGCTCATGTTCACTT GATGAAAACTCCTCCAAAAGGTCCTTCGAGAAAAAATTTATTTACAGCTCTTAATGCAGTTGAAAAG AGCAGGCAAAAGAATCCTCGAAGCTTATGTATCCAGCCACAGACAGCTCCCGATGCGCTGCCCCCTG AGAAAACACTTGAATTGACGCAATATAAAACAAAATGTGAAAACCAAAGTGGATTTATCCTGCAGCT CAAGCAGCTTCTTGCCTGTGGTAATACCAAGTTTGAGGCATTGACAGTTGTGATTCAGCACCTGCTG TCTGAGCGGGAGGAAGCACTGAAACAACACAAAACCCTATCTCAAGAACTTGTTAACCTCCGGGGAG AGCTAGTCACTGCTTCAACCACCTGTGAGAAATTAGAAAAAGCCAGGAATGAGTTACAAACAGTGTA TGAAGCATTCGTCCAGCAGCACCAGGCTGAAAAAACAGAACGAGAGAATCGGCTTAAAGAGTTTTAC ACCAGGGAGTATGAAAAGCTTCGGGACACTTACATTGAAGAAGCAGAGAAGTACAAAATGCAATTGC AAGAGCAGTTTGACAACTTAAATGCTGCGCATGAAACCTCTAAGTTGGAAATTGAAGCTAGCCACTC AGAGAAACTTGAATTGCTAAAGAAGGCCTATGAAGCCTCCCTTTCAGAAATTAAGAAAGGCCATGAA ATAGAAAAGAAATCGCTTGAAGATTTACTTTCTGAGAAGCAGGAATCGCTAGAGAAGCAAATCAATG ATCTGAAGAGTGAAAATGATGCTTTAAATGAAAAATTGAAATCAGAAGAACAAAAAAGAAGAGCAAG AGAAAAAGCAAATTTGAAAAATCCTCAGATCATGTATCTAGAACAGGAGTTAGAAAGCCTGAAAGCT GTGTTAGAGATCAAGAATGAGAAACTGCATCAACAGGACATCAAGTTAATGAAAATGGAGAAACTGG TGGACAACAACACAGCATTGGTTGACAAATTGAAGCGTTTCCAGCAGGAGAATGAAGAATTGAAAGC TCGGATGGACAAGCACATGGCAATCTCAAGGCAGCTTTCCACGGAGCAGGCTGTTCTGCAAGAGTCG CTGGAGAAGGAGTCGAAAGTCAACAAGCGACTCTCTATGGAAAACGAGGAGCTTCTGTGGAAACTGC ACAATGGGGACCTGTGTAGCCCCAAGAGATCCCCCACATCCTCCGCCATCCCTTTGCAGTCACCAAG GAATTCGGGCTCCTTCCCTAGCCCCAGCATTTCACCCAGATGACACCTCCCCAAA
Variant sequences of NOV8 are included in Example 3, Table 20. A variant sequence can include a single nucleotide polymorphism (SNP). A SNP can, in some instances, be referred to as a "cSNP" to denote that the nucleotide sequence containing the SNP originates as a cDNA.
The NOV8 protein (SEQ ID NO: 16) encoded by SEQ ID NO: 15 is 1259 amino acid residues in length and is presented using the one-letter amino acid code in Table 8B. Psort analysis predicts the NOV8 protein of the invention to be localized in the nucleus with a certainty of 0.7600.
Table 8B. Encoded NOV8 protein sequence (SEQ ID NO: 16)
MNCKPSLPVIKMEITHAYNPKSPPTQNSSASSVNWNSANPDDMVVDYETDPAVVTGENISLSLQG
VEVFGHEKSSSDFISKQVLDMHKDSICQCPALVGTEKPKYLQHSCHSLEAVEGQSVEPSLPFVWK
PNDNLNCAGYCDALELNQTFDMTVDKVNCTFISHHAIGKSQSFHTAGSLPPTGRRSGSTSSLSYS
TWTSSHSDKTHARETTYDRESFENPQVTPSEAQDMTYTAFSDWMQSEVFVSDIGNQCACSSGKV
TSEYTDGSQQRLVGEKΞTQALTPVSDGMΞVPNDSALQEFFCLSHDESNSEPHSQSSYRHKEMGQN
LRETVSYCLIDDECPLMVPAFDKSΞAQVLNPEHKVTETEDTQMVS GKDLGTQNHTSELILSSPP
GQKVGSSFGLT DANDMVISTDKTMCMSTPVLEPTKVTFSVSPIEATEKCKKVEKGNRGLK IPD
SKEAPVNLCKPSLGKSTIKTNTPIGCKVRKTEIISYPRPNFl^KAKVMSRAVLQPKDAALSKVT
PRPQQTSASSPSSVNSRQQTVLSRTPRSDLNADKKAEILINKTHKQQFNKLITSQAVHVTTHSKN
ASHRVPRTTSAVKSNQEDVDKASSSNSACETGSVSALFQKIKGILPVKMESAECLEMTYVPNIDR
ISPE KGEKENGTSMEKQELKQEIMNETFΞYGSLFLGSASKTTTTSGRNISKPDSCGLRQIAAPK
AKVGPPVSCLRRNSDNRNPSADRAVSPQRIRRVSSSGKPTSLKTAQSS VNLPRPLPKSKASLKS
PALRRTGSTPSIASTHSELSTYSNNSGNAAVIKYEEKPPKPAFQHGSSGSFYLKPLVSRAHVHLM
KTPPKGPSRK LFTALNAVEKSRQKNPRSLCIQPQTAPDALPPEKTLELTQYKTKCΞNQSGFILQ
LKQLLACGNTKFEALTWIQHLLSERΞEALKQHKTLSQELVNLRGELVTASTTCEKLEKARNELQ
TVYEAFVQQHQAEKTERENRLKEFYTRΞYEKLRDTYIEEAEKY MQLQEQFDNLNAAHETSKLEI
EASHSEKLELLKKAYEASLSEIKKGHEIEKKSLEDLLSEKQESLEKQI DLKSE DALNEKLKSE
EQKRRAREKANL^PQIMYLEQELESLKAVLEIKN^
QQENEELKARMDKHMAISRQLSTEQAVLQESLEKESKVNK-RLSMENΞELL KLHNGDLCSPKRSP
TSSAIPLQSPR SGSFPSPSISPR
A search against the Patp database, a proprietary database that contains sequences published in patents and patent publications, yielded several homologous proteins shown in Table 8C.
Table 8C. Patp results for NOV8
Smallest Sum
Reading High Prob Sequences producing High-scoring Segment Pairs: Frame Score P(N)
>patp:AAG63542 Amino acid sequence of a human ATIP isoform +1 6389 0.0
>patp:AAG635 9 Amino acid sequence of a human ATIP isoform +1 6233 0.0
>patp:AAG63541 Amino acid sequence of a human ATIP isoform +1 3928 0.0
>patp:AAG63537 Amino acid sequence of a ATIP isoform +1 3279 0.0
>patp:AAG63530 Amino acid sequence of a human ATIP isoform +1 2954 1.5e-307 In a BLAST search of public sequence databases, it was found, for example, that the nucleic acid sequence of this invention has 3751 of 3751 bases (100%) identical to a gb:GENBANK-ID:AB033114|acc:AB033114.1 mRNA from Hom sapiens (mRNA for KIAA1288 protein, partial eds). The full amino acid sequence of the protein of the invention was found to have 1245 of 1245 amino acid residues (100%) identical to, and 1245 of 1245 amino acid residues (100%) similar to, the 1245 amino acid residue ptnr:SPTREMBL- ACC:Q9ULD2 protein from Homo sapiens (KIAA1288 PROTEIN).
NOV8 also has homology to the proteins shown in the BLASTP data in Table 8D.
Figure imgf000058_0001
A multiple sequence alignment is given in Table 8E, with the NOV8 protein being shown on line 1 in Table 8E in a ClustalW analysis, and comparing the NOV8 protein with the related protein sequences shown in Table 8D. This BLASTP data is displayed graphically in the ClustalW in Table 8E.
Table 8E. ClustalW Analysis of NOV8 l) > NOV8; SEQ ID NO: 16
2) > gi|6331407|/ KIAA1288 protein [Homo sapiens]; SEQ TD NO:67
3) > gijl7865632|/ AT2 receptor-interacting protein 1 [Homo sapiens]; SEQ ID NO:68
4) > gi|10436722|/ unnamed protein product [Homo sapiens]; SEQ ID NO:69
5) > gi|3882269|/ KIAA0774 protein [Homo sapiens]; SEQ ID NO:70
6) > gijl7475630|/ KIAA0774 protein [Homo sapiens]; SEQ ID NO:71
10 20 30 40 50 60
NOV8 1 -MNCKPSLPVIKMEITΗAYNPKSPPTQNSSASSVNWNSANPDDMWDYETDPAWTG- - - 56 gi| 6331407 | 1 TΗAYNPKSPPTQNSSASSVNWNSANPDDMWDYETDPAWTG 2 gi|17865632| 1 X gi| 10436722 | 1 gi|3882269| 1 RGQIPGGGEGPQKTLPDΗAVPAAFPATDSTSEGKSVRΗPKPSTSESKQSTPSETQTVGAΗ 60 gi| 17475630 | 1 10 6
20
66 52
80
23 09
40
Figure imgf000059_0001
Figure imgf000059_0002
370 380 390 400 410 420
Figure imgf000059_0003
gi|3882269| δGDLKPfeANLYΞKFKPDLQKPRVFSSGLMVSGIjSpPGHPFSfflMSfeKFfflQfeδδDH 535 gi 1 7475630 I iGDLKPSANLYEKFKPDL'Q PRVFSSGLMVSGlBpPGHPFsfflMsfeKFfflQfefflDH 273
550 560 570 580 590 600
NOV8 524 Q^TSAS^SSVSSRQQTVLSgTPRSDfflNAD^KAEILjIHKTHKQQFNKflgT^AVHVTiTHS 583 gi| 6331407 | 510 QQTSAS^SSVSSRQQTVLS|TPRSD2NAD^KAEIL'INKTHKQQFNKgaT^AVHV3|τHS 569 gi|l7865632| 1 gi| 10436722 | 1 gi|3882269| 536 GKEEFCB 3PYA YEVPPTFY|- ILKPQLGLGASRLPSAKSRIR -RSSJASA 591 gi| 17475630 | 274 GBEEFCR JPYAHYEVPPTFYg- ILKPQLGLGAMSRLPSAKSRIR -RSSASA1 329
Figure imgf000060_0001
790 800 810 820 830 840 GSTPSIASTHSELSTYSNNSGNAAVIKYEEKPPKPA 822 GSTPSIASTHSELSTYSNNSGNAAVIKYEEKPPKPA 808 ST 35
766
Figure imgf000060_0002
504
Figure imgf000060_0003
970 980 990 1000 1010 1020
Figure imgf000060_0004
75 00
Figure imgf000061_0001
38
1030 1040 1050 1060 1070 1080
Figure imgf000061_0002
1090 1100 1110 1120 1130 1140
N0V8 1059 M*zUi<tMzHάUΛttMM^,. \ nnMHrolM- lslsfc 1116 gi I 6331407 I 1045 iSEIKKGHEIEKKSLEDLLSEKQESLEKQINDL SENDALNEKLK ■EEQKRRAREK 1102 gijl7865632| 236 JSEIKKGHEIEKKSLEDLLSEKQESLEKQINDLKSENDALNEKLK EEQKRRAREKi 293 gi 110436722 j 40 JSEIKKGHEIEKKSLEDLLSEKQESLEKQINDLKSENDALNEKLK EEOKRRAREKAli 97 gi J3882269| 961 VQ LMSTH ^EWgE FEKLRL^QIWBTfflTFQSpSffl D A RFEfi M-RKNTEgQLE 1020 gijl7475630| 699 VQ|LMSTB ]ESSB FE|:Lp;L^QD vBτ3τFQSQs|Rε) !RFEy !U-R NTEBQLE 758
1150 1160 1170 1180 1190 1200
N0V8 1117 JKNPQIMYLEQELESLKAVLEjj KNEKLHQQDIKLMKMEKLVDN TALVDKLKRFQQENE 1176 gi I 6331407 I 1103 JKNPQIMYLEQELESLKAVLE jj KNEKLHQQDIKLMKMEKLVDNNTALVDKLKRFQQENEE 1162 gij 17865632 I 294 JKNPQIMYLEQELESLKAVLE j] KNEKLHQQDIKLMKMEKLVDNNTALVDKLKRFQQENEE 353 gijl0436722 j 98 JKNPQIMYLEQELESLKAVLE nKNEKLHQQDIKLMKMEKLVDNNTALVDKLKRFQQENE1 157 giJ3882269| 1021 &ALAPYQI SILEL SlQVLSgζ 1080 gij 17475630 I 759 SALAPYQI SlLELE SlQVLβ 818
1210 1220 1230 1240 1250 1260
N0V8 1177 1236 gi| 6331407 I 1163 JKARMDKHMAISRQLSTEQAVLQESLEKESKVNKRLSMENEELLWKLHNGDLCSPKRSPT 1222 gi 117865632 I 354 UKARMDKHMAISRQLSTEQAVLQESLEKESKVNKRLSMENEELLWKLHNGDLCSPKRSP'I 413 gijl0436722| 158 JKARMDKHMAISRQLSTEQAVLQESLEKESKVNKRLSMENEELLWKLHNGDLCSPKRSPT 217 giJ3882269| 1081 QT^PTB LSP 1140 gijl7475630| 819 ggNTvy S STOEKSS-ERTIB-BBMI IOTBHPT" LSP 878
1270 1280
N0V8 1237 SSAIPLQSPRNSGSFPSPSISPR 1259 gi I 6331407 I 1223 SSAIPLQSPRNSGSFPSPSISPR 1245 g j 17865632 I 414 SSAIPLQSPRNSGSFPSPSISPR 436 gi 110436722 j 218 SSAIPLQSPRNSGSFPSPSISPR 240 giJ3882269| 1141 fSPVYRi SSSGPSgPAR PR 1163 gij 17475630 I 879 ΪSPVYRI SSSGPSBPAR' TTG 901
The presence of identifiable domains in the protein disclosed herein was determined by searches using algorithms such as PROSITE, Blocks, Pfam, ProDomain, Prints and then determining the Interpro number by crossing the domain match (or numbers) using the Interpro website (http:www.ebi.ac.uk/interpro/). Table 8F lists the domain description from DOMAIN analysis results against NOV8.
Figure imgf000061_0003
Consistent with other known members of the Nuclear Protein-like family of proteins, NOV8 has, for example, an RNA polymerase omega subunit signature sequence and homology to other members of the Nuclear Protein-like Protein Family. NOV8 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts. For example, NOV8 nucleic acids and polypeptides can be used to identify proteins that are members of the Nuclear Protein-like Protein Family. The NOV8 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOV8 activity or function. Specifically, the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular replication, and signal transduction. These molecules can be used to treat, e.g., Cardiovascular diseases, Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus , Pulmonary stenosis, Subaortic stenosis, Ventricular septal defect (VSD), valve diseases, Tuberous sclerosis, Scleroderma, Obesity, Transplantation, DiabetesNon Hippel-Lindau (VHL) syndrome , Pancreatitis, Obesity, Hyperparathyroidism, Hypoparathyroidism as well as other diseases, disorders and conditions. hi addition, various ΝOV8 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. For example, the NOV8 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the Nuclear Protein-like Protein Family.
The NOV8 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of cardiac or endocrine physiology. As such, the NOV8 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat infection, cardiovascular system, immune system, and nervous system disorders, e.g., Cardiovascular diseases, Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect,
Ductus arteriosus , Pulmonary stenosis, Subaortic stenosis, Ventricular septal defect (VSD), valve diseases, Tuberous sclerosis, Scleroderma, Obesity, Transplantation, DiabetesNon Hippel-Lindau (VHL) syndrome , Pancreatitis, Obesity, Hyperparathyroidism, Hypoparathyroidism as well as other diseases, disorders and conditions. The NOV8 nucleic acids and polypeptides are useful for detecting specific cell types. For example, expression analysis has demonstrated that a NOV8 nucleic acid is expressed in Heart, Aorta, Coronary Artery, Vein, Umbilical Vein, Adrenal Gland/Suprarenal gland, Pancreas, Islets of Langerhans, Parathyroid Gland, Thyroid, Pineal Gland, Tongue, Salivary Glands, Stomach, Liver, Small Intestine, Colon, Ascending Colon, Lymphoid tissue, Spleen, Brain, Thalamus, Hypothalamus, Temporal Lobe, Amygdala, Cerebral Medulla/Cerebral white matter, Basal Ganglia/Cerebral nuclei, Substantia Nigra, Hippocampus, Spinal Chord, Cervix, Mammary gland/Breast, Ovary, Placenta, Uterus, Prostate, Testis, Lung, Nasoepithelium, Larynx, Urinary Bladder, Kidney, Kidney Cortex, Retina, Skin, Foreskin, Epidermis, Dermis.
Additional utilities for NOV8 nucleic acids and polypeptides according to the invention are disclosed herein.
NOV9
ANOV9 polypeptide has been identified as a Hemicentin precursor-like protein (also referred to as CG94702-01). The disclosed novel NOV9 nucleic acid (SEQ ID NO:17) of 11796 nucleotides is shown in Table 9A. The novel NOV9 nucleic acid sequences maps to the chromosome 9.
An ORF begins with an ATG initiation codon at nucleotides 1-3 and ends with a TAA codon at nucleotides 11794-11796. A putative untranslated region and/or downstream from the termination codon is underlined in Table 9A, and the start and stop codons are inbold letters.
Table 9A. NOV9 Nucleotide Sequence (SEQ ID NO:17)
ATGTCTGCCTTATTTGCAGCTGTGTACCAGATGCTAAAACCACGCCTGGTCCATAACAGCCCACATC CGGTGACCTATCAAATTGAGGCAAGTTTAAAGCCAGAGCAGCCTGGTGTCACGCTGGTGTCCATCCC AGTCTTCCTGGCACCTTCCTGGCACAAAGCCTCAGAGCTGATCCCGACCCAGTCCTTCCGAGCACAG GGGGCAGGGAAGCAGCTCCTCGGCTCTCCTTGCCCCCAAGTGCCCCCCAGCATCCGGGAGGACGGGC GCAAGGCCAACGTGTCGGGTATGGCCGGGCAGTCCCTGACGCTGGAGTGTGACGCGAACGGCTTTCC AGTCCCTGAGATCGTGTGGCTGAAGGACGCGCAGCTGATTCCTAAGGTGGGCGGCCACCGCCTCCTG GACGAGGGCCAGTCCCTCCACTTCCCCAGGATCCAGGAGGGTGATTCTGGGCTCTACTCCTGCCGGG CAGAGAACCAGGCTGGCACCGCCCAGAGGGACTTCCATCTCCTTGTGCTCACCCCTCCTTCCGTGCT TGGAGCCGGGGCCGCTCAGGAGGTGCTAGGATTGGCCGGTGCAGACGTGGAGCTGCAGTGTTGGACC TCAGGGGTCCCCACGCCCCAGGTGGAGTGGACCAAGGACAGGCAGCCTGTCCTTCCGGGAGGCCCTC CCTGCAGGTCCAGGAGGATGGCCAGGTTCTCAGGATCACCGGCAGTCACGTGGGGGATGAGGGACG ATACCAGTGCGTGGCCTTCAGCCCAGCTGGTCAGCAGGCCAGGGACTTCCAGCTCCGAGTTCATGCG CCCCCCACTATCTGGGGCTCCAACGAGACAGGCGAGGTGGCCGTCATGGΆGGACCACCTAGTGCAGC TCCTGTGTGAGGCTCGAGGAGTGCCCACCCCAAACATCACCTGGTTCAAGGACGGGGCCCTGCTCCC CACCAGCACCAAGGTGGTCTACACTAGGGGCGGTCGGCAGTTGCAGCTGGGGAGGGCCCAGAGCTCC GATGCCGGCGTCTACACCTGCAAGGCCAGCAATGCTGTGGGGGCCGCAGAGAAGGCCACCAGGCTGG ATGTTTATGTCCCACCTACCATCGAGGGCGCCGGTGGAAGACCATACGTGGTGAAGGCTGTGGCTGG GAGGCCTGTGGCGCTGGAGTGCGTGGCCAGAGGCCACCCGTCCCCCACCCTCTCCTGGCACCACGAG GGGCTGGCCGTGGCAGAGAGCAACGAGTCGCGGCTGGAGACAGACGGGAGTGTGCTGAGGCTGGAGA GCCCGGGGGAGGCATCCAGTGGCCTGTACAGCTGTGTGGCCAGCAGTCCTGCCGGGGAAGCCGTCCT GCAGTACTCCGTGGAGGTTCAGGTGCCCCCACAGCTCCTGGTGGCTGAAGGCTTGGGACAGGTGACC ACCATCGTGGGACAGCCCCTGGAACTTCCCTGCCAGGCCTCAGGCTCCCCAGTACCCΆCTATCCAGT GGCTGCAGAATGGCCGCCCAGCCGAGGAGCTGGCTGGGGTGCAGGTGGCCTCGCAGGGGACCACACT GCACATTGACCATGTGGAGCTGGACCACTCAGGCCTCTTCGCCTGCCAGGCCACCAATGAGGCGGGC ACTGCCGGGGCCGAGGTGGAGGTGTCTGTGCATGAGTTCCCATCGGTCAGTATCATTGGGGGTGAGA ACATCACAGCTCCTTTCCTGCAGCCTGTGACCCTCCAGTGCATAGGGGATGGGGTGCCCACCCCAAG CCTCCGTTGGTGGAAGGATGGTGTAGCCCTGGCAGCCTTTGGGGGGAACCTACAGATTGAGAAGGTG GACCTGAGGGACGAGGGCATCTACACTTGTGCTGCTACCAACCTGGCTGGGGAGAGCAAGAGGGAAG TGGCGCTGAAAGTTTTGGTGCCCCCCAACATCGAGCCAGGCCCAGTCAΆCAAGGCAGTGCTGGAAAA GCCTCAGTGACCTTGGAGTGTCTGGCTTCGGGCGTGCCCCCTCCTGATGTCTCCTGGTTCAAGGGC CACCAACCTGTCTCTTCATGGATGGGAGTGACAGTATCAGTGGATGGGAGAGTTCTCCGCATTGAGC AAGCCCAGCTTTCTGATGCTGGGAGCTACCGCTGTGTGGCATCCAATGTGGCAGGTAGCACAGAGCT GCGGTATGGCCTACGGGTCAATGTGCCCCCTCGAATCACACTGCCACCCAGCCTGCCAGGCCCTGTG TTGGTCAACACCCCTGTCCGGCTGACCTGCAΆTGCCACCGGTGCCCCCAGCCCCACACTGATGTGGC TGAAGGATGGAAACCCTGTGTCCCCTGCAGGGACCCCTGGCCTGCAGGTCTTCCCTGGGGGCCGGGT CCTCACCTTGGCTAGTGCCCGGGCCTCCGACTCTGGGAGGTACTCCTGCGTGGCTGTGAGCGCGGTG GGCGAGGACCGCCAGGATGTTGTCCTGCAAGTCCACATGCCCCCGAGTATCCTTGGAGAAGAGCTGA ATGTGTCCGTTGTGGCCAATGAGTCAGTGGCCCTGGAGTGCCAGAGCCACGCCATGCCCCCTCCTGT GCTGAGCTGGTGGAAGGACGGGCGGCCCCTGGAACCACGGCCTGGAGTCCACCTCTCCGCAGACAAA GCCTTGCTGCAGGTGGACAGAGCCGATGTGTGGGATGCGGGCCATTACACCTGTGAGGCACTGAACC AGGCCGGCCACTCAGAGAAACACTACAATCTGAACGTCTGGGGTCAACCCCTCCCCGGGGAGGGGGC GGCCTCCAGCACGTGTCGGCTGTGGGGAGGCTGTTGTACCTGGGACAGGCCCAGCTGGCTCAGGAA GGAACATACACCTGTGAATGCAGCAACGTGGTGGGGAACAGCAGCCAGGACCTGCAGCTGGAGGTGC ACGTTCCCCCTCAGATTGCCGGTCCCCGGGAGCCTCCCACACAAGTCTCTGTGGTCCAGGATGGAGT GGCCACTCTGGAGTGCAACGCCACAGGGAΆACCCCCTCCGACAGTGACATGGGAGCGGGACGGCCAG CCCGTGGGGGCTGAACTGGGCCTGCAGCTGCAGAACCAGGGTCAGAGCCTGCATGTGGAGCGGGCCC AGGCTGCCCACACTGGACGCTACAGCTGTGTGGCCGAGAACCTGGCTGGGAGGGCAGAGAGGAAGTT GAGCTCTCCGTACTGGTGCCCCCAGAGCTCATTGGAGACTTGGACCCGCTGACCAACATCACTGCT GCCTTGCACAGCCCCTTAACTCTGCTCTGTGAAGCCATGGGGATCCCACCTCCAGCCATCCGCTGGT TCCGAGGGGAGGAGCCTGTCAGCCCCGGGGAGGACACCTACCTGCTGGCAGGTGGCTGGATGCTGAA GATGACTCAGACACAGGAGCAAGACAGTGGCCTCTACTCATGCCTGGCAAGCAΆCGAGGCTGGGGAG GCACGGAGGAACTTCAGTGTGGAGGTGCTGGTTCCTCCCAGTATTGAGAACGAGGACTTGGAGGAGG TGATCAAGGTCCTTGATGGACAGACTGCCCATCTTATGTGCAACGTCACAGGCCACCCACAGCCCAA GCTCACATGGTTCAΆAGATGGCCGGCCTCTGGCTAGGGGAGATGCTCACCACATCTCCCCAGACGGA GTCCTCCTGCAGGTCCTCCAGGCAAACCTGTCCAGTGCTGGCCACTACTCCTGCATTGCAGCCAACG CTGTTGGGGAGAAGACCAAACACTTCCAGCTCAGTGTCCTGTTGGCTCCCACCATCCTGGGAGGGGC CGAGGACAGTGCAGATGAGGAGGTGACCGTGACTGTCAACAACCCCATCTCTCTGATCTGCGAGGCC CTGGCCTTCCCTTCCCCCAACATCACCTGGATGAAGGACGGGGCCCCGTTTGAGGCCTCCAGGAACA TCCAGCTGCTCCCAGGTACCCACGGGCTGCAGATCCTGAATGCCCAGAAGGAAGATGCTGGCCAGTA CACCTGCGTGGTCACCAATGAGCTCGGGGAGGCCGTGAAAΆACTACCATGTGGAAGTGCTCATCCCC CCTTCCATCTCCAAAGACGACCCCTTGGCGGAGGTCGGCGTGAAGGAGGTGAAGACCAAGGTCAACA GCACCTTGACCTTGGAGTGTGAGAGCTGGGCTGTGCCCCCGCCCACCATCCGCTGGTACAAGGATGG ACAGCCCGTGACCCCCAGCTCGCGGCTGCAGGTCCTGGGTGAAGGGCGACTGCTCCAGATCCAGCCC ACACAGGTCTCAGACTCGGGGCGGTACCTGTGTGTGGCCACCAATGTGGCTGGCGAGGACGACCAGG ACTTCAACGTGCTCATCCAGGTGCCCCCCATGTTCCAGAAGGTGGGTGATTTCAGTGCAGCCTTCGA GATCCTGTCCCGGGAGGAGGAGGCCCGGGGCGGAGTCACGGAATACAGGGAGATCGTGGAGAACAAC CCAGCCTACCTGTACTGCGACACCAACGCGATCCCACCCCCGGACCTCACCTGGTACAGAGAGGATC AGCCCCTCTCGGCCGGGGATGAGGTGTCTGTGCTGCAAGGAGGCCGGGTCCTGCAGATCCCCCTGGT GCGGGCAGAGAACGCCGGGAGGTACTCGTGCAAGGCCTCCAACGAGGTGGGCGAGGACTGGCTGCAC TACGAGCTGCTGGTGCTGACCCCACCTGTGATCCTGGGTGACACAGAGGAGCTGGTGGAAGAGGTGA CAGTCAATGCCAGCAGCACCGTCAGCCTGCAGTGCCCGGCCCTGGGAAACCCCGTGCCCACCATCTC ATGGCTCCAGAATGGGCTGCCTTTCTCCCCGAGCCCACGGCTGCAGGTCCTGGAGGACGGGCAAGTC TTGCAGGTTTCCACGGCAGAGGTGGCCGACGCCGCCAGCTACATGTGTGTGGCCGAGAACCAGGCGG GCTCCGCTGAGAAGCTCTTCACCCTCAGGGTTCAAGGCCTGGACTTGGAGCAGGTCACTGCCATCCT CAACAGCAGCGTCTCCCTCCCTTGCGACGTCCACGCTCACCCAAACCCCGAGGTCACGTGGTACAAG GACAGCCAGGCCCTCTCCCTGGGTGAAGAGGTCTTCCTCCTGCCTGGCACCCACACGCTGCAGCTGG GGAGAGCACGGCTGTCGGACTCCGGGATGTACACATGCGAAGCCCTCAATGCTGCCGGCCGAGACCA GAAGCTGGTGCAGCTCAGTGTTCTGGTTCCCCCGGCCTTCAGGCAGGCTCCCAGAGGTCCCCAGGAT GCGGTCCTGGTGAGGGTCGGGGACAAAGCTGTCCTGAGCTGCGAGACAGATGCGCTCCCTGAGCCAA CTGTGACCTGGTACAAGGATGGGCAGCCCCTGGTCCTGGCACAGCGGACCCAGGCTCTGCGGGGTGG GCAGAGGCTGGAGATCCAGGAAGCCCAGGTATCGGATAAAGGTTTATACAGCTGTAAAGTCAGCAAC GTGGCTGGGGAGGCCGTGCGGACCTTCACCCTCACCGTCCAGGTGCCCCCAACATTTGAGAACCCCA AGACAGAGACAGTGAGCCAGGTGGCTGGGAGCCCCCTGGTCCTGACCTGTGATGTGTCCGGGGTCCC TGCACCCACGGTCACTTGGCTGAAGGACAGGATGCCTGTGGAGAGCAGCGCGGTGCACGGTGTGGTC TCCCGGGGGGGCCGCCTCCAGCTGAGCCGCCTGCAACCGGCCCAGGCGGGCACCTACACGTGCGTGG CTGAGAACACCCAGGCTGAGGCCCGCAAGGACTTCGTGGTAGCAGTGCTGGTGGCCCCCCGGATCCG GAGCTCGGGCGTGGCGCGGGAGCACCATGTCTTGGAAGGGCAGGAGGTGCGGCTGGACTGTGAGGCC GATGGGCAGCCGCCGCCGGACGTGGCCTGGCTGAAGGACGGCAGCCCGCTGGGCCAGGACATGGGCC CCCACCTCCGGTTCTACCTGGACGGCGGCTCCCTGGTGCTAAAAGGCCTGAGGGCCTCGGACGCGGG TGCCTACACCTGCGTGGCCCACAACCCAGCCGGGGAGGACGCCAGGCTGCACACGGTGAATGTGCTG GTTCCTCCCACCATCAAGCAGGGAGCAGACGGCTCGGGGACCCTGGTGAGCAGGCCTGGGGAGCTGG TGACCATGGTGTGCCCTGTGCGGGGCTCCCCGCCCATCCACGTGAGCTGGCTCAAGGACGGCCTGCC CCTCCCGCTCTCCCAGCGCACCCTCCTCCACGGCTCTGGCCACACCCTCAGGATTTCCAAGGTGCAA TTGGCAGACGCTGGCATCTTCACCTGTGTGGCCGCAAGCCCAGCTGGCGTGGCGGACAGGAACTTCA CCTTGCAGGTGCAGGTGCCCCCTGTCCTGGAGCCGGTGGAGTTCCAGAATGACGTGGTGGTGGTTCG TGGCTCCCTGGTGGAACTCCCGTGCGAGGCCCGGGGCGTTCCCCTGCCTCTCGTGTCGTGGATGAAG GATGGGGAACCCTTGTTGTCCCAGAGCCTCGAGCAGGGGCCCAGCCTGCAGCTGGAGGCAGTGGGAG CTGGTGACTCGGGGACCTACTCCTGTGTGGCCGTGAGCGAGGCGGGGGAAGCCAGGAGGCATTTCCA GCTGACCGTCATGGAGCCCCCTCACATTGAGGACTCAGGCCAGCCTACAGAGCTGTCGCTGACCCCC GGCGCCCCCATGGAGCTCCTCTGTGATGCCCAGGGCACCCCCCAGCCCAACATCACCTGGCATAAGG ACGGGCAGGCCCTGACCAGGCTGGAGAACAACAGCAGAGCCACACGGGTGCTCCGGGTGGAGAATGT GCAGGTTAGGGATGCTGGGCTGTACACTTGTCTGGCTGAAAGCCCTGCAGGTGCAATTGAGAAGAGC TTCCGGGTCAGGGTTCAAGCCCCTCCAAACATTGTTGGGCCCCGAGGCCCCCGCTTTGTGGTCGGCC TGGCCCCAGGGCAGCTGGTCCTGGAGTGTTCGGTGGAGGCAGAGCCAGCGCCCAAGATCACGTGGCA CCGAGACGGCATTGTGCTGCAGGAGGACGCCCACACACAATTCCCGGAGCGGGGCAGGTTCCTCCAG CTGCAGGCCCTGAGCACGGCTGACAGCGGCGACTACAGCTGCACAGCCCGCAACGCCGCAGGCAGCA CTAGTGTCGCCTTCCGCGTGGAGATCCACACGGTGCCCACCATCCGGTCAGGACCACCTGCAGTGAA CGTCTCAGTGAACCAGACAGCCCTGCTGCCTTGCCAGGCCGACGGCGTGCCCGCACCCCTCGTGAGC TGGCGGAAGGACAGGGTCCCCCTGGATCCCAGGAGCCCCAGGGCAACCCCCATCCATTCTAGGTTTG AAATTCTGCCTGAGGGTTCCCTGAGAATCCAGCCAGTCCTTGCCCAGGACGCCGGCCACTACCTCTG CCTGGCATCCAACTCTGCTGGCTCCGATCGTCAAGGCCGTGACCTACGGGTCTTGGAGCCTCCAGCC ATCGCCCCCAGCCCCTCCAACCTGACCCTGACCGCCCACACCCCAGCCTTGCTGCCCTGCGAGGCCA GCGGCTCCCCTAAGCCCCTGGTGGTCTGGTGGAAGGACGGACAGAAGCTGGACTTCCGCCTGCAGCA GGGCGCCTACCGGCTCCTGCCCTCCAACGCCCTGCTCCTCACGGCCCCCGGCCCCCAGGACTCAGCC CAGTTTGAATGCGTGGTGAGCAATGAGGTGGGCGAGGCCCACAGGCTCTACCAGGTGACCGTCCATG TGCCTCCCACCATTGCCGATGACCAGACAGACTTCACCGTGACCATGATGGCACCTGTGGTCCTCAC ATGTCACAGCACGGGTATACCAGCTCCGACCGTGTCCTGGAGCAAGGCAGGCGCCCAGCTAGGAGCT CGGGGGAGTGGCTATCGTGTCTCACCATCGGGCGCCCTGGAGATCGGGCAGGCCCTCCCCATCCACG CAGGCCGCTACACCTGCTCAGCCCGCAACTCTGCCGGCGTAGCCCACAAGCACGTCTTCCTCACTGT GCAAGCCTCCCCGGTGGTGAAGCCGCTGCCCAGCGTGGTTCGGGCAGTGGCAGAGGAGGAGGTGCTG CTGCCCTGCGAGGCCTCAGGCATCCCCCGGCCGACCATCACCTGGCAGAAGGAAGGGCTCAACGTCG CTACTGGAGTGAGTACCCAGGTCCTACCAGGCGGACAGCTGCGGATTGCCCATGCCAGCCCAGAGGA TGCTGGAAACTATCTCTGCATCGCTAAGAACAGTGCGGGCAGTGCCATGGGGAAGACGCGGCTGGTG GTGCAAGTCCCACCAGTGATCGAGAATGGCCTCCCAGACCTGTCCACCACCGAAGGCTCCCACGCCT TCTTGCCTTGCAAGGCGAGGGGCAGTCCTGAGCCCAACATCACCTGGGACAAAGATGGCCAGCCTGT GTCGGGCGCCGAGGGGAAGTTCACCATCCAGCCTTCTGGGGAGTTGCTGGTGAAGAACTTGGAGGGC CAGGACGCAGGCACCTATACCTGTACCGCTGAGAACGCCGTGGGCCGGGCCCGCCGCCGCGTGCACC TCACCATCCTGGTACTGCCTGTGTTCACCACCCTGCCTGGGGACCGCAGCCTGCGCCTTGGGGACAG GCTGTGGCTTCGCTGTGCAGCCCGGGGCAGCCCCACCCCTCGCATTGGCTGGACTGTCAACGACCGG CCAGTCACAGAAGGGGTGTCTGAGCAGGATGGAGGCAGCACGCTGCAGCGGGCCGCTGTCTCCAGAG AAGACAGCGGGACCTATGTCTGCTGGGCGGAGAACAGAGTGGGCCGCACGCAGGCGGTCAGCTTCGT CCACGTGAAGGAGGCTCCTGTCCTACAAGGGGAGGCTTTCTCCTACCTGGTGGAACCTGTAGGAGGC AGCATTCAGCTAGACTGTGTGGTGCGTGGAGACCCAGTGCCGGACATCCACTGGATCAAAGATGGCC TTCCACTGCGGGGCAGCCACCTCCGGCACCAGCTGCAGAATGGCTCGCTGACCATCCGCAGGACTGA GGCAAGGCGGGGCCTGGCACCTTGGAGGGACGATGCGGGACGGTACCAGTGCCTGGCAGAGAATGAG ATGGGCGTGGCGAAGAAAGTGGTGATCCTCGTCCTGCAGACCAGGATGGTGCCAGCAGAGCCCCACT TGAAGCGCCAACTCCCACCGATCCCCAGCAATAATGAGGCACCCTCCCTGTTCCCGGGTGTCCATGG AGGCCACGTGGGGAACCCGGACTTCCACTCTCATCTAGCAGAAGTTCTCGCCGTTCAGTTGCTGGCT GGGTCCCTGCTCTTCTCAGCCAGGGCCATGCCGCAGGCCAGCACAGCAGCCATTTCCCTTTTGGCTC CTACCAGTTTTGCCCCTTTTCCTGATGATATTTCTCAGGGCATACTTTCATCCTCTACTGCACATCA AGGCAGCCCCCAGGGGTGGCAAAAGCTGCTGTTTTTCACAGCCATCCCTAATAAAACCACTGTGATG GTCACGGTGGAGCCCCAGGACATGACAGTGAGATCTGGGGATGACGTGGCCCTGCGGTGCCAGGCCA CTGGAGAGCCCACACCCACCATTGAATGGCTACAGGCGGGTCAACCCTTGCGGGCCAGCCGGCGGCT CCGGACCCTGCCCGATGGGAGCCTGTGGCTGGAGAACGTGGAGACTGGGGATGCAGGCACCTACGAC TGCGTCGCTCACAACCTCCTGGGCTCTGCCACAGCCCGGGCGTTCCTGGTCTGTGCCAGCCACGCCA TCGTGGGCTCCCGGCATTTCAGAGACCCACAGGTCTTCTGTGAGTTTGTGGTCCCGCCTCCTCATTT TACAGGGGAGCCCCAGGGGAGCTGGGGCAGCATGACTGGGGTGATAAATGGCCGGAAATTTGGCGTG GCCACACTCAACACCAGCGTGATGCAGGAGGCACACTCCGGGGTCAGCAGCATCCACAGCAGCATCC GCCATGTCCCAGCAAACGTGGGGCCTCTGATGCGGGTGCTCGTGGTCACCATCGCCCCCATCTACTG GGCCCTGGCCAGAGAGAGTGGGGAAGCCCTGAATGGCCACTCTCTGACTGGGGGCAGGTTCCGGCAG GAGTCACACGTGGAGTTTGCTACAGGGGAGCTGCTCACGATGACCCAGGTGGCCCGGGGTCTGGATC CCGATGGCCTCCTGCTCCTCGACGTGGTGGTCAATGGCGTTGTCCCCGAGAGCCTGGCTGACGCAGA TCTTCAAGTGCAGGACTTTGAGGAGCACTACGTGCAAACAGGGCCTGGCCAGCTGTTCGTGGGCTCC ACACAGCGCTTCTTCCAGGGCGGCCTCCCCTCGTTCCTACGCTGCAACCACAGCATCCAGTACAACG CGGCCCGGGGCCCCCAGCCCCAGCTGGTGCAGCACCTGCGGGCCTCAGCTATCAGCTCGGCCTTTGA TCCAGAGGCCGAGGCCCTGCGCTTCCAGCTCGCTACAGCCCTGCAGGCGGAGGAGAACGAGGTCGGC TGCCCCGAGGGCTTTGAGCTGGACTCCCAGGGAGCGTTTTGTGTGGACAGGGACGAGTGCTCAGGAG GCCCTAGCCCCTGCTCCCATGCCTGCCTTAATGCACCCGGCCGCTTCTCCTGCACCTGCCCCACTGG CTTCGCCCTGGCCTGGGATGACAGGAACTGCAGAGATGTGGACGAGTGTGCGTGGGATGCTCACCTC TGCCGAGAGGGACAGCGCTGTGTGAACCTGCTCGGGTCCTACCGCTGCCTCCCCGACTGTGGGCCTG GCTTCCGGGTGGCTGATGGGGCCGGCTGTGAAGATGTGGACGAATGCCTGGAGGGGTTGGACGACTG TCACTACAACCAGCTCTGCGAGAACACCCCAGGCGGTCACCGCTGCAGCTGCCCCAGGGGTTACCGG ATGCAGGGCCCCAGCCTGCCCTGCCTAGATGTCAATGAGTGCCTGCAGCTGCCCAAGGCCTGCGCCT ACCAGTGCCACAACCTCCAGGGCAGCTACCGCTGCCTGTGCCCCCCAGGCCAGACCCTCCTTCGCGA CGGCAAGGCCTGCACCTCACTGGAGCGGAATGGACAAAATGTGACCACCGTCAGCCACCGAGGCCCT CTATTGCCCTGGCTGCGGCCCTGGGCCTCGATCCCCGGTACCTCCTACCACGCCTGGGTCTCTCTCC GTCCGGGTCCCATGGCCCTGAGCAGTGTGGGCCGGGCCTGGTGCCCTCCTGGTTTCATCAGGCAGAA CGGAGTCTGCACAGACCTTGACGAGTGCCGCGTGAGGAACCTGTGTCAGCACGCCTGCCGCAACACT GAGGGCAGCTACCAGTGCCTGTGCCCCGCCGGCTACCGTCTGCTCCCCAGCGGGAAGAACTGCCAGG ACATCAACGAGTGCGAGGAGGAGAGCATCGAGTGTGGACCCGGCCAGATGTGCTTCAACACCCGTGG CAGCTACCAGTGTGTGGACACACCCTGTCCTGCCACCTACCGGCAGGGCCCCAGCCCTGGGACGTGC TTCCGGCGCTGCTCGCAGGACTGCGGCACGGGCGGCCCCTCTACGCTGCAGTACCGGCTGCTGCCGC TGCCCCTGGGCGTGCGCGCCCACCACGACGTGGCCCGCCTCACCGCCTTCTCCGAGGTCGGCGTCCC CGCCAACCGCACCGAGCTCAGCATGCTGGAGCCCGACCCCCGCAGCCCCTTCGCGCTGCGTCCGCTG CGCGCGGGCCTTGGCGCGGTCTACACCCGTCGCGCGCTCACCCGCGCCGGCCTCTACCGGCTCACCG TGCGTGCTGCGGCACCGCGCCACCAAAGCGTCTTCGTCTTGCTCATCGCCGTGTCCCCCTACCCCTA CTAA
Variant sequences of NOV9 are included in Example 3, Table 21. A variant sequence can include a single nucleotide polymorphism (SNP). A SNP can, in some instances, be referred to as a "cSNP" to denote that the nucleotide sequence containing the SNP originates as a cDNA.
The NOV9 protein (SEQ ID NO: 18) encoded by SEQ ID NO: 17 is 3931 amino acid residues in length and is presented using the one-letter amino acid code in Table 9B. Psort analysis predicts the NOV9 protein of the invention to be localized at the plasma membrane with a certainty of 0.7300.
Table 9B. Encoded NOV9 protein sequence (SEQ ID NO:18) SALFAAVYQMLKPRLVHNSPHPVTYQIEASLKPEQPGVTLVSIPVFLAPSWHKASELIPTQSFR AQGAGKQLLGSPCPQVPPSIREDGRKANVSGMAGQSLTLECDANGFPVPEIVWL DAQLIPKVGG HRLLDEGQSLHFPRIQEGDSGLYSCRAENQAGTAQRDFHLLVLTPPSVLGAGAΆQEVLGLAGADV ELQC TSGVPTPQVE TKDRQPVLPGGPHLQVQEDGQVLRITGSHVGDEGRYQCVAFSPAGQQAR DFQLRVHAPPTI GSNETGEVAVMEDHLVQLLCEARGVPTPNIT FKDGALLPTSTKVVYTRGGR QLQLGRAQSSDAGVYTCKASNAVGAAEKATRLDVYVPPTIEGAGGRPYWKAVAGRPVALECVAR GHPSPTLS HHEGLPVAESNESRLETDGSVLRLESPGEASSGLYSCVASSPAGEAVLQYSVEVQV PPQLLVAEGLGQVTTIVGQPLELPCQASGSPVPTIQ LQNGRPAEELAGVQVASQGTTLHIDHVE LDHSGLFACQATNEAGTAGAEVEVSVHEFPSVSIIGGENITAPFLQPVTLQCIGDGVPTPSLRWW DGVALAAFGGNLQIEKVDLRDΞGIYTCAATNLAGESKREVALKVLVPPNIEPGPVNKAVLENAS VTLΞCLASGVPPPDVSWFKGHQPVSSWMGVTVSVDGRVLRIEQAQLSDAGSYRCVASNVAGSTEL RYGLRVNVPPRITLPPSLPGPVLVNTPVRLTCNATGAPSPTLM LKDGMPVSPAGTPGLQVFPGG VLTLASARASDSGRYSCVAVSAVGEDRQDWLQVHMPPSILGEEL VSWA ESVALECQSHAM PPVLSW KDGRPLEPRPGVHLSADKALLQVDRADV DAGHYTCEALNQAGHSEKHYWLNVWGQP LPGEGAGLQHVSAVGRLLYLGQAQLAQEGTYTCECS WGNSSQDLQLEVHVPPQIAGPREPPTQ SWQDGVATLECNATGKPPPTVTWERDGQPVGAELGLQLQWQGQSLHVΞRAQAAHTGRYSCVAE NLAGRAERKFΞLSVLVPPΞLIGDLDPLTNITAALHSPLTLLCEAMGIPPPAIRWFRGEΞPVSPGE DTYLLAGGWMLKMTQTQEQDSGLYSCLASNEAGΞARRNFSVEVLVPPSIENEDLEEVIKVLDGQT AHLMCNVTGHPQPKLT FKDGRPLARGDAHHISPDGVLLQVLQANLSSAGHYSCIAA AVGEKTK HFQLSVLLAPTILGGAEDSADEEVTVTVNNPISLICEALAFPSPNITWMKDGAPFEASR IQLLP GTHGLQILNAQKEDAGQYTCWTNELGEAVKNYHVEVLIPPSISKDDPLAEVGVKEVKTKV STL TLECESWAVPPPTIR YKDGQPVTPSSRLQVLGΞGRLLQIQPTQVSDSGRYLCVATNVAGEDDQD FNVLIQVPPMFQKVGDFSAAFΞILSREEEARGGVTEYREIVENNPAYLYCDTNAIPPPDLT YRE DQPLSAGDEVSVLQGGRVLQIPLVRAENAGRYSCKASNEVGED LHYELLVLTPPVILGDTEΞLV EEVTV ASSTVSLQCPALGNPVPTISWLQNGLPFSPSPRLQVLEDGQVLQVSTAEVADAASYMCV AENQAGSAEKLFTLRVQGLDLEQVTAILNSΞVSLPCDVHAHPNPEVT YKDSQALSLGEEVFLLP GTHTLQLGRARLSDSG YTCEALNAAGRDQKLVQLSVLVPPAFRQAPRGPQDAVLVRVGDKAVLS CETDALPEPTVTWYKDGQPLVLAQRTQALRGGQRLΞIQEAQVSD GLYSCKVSWVAGEAVRTFTL TVQVPPTFENPKTETVSQVAGSPLVLTCDVSGVPAPTVT LKDRMPVESSAVHGWSRGGRLQLS LQPAQAGTYTCVAENTQAEARKDFWAVLVAPRIRSSGVAREHHVLEGQEVRLDCEADGQPPPD VA LKDGSPLGQDMGPHLRFYLDGGSLVLKGLRASDAGAYTCVAH PAGEDARLHTVNVLVPPTI KQGADGSGTLVSRPGELVTMVCPVRGSPPIHVS LKDGLPLPLSQRTLLHGSGHTLRISKVQLAD AGIFTCVAASPAGVADRNFTLQVQVPPVLEPVEFQNDVWVRGSLVELPCEARGVPLPLVS MKD GEPLLSQSLEQGPSLQLEAVGAGDSGTYSCVAVSEAGEARRHFQLTVMEPPHIEDSGQPTELSLT PGAPMELLCDAQGTPQPNITWHKDGQALTRLΞNNSRATRVLRVENVQVRDAGLYTCLAESPAGAI EKSFRVRVQAPPNIVGPRGPRFWGLAPGQLVLECSVEAEPAPKIT HRDGIVLQEDAHTQFPER GRFLQLQALSTADSGDYSCTARNAAGSTSVAFRVEIHTVPTIRSGPPAVNVSVNQTALLPCQADG VPAPLVS R DRVPLDPRSPRATPIHSRFEILPEGSLRIQPVLAQDAGHYLCLASNSAGSDRQGR DLRVLEPPAIAPSPSNLTLTAHTPALLPCEASGSPKPLWW KDGQ LDFRLQQGAYRLLPSNAL LLTAPGPQDSAQFECWSNEVGEAHRLYQVTVHVPPTIADDQTDFTVTMMAPWLTCHSTGIPAP TVSWSKAGAQLGARGSGYRVSPSGALEIGQALPIHAGRYTCSARNSAGVAH HVFLTVQASPWK PLPSWRAVAEEEVLLPCEASGIPRPTIT QKEGLNVATGVSTQVLPGGQLRIAHASPEDAGNYL CIAKNSAGSAMGKTRLWQVPPVIENGLPDLSTTEGSHAFLPCKARGSPEPNITWDKDGQPVSGA EGKFTIQPSGELLVKNLEGQDAGTYTCTAENAVGRARRRVHLTILVLPVFTTLPGDRSLRLGDRL WLRCAARGSPTPRIG TVNDRPVTEGVSEQDGGSTLQRAAVSREDSGTYVC AENRVGRTQAVSF VHVKEAPVLQGEAFSYLVEPVGGSIQLDCWRGDPVPDIHWIKDGLPLRGSHLRHQLQNGSLTIR RTEARRGLAPWRDDAGRYQCLAENEMGVAKKWILVLQTRMVPAEPHLKRQLPPIPSNNEAPSLF PGVHGGHVGNPDFHSHLAEVLAVQLLAGSLLFSARAMPQASTAAISLLAPTSFAPFPDDISQGIL SSSTAHQGSPQG QKLLFFTAIPNKTTVMVTVEPQDMTVRSGDDVALRCQATGEPTPTIE LQAG QPLRASRRLRTLPDGSL LENVETGDAGTYDCVAHNLLGSATARAFLVCASHAIVGSRHFRDPQV FCEFWPPPHFTGEPQGS GSMTGVINGRKFGVATLNTSVMQEAHSGVSSIHSSIRHVPANVGPL MRVLWTIAPIY ALARESGEALNGHSLTGGRFRQESHVEFATGELLTMTQVARGLDPDGLLLLD VW GWPESLADADLQVQDFEEHYVQTGPGQLFVGSTQRFFQGGLPSFLRCNHSIQYNAARGPQ PQLVQHLRASAISSAFDPEAEALRFQLATALQAEENEVGCPEGFELDSQGAFCVDRDECSGGPSP CSHACLNAPGRFSCTCPTGFALA DDRNCRDVDECAWDAHLCREGQRCVNLLGSYRCLPDCGPGF RVADGAGCEDVDECLEGLDDCHYNQLCENTPGGHRCSCPRGYRMQGPSLPCLDVNECLQLPKACA YQCH LQGSYRCLCPPGQTLLRDGKACTSLERNGQNVTTVSHRGPLLP LRP ASIPGTSYHA V SLRPGPMALSSVGRAWCPPGFIRQNGVCTDLDECRVRNLCQHACRNTΞGSYQCLCPAGYRLLPSG KNCQDINECEEESIECGPGQMCFNTRGSYQCVTTPCPATYRQGPSPGTCFRRCSQDCGTGGPSTL QYRLLPLPLGVRAHHDVARLTAFSEVGVPANRTELSMLEPDPRSPFALRPLRAGLGAVYTRRALT RAGLYRLTVRAAAPRHQSVFVLLIAVSPYPY
A search against the Patp database, a proprietary database that contains sequences published in patents and patentpublications, yielded several homologous proteins shown in Table 9C. Table 9C. Patp results for NOV9
Smallest Sum
Reading High Prob Sequences producing High-scoring Segment Pairs: Frame Score P(N)
>patp:AAY53667 Sequence gi/3328186 +1 1529 6.5e-244
>patp:AAY87 06 Human secreted protein sequence ID NO: 245 +1 2235 6.4e-230
>patp:AAE06183 Human gene 57 encoded secreted protein +1 2235 6.4e-230
>patp:AAY87120 Human secreted protein sequence SEQ ID: 159 +1 2235 6.4e-230
>patp:AAE06097 Human gene 57 secreted protein HRACD80 +1 2235 6.4e-230
In a BLAST search of public sequence databases, it was found, for example, that the nucleic acid sequence of this invention has 625 of 1067 bases (58%) identical to a gb:GENBANK-ID:HSLTGFBP4|acc:Y13622.1 mRNA from Homo sapiens (mRNA for latent transforming growth factor-beta binding protein-4). The full amino acid sequence of the protein of the invention was found to have 502 of 1665 amino acid residues (30%) identical to, and 767 of 1665 amino acid residues (46%) similar to, the 5198 amino acid residue ρtnr:SPTREMBL-ACC:O76518 protein from Caenorhabditis elegans (HEMICENTIN PRECURSOR).
NOV9 also has homology to the proteins shown in the BLASTP data in Table 9D.
Table 9D. BLAST results for NOV9
Gene Index/ Protein/ Organism Length Identity Positives Expect Identifier (aa) (%) (%) gi 114575679 | gb | AAK6 hemicentin 5636 1230/3017 1785/3017 0.0
8690.l|AF156100_l [Homo sapiens] (40%) (58%)
(AF156100) gi 118547943 | ref |XP_ hemicentrin 3645 979/2379 1413/2379 0.0 053531.31 (XM 053531 [Homo sapiens] (41%) (59%) gi 117568539 I ref |NP_ Ig superfamily 5175 857/3077 1348/3077 0.0 509636.11 (NM_077235 repeats (I-type) (27%) (42%) ) [CaenorhaJbdi tis elegans] gi 1175685411 ref |NP_ IG 5198 857/3077 1348/3077 0.0 509635. ll (NM 077234 (immunoglobulin) (27%) (42%) superfamily (47 domains)
[ Caenorhabdi ti s elegans] gi 113872813 | emb| CAC fibulin-6 2673 552/1399 796/1399 0.0 37630. l| (AJ306906) [Homo sapiens] (39%) (56%)
A multiple sequence alignment is given in Table 9E, with the NOV9 protein being shown on line 1 in Table 9E in a ClustalW analysis, and comparing the NOV9 protein with the related protein sequences shown in Table 9D. This BLASTP data is displayed graphically in the ClustalW in Table 9E. Table 9E. ClustalW Analysis of NOV9
1) > N0V9; SEQ ID NO: 18
2) > gi| 14575679|/ hemicentin [Homo sapiens]; SEQ ID NO:72
3) > gij 18547943|/ hemicentrin [Homo sapiens]; SEQ ID NO:73
4) > gijl7568539J/ Ig superfamily repeats (I-type) [Caenorhabditis elegans]; SEQ ID NO:74
5) > gij 1756854 lj/ IG (immunoglobulin) superfamily (47 domains) [Caenorhabditis elegans]; SEQ ID NO:75
6) > gi|13872813|/ fibulin-6 [Homo sapiens]; SEQ ID NO:76
4090 4100 4110 4120 4130 4140
Figure imgf000069_0001
4150 4160 4170 4180 4190 4200
Figure imgf000069_0002
4210 4220 4230 4240 4250 4260
Figure imgf000069_0003
4270 4280 4290 4300 4310 4320
Figure imgf000069_0004
4390 4400 4410 4420 4430 4440
Figure imgf000069_0005
4450 4460 4470 4480 4490 4500
NOV9 2784 T^^GQPV AE^FJSl' J s^-jJL ^ EGpl^TggTgEEvgR RRRgHgglL^j 2842
Figure imgf000070_0001
4510 4520 4530 4540 4550 4560
Figure imgf000070_0002
4570 4580 4590 4600 4610 4620
Figure imgf000070_0003
4630 4640 4650 4660 4670 4680
Figure imgf000070_0004
4690 4700 4710 4720 4730 4740
Figure imgf000070_0005
4750 4760 4770 4780 4790 4800
Figure imgf000070_0006
4810 4820 4830 4840 4850 4860
Figure imgf000070_0007
4870 4880 4890 4900 4910 4920
Figure imgf000070_0008
4930 4940 4950 4960 4970 4980 NOV9 3155 D MTVRSBDDVALi3c ?ATGEJa THTI 3179 gi 114575679 I 4646 WSAWQPWGTCSESCgKGTQTgARLCjS g AF 4678 gij 18547943 j 2655 SAWpP GTCgESCgKGTQTgARLCgNg P AF 2687 gijl7568539J 4079 RNGNRVETGVQGVRY DGRMLTIIEARSLDSGIYLCSATGEAGSAQQAYTLEVLVS KI 4138 gij 17568541 j 4079 RNGNRVETGVQGVRYVTDGRMLTIIEARSLDSGI LCSATBEAGSAQQAYTLEVLVS KI 4138 gi 1 13872813 I 1683 WSAWpPWG ESCgKGTQTJgA C jE AF 1715
4990 5000 5010 5020 5030 5040
NOV9 3179 -- LQAGQPLRASRgLl LPDGSL LEN 3206 gijl4575679| 4679 GG SYCDGAETQMQVCNEF ΪPIHgKWATW2 SACSVSCGGfflAaQg RHCSDPVPQYGGR 4738 gij 18547943 j 2688 GG SYCDGAETQMQVCNEΪ «PIH@KWATW2 SACSVSCGGjAgQg RSCSDPVPQYGGR 2747 gijl7568539J 4139 IT TPGVLTPSSGSKFSLPlAVRgYPDPII! jJLNGNDIKDjENG] ISADGTLHIEKAE 4198 gij 17568541 j 4139 iτ] TPGVLTPSSGSKFSLp"' '" TLNGNDIKDϊENGH IBADGTLHIΞKAE 4198 gij 13872813] 1716 GG§ YCDGAETQMQVCNE: SACSVSCGGgAgQ] CSDPVPQYGGR 1775
5050 5060 5070 5080 5090 5100
Figure imgf000071_0001
5110 5120 5130 5140 5150 5160
Figure imgf000071_0002
5170 5180 5190 5200 5210 5220
Figure imgf000071_0003
5230 5240 5250 5260 5270 5280
Figure imgf000071_0004
5290 5300 5310 5320 5330 5340
Figure imgf000071_0005
5410 5420 5430 5440 5450 5460
Figure imgf000072_0001
5470 5480 5490 5500 5510 5520
Figure imgf000072_0002
5530 5540 5550 5560 5570 5580
Figure imgf000072_0003
5590 5600 5610 5620 5630 5640
Figure imgf000072_0004
5650 5660 5670 5680 5690 5700
Figure imgf000072_0005
5710 5720 5730 5740 5750 5760
Figure imgf000072_0006
5830 5840 5850 5860 5870 5880
Figure imgf000072_0007
gi 1175685411 4976 TG-Efc HFAMNPHTRlgEgLjβj gij 13872813 j 2451 RTIjgKTg" βSEA- -S DTB SIB
5890 5900 5910 5920 5930 5940
Figure imgf000073_0001
5950 5960 5970 5980 5990 6000
Figure imgf000073_0002
The NOV9 Clustal W alignment shown in Table 9E was modified to begin at amino residue 4080. The data in Table 9E includes all of the regions overlapping with the NOV9 protein sequences.
The presence of identifiable domains in the protein disclosed herein was determined by searches using algorithms such as PROSITE, Blocks, Pfam, ProDomain, Prints and then determining the Interpro number by crossing the domain match (or numbers) using the Interpro website (http:www.ebi.ac.uk interpro ). Table 9F lists the domain description from DOMAIN analysis results against NOV9.
Figure imgf000073_0003
Figure imgf000074_0001
Consistent with other known members of the Hemicentin Precursor-like family of proteins, NON9 has, for example, thirty-three immunoglobulin (ig) signature sequences and four epidermal growth factor (EGF) signature sequences, as well as homology to other members of the Hemicentin Precursor-like Protein Family. ΝON9 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts. For example, ΝON9 nucleic acids and polypeptides can be used to identify proteins that are members of the Hemicentin Precursor-like Protein Family. The ΝON9 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance ΝON9 activity or function. Specifically, the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular differentiation, and signal transduction. These molecules can be used to treat, e.g., Cardiovascular diseases, Hyperparathyroidism, Hypoparathyroidism, Lymphedema, Allergies as well as other diseases, disorders and conditions..
In addition, various ΝON9 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. For example, the ΝON9 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the Hemicentin Precursor-like Protein Family.
Hemicentrin is an extracellular matrix protein with a modular sturcture. Like ΝON9, the hemicentrin structure includes many immunoglobulin domains flanked by EGF domains. The protein is likely involved in cellular differentiation of epithelial tissue. The NON9 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of cardiac, immune and endocrine physiology. As such, the ΝON9 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat cardiovascular, immune, and endocrine disorders, e.g., Cardiovascular diseases,
Hyperparathyroidism, Hypoparathyroidism, Lymphedema, Allergies as well as other diseases, disorders and conditions.
The ΝON9 nucleic acids and polypeptides are useful for detecting specific cell types. For example, expression analysis has demonstrated that a ΝON9 nucleic acid is expressed in Adipose, Thyroid, Colon, Lymph node, Bone, Myometrium, Prostate, Testis, Aorta, Nein.
Additional utilities for ΝON9 nucleic acids and polypeptides according to the invention are disclosed herein.
ΝOV10 A ΝON10 polypeptide has been identified as a Selectin-like protein. The novel ΝON10 nucleic acid sequences maps to the chromosome 9. Two alternative novel ΝOV10, ΝONlOa and ΝONlOb, nucleic acids and encoded polypeptides are provided.
ΝOVlOa A ΝON10 variant is ΝONlOa (alternatively referred to herein as CG94661-01), which includes the 1268 nucleotide sequence (SEQ D ΝO:19) shown in Table 10A. A NOVlOa ORF begins with a ATG initiation codon at nucleotides 145-147 and ends with a TGA codon at nucleotides 871-873. Putative untranslated regions upstream from the initiation codon and downstream from the termination codon are underlined in Table 10A, and the start and stop codons are in bold letters.
Table 10A. NOVlOa Nucleotide Sequence (SEQ ID NO:19)
GCGGCCGCCACCCTCCGTGGCAAGGCGAGGCCCCGGGGGCGGGCCGGGGTCACCACGCCTGTCCCAG GGAACCGCACAGACGGTACTCACCCTTCTTGCGATGATGTGAGATGATAAAATGCCTACATGATGAG ATGAAGTGAGATGAAAAACATAGGCCTTGTGATGGAATGGGAAATTCCAGAGATAATTTGCACGTGC GCTAAGCTGCGGCTACCCCCGCAAGCAACCTTCCAAGTCCTTCGTGGCAATGGTGCTTCCGTGGGGA CCGTGCTCATGTTCCGCTGCCCCTCCAACCACCAGATGGTGGGGTCTGGGCTCCTCACCTGCACCTG GAAGGGGAGCATCGCTGAGTGGTCTTCAGGGTCCCCAGTGTGCAAACTGGTGCCACCACACGAGACC TTTGGCTTCAAGGTGGCCGTGATCGCCTCCATTGTGAGCTGTGCCATCATCCTGCTCATGTCCATGG CCTTCCTCACCTGCTGCCTCCTCAAGTGCGTGAAGAAGAGCAAGCGGCGGCGCTCCAACAGGTCAGC CCAGCTGTGGTCCCAGCTGAAAGATGAGGACTTGGAGACGGTGCAGGCCGCATACCTTGGCCTCAAG CACTTCAACAAACCCGTGAGCGGGCCCAGCCAGGCGCACGACAACCACAGCTTCACCACAGACCATG GTGAGAGCACCAGCAAGCTGGCCAGTGTGACCCGCAGCGTGGACAAGGACCCTGGGATCCCCAGAGC TCTAAGCCTCAGTGGCTCCTCCAGCTCACCCCAAGCCCAGGTGATGGTGCACATGGCAAACCCCAGA CAGCCCCTGCCTGCCTCTGGGCTGGCCACAGGAATGCCACAACAGCCCGCAGCATATGCCCTAGGGT GACCACGCAGTGAGGCTGGTGCCCATGCTCCACACTGGGAGGCCAGGCTGACCCCACCAGCCAGTCA GCTACAACTCCACATCAACTCCACATGCGCCCAGCTCGAGACTGATGAGTGGAATCAGCTTCCAGGT GTAGGGACCCCTTGAGGGGCCGAGCTGACATCCAAGGCTGAGGACCCCAGTGGGGAGTGTTCTGTTC CGGCATATCCTGGCCGTAACGATTTTTATAGTTATGGACTACTTGAAACCACTACTGAGGGTAATTT ACTAGCTGTGGCCTCCCACTAACTAGCATTCCTTTAAAGAGACTGGGAAATGTTTTAAGCAAATCTA GTTTTGTATAATAAAATAAGAAAATAGCAATAAACTTCTTTTCAGCAACTACAAAAAAAAAA
The NOVlOa polypeptide (SEQ LD ΝO:20) encoded by SEQ ID NO:19 is 242 amino acid residues in length and is presented using the one-letter amino acid code in Table 10B. The Psort profile for the NOVlOa predicts that this peptide is likely to be localized at the plasma membrane with a certainty of 0.7000.
Table 10B. NOVlOa protein sequence (SEQ ID NO:20)
MKNIGLVME EIPEIICTCAKLRLPPQATFQVLRGNGASVGTVLMFRCPSNHQMVGSGLLTCT GS IAE SSGSPVCKLVPPHETFGFKVAVIASIVSCAIILLMSMAFLTCCLLKCVKKS RRRSNRSAQL SQLKDEDLETVQAAYLGLKHFNKPVSGPSQAHDNHSFTTDHGESTSKLASVTRSVDKDPGIPRALSL SGSSSSPQAQVMVHMANPRQPLPASGLATGMPQQPAAYALG
NOVlOb
Alternatively, a NOV10 variant is the novel NOV 10b (alternatively referred to herein as CG94661-02), which includes the 887 nucleotide sequence (SEQ ID NO:21) shown in Table IOC. NOV 10b was created by polymerase chain reaction (PCR) using the primers detailed in Example 1, Table 17. Primers were designed based on in silico predictions of the full length or some portion (one or more exons) of the cDNA/protein sequence of the invention. The PCR product derived by exon linking, covering the entire open reading frame, was cloned into the pCR2.1 vector from Invitrogen to provide clone 143260::COR100348691_extn.698976.C20.
The NOVl 0b ORF begins with a Kozak consensus ATG initiation codon at nucleotides 72-74 and ends with a TGA codon at nucleotides 1958-1960. Putative untranslated regions upstream from the initiation codon and downstream from the termination codon are underlined in Table 10C, and the start and stop codons are in bold letters.
Table IOC. NOVl 0b Nucleotide Sequence (SEQ ID NO:21)
GCACAGACGGTACTCACCCTTCTTGCGATGATGTGAGATGATAAAATGCCTACATGATGAGATGAAG TGAGATGAAAAACATAGGCCTTGTGATGGAATGGGAAATTCCAGAGATAATTTGCATGTGCGCTAAG CTGCGGCTACCCCCGCAAGCAACCTTCCAAGTCCTTCGTGGCAATGGTGCTTCCGTGGGGACCGTGC TCATGTTCCGCTGCCCCCCCAACCACCAGATGGTGGGGTCTGGGCTCCTCACCTGCACCTGGAAGGG GAGCATCGCTGAGTGGTCTTCAGGGTCCCCAGTGTGCAAACTGGTGCCACCACACGAGACCTTTGGC TTCAAGGTGGCCGTGATCGCCTCCATTGTGAGCTGTGCCATCATCCTGCTCATGTCCATGGCCTTCC TCACCTGCTGCCTCCTCAAGTGCGTGAAGAAGAGCAAGCGGCGGCGCTCCAACAGGTCAGCCCAGCT GTGGTCCCAGCTGAAAGATGAGGACTTGGAGACGGTGCAGGCCGCATACCTTGGCCTCAAGCACTTC AACAAACCCGTGAGCGGGCCCAGCCAGGCGCACGACAACCACAGCTTCACCACAGACCATGGTGAGA GCACCAGCAAGCTGGCCAGTGTGACCCGCAGCGTGGACAAGGACCCTGGGATCCCCAGAGCTCTAAG CCTCAGTGGCTCCTCCAGCTCACCCCAAGCCCAGGTGATGGTGCACATGGCAAACCCCAGACAGCCC CTGCCTGCCTCTGGGCTGGCCACAGGAATGCCACAACAGCCCGCAGCATATGCCCTAGGGTGACCAC GCAGTGAGGCTGGTGCCCATGCTCCACACTGGGAGGCCAGGCTGACCCCACCAGCCAGTCAGCTACA ACTCCACATCAACTCC
Variant sequences of NOV 10b are included in Example 3, Table 22. A variant sequence can include a single nucleotide polymorphism (SNP). A SNP can, in some instances, be referred to as a "cSNP" to denote that the nucleotide sequence containing the SNP originates as a cDNA.
The NOVlOb protein (SEQ ID NO:22) encoded by SEQ ID NO:21 is 242 amino acid residues in length and is presented using the one-letter code in Table 10D. The Psort profile for NOVl Ob predicts that this sequence is likely to be localized at the plasma membrane with a certainty of 0.7000.
Table 10D. NOVlOb protein sequence (SEQ ID NO:22)
MKNIGLVMΞWEIPEIICMCAKLRLPPQATFQVLRGNGASVGTVLMFRCPPNHQMVGSGLLTCTWKGS lAEWSSGSPVCKLVPPHETFGFKVAVIASIVSCAIILLMSMAFLTCCLLKCVKKSKRRRSNRSAQL SQLKDEDLETVQAAYLGLKHFNKPVSGPSQAHDNHSFTTDHGESTSKLASVTRSVDKDPGIPRALSL SGSSSSPQAQVMVHMANPRQPLPASGLATGMPQQPAAYALG
NOV10 Clones
Unless specifically addressed as NOVlOa or NOVlOb, any reference to NOV10 is assumed to encompass all variants. NOVlOa differs from NOVlOb at amino acid position 18 (T>M) and amino acid position 50 (S>P) as shown in Tables 10B and 10D.
A search against the Patp database, a proprietary database that contains sequences published in patents and patent publications, yielded several homologous proteins shown in Table 10E.
Table 10E. Patp results for NOV10
Smallest Sum
Reading High Prob
Sequences producing High-scoring Segment Pairs: Frame Score P(N)
>patp:AAM93054 Human digestive system antigen +1 210 7.2e-17
>patp:AAR05 94 Endothelial leukocyte adhesion molecule-1 +1 113 0.0016
>patp:AAR08116 Endothelial leucocyte adhesion molecule-1 +1 113 0.0016
>patp:AAW18839 E-selectin +1 113 0.0016
>patp:AA 46733 Endothelial leukocyte adhesion molecule-1 +1 113 0.0016
h a BLAST search of public sequence databases, it was found, for example, that the
NOVlOa nucleic acid sequence of this invention has 438 of 447 bases (97%) identical to a gb:GENBANK-ID:HSM802384|acc:AL137623.1 mRNA from Homo sapiens (cDNA DKFZp434J1812 (from clone DKFZp434J1812)). The full amino acid sequence of the protein of the invention was found to have 110 of 139 amino acid residues (79%) identical to, and 123 of 139 amino acid residues (88%) similar to, the 269 amino acid residue ptm:SPTREMBL- ACC:Q9D176 protein from Mus musculus (170001711 IRK PROTEIN).
Similarly, it was found, for example, the NOV 10b nucleic acid sequence of this invention has 438 of 447 bases (97%) identical to a gb:GENBANK-
ID:HSM802384|acc: ALl 37623.1 mRNA from Homo sapiens (cDNA DKFZp434J1812 (from clone DKFZp434J1812)). The full amino acid sequence of the protein of the invention was found to have 108 of 138 amino acid residues (78%) identical to, and 121 of 138 amino acid residues (87%) similar to, the 269 amino acid residue ptnr:SPTREMBL-ACC:Q9D176 protein from Mus musculus (1700017111PJK PROTEIN).
Additional BLAST results are shown in Table 10F.
Figure imgf000078_0001
Figure imgf000079_0002
A multiple sequence alignment is given in Table 10G, with the NOV10 protein of the mvention being shown on line 1, in a ClustalW analysis comparing NOV10 with related protein sequences disclosed in Table 10F.
Table 10G. Information for the ClustalW proteins:
1) > NOVlOa; SEQ ID NO:20
2) > NOVlOb; SEQ ID NO:22
3) > gi|1577905/ similar to RTKEN cDNA 1700017111 gene [Homo sapiens]; SEQ ID NO:77 4) > gi| 1283478/ Sushi domain (SCR repeat) containing protein-data source: Pfam, source key:
PF00084, evidence:ISS-ρutative [Mus musculus]; SEQ ID NO:78
5) > gi| 1285054/ Sushi domain (SCR repeat) containing protein-data source: Pfam, source key:
PF00084, evidence:ISS-putative [Mus musculus]; SEQ ID NO:79
6) > gi|1283897/ Sushi domain (SCR repeat) containing protein-data source: Pfam, source key: PF00084: ISS-putative [Mus musculus]; SEQ ID NO:80
7) > gi|7494498/ scavenger receptor cysteine-rich protein homolog sxctvX2-Geodia cydonium; SEQ ID
NO:81
Figure imgf000079_0001
Figure imgf000080_0001
NOVlOa
Figure imgf000080_0002
Figure imgf000080_0003
TPSSHVIPSTgDNSVGAEVSFQCEDGYTLQGEKKITCLPTQKWSANPPSC
Figure imgf000080_0004
The NOVIO Clustal W alignment shown in Table 10F was modified to begin at amino residue 1600 and end at amino acid residue 2000. The data in Table 10F includes all of the regions overlapping with the NOVIO protein sequences.
The NOVIO Clustal W alignment shown in Table 10G was modified to begin at amino residue 1601. The data in Table 10G includes all of the regions overlapping with the NOVIO protein sequences.
The presence of identifiable domains in the protein disclosed herein was determined by searches using algorithms such as PROSITE, Blocks, Pfam, ProDomain, Prints and then determining the Interpro number by crossing the domain match (or numbers) using the Interpro website (http:www.ebi.ac.uk/interpro ). Table 10H lists the domain description from DOMAIN analysis results against NOVIO.
Figure imgf000081_0001
Consistent with other known members of the Selectin-like family of proteins, NOV10 has, for example, a Sushi domain (SCR repeat) signature sequences and homology to other members of the Selectin-like Protein Family. NOV10 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts. For example, NOV10 nucleic acids and polypeptides can be used to identify proteins that are members of the Selectin-like Protein Family. The NOV10 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOV10 activity or function. Specifically, the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular adhesion and signal transduction. These molecules can be used to treat, e.g., Cardiovascular diseases, Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus , Pulmonary stenosis, Subaortic stenosis, Ventricular septal defect (VSD), valve diseases, Tuberous sclerosis, Scleroderma, Obesity, Transplantation, Von Hippel-Lindau (VHL) syndrome,
Cirrhosis, Transplantation, Diabetes, Autoimmune disease, Renal artery stenosis, Interstitial nephritis, Glomerulonephritis, Polycystic kidney disease, Systemic lupus erythematosus, Renal tubular acidosis, IgA nephropathy, Hypercalceimia, Lesch-Nyhan syndrome, Systemic lupus erythematosus, Autoimmune disease, Asthma, Emphysema, Scleroderma, allergy as well as other diseases, disorders and conditions.
In addition, various NOV10 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. For example, the NOV10 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of protems belonging to the Selectin-like Protein Family.
The NOV10 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of cardiac and immune physiology. As such, the NOV10 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat cardiovascular and immune disorders, e.g., Cardiovascular diseases, Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus , Pulmonary stenosis, Subaortic stenosis, Ventricular septal defect (VSD), valve diseases, Tuberous sclerosis, Scleroderma, Obesity, Transplantation, Von Hippel-Lindau (VHL) syndrome, Cirrhosis, Transplantation, Diabetes, Autoimmune disease, Renal artery stenosis, Interstitial nephritis, Glomerulonephritis, Polycystic kidney disease, Systemic lupus erythematosus, Renal tubular acidosis, IgA nephropathy, Hypercalceimia, Lesch-Nyhan syndrome, Systemic lupus erythematosus , Autoimmune disease, Asthma, Emphysema, Scleroderma, allergy as well as other diseases, disorders and conditions.
The NOVIO nucleic acids and polypeptides are useful for detecting specific cell types. For example, expression analysis has demonstrated that a NOVIO nucleic acid is expressed in Heart, Thyroid, Parotid Salivary glands, Liver, Colon, Ascending Colon, Bone Marrow, Peripheral Blood, Lymphoid tissue, Spleen, Lymph node, Tonsils, Thymus, Cerebellum, Spinal Chord, Cervix, Mammary gland/Breast, Ovary, Placenta, Uterus, Oviduct/Uterine Tube/Fallopian tube, Vulva, Prostate, Testis, Lung, Kidney, Kidney Cortex, Retina, Skin.
Additional utilities for NOVIO nucleic acids and polypeptides according to the invention are disclosed herein.
NOV11
A NOVl 1 polypeptide has been identified as a Nuclear Protein-like protein (also referred to as CG94325-01). The disclosed novel NOV11 nucleic acid (SEQ ID NO:23) of 8670 nucleotides is shown in Table 11 A. The novel NOVl 1 nucleic acid sequences maps to the chromosome 15.
An ORF begins with an ATG initiation codon at nucleotides 204-206 and ends with a TAA codon at nucleotides 7152-7154. A putative untranslated region and/or downstream from the termination codon is underlined in Table 11 A, and the start and stop codons are in bold letters.
Table 11 A. NOV11 Nucleotide Sequence (SEQ ID NO:23)
ACGCGTAGAGCCGCTTTGCGCGTGCGCATCACCTAGGCGGTTAGATTTGAATACTTCACTGAGGCGA GCCGGGCGTTGTGAGCGGACTGCTAGAGGCGGCTGTCTGTTTCCGCTCTAAGGAAACTCAGAGCGTG TGGACCCCAAACAAGTCTGCGCAAAATTTGTCGAGGAGGTTTGCCGCGGCAGAAAAGTTTTCTTCAA AAATGGATGGGGTGTCTTCAGAGGCTAATGAAGAAAATGACAATATAGAGAGACCTGTTAGAAGACG GCATTCTTCAATATTGAAACCCCCAAGGAGTCCTCTTCAGGACCTCAGAGGTGGGAATGAAAGAGTT CAGGAATCCAATGCTTTGAGAAATAAGAAAAACTCTCGTCGAGTCAGCTTTGCAGATACTATAAAGG TATTCCAGACGGAGTCTCATATGAAAATAGTGAGAAAGTCAGAAATGGAAGAAACAGAAACAGGAGA AAATCTTCTTTTGATACAGAATAAGAAATTAGAAGATAATTACTGTGAAATTACTGGGATGAACACA TTGCTTTCTGCTCCCATTCATACCCAGATGCAACAGAAGGAGTTTTCAATTATAGAACATACCCGTG AAAGGAAACATGCAAATGACCAGACAGTCATTTTTTCAGATGAAAACCAGATGGACCTGACATCAAG TCACACTGTAATGATTΆCCAAΆGGCCTTTTAGATAΆTCCCATAAGTGΆAAAGTCCACCAAGATAGΆT ACCACATCATTTCTAGCTAATTTAAAGCTTCACACCGAGGACTCAAGAATGAAΆΆAAGAAGTAAATT TTTCCGTGGATCAAAACACTTCTTCAGAAAATAAAATAGATTTCAATGACTTCATAAAAAGATTGAA AACAGGAΆAATGTAGTGCTTTTCCTGATGTGCCTGATAAAGAAAΆTTTTGAGATACCTATTTATTCC AAGGAACCGAACAGTGCCTCTTCTACACATCAAATGCATGTATCTCTTAAGGAAGATGAAAATAACA GTAATATTACTAGGCTCTTTAGAGAAAAAGATGATGGGATGAATTTCACCCAGTGTCATACAGCCAA TATTCAGACATTGATTCCCACATCCAGTGAGACCAACTCACGGGAATCTAAAGGTAΆTGATATTACA ATTTATGGCAATGACTTTATGGACTTGACATTTAACCACACTTTGCAGATCTTACCTGCAACAGGTA ATTTTTCTGAAATAGAAAATCAAACTCAGAATGCCATGGATGTAACAACAGGTTATGGAACTAAAGC TTCAGGAAATAAAACAGTTTTTAAGAGTAAACAAAATACTGCTTTTCAAGACCTTTCCATAAACTCT GCAGACAAAATACATATTACCAGAAGTCATATTATGGGGGCAGAAACTCACATAGTCTCACAGACTT GTAATCAGGATGCCAGAATATTAGCCATGACCCCAGAATCTATATATTCTAATCCATCTATTCAAGG TTGTAAGACTGTTTTCTATTCTAGTTGTAATGATGCCATGGAAATGACCAAATGTCTCTCAAATATG AGAGAGGAGAAAAATTTGCTAAAGCATGACAGTAATTATTCTAAAATGTATTGCAATCCAGATGCTA TGTCTTCTCTCACAGAGAAAACTATTTATTCCGGAGAGGAGAACATGGACATTACCAAGAGTCATAC AGTTGCAATAGATAATCAAATTTTTAAACAAGATCAATCAAATGTGCAAATAGCAGCTGCACCAACA CCCGAAAAAGAAATGATGCTCCAAAATCTTATGACCACATCAGAAGATGGGAAAATGAATGTAAATT GTAACTCAGTTCCTCATGTATCTAAGGAAAGAATACAGCAGAGCCTGTCAAATCCTTTGTCTATTTC ATTGACTGATAGAAAGACTGAACTCTTATCAGGTGAAAATACGGATTTGACTGAAAGTCACACAAGT AACTTAGGAAGTCAGGTTCCTCTTGCAGCTTATAATCTAGCACCGGAGAGTACCAGTGAATCTCACT CTCAGAGCAAAAGCTCTTCAGATGAATGTGAAGAAATTACCAAAAGTCGTAATGAACCATTTCAGCG ATCAGACATAATAGCCAAAAACAGCTTAACCGACACCTGGAACAAAGACAAAGATTGGGTTTTGAAG ATTTTGCCCTACCTTGATAAAGATTCTCCTCAGTCAGCTGATTGTAATCAGGAGATAGCAACAAGCC ATAATATAGTCTACTGTGGTGGAGTTCTTGATAAACAAATAACTAATAGAAATACAGTATCATGGGA ACAATCTTTGTTTTCTACCACAAAGCCATTATTTTCATCAGGACAGTTCTCTATGAAAAATCATGAT ACTGCTATAAGTAGTCATACAGTGAAATCTGTACTAGGCCAGAATTCTAAACTGGCTGAGCCACTGA GGAAAAGTTTAAGCAATCCCACACCTGACTATTGCCATGACAAGATGATTATATGTTCAGAGGAAGA GCAAAATATGGATCTAACAAAGAGCCACACTGTCGTCATTGGATTTGGTCCTTCTGAACTACAAGAA CTTGGTAAAACTAATTTAGAACACACTACTGGCCAGCTAACAACAATGAACAGACAGATAGCTGTAA AAGTTGAAAAATGTGGTAAAAGTCCCATAGAAAAAAGTGGAGTGCTTAAATCTAACTGTATTATGGA TGTGTTAGAGGACGAAAGTGTACAGAAACCTAAATTTCCAAAGGAAAAGCAAAATGTCAAAATTTGG GGAAGGAAAAGTGTTGGTGGACCAAAAATTGATAAGACTATTGTATTTTCAGAAGACGATAAGAATG ATATGGATATCACTAAGAGTTATACAATAGAAATAAACCATAGACCTTTATTAGAGAAACGTGATTG TCATTTGGTGCCATTGGCAGGAACTTCTGAAACTATTTTATATACATGTGGGCAGGATGACATGGAG ATCACTAGAAGTCACACAACTGCCTTAGAATGTAAAACTGTCTCACCAGATGAAATAACTACTAGGC CTATGGACAAAACTGTAGTGTTTGTAGATAATCATGTTGAACTAGAAATGACAGAGTCCCATACTGT TTTCATTGACTACCAAGAAAAGGAAAGAACAGACAGACCTAACTTTGAACTATCCCAAAGGAAAAGC CTAGGAACACCAACAGTGATATGTACTCCTACTGAGGAGAGTGTTTTCTTTCCAGGAAATGGTGAAA GTGACCGTCTAGTAGCAAATGACAGCCAGCTAACCCCTCTGGAGGAATGGTCTAATAATAGGGGCCC TGTAGAGGTAGCTGATAACATGGAATTGTCTAAATCAGCCACTTGCAAAAACATCAAAGATGTACAA AGTCCTGGATTTCTGAATGAACCTCTATCAAGCAAAAGTCAGAGAAGAAAAAGCCTTAAGCTAAAAA ATGACAAGACCATTGTATTTTCAGAGAATCATAAAAATGATATGGATATTACCCAGAGTTGTATGGT GGAAATAGATAACGAAAGTGCCCTGGAGGATAAAGAGGACTTCCATTTGGCAGGGGCTTCTAAAACT ATTTTGTATTCATGTGGGCAGGATGACATGGAGATCACTAGGAGTCACACAACTGCCTTAGAATGTA AAACTCTCCTGCCAAACGAAATAGCTATTAGGCCCATGGACAAAACCGTATTGTTCACAGATAATTA CAGTGATCTGGAAGTCACCGATTCCCATACTGTTTTCATTGACTGTCAAGCCACAGAGAAAATACTT GAAGAAAACCCTAAATTTGGAATAGGAAAAGGAAAAAACTTGGGTGTTTCCTTTCCTAAGGATAATA GCTGTGTTCAAGAAATCGCTGAAAAACAAGCACTGGCTGTAGGAAACAAAATAGTTCTTCACACCGA GCAAAAGCAACAACTCTTTGCTGCTACTAATAGAACTACTAATGAAATCATCAAATTTCATAGTGCT GCTATGGATGAAAAGGTCATAGGGAAAGTTGTAGACCAGGCCTGTACATTGGAAAAAGCGCAAGTTG AAAGCTGTCAGTTAAATAATAGAGATAGAAGAAATGTGGACTTTACAAGTAGTCATGCAACTGCTGT TTGTGGATCCAGTGATAATTATTCCTGTTTACCAAATGTTATTTCCTGTACTGATAATTTGGAGGGT AGTGCCATGCTCTTATGTGATAAAGATGAGGAAAAAGCCAATTATTGCCCAGTGCAAAATGATCTTG CTTATGCAAATGATTTTGCCAGTGAATATTACTTGGAATCTGAGGGACAGCCTCTCTCTGCTCCTTG TCCTTTGTTAGAGAAGGAAGAAGTTATTCAAACCAGTACCAAAGGACAGTTAGACTGTGTTATAACA CTGCACAAAGATCAAGATCTGATTAAGGATCCACGAAATCTATTGGCTAATCAAACTTTAGTATATA GTCAAGATCTGGGGGAGATGACTAAACTTAATTCAAAGCGAGTATCTTTTAAGCTTCCAAAGGATCA AATGAAAGTCTATGTTGATGACATTTATGTTATTCCTCAGCCTCATTTCTCAACCGACCAACCTCCA TTACCTAAAAAAGGACAGAGTAGTATCAATAAAGAAGAAGTAATACTGTCTAAAGCTGGAAATAAGA GTTTAAATATTATAGAAAATTCCTCTGCACCCATATGTGAAAACAAGCCCAAAATACTCAATAGTGA GGAATGGTTTGCTGCAGCCTGTAAAAAAGAACTGAAGGAAAATATTCAAACAACTAACTATAATACA GCTCTAGATTTCCACAGTAACTCAGACGTAACTAAGCAAGTCATTCAAACTCATGTCAATGCTGGAG AAGCACCAGATCCTGTAATTACATCTAATGTTCCATGTTTTCATAGTATCAAACCAAATCTGAATAA TTTGAATGGAAAAACTGGAGAGTTTTTAGCCTTTCAAACTGTTCATCTACCACCCCTTCCAGAGCAA TTACTTGAATTAGGAAATAAGGCACACAATGATATGCATATAGTGCAAGCTACAGAAATACATAATA TTAACATAATCTCCAGCAATGCTAAAGATAGTAGAGATGAGGAAAATAAAAAGTCTCATAATGGAGC TGAAACCACCTCTCTACCGCCAAAGACAGTTTTTAAAGATAAAGTAAGGAGATGTTCTTTGGGAATC TTTTTGCCTAGATTGCCCAACAAGAGAAATTGTAGTGTCACTGGTATTGATGACCTGGAACAGATTC CAGCAGACACAACTGATATAAATCACTTAGAAACTCAGCCGGTCTCTAGCAAAGATTCAGGCATTGG ATCTGTTGCAGGTAAACTGAACCTAAGTCCTTCTCAATATATAAATGAGGAAAATCTTCCTGTATAT CCTGATGAGATCAATTCTTCAGACTCTATTAACATAGAAACTGAGGAAAAGGCCTTGATTGAGACAT ACCAAAAAGAGATTTCACCATATGAAAATAAAATGGGAAAAACTTGCAATAGCCAAAAAAGAACGTG GGTACAAGAAGAAGAAGATATTCATAAGGAGAAAAAAATCAGAAAAAATGAGATTAAGTTTAGTGAT ACGACACAAGATCGGGAGATTTTTGATCACCATACTGAAGAGGATATAGATAAAAGTGCTAACAGTG TATTGATAAAAAACCTGAGCAGGACCCCATCTAGTTGCAGCAGCTCTCTGGATTCAATCAAGGCTGA TGGGACCTCTCTGGACTTCAGCACTTACCGCAGTAGTCAAATGGAATCACAGTTTCTCAGAGATACT ATTTGTGAAGAGAGCTTGAGGGAGAAACTCCAAGATGGGAGAATAACAATAAGGGAGTTCTTTATAC TTCTCCAGGTCCACATCTTGATACAGAAACCCCGACAGAGCAATCTCCCAGGCAATTTTACTGTAAA CACACCACCTACTCCAGAAGACCTGATGTTAAGTCAATATGTTTACCGACCCAAGATACAGATTTAT AGAGAAGATTGTGAGGCTCGTCGCCAAAAGATTGAAGAATTAAAGCTTTCTGCATCGAACCAAGATA AGCTGTTGGTTGATATAAATAAGAACCTGTGGGAAAAAATGAGACACTGCTCTGACAAAGAGCTGAA GGCCTTTGGAATTTATCTTAACAAAATAAAGTCATGTTTTACCAAGATGACTAAAGTCTTCACTCAC CAAGGAAAAGTGGCTCTGTATGGCAAGCTGGTGCAGTCAGCTCAGAATGAGAGGGAGAAACTTCAAA TAAAGATAGATGAGATGGATAAAATACTTAAGAAGATCGATAACTGCCTCACTGAGATGGAAACAGA AACTAAGAATTTGGAGGATGAAGAGAAAAACAATCCTGTGGAAGAATGGGATTCTGAAATGAGAGCT GCAGAAAAAGAATTGGAACAGCTGAAAACTGAAGAAGAGGAGCTTCAAAGAAATCTCTTAGAACTGG AGGTACAAAAAGAGCAGACCCTTGCTCAAATAGACTTTATGCAAAAACAAAGAAATAGAACTGAAGA GCTACTGGATCAGTTGAGCTTGTCTGAGTGGGATGTCGTTGAGTGGAGTGATGATCAAGCTGTATTC ACCTTTGTTTATGACACGATACAACTCACCATCACCTTTGAAGAGTCAGTTGTTGGTTTCCCTTTCC TGGACAAGCGTTATAGGAAGATTGTTGATGTCAATTTTCAATCTCTGTTAGATGAGGATCAAGCTCC TCCTTCCTCCCTTTTAGTTCATAAGCTTATTTTCCAGTACGTTGAAGAAAAGGAATCCTGGAAGAAG ACATGTACAACCCAGCATCAGTTACCCAAGATGCTTGAAGAATTCTCACTGGTAGTGCACCATTGCA GACTCCTTGGAGAGGAGATTGAGTATTTAAAGAGATGGGGACCAAATTATAACCTAATGAACATAGA TATTAATAATAATGAATTGAGACTTTTATTCTCTAGCTCCGCAGCATTTGCAAAGTTTGAAATAACT TTGTTTCTCTCAGCCTATTATCCATCTGTACCATTACCTTCCACCATTCAGAATCACGTTGGGAACA CTAGCCAAGATGATATTGCTACCATTCTATCTAAAGTGCCACTGGAGAACAACTACCTGAAGAATGT AGTCAAGCAAATTTACCAAGATCTGTTTCAGGACTGCCATTTCTACCACTAGACCCTTGGACCACCA TTGGAACAACCAAGCAGAATGTACTTGATATTATTTCAGGGTCCCATTGCTGTTCAGCCTTTGTTTT TACGTCATTACAAGCTGAGTAAAATTCCTTCTGATGATGTTATAGTTAATCTGTATGTTTTTTATAT CTCTGCAGAATGATGGTGATGAAGTCTGGATGGTAGGCCTCATAGCCTACTATCAACTTACTCATCT TTGTACCAAAGGTTTAAGTAATAGGACACTTAGGAAAAATGTCTCCTAACTAAACTAGTGCTTTCTG CTTTAGTACAAGCCCTAAGGATTAACTTAAGTATAAGAAGTGTTATCACTGACAAGAACATTAGCCA TTTTCCCATAACTAGATAGAGCTATGATTTTTTAGGTTGCCTGGCTTCTGCCTAGCAGATATTTCTG GAGTAGAAATGTATCTGTCTACAAACTATTATCCTTTTTCTCCGTTACTAAAATGCTATTAAGAGAA AGTAGGGCTGGGTGTGAGCCACCACACCCAGCAATGTTTTCTTAATAAGTATAGTTTTTCTAGGGAA AGTTAATTCATTTTTGTCTAGTACATATATGTAAATATATTAATGTTGTTTTTGTGTTTGTGATGTA GTAAGGAGATGTACATAGAAATTCATTGAGGTATATAGATACTCATCTGTCTAGGCAGTTCCCAATT TTCTGAAGAATGTTTTACAGCAAAATTTTCTATTTTCTTTTATTAAATAGTGACACGTCAAACAATG TCACATCCAAAACACTAGTTTCATCAATTTCTAGCAGTAATAATAGACTTGCTGTAAGTATTGTTTT CTGATGCCATACCCTTGTCATACATATTATTAAATGACCAATATTATGTATGAAGTAGACAAAAAAA TTTACTCAAACTTCATTCAAATCCTAATTGTGATAATTTTTGTTTTATATTTAATTATAAACCAAAA TACATTTGCATTTTTAAGCTAATTTGTCTCAAAATTTTGCTTTATATTTTTGGATCAGGTTAAAGTC CTGTGGATCCCCTGAATGTTATTGTCCCTCTTGATTGGTTTTTACTTCTGAGCTATACGTCAAAAGA CACATAAGCTTCAAAAGTCAAGACAAACCTCATTTGCCATAAAAATCAAGATATAGATGTTCTGTTC CGTAAACTCCTTGAAAAACATTTTAAAGTCATCAATATGATCTGTTTCCCATGAAACTTAAGTTAGC TTTCTTATTGGAGTTATTTCTTTTCTGTAAGTCTGAAAAGTAGAGATTTTGTTTTACGCATTTTAGT AACCTGCAACAACCAACτCTAAAAAAGATTTGGCTTGTAATGACGGTCτCTGCTTTTTTGGGTTTGG AGTACACAATTGTAATATTTACTTAGTTATTTGTGTTTTTCTTTGTTCAAGGTATTGACTAGTTTCA TAAATTTTTTGCAAGTTTTTCTTTCATTGGTTGGAAAGCAGATTACATTTTGCACTATTAAAATAAG TTTATTACTTTAAAAAAAAAGTCGACG Variant sequences of NOVl 1 are included in Example 3, Table 23. A variant sequence can include a single nucleotide polymorphism (SNP). A SNP can, in some instances, be referred to as a "cSNP" to denote that the nucleotide sequence containing the SNP originates as a cDNA.
The NOVl 1 protein (SEQ ID NO:24) encoded by SEQ ID NO:23 is 2316 amino acid residues in length and is presented using the one-letter amino acid code in Table 1 IB. Psort analysis predicts the NOVl 1 protein of the invention to be localized at the nucleus with a certainty of 0.8800.
Table 11B. Encoded NOV11 protein sequence (SEQ ID NO:24)
MDGVSSEANEENDNIERPVRRRHSSILKPPRSPLQDLRGGNERVQESNA RNKKNSRRVSFADTI KVFQTΞSHMKIVRKSEMEETETGENLLLIQNKKLEDNYCEITGMNTLLSAPIHTQMQQKEFSIIE HTRERKHANDQTVIFSDENQMDLTSSHTV ITKGLLDNPISEKSTKIDTTSFLA LKLHTEDSRM KKEVNFSVDQNTSSENKIDFNDFIKRLKTGKCSAFPDVPDKENFEIPIYS EPNSASSTHQMHVS LKEDΞLRØSNITRLFREKDDGMNFTQCHTANIQTLIPTSSETNSRESKGNDITIYGNDF DLTFNH T QILPATGNFSEIENQTQNAMDVTTGYGTKASGNKTVFKSKQNTAFQDLSINSADKIHITRSHI MGAETHIVSQTCNQDARI AMTPESIYSNPSIQGCKTVFYSSCNDAMEMTKCLSNMREEKNLLKH DSNYSKMYCNPDAMSSLTEKTIYSGEENMDITKSHTVAIDNQIFKQDQSNVQIAAAPTPEKEMML QNLMTTSEDGKMNVNCKTSVPHVSKERIQQSLSNPLSISLTDRKTELLSGENTDLTESHTSNLGSQ VPLAAYNLAPESTSESHSQSKSSSDECEEITKSRNEPFQRSDIIAKNSLTDTWNKDKDWVLKILP YLDKDSPQSADCNQEIATSH IVYCGGVLDKQITNRNTVS EQSLFSTTKPLFSSGQFSMK HDT AISSHTVKSVLGQNSKLAEPLRKSLSNPTPDYCHDKMIICSΞEEQNMD TKSHTWIGFGPSELQ ELGKTNLΞHTTGQLTTMNRQIAVKVEKCGKSPIEKSGVLKSNCIMDVLEDESVQKP FPKEKQNV KIWGRKSVGGPKID TIVFSEDDKNDMDITKSYTIΞI HRP LEKRDCHLVPLAGTSETI YTCG QDDMEITRSHTTALECKTVSPDEITTRPMDKTWFVDNHVE EMTESHTVFIDYQEKERTDRPNF E SQRKSLGTPTVICTPTEESVFFPGNGESDRLVANDSQLTPLEE S RGPVEVADN ELSKSA TCKNIKDVQSPGFLNEP SSKSQRRKSLKLIOROKTIVFSE HKNDMDITQSCMVEIDNESALEDK EDFHLAGASKTI YSCGQDDMEITRSHTTALECKTLLPNEIAIRPMDKTVLFTDNYSDLEVTDSH TVFIDCQATEKI EENPKFGIGKGKNLGVSFPKDNSCVQEIAEKQA AVGNKIVLHTEQKQQLFA ATNRTTNEIIKFHSAAMDEKVIGKVVDQACTLEKAQVESCQLNNRDRRNVDFTSSHATAVCGSSD NYSCLPNVISCTDNLEGSA LLCDKDEEKANYCPVQNDLAYANDFASEYY ESEGQPLSAPCPLL EKEEVIQTSTKGQ DCVITLHKDQD IKDPRNLDANQTLVYSQDLGEMTKLNSKRVSFKLP DQM KVYVDDIYVIPQPHFSTDQPPLPKKGQSSINKEEVILS AGNKSLNIIENSSAPICENKPKILNS EE FAAACKKELKENIQTTNYNTALDFHSNSDVTKQVIQTHV AGEAPDPVITSNVPCFHSIKPN LN LNGKTGEFLAFQTVHLPPLPEQLLELGNKAH DMHIVQATEIHNINIISSNAKDSRDEENKK SHNGAETTSLPPKTVF DKVRRCSLGIFLPRLPNKRNCSVTGIDDLEQIPADTTDINHLETQPVS SKDSGIGSVAGK NLSPSQYINEENLPVYPDEINSSDSINIETEEKALIETYQKEISPYENKMGK TCNSQKRT VQΞEΞDIHKEKKIRKNΈIKFSDTTQDREIFDHHTEΞDIDKSANSVLI NLSRTPSS CSSSLDSIKADGTSLDFSTYRSSQ ESQFLRDTICEESLREKLQDGRITIREFFILLQVHILIQK PRQSNLPGNFTVNTPPTPED MLSQYVYRPKIQIYREDCEARRQKIEELK SASNQDKLLVDINK NL EKMRHCSDKE KAFGIYLNKIKSCFTKMTKVFTHQGKVALYGKLVQSAQNEREKLQIKIDEM DKILKKIDNCLTEMETETKNLEDEEK NPVΞE DSEMRAAEKELEQLKTEEEE QR LLELEVQK EQTLAQIDFMQ QRNRTEELLDQLSLSΞWDWEWSDDQAVFTFVYDTIQLTITFEESWGFPF D KRYRKIVDVNFQSLLDEDQAPPSSLLVHKLIFQYVEEKESWKKTCTTQHQLPKMLEEFSLWHHC RL GEEIEYLKR GPNYN MNIDINMNELRLLFSSSAAFAKFEITLF SAYYPSVPLPSTIQ HV GNTSQDDIATILSKVP E NYLKNWKQIYQDLFQDCHFYH
A search against the Patp database, aproprietary database that contains sequences published inpatents and patent publications, yielded several homologous proteins shown in Table llC. Table 11C. Patp results for NOV11
Smallest Sum
Reading High Prob
Sequences producing High-scoring Segment Pairs: Frame Score P(N)
>patp:AAW88398 Human testis secreted protein dol5_4 +1 2444 1.7e-253
>patp:AAU71933 Human bone marrow tissue polypeptide #11 +1 2444 1.7e-253
>patp:AAU71961 Human bone marrow tissue polypeptide #39 +1 2444 1.7e-253
>patp:AAU71933 Human bone marrow tissue polypeptide #11 +1 2444 1.7e-253
>patp:AAU71961 Human bone marrow tissue polypeptide #39 +1 2444 1.7e-253
In a BLAST search of public sequence databases, it was found, for example, that the nucleic acid sequence of this invention has 5584 of 5584 bases (100%) identical to a gb:GENBANK-ID:AB046790|acc:AB046790.1 mRNA from Homo sapiens (mRNA for KIAA1570 protein, partial eds). The full amino acid sequence of the protein of the invention was found to have 1790 of 1793 amino acid residues (99%) identical to, and 1792 of 1793 amino acid residues (99%) similar to, the 1833 amino acid residue ptnr:SPTREMBL- ACC:Q9NR92 protein from Homo sapiens (AF15Q14 PROTEIN).
NOVl 1 also has homology to the proteins shown in the BLASTP data in Table 1 ID.
Table 11D. BLAST results for NOV11
Gene Index/ Protein/ Organism Length Identity Positives Expect Identifier (aa) (%) (%) gi I 18308012 |gb|AAL6 AF15ql4 isoform 2 2316 2316/2316 2316/2316 0.0 7803.l|AF461041_l(A [Homo sapiens] (100%) (100%) F461041) gi| 9966807 I ref I NP_0 AF15ql4 protein 1833 1790/1793 1792/1793 0.0 65113. l| (NM 020380) [Homo sapiens] (99%) (99%) gi I 14749154 | ref | XP_ AF15ql4 protein 1833 1789/1793 1791/1793 0.0 031524. ll (XM 031524 [Homo sapiens] (99%) (99%) gi 110047205 |dbj |BAB KIAA1570 protein 1360 1360/1360 1360/1360 0.0 13396. ll (AB046790) [Homo sapiens] (100%) (100%) gi 114749150 I ref |XP_ similar to 915 900/900 900/900 0.0 012461.3 I KIAA1570 protein (100%) (100%) (XM 012461) [Homo sapiens]
A multiple sequence alignment is given in Table 1 IE, with the NOVl 1 protein being shown on line 1 in Table 1 IE in a ClustalW analysis, and comparing the NOVl 1 protein with the related protein sequences shown in Table 1 ID. This BLASTP data is displayed graphically in the ClustalW in Table 1 IE.
Table HE. ClustalW Analysis of NOVl 1
1) > NOVl 1; SEQ ID NO:24
2) > gi|18308012]/ AF15ql4 isoform 2 [Homo sapiens]; SEQ ID NO:82
3) > gi|9966807|/ AF15ql4 protein [Homo sapiens]; SEQ ED NO:83
4) > gijl4749154|/ AF15ql4 protein [Homo sapiens]; SEQ ID NO:84
5) > gijl0047205|/ KIAA1570 protein [Homo sapiens]; SEQ ID NO:85 6) > gi|14749150|/ similar to K1AA1570 protein [Homo sapiens]; SEQ ID NO:86
Figure imgf000087_0001
130 140 150 160 170 180
Figure imgf000087_0002
NOVll 421
Figure imgf000087_0003
480
Figure imgf000088_0001
790 800 810 820 830 840
Figure imgf000088_0002
850 860 870 880 890 900
Figure imgf000088_0003
910 920 930 940 950 960
Figure imgf000089_0001
1030 1040 1050 1060 1070 1080
I
NOVll 1021 WSNNRGPVEVADNMELSKSATCKNIKDVQSPGFLNEPLSSKSQRRKSLKLKNDKTIVFSE gi | 18308012 | 1021 WSNNRGPVEVADNMELSKSATCKNIKDVQSPGFLNEPLSSKSQRRKSLKLKNDKTIVFSE giJ9966807| 1021 WSNNRGPVEVADNMELSKSATCKNIKDVQSPGFLNEPLSSKSQRRKSLKLKNDKTIVFSE gij 1474915 | 1021 WSNNRGPVEVADNMELSKSATCKNIKDVQSPGFLNEPLSSKSQRRKSLKLKNDKTIVFSE gij 10047205 | 65 HSNNRGPVEVADNMELSKSATCKNIKDVQSPGFLNEPLSSKSQRRKSLKLKNDKTIVFSE gi j 14749150 j 1 lELSKSATCKNIKDVQSPGFLNEPLSSKSQRRKSLKLKNDKTIVFSr
1090 1100 1110 1120 1130 1140
NOVll 1081 NHKNDMDITQSCMVEIDNESALEDKEDFHLAGASKTILYSCGQDDMEITRSHTTALECK1 gi 118308012 I 1081 NHKNDMDITQSCMVEIDNESALEDKEDFHLAGASKTILYSCGQDDMEITRSHTTALECKl gij 9966807 I 1081 MHKNDMDITQSCMVEIDNESALEDKEDFHLAGASKTILYSCGQDDMEITRSHTTALECKT gij 14749154 I 1081 NΗKNDMDITQSCMVEIDNESALEDKEDFHLAGASKTILYSCGQDDMEITRSHTTALECKI gij 10047205 j 125 NHKNDMDITQSCMVEIDNESALEDKEDFHLAGASKTILYSCGQDDMEITRSHTTALECKl gij 14749150 48 MHKNDMDITQSCMVEIDNESALEDKEDFHLAGASKTILYSCGQDDMEITRSHTTALECKT
1150 1160 1170 1180 1190 1200 ..I... ..I
NOVll 1141 LLPNEIAlRPMDKTVLFTDNYSDLEVTDSHTVFIDCQATEKILEENPKFGIGKGKNLGV gi | 18308012 | 1141 LLPNEIAIRPMDKTVLFTDNYSDLEVTDSHTVFIDCQATEKILEENPKFGIGKGKNLGVΣ gi|9966807| 1141 LLPNEIAIRPMDKTVLFTDNYSDLEVTDSHTVFIDCQATEKILEENPKFGIGKGKNLGV gi| 1474915 | 1141 LLPNEIAIRPMDKTVLFTDNYSDLEVTDSHTVFIDCQATEKILEENPKFGIGKGKNLGV gi| 10047205 | 185 LLPNEIAIRPMDKTVLFTDNYSDLEVTDSHTVFIDCQATEKILEENPKFGIGKGKNLGV gi | 14749150 | 108 LLPNEIAlRPMDKTVLFTDNYSDLEVTDSHTVFIDCQATEKILEENPKFGIGKGKNLGV
1210 1220 1230 1240 1250 1260
NOVll 1201 FPKDNSCVQEIAKKQALAVGNKIVLHTEQKQQLFAATNRTTNEIIKFHSAAMDEKVIGKV gi | 18308012 | 1201 FPKDNSCVQEIAEKQALAVGNKIVLHTEQKQQLFAATNRTTNEIIKFHSAAMDEKVIG gi|9966807| 1201 FPKDNSCVQEIAEKQALAVGNKIVLHTEQKQQLFAATNRTTNEIIKFHSAAMDEKVIG gi | 14749154 | 1201 FPKDNSCVQEIAEKQALAVGNKIVLHTEQKQQLFAATNRTTNEIIKFHSAAMDEKVIGKV gi| 10047205] 245 FPKDNSCVQEIAEKQALAVGNKIVLHTEQKQQLFAATNRTTNE11KFHSAAMDEKVIGKV gi| 14749150 | 168 FPKDNSCVQEIAEKQALAVGNKIVLHTEQKQQLFAATNRTTNEIIKFHSAAMDEKVIGKΛ/
1270 1280 1290 1300 1310 1320
I ..I
NOVll 1261 roQACTLEKAQVESCQLNNRDRRNVDFTSSHATAVCGSSDNYSCLPNVISCTDNLEGSAl gi] 18308012 | 1261 DQACTLEKAQVES CQLNNRDRRNVDFTS SHATAVCGSSDNYS CLPNVI S CTDNLEGS At gi] 9966807 | 1261 DQACTLEKAQVESCQLNNRDRRNVDFTSSHATAVCGSSDNYSCLPNVISCTDNLEGSAM gi| 14749154] 1261 TDQACTLEKAQVESCQLNNRDRRNVDFTSSHATAVCGSSDNYSCLPNVISCTDNLEGSAM gi| 10047205 | 305 QACTLEKAQVESCQLNNRDRRNVDFTSSHATAVCGSSDNYSCLPNVISCTDNLEGSAlV gi| 14749150 | 228 DQACTLEKAQVESCQLNNRDRRNVDFTSSHATAVCGSSDNYSCLPNVISCTDNLEGSAT
1330 1340 1350 1360 1370 1380
NOΛ Til 1321 LLCDKDEEKANYCPVQNDLAYANDFASEYYLESEGQPLSAPCPLLEKEEVIQTSTKGQLD gi 183080121 1321 LLCDKDEEKANYCPVQNDLAYANDFASEYYLESEGQPLSAPCPLLEKEEVIQTSTKGQLD gi 9966807| 1321 LLCDKDEEKANYCPVQNDLAYANDFASEYYLESEGQPLSAPCPLLEKEEVIQTSTKGQLD gi 14749154 | 1321 LLCDKDEEKANYCPVQNDLAYANDFASEYYLESEGQPLSAPCPLLEKEEVIQTSTKGQLD gi 10047205 | 365 LLCDKDEEKANYCPVQNDLAYANDFASEYYLESEGQPLSAPCPLLEKEEVIQTSTKGQLD gi 14749150 | 288 LLCDKDEEKANYCPVQNDLAYANDFASEYYLESEGQPLSAPCPLLEKEEVIQTSTKGQLD 1390 1400 1410 1420 1430 1440
NOVll 1381 VITLHKDQDLIKDPRNLLANQTLVYSQDLGEMTKLNSKRVSFKLPKDQMKVYVDDIYVI gi] 18308012 | 1381 VITLHKDQDLIKDPRNLLANQTLVYSQDLGEMTKLNSKRVSFKLPKDQMKVYVDDIYVI gi|9966807| 1381 VI LHKDQDLIKDPRNLLANQTLVYSQDLGEMTKLNSKRVSFKLPKDQMKVYVDDIYVI gi 114749154] 1381 VITLHKDQDLIKDPRNLLANQTLVYSQDLGEMTKLNSKRVSFKLPKDQMKVYVDDIYVI gi|l0047205| 425 VITLHKDQDLIKDPRNLLANQTLVYSQDLGEMTKLNSKRVSFKLPKDQMKVYVDDIYVI gi| 14749150 | 348 VITLHKDQDLIKDPRNLLANQTLVYSQDLGEMTKLNSKRVSFKLPKDQMKVYVDDI VI
1450 1460 1470 1480 1490 1500
NOVll 1441 'QPHFSTDQPPLPKKGQSSINKEEVILSKAGNKSLNIIENSSAPICENKPKILNSEEWFΛ gi| 18308012 | 1441 PQPHFSTDQPPLPKKGQSSINKEEVILSKAGNKSLNIIENSSAPICENKPKILNSEEWFA gi| 9966807] 1441 'QPHFSgJDQPPLPKKGQSSINKEEVILSKAGNKSLNIIENSSAPICENKPKILNSEEWFA gi| 14749154 | 1441 'QPHFSTDQPPLPKKGQSSINKEEVILSKAGNKSLNIIENSSAPICENKPKILNSEEWF.fi gi| 10047205 | 485 PQPHFSTDQPPLPKKGQSSINKEEVILSKAGNKSLNIIENSSAPICENKPKILNSEEWFΛ gi| 14749150 | 408 >QPHFSTDQPPLPKKGQSSINKEEVILSKAGNKSLNIIENSSAPICENKPKILNSEEWF£
1510 1520 1530 1540 1550 1560
NOVll 1501 CKKELKENIQTT YNTALDFHSNSDVTKQVIQTHVNAGEAPDPVITSNVPCFHSIKPI gi| 18308012] 1501 CKKELKENIQTTNYNTALDFHSNSDVTKQVIQTHVNAGEAPDPVITSNVPCFHSIKPl gi|9966807| 1501 AACKKELKENIQTTNYNTALDFHSNSDVTKQVIQTHVNAGEAPDPVITSNVPCFHSIKPN gi| 14749154 | 1501 AACKKELKENIQTTNYNTALDFHSNSDVTKQVIQTHVNAGEAPDPVITSNVPCFHSIKPN gi | 10047205 | 545 LCKKELKENIQTTNYNTALDFHSNSDVTKQVIQTHVNAGEAPDPVITSNVPCFHSIKPN gi | 14749150 | 468 AACKKELKENIQTTNYNTALDFHSNSDVTKQVIQTHVNAGEAPDPVITSNVPCFHSIKPN
1570 1580 1590 1600 1610 1620
NOVll 1561 gi| 18308012] 1561 NLNGKTGEFLAFQTVHLPPLPEQLLELGNKAHNDMHIVQATEIHNINIISSNAKDSR gi | 9966807 | 1561 NNLNGKTGEFLAFQTVHLPPLPEQLLELGNKAHNDMHIVQATΞIHNINIISSNAKDSR_ gi| 14749154 | 1561 JNNLNGKTGEFLAFQTVHLPPLPEQLLELGNKAHNDMHIVQATEIHNINIISSNAKDSRI gi| 10047205 | 605 JNNLNGKTGEFLAFQTVHLPPLPEQLLELGNKAHNDMHIVQATEIHNINI ISSNAKDSR gi] 14749150 | 528 JNNLNGKTGEFLAFQTVHLPPLPEQLLELGNKAHNDMHIVQATEIHNINIISSNAKDSRI
1630 1640 1650 1660 1670 1680
NOVll 1621 EENKKSHNGAETTSLPPKTVFKDKVRRCSLGIFLPRLPNKRNCSVTGIDDLEQIPADTT gi| 18308012 | 1621 EENKKSHNGAETTSLPPKTVFKDKVRRCSLGIFLPRLPNKRNCSVTGIDDLEQIPADTT gi|9966807| 1621 EENKKSHNGAETTSLPPKTVFKDKVRRCSLGIFLPRLPNKRNCSVTGIDDLEQIPADTTD gi| 1474915 | 1621 EENKKSHNGAETTSLPPKTVFKDKVRRCSLGIFLPRLPNKRNCSVTGIDDLEQIPADTTD gi| 10047205 | 665 EENKKSHNGAETTSLPPKTVFKDKVRRCSLGIFLPRLPNKRNCSVTGIDDLEQIPADTTI gi| 14749150 | 588 EENKKSHNGAETTSLPPKTVFKDKVRRCSLGIFLPRLPNKRNCSVTGIDDLEQIPADTTI
1690 1700 1710 1720 1730 1740
NOT 711 1681 INHLETQPVSSKDSGIGSVAGKLNLSPSQYINEENLPVYPDEINSSDSINIETEEKALIE gi 18308012] 1681 INHLETQPVSSKDSGIGSVAGKLNLSPSQYINEENLPVYPDEINSSDSINIETEEKALIE gi 9966807 | 1681 INHLETQPVSSKDSGIGSVAGKLNLSPSQYINEENLPVYPDEINSSDSINIETEEKALIE gi 1474915 | 1681 INHLETQPVSSKDSGIGSVAGKLNLSPSQYINEENLPVYPDEINSSDSINIETEEKALIE gi 10047205] 725 INHLETQPVSSKDSGIGSVAGKLNLSPSQYINEENLPVYPDEINSSDSINIETEEKALIE gi 14749150 | 648 INHLETQPVSSKDSGIGSVAGKLNLSPSQYINEENLPVYPDEINSSDSINIETEEKALIE
1750 1760 1770 1780 1790 1800
NOVll 1741 TYQKEISPYENKMGKTCNSQKRT VQEEEDIHKEKKIRKNEIKFSDTTQDREIFDHHTEE 1800 gi 18308012 | 1741 TYQKEISPYENKMGKTCNSQKRT VQEEEDIHKEKKIRKNEIKFSDTTQDREIFDHHTEE 1800 gi 9966807| 1741 TYQKEISPYENKMGKTCNSQKRT VQEEEDIHKEKKIRKNEIKFSDTTQDRE |VS 1794 i 14749154] 1741 TYQKEISPYENKMGKTCNSQKRTWVQEEEDIHKEKKIRKNEIKFSDTTQDRE is 1794 gi 10047205 | 785 TYQKEISPYENKMGKTCNSQKRT VQEEEDIHKEKKIRKNEIKFSDTTQDREIFDHHTEEj 44 gi 14749150 | 708 TYQKEISPYENKMGKTCNSQKRT VQEEEDIHKEKKIRKNEIKFSDTTQDREIFDHHTEE 67
1810 1820 1830 1840 1850 1860
NOVll 1801 DIDKSANSVLIKNLSRTPSSCSSSLDSIKADGTSLDFSTYRSSQMESQFLRDTICEESLR 1860 gi 18308012] 1801 DIDKSANSVLIKNLSRTPSSCSSSLDSIKADGTSLDFSTYRSSQMESQFLRDTICEESLR 1860 gi 9966807| 1794 SVLΪ >TQ.RMFLNFGFfflFVFffl -CGYS- 1817 gi 1474915 | 1794 RMFLNFGFgFVFg -CGYS- 1817 gi 110047205 I 845 JIDKSANSVLIKNLSRTPSSCSSSLDSIKADGTSLDFSTYRSSQMESQFLRDTICEESLR 904 gi] 14749150] 768 )IDKSANSVLIKNLSRTPSSCSSSLDSIKADGTSLDFSTYRSSQMESQFLRDTICEESLR 327
1870 1880 1890 1900 1910 1920
NOVll 1861 EKLQDGRITIREFFILLQVHILIQKPRQSNLPG FTVNTPPTPEDLMLSQYVYRPKIQIY 1920 gi 118308012 I 1861 EKLQDGRITIREFFILLQVHILIQKPRQSNLPGNFTVNTPPTPEDLMLSQYVYRPKIQIY 1920 giJ9966807| 1817 ll0S 1832 gij 14749154 I 1817 ,IBS 1832 gij 10047205 j 905 IKLQDGRITIREFFILLQVHILIQKPRQSNLPGNF VNTPPTPEDLMLSQYVYRPKIQIY 4 gij 14749150 j 828 ΪKLQDGRITIREFFILLQVHILIQKPRQSNLPGNFTVNTPPTPEDLMLSQYVYRPKIQIY
1930 1940 1950 1960 1970 1980
NOVll 1921 (3?πτ!Rr rκrawnπ |LKLSASNQDKLLVDINKNLWEKMRHCSDKELKAFGIYLNKIKSCFTK 1980 g 118308012 I 1921 L LSASNQDKLLVDINKNLWEKMRHCSDKELKAFGI YLNKI KS CFTK 1980 gij 9966807 I 1833 T- X833 gij 14749154 I 1833 T- 1833 gi 110047205 j 965 iaaww-fe aawwiaa iLfeLSASNQDKLLVDINKNLWEKMRHCSDKELKAFGI YLNKI KSCFTK 1024 gi 114749150 j 888 SSΪHEE.ESΞ.M \ήss 903
1990 2000 2010 2020 2030 2040
Figure imgf000091_0001
2170 2180 2190 2200 2210 2220
Figure imgf000091_0002
2290 2300 2310
NOVll 2281 DDIATILSKVPLENNYLKNWKQIYQDLFQDCHFYH 2316 gi]l8308012| 2281 DDIATILSKVPLENNYLKNWKQIYQDLFQDCHFYH 2316 gi I 9966807 | 1833 1833 gi j l4749154 | 1833 1833 gi j 10047205 I 1325 DDIATILSKVPLENNYLKNWKQIYQDLFQDCHFYH 1360 gi 14749150 915 915
NOVll nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts. For example, NOVl 1 nucleic acids and polypeptides can be used to identify proteins that are members of the Nuclear Protein-like Protein Family. The NOVl 1 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOVl 1 activity or function. Specifically, the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular replication, and signal transduction. These molecules can be used to treat, e.g. , Von Hippel-Lindau (VHL) syndrome, Cirrhosis, Transplantation, Hemophilia, hypercoagulation, Idiopathic thrombocytopenic purpura, autoimmume disease, allergies, immunodeficiencies, transplantation, Graft vesus host, Cardiovascular diseases, Von Hippel-Lindau (VHL) syndrome , Alzheimer's disease, Stroke, Tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, Cerebral palsy, Epilepsy, Lesch-Nyhan syndrome, Multiple sclerosis, Ataxia-telangiectasia, Leukodystrophies, Behavioral disorders, Addiction, Anxiety, Pain, Neuroprotection, Systemic lupus erythematosus , Autoimmune disease, Asthma, Emphysema, Scleroderma, allergy, as well as other diseases, disorders and conditions. h addition, various NOVl 1 nucleic acids and polypeptides according to the invention are useful, ter alia, as novel members of the protein families according to the presence of sequence relatedness to previously described proteins. The NOVll nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of cardiac, immune, and nerve physiology. As such, the NOVl 1 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat cardiovascular, immune, and nervous system disorders, e.g., Von Hippel-Lindau (VHL) syndrome, Cirrhosis,
Transplantation, Hemophilia, hypercoagulation, Idiopathic thrombocytopenic purpura, autoimmume disease, allergies, immunodeficiencies, transplantation, Graft vesus host, Cardiovascular diseases, Von Hippel-Lindau (VHL) syndrome , Alzheimer's disease, Stroke, Tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, Cerebral palsy, Epilepsy, Lesch-Nyhan syndrome, Multiple sclerosis, Ataxia-telangiectasia,
Leukodystrophies, Behavioral disorders, Addiction, Anxiety, Pain, Neuroprotection, Systemic lupus erythematosus , Autoimmune disease, Asthma, Emphysema, Scleroderma, allergy, as well as other diseases, disorders and conditions.
The NOVl 1 nucleic acids and polypeptides are useful for detecting specific cell types. For example, expression analysis has demonstrated that a NOVl 1 nucleic acid is expressed in Adipose, Aorta, Artery, Coronary Artery, Umbilical Vein, Thyroid, Liver, Small Intestine, Duodenum, Colon, Ascending Colon, Bone Marrow, Lymph node, Tonsils, Thymus, Cartilage, Muscle, Brain, Cervix, Uterus, Vulva, Prostate, Testis, Lung, Bronchus, Urinary Bladder, Kidney, Skin, Epidermis, Dermis.
Additional utilities for NOVl 1 nucleic acids and polypeptides according to the invention are disclosed herein.
NOV12
A NOV 12 polypeptide has been identified as a Plasma Membrane Protein-like protein (also referred to as CG94282-01). The disclosed novel NOV12 nucleic acid (SEQ ID NO:25) of 8811 nucleotides is shown in Table 12 A. The novel NOV 12 nucleic acid sequences maps to the chromosome 12.
An ORF begins with an ATG initiation codon at nucleotides 1-3 and ends with a TAG codon at nucleotides 4378-4380. A putative untranslated region and/or downstream from the termination codon is underlined in Table 12 A, and the start and stop codons are in bold letters.
Table 12A. NOV12 Nucleotide Sequence (SEQ ID NO:25)
ATGCTGTTCAAGCTCCTGCAGAGACAAACCTATACCTGCCTGTCCCACAGGTATGGGCTCTACGTGTGCTTCTT GGGCGTCGTTGTCACCATCGTCTCCGCCTTCCAGTTCGGAGAGTGGGTAGAAGCCAGGGATCCTGCCAAACATC CTATAGTGCACAGGACAGCCCCTACAACAAAGAATCATCCAGCCCAAAATGTCGATAGTGCTGAAGTTGAGAAA TCCGGAATTAGAAGGGGCAAGAATGGCTGCAGGGCAGTTAGTCTACAGGACTGGCCTGGGACTAGAGGATGTGC CAATTTCACCTTCGCCTTCTGCCATGATTGTAAGTTTTCTGAGGTCTCCCAGAAACGCTTCCTGTACATCCTGC AGAACTGTCATTGGTTAACTGATTGGGGTTGGACTTGGTTGGCTCTGCTCCACGGGTCTCTCATCCTCCAGGGA CCAGCCAGCGAACCTGGTTGTGTTCTTCTCAAGGCAAAGGTGGTTCTGGAATGGAGCCGAGATCAATACCATGT TTTGTTTGATTCCTATAGAGACAATATTGCTGGAAAGTCCTTTCAGAATCGGCTTTGTCTGCCCATGCCGATTG ACGTTGTTTACACCTGGGTGAATGGCACAGATCTTGAACTACTGAAGGAACTACAGCAGGTCAGAGAACAGATG GAGGAGGAGCAGAAAGCAATGAGAGAAATCCTTGGGAAAAACACAACGGAACCTACTAAGAAGAGTGAGAAGCA GTTAGAGTGTTTGCTAACACACTGCATTAAGGTGCCAATGCTTGTCCTGGACCCAGCCCTGCCAGCCAACATCA CCCTGAAGGACCTGCCATCTCTTTATCCTTCTTTTCATTCTGCCAGTGACATTTTCAATGTTGCAAAACCAAAA AACCCTTCTACCAATGTCTCAGTTGTTGTTTTTGACAGTACTAAGGATGGGACATTGCTCACTCAGAAGGTGAC TTTTGAGTGGAAATGTGAAGAAGGTGAGGTAGCCAGCAATGCGAATATCTGGGGAAAGACTGATCTGGGTTCGC CCAGGAGGCCTTTGCCATGGCCTGTGGCCCTGGAGCCACCTAGGGCTCAGCTCAGCTCTGCCCTACAGATTCTC ACTAGGCCACGGGTATCTCAGGACAGAGCCAACACAAGTTATGAAATTAAACTAGACACACCCCTTCTTCGAGG TTACGCCAAGCCAGTGCCTGGGCCTGAAACTGGCCTGCAGCCCCTCAGCTTCGCCCACTGCCTTCCGACCCTGG ACCTTCGCAAAGTGAACGAGCTTCGGGACTTCGTGAAAATGTATAAGCAGGATCCGAGCATTCTGCATACCAAG GAAACGTGCTTTCTGAGGGAGCAGGTGGAGAGCATGGGGGAAAGCTATTATAAATCAGAAGAAAATATCAAGGA ATTAAAAACAGGTAGTAAGAAGGTGGAGGAAAACATAAGCACAGACGAACTATCAAGTGAGGAAAGTGATCTAG AAATTGATAACGAAGCTGTGATTGAACCAGACACTGATTCCCCTCAAGAAATGGGAGATGGAGAGGCCAGTGTA GCGCTTCTAAAACTGAATAACCCCAAGGATTTTCAAGAATTGAATAAGCAAACTAAGAAGAACATGACCATTGA TGGAAAAGAACTGACCATAAGTCCTGCATATTTATTATGGGATCTGAGCGCCATCAGCCAGTCTAAGCAGGATG AAGACATCTCTGCCAGTCGTTTTGAAGATAACGAAGAACTGAGGTACTCATTGCGATCTATCGAGAGGCATGCA CCATGGGTTCGGAATATTTTCATTGTCACCAACGGGCAGATTCCATCCTGGCTGAACCTTGACAATCCTCGAGT GACAATAGTAACACACCAGGATGTTTTTCGAAATTTGAGCCACTTGCCTACCTTTAGTTCACCTGCTATTGAAA GTCACATTCATCGCATCGAAGGGCTGTCCCAGAAGTTTATTTACCTAAATGATGATGTCATGTTTGGGAAGGAT GTCTGGCCAGATGATTTTTACAGTCACTCCAAAGGCCAGAAGGTTTATTTGACATGGCCTGTGCCAAACTGTGC CGAGGGCTGCCCAGGTTCCTGGATTAAGGATGGCTATTGTGACAAGGCTTGTAATAATTCAGCCTGCGATTGGG ATGGTGGGGATTGCTCTGGAAACAGTGGAGGGAGTCGCTATATTGCAGGAGGTGGAGGTACTGGGAGTATTGGA GTTGGACAGCCCTGGCAGTTTGGTGGAGGAATAAACAGTGTCTCTTACTGTAATCAGGGATGTGCGAATTCCTG GCTCGCTGATAAGTTCTGTGACCAAGCATGCAATGTCTTGTCCTGTGGGTTTGATGCTGGCGACTGTGGGCAAG AAAACTCAGACTCAAAGAATAGGAAAACAGAGGAAAAATGCCCAGTTAAAAAAAAAAAAATCATGTTTCTGTTT TTTCCTCTAGATCATTTTCATGAATTGTATAAAGTGATCCTTCTCCCAAACCAGACTCACTATATTATTCCAAA AGGTGAATGCCTGCCTTATTTCAGCTTTGCAGAAGTAGCCAAAAGAGGAGTTGAAGGTGCCTATAGTGACAATC CAATAATTCGACATGCTTCTATTGCCAACAAGTGGAAAACCATCCACCTCATAATGCACAGTGGAATGAATGCC ACCACAATACATTTTAATCTCACGTTTCAAAATACAAACGATGAAGAGTTCAAAATGCAGATAACAGTGGAGGT GGACACAAGGGAGGGACCAAAACTGAATTCTACAGCCCAGAAGGGTTACGAAAATTTAGTTAGTCCCATAACAC TTCTTCCAGAGGCGGAAATCCTTTTTGAGGATATTCCCAAAGAAAAACGCTTCCCGAAGTTTAAGAGACATGAT GTTAACTCAACAAGGAGAGCCCAGGAAGAGGTGAAAATTCCCCTGGTAAATATTTCACTCCTTCCAAAAGACGC CCAGTTGAGTCTCAATACCTTGGATTTGCAACTGGAACATGGAGACATCACTTTGAAAGGATACAATTTGTCCA AGTCAGCCTTGCTGAGATCATTTCTGATGAACTCACAGCATGCTAAAATAAAAAATCAAGCTATAATAACAGAT GAAACAAATGACAGTTTGGTGGCTCCACAGGAAAAACAGGTTCATAAAAGCATCTTGCCAAACAGCTTAGGAGT GTCTGAAAGATTGCAGAGGTTGACTTTTCCTGCAGTGAGTGTAAAAGTGAATGGTCATGACCAGGGTCAGAATC CACCCCTGGACTTGGAGACCACAGCAAGATTTAGAGTGGAAACTCACACCCAAAAAACCATAGGCGGAAATGTG ACAAAAGAAAAGCCCCCATCTCTGATTGTTCCACTGGAAAGCCAGATGACAAAAGAAAAGAAAATCACAGGGAA AGAAAAAGAGAACAGTAGAATGGAGGAAAATGCTGAAAATCACATAGGCGTTACTGAAGTGTTACTTGGAAGAA AGCTGCAGCATTACACAGATAGTTACTTGGGCTTTTTGCCATGGGAGAAAAAAAAGTATTTCCAAGATCTTCTC GACGAAGAAGAGTCATTGAAGACACAATTGGCATACTTCACTGATAGCAAAAATACTGGGAGGCAACTAAAAGA TACATTTGCAGATTCCCTCAGATATGTAAATAAAATTCTAAATAGCAAGTTTGGATTCACATCGCGGAAAGTCC CTGCTCACATGCCTCACATGATTGACCGGATTGTTATGCAAGAACTGCAAGATATGTTCCCTGAAGAATTTGAC AAGACGTCATTTCACAAAGTGCGCCATTCTGAGGATATGCAGTTTGCCTTCTCTTATTTTTATTATCTCATGAG TGCAGTGCAGCCACTGAATATATCTCAAGTCTTTGATGAAGTTGATACAGATCAATCTGGTGTCTTGTCTGACA GAGAAATCCGAACACTGGCTACCAGAATTCACGAACTGCCGTTAAGTTTGCAGGATTTGACAGGTCTGGAACAC ATGCTAATAAATTGCTCAAAAATGCTTCCTGCTGATATCACGCAGCTAAATAATATTCCACCAACTCAGGAATC CTACTATGATCCCAACCTGCCACCGGTCACTAAAAGTCTAGTAACAAACTGTAAACCAGTAACTGACAAAATCC ACAAAGCATATAAGGACAAAAACAAATATAGGTTTGAAATCATGGGAGAA.GAAGAAATCGCTTTTAAAATGATT CGTACCAACGTTTCTCATGTGGTTGGCCAGTTGGATGACATAAGAAAAAACCCTAGGATCTCACTCTGTTGTCC AAGCTGGAATGCAGTAATGCAAACATGGCTCACTGTAGCCTCGACCTCGTGGGCTCAAGCAATCCTCCCACCTC AGCCTCCTGACTAGTGGAACCACAGACATGAGCTGCTGCACCCAGCTAAAATGGAGTATTTTTAATTTCTGGGT CTTTTAAATGCATTTGGAGGTCTTTAGTTTTACCTCACTGAAa.TTAGGATTTTAATTATAAATAATCAAAGATG TGAACCTTACAGACATTTTAAAGCCATTATATTTTTTCTATAAfi.CCCTGTTCTCGTTTGGAGGAGAAAGAAfi.TT GGAATTTTCAAAAAAAATAAAAATACCTTTAACACCTATTTAGTGTCTTTAGTAATCCAGTAAAATACTTGATT TTTTACTAAATGTTTCCCACAAGCCAAGCAAACCATAAGCTACAATAATAATTACCTAGCGTACAGCCCTCTTT GCATATGCTGTTCCCTCCACTTGAAGTGTACTGTTTAATTTCTTAAfiATAACTTTAGCTTTTAAGAACCAATTT TGATGGGAGTACAGACTTCCCCCATTTTCTTGATGAGTTCTCTCCGTCATGTGTAGTAATAATGTGAGAATTTG CAGTTTTTAGTTGTAGCCTATACTTTTAGGTCTTTGTGCCAATTTGAAAGTTATTGGGTTAGAGTATTCATAGA CATTTTCATGGTACTTAAAGGGACAGGGGTTTAGTAAAAAGACACATGGCAAGCCAGGCTTTTTCCACAGTTTG CCAGGCCCAGCTGCCTCTTGTGTACCTGAACAGATTTTATCATTAACCCTTGTTTATGTTGTTTTGTTTTATTT CGACGAAGGCTTATTTTAAGTCAGGCATGGAAAACTAGACTTCAGACTGACTTCAGCTTTAAGGACATGTTTAT CCCGTTAACAGGGAGTCTGGGATAGACAATCTCCAGGCTTTGTTTTTCTCTGAATTTCTTAGCTCTGCTTGTGA TGGCTTCATCATCAGGCCACAGACCATTAACACATTCTAGAACTTTAACATTGGTTAAATAATACCATCTAATA GCCTGTCTTCAGCATTTCCCCAGTTGCCTCCAAATGCCCTTCATAGCTGTTCTCTGCCTCTGTTTGTTTTTAAT CCAAGATACACTCAAGGCTCATATATTAGGTTGACATAGCTCTTTAGTATCCTTTAATTTAAAGCAGTCTCCAG GTTTAGAGAAAGATGAATGAGCTTTCACATACCCCTCACTTGTCTTCTTCAGAAGTGTAGGCTACAACTAAAAC TTCCTTCTTCAGAAGGAAGACAAGTGATTTATATTTATTTACTTCCATTTCTATTTGACCTTGTTTCATCTAAA CACACCCACTCCACCACTGCTACCTGATTAATATTAAGTGAATCTCAAACATTGTATCATTTTAGCTCCACGTT TTTTGTATGTATCTCCAAAATATAAAGATTCTTAAAAATATAACCACAATACCATTATCACCCTAAAAAAATCA ATAATGATTCCTTAATATGACCAACTACTTTGTCAATGTACACCTTTCACTCCTCTTAACTTTCATAAAGACTT ATGTGTTTTTTTGGTTTTTAAGTTTGTTGGTTTGAAGTTAAATCTATGGGTTTTCCCTCCATCTCTCTTTTTTT AACCGTATAATTTTTTGCATGTGTATATGAATAAATCTGATTATAGATTCTATAGCTATCTTACACTTTGTCCC TCTCTGATTGAATCCTAGTTAACAAGTTTCTATGTCTCTTGTATTTCCCATAAATTGGTAGTTGGATCTGAAGG CTTTATCAGGTTTGTTTGATTTTTTTTTTTTTAATTTTGGCGAATCTACTTCAAAAGTATTGGCCTACCTACAA GCCACTTTAATGGGCCCTTAGTTTAGTGACCTTTGCCTTGAAAGGAACTTGAAACAAGCAAGGAAGCACCACTG TAATCTGCTTTTTTGCCAGAACTGTAGCATCTTACAGCTTGGTTAGAGACATAGTAAGCAGAAATTATCAAATT CATATAATCTGTAGCTATAAGGCACTGTCTCTCTCTCTCAATTATTTACATGATTTTTCTTTGTAATATAACTA TCATTTCAGAGAACTTGGTTTTGATTTTTTTTTTTTAATCTTTTTGAGACAGAGTCTCGCTTTATCACCCAGGC TGGAGTGCAGTGGTGCAATCTAAAGATTGCTGACTGCAACCTCTGCCTCCCGAGTTCAGCAATTCTAGTGCCTC AGCCTCTCGAGTAGCTGGGATTACAGGCATGCCACCACACCCGGCTAATTTTTTTGTATTTTTAGTAAAGACAG GGTTTCACCATGTTGGCTAGGCTGGTCTCAAATTTTTGACCTCAAGTAATCAGCCTACCTTGATCTCCCAAAGT GCTGGGATTACAGGCATGAGCCACCATGCATGGCCTTCAGAGAACTTGGTTTTAGGTACTTACGGATTGTCTTT CTTTTTTTTCCTCACTGCAGCCTCTCCCTCCCAGGTTCAAGCGATTCTCCTACCTCAGCTTCCTGAAGAGCTGG GACCACAGGAAGTTTGTTTGCCTGAATGACAACATTGACCACAATCATAAAGATGCTCAGACAGTGAAGGCTGT TCTCAGGGACTTCTATGAATCCATGTTCCCCATACCTTCCCAATTTGAACTGCCAAGAGAGTATCGAAACCGTT TCCTTCATATGCATGAGCTGCAGGAATGGAGGGCTTATCGAGACAAATTGAAGTTTTGGACCCATTGTGTACTA GCAACATTGATTATGTTTACTATATTCTCATTTTTTGCTGAGCAGTTAATTGCACTTAAGCGGAAGATATTTCC CAGAAGGAGGATACACAAAGAAGCTAGTCCCAATCGAATCAGAGTATAGAAGATCTTCATTTGAAAACCATCTA CCTCAGCATTTACTGAGCATTTTAAAACTCAGCTTCACAGAGATGTCTTTGTGATGTGATGCTTAGCAGTTTGG CCCGAAGAAGGAAAATATCCAGTACCATGCTGTTTTGTGGCATGAATATAGCCCACTGACCAGGAATTATTTAA CCAACCCACTGAAAACTTGTGTGTTGAGCAGCTCTGAACTGATTTTACTTTTAAAGAATTTGCTCATGGACCTG TCATCCTTTTTATAAAAAGGCTCACTGACAAGAGACAGCTGTTAATTTCCCACAGCAATCATTGCAGACTAACT TTATTAGGAGAR.GCCTATGCCAGCTGGGAGTGATTGCTAAGAGGCTCCAGTCTTTGCATTCCAAAGCCTTTTGC TAAAGTTTTGCACTTTTTTTTTTTCATTTCCCATTTTTAAGTAGTTACTAAGTTAACTAGTTATTCTTGCTTCT GAGTATAACGAATTGGGATGTCTAAACCTATTTTTATAGATGTTATTTAAATAATGCAGCAATATCACCTCTTA TTGACAATACCTAAATTATGAGTTTTATTAATATTTAAGACTGTAAATGGTCTTAAACCACTAACTACTGAAGA GCTCAATGATTGACATCTGAAATGCTTTGTAATTATTGACTTCAGCCCCTAAGAATGCTATGATTTCACGTGCA GGTCTAATTTCAAAGGGCTAGAGTTAGTACTACTTACCAGATGTAATTATGTTTTGGAAATGTACATATTCAAA CAGAAGTGCCTCATTTTAGAAATGAGTAGTGCTGATGGCAGTGGCACATTACAGTGGTGTCTTGTTTAATACTC ATTGGTATATTCCAGTAGCTATCTCTCTCAGTTGGTTTTTGATAGAACAGAGGCCAGCAAACTTTCTTTGTAAA AGGCTGGTTAGTAAATTATTGCAGGCCACCTGTGTCTTTGTCATACATTCTTCTTGCTGTTGTTTAGTTTGTTT TTTTTCAAACAACCCTCTAAAAATGTAAAAACCATGTTTAGCTTGCAGCTGTACAAAAACTGCCCACCAGCCAG ATGTGACCCTCAGGCCATCATTTGCCAATCACTGAGAATTAGTTTTTGTTGTTGTTGTTGTTGTTGTTTTTGAG ACAGAGTCTCTCTCTGTTGCCCAGGCTGGAGTGCAGTGGCGCAATCTCAGCTCACTGCAACCTCCGCCTCCCGG GTTCAAGCAGTTCTGTCTCAGCCTTCTGAGTAGCTGGGACTACAGGTGCATGCCACCACACCCTGCTAATTTTT GTATTTTTAGTAGAGACGGGGGTTCCACCATATTGGTCAGGCTTATCTTGAACTCCTGACCTCAGGTGATCCAC CTGCCTCTGCCTCCCAAAGTGCTGAGATTACAGGCATAAGCCAGTGCACCCAGCCGAGAATTAGTATTTTTATG TATGGTTAAACCTTGGCGTCTAGCCATATTTTATGTCATAATACAATGGATTTGTGAAGAGCAGATTCCATGAG TAACTCTGACAGGTATTTTAGATCATGATCTCAACAATATTCTTCCAAAATGGCATACATCTTTTGTACAAAGA ACTTGAAATGTAAATACTGTGTTTGTGCTGTAAGAGTTGTGTATTTCAAAAACTGAAATCTCATAAAAAGTTAA ATTTT
Variant sequences of NOV12 are included in Example 3, Table 24. A variant sequence can include a single nucleotide polymorphism (SNP). A SNP can, in some instances, be referred to as a "cSNP" to denote that the nucleotide sequence containing the SNP originates as a cDNA.
The NOV12 protein (SEQ ID NO:26) encoded by SEQ ID NO:25 is 1459 amino acid residues in length and is presented using the one-letter amino acid code in Table 12B. Psort analysis predicts the NOV12 protein of the invention to be localized at the plasma membrane with a certainty of 0.6500.
Table 12B. Encoded NOV12 protein sequence (SEQ ID NO:26)
MLFKLLQRQTYTCLSHRYGLYVCFLGVWTIVSAFQFGEWVEARDPAKHPIVHRTAPTTK HPAQ l^SAEVEKSGIRRGKNGCRAVSLQD PGTRGCANFTFAFCHDCKFSEVSQKRFLYI QNCHWLT D G TWLALLHGSLI QGPASEPGCVLLKAKWLE SRDQYHVLFDSYRDNIAGKSFQNRLCLPM PIDWYT V GTDLELLKE QQVREQMEEEQKAMREI GKNTTEPTKKSEKQLECLLTHCIKVPM VLDPA PANIT D PSLYPSFHSASDIF1WAKPKNPSTNVSVVVFDSTKDGT LTQKVTFEW CEEGEVASNANI GKTDLGSPRRPLPWPVALEPPRAQ SSALQI TRPRVSQDRANTSYEIKLDT P LRGYAKPVPGPETGLQPLSFAHCLPT DLRKVNELRDFVKMYKQDPSILHTKETCFLREQVES MGESYYKSEENIKEL TGSKKVEENISTDELSSEESDLEIDNEAVIEPDTDSPQEMGDGEASVAL LKLNNPKDFQE NKQTKKNMTIDGKELTISPAY LWDLSAlSQSKQDEDISASRFEDNEΞLRYSL RSIERHAP VRNIFIVTNGQIPSWLN DNPRVTIVTHQDVFRN SHLPTFSSPAIESHIHRIEG SQKFIYL1TODVMFGKDVWPDDFYSHSKGQKVYLTWPVPNCAEGCPGS IKDGYCDKACNNSACD DGGDCSGNSGGSRYIAGGGGTGSIGVGQPWQFGGGINSVSYCNQGCANS LADKFCDQACNV SC GFDAGDCGQENSDSKNRKTEEKCPVKKK IMF FFP DHFHELYKVILLPNQTHYIIPKGEC PY FSFAEVAKRGVEGAYSDNPIIRHASIANKWKTIHLIMHSGM ATTIHFNLTFQNT DEEFKMQIT VEVDTREGPK NSTAQKGYENLVS IT LPEAEILFEDIPKEKRFPKFKRHDVMSTRRAQEEVKI P VNISLLPKDAQLSLNT DLQ EHGDIT KGYN SKSA LRSFLMNSQHAKI NQAIITDETND SLVAPQEKQVH SILPNSLGVSERLQRLTFPAVSVKV GHDQGQNPP DLETTARFRVETHTQKT IGGNVT EKPPSLIVPLESQMTKEKKITGKEKENSRMEENAENHIGVTEVLLGRK QHYTDSYLG FLP EKKKΪFQDLLDEEES KTQLAYFTDSKNTGRQ KDTFADS RYVNKILNSKFGFTSRKVPA HMPHMIDRIVMQELQDMFPEEFDKTSFHKVRHSEDMQFAFSYFYY MSAVQP NISQVFDEVDTD QSGVLSDREIRTLATRIHELPLSLQD TGLEHMLINCSKMLPADITQLNWIPPTQESYYDPNLPP VTKSLVTNCKPVTDKIHKAYKDKNKYRFEIMGEEEIAFKMIRTNVSHVVGQLDDIRKNPRISLCC PSWNAVMQTWLTVASTSWAQAILPPQPPD
A search against the Patp database, a proprietary database that contains sequences published in patents and patent publications, yielded several homologous proteins shown in Table 12C.
Table 12C. Patp results for NOV12
Smallest Sum
Reading High Prob
Sequences producing High-scoring Segment Pairs: Frame Score P(N)
>patp:ABB30279 Peptide #2930 encoded by breast cell +1 1900 7.5e-196
>patp:AAM56268 Human brain expressed single exon probe +1 1900 7.5e-196
>patp:AAM16457 Peptide #2891 encoded by probe +1 1900 7.5e-196
>patp:AAM28952 Peptide #2989 encoded by probe +1 1900 7.5e-196
>patp:AAM0 186 Peptide #2868 encoded by probe +1 1900 7.5e-196
In a BLAST search of public sequence databases, it was found, for example, that the nucleic acid sequence of this invention has 6444 of 6447 bases (99%) identical to a gb:GENBANK-ID:AB033034|acc:AB033034.1 mRNA from Homo sapiens (mRNA for
KIAA1208 protein, partial eds). The full amino acid sequence of the protein of the invention was found to have 663 of 663 amino acid residues (100%) identical to, and 663 of 663 amino acid residues (100%) similar to, the 663 amino acid residue ptnr:SPTREMBL-ACC:Q9ULL2 protein from Homo sapiens (KIAA1208 PROTEIN). NOV 12 also has homology to the proteins shown in the BLASTP data in Table 12D.
Figure imgf000096_0001
Figure imgf000097_0004
A multiple sequence alignment is given in Table 12E, with the NOV12 protein being shown on line 1 in Table 12E in a ClustalW analysis, and comparing the NOV 12 protein with the related protein sequences shown in Table 12D. This BLASTP data is displayed graphically in the ClustalW in Table 12E.
Table 12E. ClustalW Analysis of NOV12
1) > NOV12; SEQ ID NO:26
2) > gi|6382022|/ KIAA1208 protein [Homo sapiens]; SEQ ID NO:87
3) > gi|16551459|/ unnamed protein product [Homo sapiens]; SEQ ID NO:88
4) > gi|2137411|/ hypothetical protein - mouse (fragment); SEQ ID NO:89
5) > gijl 1360271|/ hypothetical protein DKFZp762B226.1 - human (fragment); SEQ ID NO:90
6) > gi|7303923|/ CG8027 gene product [Drosophila melanogaster]; SEQ ID NO:91
10 20 30 40 50 60
NOV12 1 M FKLLQRQTYTCLSHRYGLYVCF GλrWTIVSAFQFGEWVEARDPAKHPIVHRTA 56 gi I 6382022 I 1 RKTEEKCPVKKKKIMFLFFPLDHFHE YKVILLPNQTHYIIPKGECLP 48 gi 116551459 I 1 M FKLLQRQTYTCLSHRYG YVCF GVWTIVSAFQF 37 gi j 2137411 1 gi j 11360271 1 gi J 7303923 |
6
Figure imgf000097_0001
130 140 150 160 170 180
NOV12 117 KRFLYILQNCHWLTD GWTW ALLHGS ILQGPASEPGCVLLKAKWLE SRDQYHVIJFD 176
48 YFSFAEVA--KRGVEG 62 | 38 ΞW E SRDQYHV FD 54 |
Figure imgf000097_0002
36 21 14 0
Figure imgf000097_0003
Figure imgf000098_0001
91
Figure imgf000099_0001
970 980 990 1000 1010 1020
Figure imgf000099_0002
1090 1100 1110 1120 1130 1140 1
Figure imgf000099_0003
1150 1160 1170 1180 1190 1200
N0V12 1132 ITGKEKENSRMEENAENHIGVTEVLLGRKLQHYTDSYLGFLPWEKKKYFQDLLDEEESLK 1191 gi|6382022| 663 663 gi | l6551459 | 847 847 gi j 2137411 1 384 384 gi | H36027l | 248 248 gi j 7303923 I 598 YRNRFESWRDFQR KRRKRAVLVIGYGVSLLLWCLLRFMCHHKAKLVRRCVQRL 652
The NOV 12 Clustal W alignment shown in Table 12E was modified to end at amino residue 1200. The data in Table IE includes all of the regions overlapping with the NOV12 protein sequences.
NOV12 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts. For example, NOV12 nucleic acids and polypeptides can be used to identify proteins that are members of the Plasma Membrane Protein-like Protein Family. The NOVl 2 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOVl 2 activity or function. Specifically, the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation and signal transduction. These molecules can be used to treat, e.g., DiabetesNon Hippel-Lindau (VHL) syndrome , Pancreatitis, Obesity, Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus , Pulmonary stenosis, Subaortic stenosis, Ventricular septal defect (VSD), valve diseases, Tuberous sclerosis, Scleroderma, Obesity, Transplantation, Von Hippel-Lindau (NHL) syndrome, Cirrhosis, Transplantation, Non Hippel-Lindau (NHL) syndrome , Alzheimer's disease, Stroke, Tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, Cerebral palsy, Epilepsy, Lesch-Νyhan syndrome, Multiple sclerosis, Ataxia-telangiectasia, Leukodystrophies, Behavioral disorders, Addiction, Anxiety, Pain, Neuroprotection as well as other diseases, disorders and conditions.
In addition, various NOV 12 nucleic acids and polypeptides according to the invention are useful, ter alia, as novel members of the protein families according to the sequence relatedness to previously described proteins. The NON12 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of cardiac, nerve, and immune physiology. As such, the ΝON12 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat cardiovascular, immune, and nervous system disorders, e.g., DiabetesNon Hippel-Lindau (VHL) syndrome, Pancreatitis, Obesity, Cardiomyopathy, Atherosclerosis, Hypertension, Congenital heart defects, Aortic stenosis, Atrial septal defect (ASD), Atrioventricular (A-V) canal defect, Ductus arteriosus , Pulmonary stenosis, Subaortic stenosis, Ventricular septal defect (VSD), valve diseases, Tuberous sclerosis, Scleroderma, Obesity, Transplantation, Von Hippel-Lindau (VHL) syndrome, Cirrhosis, Transplantation, Von Hippel-Lindau (VHL) syndrome , Alzheimer's disease, Stroke, Tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, Cerebral palsy, Epilepsy, Lesch- Nyhan syndrome, Multiple sclerosis, Ataxia-telangiectasia, Leukodystrophies, Behavioral disorders, Addiction, Anxiety, Pain, Neuroprotection as well as other diseases, disorders and conditions.
The NOV12 nucleic acids and polypeptides are useful for detecting specific cell types. For example, expression analysis has demonstrated that a NOV 12 nucleic acid is expressed in Pancreas, Uterus, Epidermis, Heart, Coronary Artery, Adrenal Gland/Suprarenal gland, Pancreas, Parathyroid Gland, Salivary Glands, Liver, Small Intestine, Bone Marrow, Peripheral Blood, Lymphoid tissue, Lymph node, Cartilage, Brain, Hypothalamus, Spinal Chord, Mammary gland/Breast, Uterus, Prostate, Testis, Lung, Kidney, Epidermis, Hair Follicle.
Additional utilities for NOV12 nucleic acids and polypeptides according to the invention are disclosed herein.
NOV13
A NOVl 3 polypeptide has been identified as a BHLH Factor MATH6-like protein (also referred to as CG94399-01). The disclosed novel NOV13 nucleic acid (SEQ ID NO:27) of 2244 nucleotides is shown in Table 13 A. The novel NOV 13 nucleic acid sequences maps to the chromosome 2.
An ORF begins with an ATG initiation codon at nucleotides 105-107 and ends with a TGA codon at nucleotides 1062-1064. A putative untranslated region and/or downstream from the termination codon is underlined in Table 13 A, and the start and stop codons are in bold letters.
Table 13A. NOV13 Nucleotide Sequence (SEQ ID NO:27)
ACGCGTGAAGGGCGGGCGAAGCGGGAGAGCCAGAGACTCCTCGGCGCTGAGCGCGGCGGCGGCCCGG GCAGCCCCACGCCCCTGCCTCGCGCGCCGCCCGCGCCATGAAGCACATCCCGGTCCTCGAGGACGGG CCGTGGAAGACCGTGTGCGTGAAGGAGCTGAACGGCCTTAAGAAGCTCAAGCGGAAAGGCAAGGAGC CGGCGCGGCGCGCGAACGGCTATAAAACTTTCCGACTGGACTTGGAAGCGCCCGAGCCCCGCGCCGT AGCCACCAACGGGCTGCGGGACAGGACCCATCGGCTGCAGCCGGTCCCGGTACCGGTCCGGTGCCAG TCCCAGTGGCGCCGGCCGTTCCCCCAAGAGGGGGCACGGACACAGCCGGGGAGCGCGGGGGCTCTCG GGCGCCCGAGGTCTCCGACGCGCGGAAACGTGCTTCGCCCTAGGCGCAGTGGGGCCAGGACTCCCCA CGCCGCCGCCGCCGCCGCCTCCTGCGCCCCAGAGCCAGGCACCTGGGGGCCCAGAGGCACAGCCTTT CGGGAGCCGGGTCTGCGTCCTCGCATCTTGCTGTGCGCACCGCCCGCGCCCCGCGCCGTCAGCACCC CCAGCACCGCCAGCGCCCCCGGAGTCCACTGTGCGCCCTGGCCCCCGACGCGCCCCGGGGAAAGTTC CTACTCGTCAATTTCACACGTAATTTACAATAACCACCAGGATTCCTCCGCGTCGCCTAGGAAACGA CCGGGCGAAGCGACTGCCGCCTCCTCCGAGATCAAAGCCCTGCAGCAGACCCGGAGGCTCCTGGCGA ACGCCAGGGAGCGGACGCGGGTGCACACCATCAGCGCAGCCTTCGAGGCGCTCAGGAAGCAGGTGCC GTGCTACTCATATGGGCAGAAGCTGTCCAAACTGGCCATCCTGAGGATCGCCTGTAACTACATCCTG TCCCTGGCGCGGCTGGCTGACCTTGACTACAGTGCCGACCACAGCAACCTCAGCTTCTCCGAGTGTG TGCAGCGCTGCACCCGCACCCTGCAGGCCGAGGGACGTGCCAAGAAGCGCAAGGAGTGACTGGCTGC AGGCAAGACCAAGGCCACCACTGTGGGCCCTCCTTCCAGTCAGGCCTGAGGACAAGGTGAGCTCGCT GAGTCCAGCCTCGTGGTCTTCTCCAAGATGGCGCCCCACTTGGAGCCTACAGCCTCTCAGGGTCGGA TCGGAGCACGCCTGCCTCCCTCTCCCCTCCGCCCTCACCCAGCCAATCCGAGGCTGGTTCGCACGTT GCCCTCTGCCTGGTGGGGAGGGGAGAGCTCAGCCCCCGACTCACTCAGACCCCAAGGCCCACTGTCC AGCTGCAGAAATTCGTTGCCAAAGATTGGACAGAGACACCGAAGGAAATGGGGTGGTGAAACCCCAC AGCGAAAAGCCACACCGTTGCTCTGTGACTTTTGCTCCTCCTGTTGCCTGAGCCCCATCTCAAGCCA AAGATGAGTCAGTGGTTCTGCTAGGAACTCATGGAATGGATGGGCATTTGATGACCCCTGGGGGTCA TCTTGGCCCTCTGACCTGGTGCTCTCTCTCCACTGGGCCTTGTGCTGGTTGAGTGCAAGACAAGCCT TAGGGGCTGTGAGAGGGAGGCTGGGGTGCCTGGGCGGGGCTGGGAGTGGGACCTGAGATCCCTGCCC ACTCTCTCCCCTTCATTGGCTTGCCCAGGGCACTGGCCCCAGTTCTCAGTGTCCCTTGGGGTCCAGG CTCCTTGGGCCCTAAGCATCACCAGAAGGGAGTAAGCAGGGAGAGAAGCAATATTACTCCCTCCCCT ACACCAGGGACTTGCCCCAGGGCGGCTACCTATGGGTCTTTGCTTCCCCAGCCAGCCTCTCCTCACT GTGACCCACCCCCATGGGCCCCCGTCCCAGGCAGCCAGCACCATGGGCAGGCCCTGCCATGGACAGA AAAAGAGTTTTTCTCTTGTTCAGCCTGCACGTGGCCTGAGGAAGGAGTAGAGGCTGGGTTGGCTGGA GCCGTCCTACTGGGCAAGATGGCGCCCCACTTGGAGGGCGGTGGTCTGTTACAGGGTGTGCAGGGGC AGAGAAGGAAGGGACCAGGGGACTGGGCCAGTATGTGGAGGATGGGGCCTGCGTGTTCAAAGCCAAG GCCCGCCCCTTCCTTGTGCTCAAATGGCCAAAGCTGTTCACGTCTGTGCTCAACCATCTGCTTCAAA TTGAAGTAAAAGCCCCAAAATGTCAAGAAAAAA
Variant sequences of NOV13 are included in Example 3, Table 25. A variant sequence can include a single nucleotide polymorphism (SNP). A SNP can, in some instances, be referred to as a "cSNP" to denote that the nucleotide sequence containing the SNP originates as a cDNA.
The NOV13 protein (SEQ D NO:28) encoded by SEQ ID NO:27 is 319 amino acid residues in length and is presented using the one-letter amino acid code in Table 13B. Psort analysis predicts the NOVl 3 protein of the invention to be localized at the nucleus with a certainty of 0.7000.
Table 13B. Encoded NO 13 protein sequence (SEQ ID NO:28)
MKHIPV EDGP KTVCVKELNGLKKLKRKGKEPARRANGYKTFRLDLEAPEPRAVATNGLRDRTH RLQPVPVPVRCQSQWRRPFPQEGARTQPGSAGALGRPRSPTRGVRPRRSGARTPHAAAAAASC APEPGTWGPRGTAFREPGLRPRI CAPPAPRAVSTPSTASAPGVHCAPWPPTRPGESSYSSISH VIYNHQDSSASPRKRPGEATAASSEIKALQQTRRLLAARERTRVHTISAAFEALRKQVPCYSY GQKLSKLAI RIACNYILSLARLADLDYSADHSN SFSECVQRCTRT QAEGRA KRKE
A search against the Patp database, a proprietary database that contains sequences published in patents and patent publications, yielded several homologous proteins shown in Table 13C. Table 13C. Patp results for NOV13
Smallest Sum
Reading High Prob
Sequences producing High-scoring Segment Pairs: Frame Score P(N)
>patp:AAM93743 Human polypeptide, SEQ ID NO: 3717 +1 1197 2.3e-121
>patp:AAB95274 Human protein sequence SEQ ID NO: 17476 +1 1197 2.3e-121
>patp:AAU16607 Human novel secreted protein, Seq ID 1560 4-1 534 4.2e-51
>patp:ABG00300 Novel human diagnostic protein #291 +1 334 6.8e-33
>patp:ABG00300 Novel human diagnostic protein #291 +1 334 6.8e-33
hi a BLAST search of public sequence databases, it was found, for example, that the nucleic acid sequence of this invention has 372 of 657 bases (56%) identical to a gb:GENBANK-ID:HSBBICP4A|acc:L14320.1 mRNA from Bovine herpesvirus 1 (Bovine herpesvirus type 1 early-intermediate transcription control protein (BICP4) gene, complete eds). The full amino acid sequence of the protein of the invention was found to have 238 of 322 amino acid residues (73%) identical to, and 244 of 322 amino acid residues (75%) similar to, the 322 amino acid residue ρtnr:TREMBLNEW-ACC:BAB39468 protein from Mus musculus (BHLH FACTOR MATH6)..
NOV 13 also has homology to the proteins shown in the BLASTP data in Table 13D.
Table 13D. BLAST results for NOV13
Gene Index/ Protein/ Organism Length Identity Positives Expect Identifier (aa) (%) (%)
14249530 | ref | P_ hypothetical 321 246/321 246/321 2e-93
116216. l| (NM 032827 protein FLJ14708 (76%) (76%) ) [■HOπio sapiens] gi 113383235 |dbj |BAB bHLH factor Math6 322 233/329 240/329 3e-86 39468. l| (AB049066) [Mus musculus] (70%) (72%) gi 17864454 I ref |NP_ net [Drosophila 365 55/97 76/97 5e-21
524820. ll (NM 080081 melanogaster] (56%) (77%)
) gi I 7296271 |gb|AAF51 CG11450 gene 261 55/97 76/97 2e-20 562.1| (AE003590) product (56%) (77%) [Drosophila mel anogaster] gi 118858289 I ref |NP_ atonal homolog 2a 325 36/82 51/82 2e-10 571891. ll (NM 131816 [Danio rerio] (43%) (61%)
A multiple sequence alignment is given in Table 13E, with the NOVl 3 protein being shown on line 1 in Table 13E in a ClustalW analysis, and comparing the NOVl 3 protein with the related protein sequences shown in Table 13D. This BLASTP data is displayed graphically in the ClustalW in Table 13E.
Table 13E. ClustalW Analysis of NOV13
1) > N0V13; SEQ 1 N0:28
2) > gij 14249530]/ hypothetical protein FLJ14708 [Homo sapiens]; SEQ ID NO:92 3) > gi|13383235|/ bHLH factor Math6 [Mus musculus]; SEQ ID NO:93
4) > gι]17864454|/ net [Drosophila melanogaster]; SEQ ID NO:94
5) > gi|7296271|/ CGI 1450 gene product [Drosophila melanogaster]; SEQ ID NO:95
6) > gi|18858289|/ atonal homolog 2a [Danio rerio]; SEQ ID NO:96
Figure imgf000104_0001
The presence of identifiable domains in the protein disclosed herein was determined by searches using algorithms such as PROSITE, Blocks, Pfam, ProDomain, Prints and then determining the Interpro number by crossing the domain match (or numbers) using the Interpro website (http:www.ebi.ac.uk/interpro/). Table 13F lists the domain description from DOMAIN analysis results against NOVl 3.
Figure imgf000105_0001
Consistent with other known members of the BHLH Factor MATH6-like family of proteins, NOV 13 has, for example, a Helix-loop-helix domain and a Helix-loop-helix DNA binding domain (HLH) signature sequence as well as homology to other members of the BHLH Factor MATH6-like Protein Family. NOV 13 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts. For example, NOV 13 nucleic acids and polypeptides can be used to identify proteins that are members of the BHLH Factor MATH6-like Protein Family. The NOV13 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOVl 3 activity or function. Specifically, the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular replication, and signal transduction. These molecules can be used to treat, e.g., DiabetesNon Hippel-Lindau (VHL) syndrome , Pancreatitis, Obesity, Inflammatory bowel disease, Diverticular disease, Non Hippel-Lindau (NHL) syndrome , Alzheimer's disease, Stroke, Tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, Cerebral palsy, Epilepsy, Lesch-Νyhan syndrome, Multiple sclerosis, Ataxia-telangiectasia, Leukodystrophies, Behavioral disorders, Addiction, Anxiety, Pain, Neuroprotection, Systemic lupus erythematosus , Autoimmune disease, Asthma, Emphysema, Scleroderma, allergy as well as other diseases, disorders and conditions.
In addition, various NOV13 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. For example, the NOVl 3 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of protems belonging to the BHLH Factor MATH6-like Protein Family.
A number of eukaryotic proteins, probably sequence specific DNA- binding proteins that act as transcription factors belong to this family. They share a conserved domain that is formed of two amphipathic helices joined by a variable length linker region that could form a loop (Littlewood and Evan, Protein Prof. 2: 621-702 (1995).) This 'helix-loop-helix' (HLH) domain mediates protein dimerization and has been found in a large variety of proteins (Garrell and Campuzano, Bioessays 13: 493-498 (1991); Kato and Dang, FASEB J. 6: 3065- 72 (1992).) Most of these proteins have an short basic region adjacent to the HLH domain that specifically binds to DNA. They are referred as basic helix-loop-helix proteins (bHLH), and are classified in two groups: class A (ubiquitous) and class B (tissue-specific). The HLH proteins lacking the basic domain (Emc, Id) function as negative regulators since they form heterodimers, but fail to bind DNA. The hairy-related proteins (hairy, E(spl), deadpan) also repress transcription although they can bind DNA. The proteins of this subfamily act together with co-repressor proteins, like groucho, through their C-terminal motif WRPW.
MATH6 (Inoue, et al, Genes to Cells 6: 977-86 (2001)) is a distant homolog of Drosophila proneuronal gene Atonal Murine expression is higest in developing nervous system (ventricular zone and mantle layer, spinal cord, dorsal root ganglia). MATH6 is expressed by neuronal precursor cells and designated neurons, e.g., cerebellar Purkinje cells. The closest mammalian homolog to MATH6 is NeuroD. NeuroD point mutations and
NeuroD gene knockout animals have severe diabetes and die perinatally. The NeuroD knockout animals lack beta-Islet cells and could not be rescued with insulin administration. Also, the NeuroD knockout animals are deaf due to a loss of inner ear sensory neurons. The NON 13 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of metabolism as well as nerve and immune physiology. As such, the ΝON13 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat metabolic, hearing, nervous system, immune disorders, e.g., DiabetesNon Hippel-Lindau (NHL) syndrome , Pancreatitis, Obesity, Inflammatory bowel disease, Diverticular disease, Non Hippel-Lindau (NHL) syndrome , Alzheimer's disease, Stroke, Tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, Cerebral palsy, Epilepsy, Lesch-Νyhan syndrome, Multiple sclerosis, Ataxia-telangiectasia, Leukodystrophies, Behavioral disorders, Addiction, Anxiety, Pain, Neuroprotection, Systemic lupus erythematosus , Autoimmune disease, Asthma, Emphysema, Scleroderma, allergy as well as other diseases, disorders and conditions.
The NON13 nucleic acids and polypeptides are useful for detecting specific cell types. For example, expression analysis has demonstrated that aΝON13 nucleic acid is expressed in Pancreas, Umbilical Nein, Small Intestine, Cartilage, Synovium Synovial membrane, Brain, Placenta, Oviduct/Uterine Tube/Fallopian tube, Lung, Brain, Uterus.
Additional utilities for ΝON13 nucleic acids and polypeptides according to the invention are disclosed herein.
ΝOV14
A NOV14 polypeptide has been identified as a Putative Protein-Tyrosine Phosphatase- like protein (also referred to as CG94366-01). The disclosed novel NOV14 nucleic acid (SEQ ID NO:29) of nucleotides is shown in Table 14A. The novel NOV14 nucleic acid sequences maps to the chromosome 22.
An ORF begins with an ATG initiation codon at nucleotides 248-250 and ends with a TAA codon at nucleotides 1679-1681. A putative untranslated region and/or downstream from the termination codon is underlined in Table 14 A, and the start and stop codons are in bold letters.
Table 14A. NOV14 Nucleotide Sequence (SEQ ID NO:29)
ATTGAGTTTGAAATAACTGCCACCACAAAGTCTGTCACACATTGAGACTGAGGTCATAATAAAGAGG TTTACTTAAATAGGGAAGCATTACTATTTTCCCCCGCCTAAGATTTTGGTTGTCGCCATATAAATCC TCATTTCTAATAAAGAGAAAAAGACATTCCAGGTTCCAATAGTGCTATACACATGAATAGTCAGAAA TTAATTGGTTTCTGTCTAGAATAATGAAAAGTAATTTTTCCAAAATATGAATTCAGAATTAAGTCTC CTCTCTGACTGTTTTCTCTTATCATCCGCTAGTCCACAGACAAACGAATTTAAAGGAGCAACCGAGG AGGCACCTGCGAAAGAAAGCCCACACACAGGTGAATTTAAAGGAGCAGCCCTGGTGTCACCTATCAG TAAAAGAATGTTAGAACGACTTTCCAAGTTTGAAGTTGGAGATGCTGAAAATGTTGCTTCATATGAC AGCAAGATTAAGAAAATTGTTCATTCAATTGTGTCATCCTTTGCAGTTGGGATATTTGGAGTTTTCC TGATCTTGTTGGATGTGGCTCTGATCTTTGCTGACCTAATTTTCACTGATAGCAAAGTTTATATTCC TTTGGAGTATCGTTCTATTTCTCTAGCTATTGCTTTATTTTTTCTCATGGATGTTCTTCTTCGAGTA TTTGTAGAAAGGAGACAGCATTATTTTTCTGATTTACTTAACATTTTAGATACTGCCGTTACTGTGA TTATTCTGCTGGTTGATGTCGTTTACATTTTTTTTGACGTTAAGTTTCTTAAGGATATTCCCAGATG GACACGTTTATTTCGACTTCTACGACTTATCATTCTGATAAGAGTTTTTCGTCTGGCTCATCTAAAA AGACAACTTGGAAAGCTGATAAGAAGGCTGGTAAGTAGGNGATACGAAAGGGATGGATTTGACCTAG ACCTCACTTATATTACAGAACGTATTGTCGCTATGTCATTTCCATCTTCGGGAGGCCAGTCTTTCTA TCGGAATCCAATTAAGGAAGTCGTACAGTTTCTAGACAAGAAACATCCAAACCACTATCGAGTCTAC AATCTATGCAGTGAAAGAGCTTATGATCCTAAGCACTTCCATAATAGGGTCAGTAGAATCATGATCG ATGATCATAATGTCCCCACTCTAAGGGAGATGGTAGCATTCTCCAAGGAAGTGTTGGAGTGGATGGC TCAAGATTCTGAAAACATCGTAGTGATTCACTGTAAAGGAGGCAAAGGTAGAACCGGAACTATGGTT TGTGCCTGCCTGATTGCCAGTGAAATATTTTTAACTGCAGAGGAAAGATTGTACTATTTTGGAGAAC GGCGAACAGATAAAACCAATGGCACTAAATATCAGGGAGTAGAAACTCCTTCTCAGAATAGATATGT TGGATATTTTGCACAAGTGAAACATAGCTACAACTGGAATCTCCCTCCAAGAAAAACACTGTTTATA AAAAGATTAGTTATTTATTCGATTCATGGTAAGTGTTTAGATCTAAAAGTCCAAATAGTAATGAAGA AAAAGATTGTCTTTTCCTGCACTTCCTTAAACAGTTGTCGGGTAAGAGAAAACATGGAAACAGACAG GGTAATAATTGATGTGTTCAACTGTCCACCTCTGTATGATGATGTGAAAGTGCAATTTTTTTTTTCT TTTTAGGATTTTCCTAAATACTATCACAACTACCCTTTTTTCTTCTGGTTTAACACATCTTTAATAC AAAΆTAACAGGCTTTATCTACAAAGAAATGAATTGGATAATCTTCATAAACAΆAAAACATGGAΆAAT TTATCAACCAGAATATGCAGTAGAGATATATTTTGATGAGAΆATGACTTAAGTTATGTTGTAΆCTGG TAGCTGATTAAGTATAGTTCCCTGCACCCCTTCTGGGAAAGAATTATGTTCTTTCTAACCCTGCCAC ATAGTTATATGTTCTAAATCTTCCTTGCTGGTACATCTATATTGATATATGTATACACATGTTCTTT ATAAATCTATTAAATATATACAGATAAA
The NOV14 protein (SEQ ID NO:30) encoded by SEQ D NO:29 is 477 amino acid residues in length and is presented using the one-letter amino acid code in Table 14B. Psort analysis predicts the NOV14 protein ofthe invention to be localized at the plasma membrane with a certainty of0.6000.
Table 14B. Encoded NOV14 protein sequence (SEQ ID NO:30)
MNSELSLLSDCFLLSSASPQTNEFKGATEEAPAKESPHTGEFKGAALVSPISKRMLERLSKFEVG DAENVASYDSKIKKIVHSIVSSFAVGIFGVF ILLDVALIFADLIFTDSKVYIPLEYRSISLAIA LFFLMDVLLRVFVERRQHYFSDLLNILDTAVTVIILLVDWYIFFDVKFLKDIPR TRLFRLLRL IILIRVFRLAHLKRQLGKLIRRLVSRXYERDGFDLDLTYITERIVAMSFPSSGGQSFYRNPIKEV VQFLDKKHPNHYRVYNLCSERAYDPKHFHNRVSRIMIDDHNVPTLREMVAFSKEVLEWMAQDSEN IWIHCKGGKGRTGTMVCACLIASEIFLTAEERLYYFGERRTDKTMGTKYQGVETPSQNRYVGYF AQVKHSY LPPRKT FIKRLVIYSIHGKCLDLKVQIVM KKIVFSCTSLNSCRVRENMETDRV IIDVFNCPP YDDVKVQFFFSF
A search against the Patp database, a proprietary database that contains sequences published in patents and patent publications, yielded several homologous proteins shown in Table 14C.
Table 14C. Patp results for NOV14
Smallest
Sum
Reading High Prob
Sequences producing High-scoring Segment Pairs: Frame Score P(N)
>patp :AAGβ7459 Amino acid sequence of a human polypeptide +1 1895 2 .5e-195
■patp :AAG67638 Amino acid sequence of a human protein +1 1895 2 .5e-195
>patp :AAB73230 Human phosphatase AA493915_h +1 574 1.8e-lll
>patp:AA 3 402 Protein encoded by gene IMAGE clone 264611 +1 473 1.2e-44
>patp:AAY07450 Human TS10q23.3 gene bases 453-2243 +1 473 1.2e-44
In a BLAST search of public sequence databases, it was found, for example, that the nucleic acid sequence of this invention has 1105 of 1427 bases (77%) identical to a gb:GENBANK-ID:AF007118|acc:AF007118.1 mRNA from Homo sapiens (putative tyrosine phosphatase mRNA, complete eds). The full amino acid sequence of the protein of the invention was found to have 369 of 462 amino acid residues (79%) identical to, and 402 of 462 amino acid residues (87%) similar to, the 551 amino acid residue ptnπSWISSNEW- ACC:P56180 protein from Homo sapiens (PUTATIVE PROTETN-TYROSINE PHOSPHATASE TPTE (EC 3.1.3.48)).
NOVl 4 also has homology to the proteins shown in the BLASTP data in Table 14D.
Table 14D. BLAST results for NOV14
Gene Index/ Protein/ Organism Length Identity Positives Expect Identifier (aa) (%) (%) gi I 7019559 I ref |NP_0 transmembrane 551 369/462 402/462 0.0 37447. l| (NM 013315) phosphatase with (79%) (86%) tensin homology; tensin, putative protein-tyrosine phosphatase [Homo sapiens] gi 116166555 | ref | XP_ similar to 551 367/462 401/462 0.0 055073.l| (XM 055073 transmembrane (79%) (86%) phosphatase with tensin homology [Homo sapiens] gi 118640756 I ref |NP_ similar to 445 316/462 343/462 e-156 570141. l| (NM 130785 PUTATIVE PROTEIN- (68%) (73%) TYROSINE PHOSPHATASE TPTE [Homo sapien] gi 114787415 I emb I CAC tyrosine 664 275/432 336/432 e-141 44243.1| (AJ311311) phosphatase (63%) (77%) isoform A [Mus musculus] gi 114787417 | emb | CAC tyrosine 645 275/432 336/432 e-141 44244.1] (AJ311312) phosphatase (63%) (77%) isoform B [Mus musculus]
A multiple sequence alignment is given in Table 14E, with the NOV 14 protein being shown on line 1 in Table 14E in a ClustalW analysis, and comparing the NOV14 protein with the related protein sequences shown in Table 14D. This BLASTP data is displayed graphically in the ClustalW in Table 14E.
Table 14E. ClustalW Analysis of NOV14
1) >NOV14; SEQ ID NO:30
2) > gi|7019559|/ transmembrane phosphatase with tensin homology; tensin, putative protein-tyrosine phosphatase [Homo sapiens]; SEQ ID NO:97 3) > gi|16166555|/ similar to transmembrane phosphatase with tensin homology [Homo sapiens]; SEQ ID NO:98
4) > gi|18640756|/ similar to PUTATIVE PROTEIN-TYROSINE PHOSPHATASE TPTE; similar to transmembrane phosphatase with tensin homology; tensin, putative protein-tyrosine phosphatase
[Homo sapiens]; SEQ ID NO:99 5) > gi|14787415|/ tyrosine phosphatase isoform A [Mus musculus]; SEQ ID NO: 100
6) > gi|14787417|/ tyrosine phosphatase isoform B [Mus musculus]; SEQ ID NO: 101
NOV14 gi|7019559]
Figure imgf000110_0001
490 500 510 520 530 540
Figure imgf000111_0001
670
Figure imgf000111_0002
The presence of identifiable domains in the protein disclosed herein was determined by searches using algorithms such as PROSITE, Blocks, Pfam, ProDomain, Prints and then determining the Inteφro number by crossing the domain match (or numbers) using the Inteφro website (http:www.ebi.ac.uk/inteφro/). Table 14F lists the domain description from DOMAIN analysis results against NOV14.
Figure imgf000111_0003
Consistent with other known members of the Putative Protein-Tyrosine Phosphatase- like family of proteins, NOVl 4 has, for example, a dual specificity protein phosphatase signature sequence and homology to other members of the Putative Protein-Tyrosine Phosphatase-like Protein Family. NOV14 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts. For example, NOV14 nucleic acids and polypeptides can be used to identify proteins that are members of the Putative Protein-Tyrosine Phosphatase-like Protein Family. The NOV 14 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOV14 activity or function. Specifically, the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular replication, and signal transduction. These molecules can be used to treat, e.g., Cardiovascular diseases, cystitis, incontinence as well as other diseases, disorders and conditions hi addition, various NOV 14 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. For example, the NOV14 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the Putative Protein-Tyrosine Phosphatase-like Protein Family.
Cellular processes involving growth, differentiation, transformation and metabolism are often regulated in part by protein phosphorylation and dephosphorylation. The protein tyrosine phosphatases (PTPs), which hydrolyze the phosphate monoesters of tyrosine residues, all share a common active site motif and are classified into 3 groups. These include the receptor-like PTPs, the intracellular PTPs, and the dual-specificity PTPs, which can dephosphorylate at serine and threonine residues as well as at tyrosines. Diamond et al (1994) described a PTP from regenerating rat liver that is a member of a fourth class. The gene, which they designated Prll, was one of many immediate-early genes. Overexpression of Prll in stably transfected cells resulted in a transformed phenotype, which suggested that it may play some role in tumorigenesis. By using an in vitro prenylation screen, Gates et al. (1996) isolated 2 human cDNAs encoding PRL1 homologs, designated PTP(CAAXl) and PTP(CAAX2)(PRL2), that are farnesylated in vitro by mammalian farnesyhprotein transferase. Overexpression of these PTPs in epithelial cells caused a transformed phenotype in cultured cells and tumor growth in nude mice. The authors concluded that PTP(CAAXl) and PTP(CAAX2) represent a novel class of isoprenylated, oncogenic PTPs. Peng et al. (1998) reported that the human PTP(CAAXl) gene, or PRL1, is composed of 6 exons and contains 2 promoters. The predicted mouse, rat, and human PRL1 proteins are identical. Zeng et al (1998) determined that the human PRL1 and PRL2 proteins share 87% amino acid sequence identity.
The NOV14 nucleic acids and polypeptides, antibodies and related compounds according to the mvention will be useful in therapeutic and diagnostic applications in the mediation of cardiac and renal physiology. As such, the NOVl 4 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat cardiovascular and urogenital system disorders, e.g., Cardiovascular diseases, cystitis, incontinence as well as other diseases, disorders and conditions. The NOV 14 nucleic acids and polypeptides are useful for detecting specific cell types. For example, expression analysis has demonstrated that a NOVl 4 nucleic acid is expressed in Urinary bladder.
Additional utilities for NOV14 nucleic acids and polypeptides according to the invention are disclosed herein.
NOV15
A NOVl 5 polypeptide has been identified as a Leucine Rich Repeat (LRR)-like protein (also referred to as CG95387-02). The disclosed novel NOVl 5 nucleic acid (SEQ ID NO:31) of 3136 nucleotides is shown in Table 15A. The novel NOV15 nucleic acid sequences maps to the chromosome 19.
An ORF begins with an ATG initiation codon at nucleotides 330-332 and ends with a TAA codon at nucleotides 2331-2333. A putative untranslated region and/or downstream from the termination codon is underlined in Table 15 A, and the start and stop codons are in bold letters.
Table 15A. NOV15 Nucleotide Sequence (SEQ ID NO:31)
ACTCCTGACCTAAAGTGATCCACTCGCCTTGGCCTCCCAAAGTGCTAGGATTACAGCCCTCATTCTC TTTTGCTCCTCAGGTGACACAGGACAAGATCATCTGTCTACCCAATCATGAGCTCCAGGAGAACTTA TCAGAGGCCCCGTGCCAGCAATTGCTGCCTCGGGGGATCCCTGAGCAGATTGGGGCCCTGCAGGAGG TTAAAGGCCTTAAGAACAATTTGGACCTGCAGCAATACAGCTTTATTAACCAGCTGTGTTATGAGAC GGCCCTGCACTGGTATGCCAAGTACTTCCCTTACCTCGTGGTCATTCACACACTCATCTTCATGGTC TGCACCAGTTTCTGGTTCAAGTTCCCTGGCACCAGCTCCAAGATTGAACACTTCATCTCCATCCTGG GCAAGTGTTTCGACTCTCCATGGACCACCAGGGCCCTATCCGAGGTCTCCGGGGAGAACCAGAAGGG CCCAGCAGCCACCGAACGGGCTGCGGCATCCATAGTGGCCATGGCAGGGACCGGGCCGGGGAAGGCA GGGGAGGGTGAGAAGGAGAAAGTGCTGGCGGAACCGGAGAAGGTGGTGACCGAGCCTCCAGTTGTCA CCCTGTTGGACAAGAAGGAGGGTGAGCAAGCCAAAGCCCTGTTTGAGAAGGTGAAGAAGTTCCGCAT GCACGTGGAAGAGGGCGACATCCTGTACACCATGTACATCCGACAGACGGTGCTGAAAGTGTGTAAG TTCCTGGCCATCCTGGTCTACAACCTGGTCTATGTGGAGAAGATCAGTTTCCTGGTGGCCTGTAGGG TGGAGACGTCAGAGGTCACGGGCTACGCCAGCTTCTGCTGCAACCACACCAAGGCCCACCTCTTCTC CAAGCTGGCCTTCTGTTACATCTCCTTTGTGTGCATCTACGGACTTACCTGCATCTACACGCTCTAC TGGCTCTTCCACCGGCCCCTCAAGGAGTACTCCTTCCGTTCCGTGCGGGAGGAGACTGGCATGGGGG ACATTCCTGACGTCAAGAATGACTTCGCCTTCATGCTGCACCTCATCGATCAGTACGACTCCCTCTA CTCCAAGCGCTTCGCCGTCTTCCTGTCCGAGGTCAGCGAAAGCCGTCTAAAGCAGCTCAATCTCAAC CACGAGTGGACCCCCGAGAAGCTTCGACAGAAGCTGCAGCGCAATGCCGCGGGCCGGCTGGAGCTGG CCCTCTGCATGCTGCCGGGTCTGCCCGACACCGTCTTTGAGCTCAGTGAGGTGGAGTCACTCAGGCT GGAGGCCATCTGCGATATCACCTTCCCCCCGGGGCTGTCACAGCTGGTGCACTTGCAGGAGCTCAGC TTGCTCCACTCGCCCGCCAGGCTACCCTTCTCCTTGCAGGTCTTCCTGCGGGACCACCTGAAGGTGA TGCGCGTCAAATGCGAGGAGCTCCGCGAGGTGCCGCTTTGGGTGTTTGGGCTGCGGGGCTTGGAGGA GCTGCACCTGGAGGGGCTTTTCCCCCAGGAGCTAGCTCGGGCAGCCACCCTGGAGAGCCTCCGGGAG CTGAAGCAGCTCAAGGTGTTGTCCCTCCGGAGCAACGCCGGGAAGGTGCCAGCCAGTGTGACCGACG TTGCTGGCCACCTGCAGAGGCTCAGCCTGCACAACGATGGGGCCCGTCTGGTTGCCCTGAACAGCCT CAAGAAGCTGGCGGCATTGCGGGAGCTGGAGCTGGTGGCCTGCGGGCTGGAGCGCATCCCCCATGCA GTGTTCAGCCTGGGTGCGCTGCAGGAACTTGACCτCAAGGACAACCACCTGCGCTCCATCGAGGAAA TCCTCAGCTTCCAGCACTGCCGGAAGCTGGTCACGCTCAGGCTGTGGCACAACCAGATCGCCTACGT CCCTGAGCACGTGCGGAAGCTCAGGAGCCTGGAGCAGCTCTACCTCAGCTACAACAAGCTGGAGACC CTGCCCTCCCAGCTCGGCCTGTGCTCAGGCCTCCGTCTGCTGGATGTGTCCCACAATGGGCTACACT CCCTGCCACCCGAGGTGGGCCTCCTGCAGAACCTACAGCACCTGGCCCTCTCCTACAATGCCCTGGA GGCCCTGCCCGAAGAGCTCTTCTTCTGCCGCAAGCTGCGGACGTTGCTTCTGGGCGACAACCAACTG AGCCAGCTCTCGCCCCACGTGGGTGCCCTCAGAGCCCTCAGCCGCCTGGAGCTCAAAGGCAACCGCT TAGAGGCGCTGCCAGAAGAACTTGGCAACTGTGGGGGGCTCAAGAAGGCGGGGCTCCTGGTGGAAGA CACGCTTTACCAGGGTCTGCCGGCAGAAGTGCGGGACAAGATGGAGGAGGAATGAAGCTGGGGTGGG GCCGTTTTAGGTAGAGCCTTAAAAATGCTTCTGCCCTGGAATCTCAACCATTATCTTCCAAGATAGG AAGCCAAGTGGGTCTAGGCCAGGAGATGGGGGGGGGCGGGGGCAGCTGTGTCATCTTTCTGGGGCCC AGGAGGATCTGGGCTGGTTTGTCTGGGGAGACAGACAGGATGTTGTGGAGGTGGGGTGGAACCTGGT ATGGAGGGATTAACTCAGTCATGGCATTCTCCGACCAAAACCACACCTGTGTCTCTGGCAGGCTGGC TGGCCTTGCTCCCATCCCTAGAACTGCTGCCTCTCCCTGGATATTCCAGCTCAATTAGTGCCACATA TGGGGGAAACGACACATCCCAGTGGGATTTCCAACACTCCCCCTCCCCATGCAACAAAGCAACTTAC TTCTGGAGTTCTCTCCCAAGGAGAGGACACAGACACAGTTGTTTGCTGTGTTATATGTTAGCTCCGA ACAATGGTTCTCATTTGGCTAAGCATCAAAATCACCTAGGGAGCCGGTGCAAAACAAAATATCCCAG TCCCCTCCCCTGAAACACTGACTCAGGAGGTTTGGTTGGGGGCCAGGAGTCTGTTCCTAAATATTCC AGGTAGTTCTGGTGCAGGTAAGTGGCCCTGAGACAGTATGTTGGGAAATGCTGACGTAAAGGTATCA GGGCCGGGCGCTGTGGCTCATGACTATAATCCCAGCTGTTTGAGAGGCCAATGCAGGAGGATGGTTG AGCTCAGGAGTTCGAGATCAGCCTGGGTAACATAGCGAGACCCCACCTCTGCCA
Variant sequences of NOV15 are included in Example 3, Table 26. A variant sequence can include a single nucleotide polymoφhism (SNP). A SNP can, in some instances, be referred to as a "cSNP" to denote that the nucleotide sequence containing the SNP originates as a cDNA.
The NOVl 5 protein (SEQ D NO:32) encoded by SEQ ID NO:31 is 667 amino acid residues in length and is presented using the one-letter amino acid code in Table 15B. Psort analysis predicts the NOVl 5 protein of the invention to be localized at the plasma membrane with a certainty of 0.7900.
Table 15B. Encoded NOV15 protein sequence (SEQ ID NO:32)
MVCTSFWFKFPGTSSKIEHFISILGKCFDSPWTTRALSEVSGENQKGPAATERAAASIVAMAGTG PGKΆGEGEKΞKVLAEPΞKWTEPPWTLLD KEGEQAKALFEKVKKFRMHVEEGDILYTMYIRQT V KVCKFLAILVYNLVYVEKISFLVACRVETSEVTGYASFCCNHTKAHLFSK AFCYISFVCIYG LTCIYTLY LFHRPLKEYSFRSVREETGMGDIPDVKNDFAFMLHLIDQYDSLYSKRFAVFLSEVS ESRLKQLNL HEWTPEKLRQKLQR AAGRLELALCMLPGLPDTVFELSEVESLRLEAICDITFPP GLSQLVHLQELSLLHSPARLPFSLQVFLRDHLKVMRVKCEE REVPLWVFGLRGLEELHLEGLFP QELARAATLESLRELKQLKVLSLRSNAGKVPASVTDVAGHLQRLSLH DGARLVALNSLKKLAAL RELELVACGLERIPHAVFSLGALQELDLKDNHLRSIEEI SFQHCR LVTLRL HKΓQIAYVPEHV RKLRSLEQLYLSYNKLETLPSQLGLCSGLRLLDVSHNGLHSLPPEVGLLQNLQH ALSYNALEAL PEE FFCRK RTL LGDNQLSQLSPHVGALRALSRLE KGNR EA PEELGNCGG AG LVED TLYQGLPAEVRD MEEE
A search against the Patp database, a proprietary database that contains sequences published in patents and patent publications, yielded several homologous proteins shown in Table 15C. Table 15C. Patp results for NOV15
Smallest Sura
Reading High Prob
Sequences producing High- scoring Segment Pairs: Frame Score P(N)
>patp:AAY70473 Human CNAP-1 +1 2147 5.0e-222
>patp:AAG75 13 Human colon cancer antigen protein +1 1882 6.0e-194
>patp:AAM41692 Human polypeptide SEQ ID NO 6623 +1 1878 1.6e-193
>patp:AAU20426 Human secreted protein, Seq ID No 418 +1 1835 5.8e-189
>patp-.AAB92855 Human protein sequence SEQ ID NO: 11424 +1 1795 1.0e-184
In a BLAST search of public sequence databases, it was found, for example, that the nucleic acid sequence of this invention has 2227 of 2228 bases (99%) identical to a gb:GENBANK-ID:AK027073|acc:AK027073.1 mRNA from Homo sapiens (cDNA: FLJ23420 fis, clone HEP22352). The full amino acid sequence of the protein of the invention was found to have 444 of 444 amino acid residues (100%) identical to, and 444 of 444 amino acid residues (100%) similar to, the 444 amino acid residue ptnr:SPTREMBL-ACC:Q9H5H8 protein from Homo sapiens (CDNA: FLJ23420 FIS, CLONE HEP22352).
NOV 15 also has homology to the proteins shown in the BLASTP data in Table 15D.
Table 15D. BLAST results for NOV15
Gene Index/ Protein/ Organism Length Identity Positives Expect Identifier (aa) (%) (%) gi 113376597 I ef |NP_ hypothetical 444 444/444 444/444 0.0 079337. l| (NM 025061 protein FLJ23420 (100%) (100%) [Homo sapiens] gi 114150009 I ref |NP_ hypothetical 708 404/667 520/667 0.0 115646. ll (NM 032270 protein (60%) (77%) DKFZp586J1119 [Homo sapiens] gi 119343671 |gb |AAH2 Similar to 708 404/671 526/671 0.0 5473. ll (BC025473) hypothetical (60%) (78%) protein DKFZp586J1119 [Mus musculus] gi|7243272|dbj | BAA9 KIAA1437 protein 811 367/666 84/666 0.0 2675. l| (AB037858) [Homo sapiens] (55%) (72%) gi I 8922442 I ref |NP_0 hypothetical 682 345/673 470/673 0.0 60573.1] (NM 018103) protein FLJ10470 (51%) (69%) [Homo sapiens]
A multiple sequence alignment is given in Table 15E, with the NOV 15 protein being shown on line 1 in Table 15E in a ClustalW analysis, and comparing the NOVl 5 protein with the related protein sequences shown in Table 15D. This BLASTP data is displayed graphically in the ClustalW in Table 15E. Table 15E. ClustalW Analysis of NOV15
1) > NOV15; SEQ ID NO:32
2) > gi| 13376597|/ hypothetical protein FLJ23420 [Homo sapiens]; SEQ ID NO: 102
3) > gi| 14150009|/ hypothetical protein DKFZp586Jl 119 [Homo sapiens]; SEQ ID NO: 103
4) > gi| 19343671|/ Similar to hypothetical protein DKFZp586Jl 1 19 [Mus musculus]; SEQ ID NO: 104
5) > gi|7243272|/ KIAA1437 protein [Homo sapiens]; SEQ ID NO: 105
6) > gi|8922442|/ hypothetical protein FLJ10470 [Homo sapiens]; SEQ ID NO: 106
Figure imgf000116_0001
Figure imgf000117_0001
The NOVl 5 Clustal W alignment shown in Table 15E was modified to begin at amino residue 121. The data in Table 15E includes all of the regions overlapping with the NOVl 5 protein sequences.
The presence of identifiable domains in the protein disclosed herein was determined by searches using algorithms such as PROSITE, Blocks, Pfam, ProDomain, Prints and then determining the Inteφro number by crossing the domain match (or numbers) using the Inteφro website (http:www.ebi.ac.uk/inteφro/). Table 15F lists the domain description from DOMAIN analysis results against NOV15.
Figure imgf000118_0001
Consistent with other known members of the LRR-like family of proteins, ΝOV15 has, for example, eight Leucine Rich Repeat (LRR) signature sequences and homology to other members of the LRR-like Protein Family. NOVl 5 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts. For example, NOVl 5 nucleic acids and polypeptides can be used to identify proteins that are members of the LRR-like Protein Family. The NOV 15 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOVl 5 activity or function. Specifically, the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular replication, and signal transduction. These molecules can be used to treat, e.g., cancer, trauma, regeneration (in vitro and in vivo), viral/bacterial/parasitic infections, Alzheimer's disease, stroke, tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, cerebral palsy, epilepsy, Lesch-Nyhan syndrome, multiple sclerosis, ataxia-telangiectasia, leukodystrophies, behavioral disorders, addiction, anxiety, pain, neurodegeneration, Von Hippel-Lindau (VHL) syndrome, cirrhosis, transplantation, Hirschsprung's disease , Crohn's Disease, appendicitis, osteoporosis, hypercalceimia, arthritis, ankylosing spondylitis, scoliosis, systemic lupus erythematosus, autoimmune disease, xerostomia as well as other diseases, disorders and conditions. In addition, various NOVl 5 nucleic acids and polypeptides according to the invention are useful, ter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. For example, the NOVl 5 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the LRR-like Protein Family. LRR Proteins are a family of proteins characterized by a structural motif rich in leucine residues. They are either transmembrane or secreted proteins and are involved in protein- protein interactions. Members of this family have been implicated in extracellular matrix assembly and cellular growth. In addition, several proteins belonging to this family, such as slit, Toll and robo have been shown to mediate key roles in central nervous system development and organo genesis in Drosophila. Vertebrate orthologs of these proteins have also been shown to have similar roles in the CNS as well as other organ systems like kidney..
LRRs are relatively short motifs (22-28 residues in length) found in a variety of cytoplasmic, membrane and extracellular proteins. Although these proteins are associated with widely different functions, a common property involves protein-protein interaction. Little is known about' the 3D structure of LRRs, although it is believed that they can form amphipathic structures with hydrophobic surfaces capable of interacting with membranes. In vitro studies of a synthetic LRR from Drosophila Toll protein have indicated that the peptides form gels by adopting beta-sheet structures that form extended filaments (Packman et al. FEBS Lett.1991; 291 : 87-91). These results are consistent with the idea that LRRs mediate protein-protein interactions and cellular adhesion. Other functions of LRR-containing proteins include, for example, binding to enzymes and vascular repair.
The NOVl 5 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of cardiac, immune, and nerve physiology. As such, the NOV 15 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat cardiovascular, nervous, and immune system disorders, e.g., cancer, trauma, regeneration (in vitro and in vivo), viral/bacterial/parasitic infections, Alzheimer's disease, stroke, tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, cerebral palsy, epilepsy, Lesch-Nyhan syndrome, multiple sclerosis, ataxia-telangiectasia, leukodystrophies, behavioral disorders, addiction, anxiety, pain, neurodegeneration, Von Hippel-Lindau (VHL) syndrome, cirrhosis, transplantation, Hirschsprung's disease, Crohn's Disease, appendicitis, osteoporosis, hypercalceimia, arthritis, ankylosing spondylitis, scoliosis, systemic lupus erythematosus, autoimmune disease, xerostomia as well as other diseases, disorders and conditions. The NOVl 5 nucleic acids and polypeptides are useful for detecting specific cell types.
For example, expression analysis has demonstrated that a NOV 15 nucleic acid is expressed in Coronary Artery, Parotid Salivary glands, Liver, Colon, Bone, Synovium/Synovial membrane, and Brain.
Additional utilities for NOV 15 nucleic acids and polypeptides according to the invention are disclosed herein. NOV16
A NOVl 6 polypeptide has been identified as a RhoGEF-like protein (also referred to as CG95419-02). The disclosed novel NOV16 nucleic acid (SEQ ID NO:33) of 5372 nucleotides is shown in Table 16A. The novel NOVl 6 nucleic acid sequences maps to the chromosome 5.
Ail ORF begins with an ATG initiation codon at nucleotides 61-63 and ends with a TAA codon at nucleotides 5179-5181. A putative untranslated region and/or downstream from the termination codon is underlined in Table 16 A, and the start and stop codons are in bold letters.
Table 16A. NOV16 Nucleotide Sequence (SEQ ID NO:33)
CCATGGGGCCTCCTGCAATAACTTCTCTTGTTTATTATTTTCATTGCAGATGCGAAAGCCATGGAGT TGAGCTGCAGCGAAGCACCTCTTTACCAGGGGCAGATGATGATCTATGCGAAGTTTGACAAAAATGT GTATCTTCCTGAAGATGCTGAGTTTTACTTTACTTATGACGGATCTCATCAGCGACATGTCATGATT GCAGAGCGCATCGAGGATAACGTTCTCCAGTCCAGCGTCCCAGGCCATGGGCTTCAGGAGACGGTGA CGGTATCTGTGTGCCTCTGCTCGGAAGGTTACTCTCCGGTGACCATGGGCTCTGGCTCAGTGACCTA CGTGGACAACATGGCTTGCAGGCTGGCTCGTCTGCTGGTGACGCAGGCCAATCGCCTCACAGCCTGC AGCCACCAGACCCTGCTGACCCCATTTGCCTTGACGGCAGGAGCACTGCCTGCCTTGGATGAGGAGC TCGTGCTGGCTCTGACCCATCTGGAATTGCCTCTAGAGTGGACTGTGTTGGGAAGTTCTTCACTTGA AGTATCTTCTCACAGAGAATCTCTTCTACACCTGGCTATGAGATGGGGCCTGGCTAAACTTTCCCAG TTCTTCTTGTGTCTCCCGGGGGGAGTCCAGGCCTTGGCTTTACCCAACGAAGAGGGTGCCACACCAT TAGACTTAGCTTTACGTGAAGGACACTCCAAGCTGGTGGAAGACGTCACAAGTTTTCAGGGCAGATG GTCCCCAAGCTTCTCCCGAGTGCAGCTCAGTGAAGAAGCCTCCTTGCATTACATTCACTCATCGGAA ACGCTGACCCTGACCCTGAACCACACAGCCGAGCATTTGTTGGAGGCAGATATTAAACTCTTCCGGA AATACTTTTGGGATAGAGCCTTTCTTGTCAAGGCCTTTGAGCAAGAAGCCAGGCCAGAGGAAAGAAC AGCTATGCCCTCCAGCGGTGCAGAAACTGAAGAAGAGATTAAGAATTCAGTGTCCAGCAGATCAGCA GCCGAAAAGGAAGATATAAAGCGTGTCAAAAGCCTGGTGGTTCAACACAATGAACATGAAGACCAGC ACAGCCTAGATTCTAGATCGCTCCTTCGATATCCTAAAAAATCCAAGCCGCCCTCGACATTGCTTGC TGCAGGCCGGCTTTCAGACATGCTGAATGGAGGTGATGAAGTCTACGCTAACTGTATGGTGATTGAT CAGGTTGGTGATTTGGATATCAGCTATATTAATATAGAGGGAATCACTGCCACTACCAGCCCTGAAT CCAGAGGTTGCACTCTGTGGCCTCAGAGCAGCAAACACACCCTTCCTACAGAAACCAGTCCCAGTGT GTACCCACTTAGTGAAAATGTCGAAGGGACAGCACACACTGAAGCCCAGCAGTCCTTCATGTCACCA TCAAGTTCGTGTGCTTCCAACTTGAATCTTTCTTTTGGTTGGCATGGATTTGAAAAGGAACAAAGTC ATCTAAAGAAAAGAAGTTCTAGCCTTGATGCCTTGGACGCCGACAGTGAAGGGGAAGGGCATTCTGA GCCATCCCACATCTGTTACACTCCAGGGTCTCAGAGCTCCTCAAGAACTGGGATTCCTAGTGGGGAT GAATTGGACTCTTTTGAGACTAACACTGAACCGGATTTTAATATCTCCAGGGCTGAATCCCTTCCTC TATCAAGTAATCTACAGTTGAAGGAATCACTGCTTTCTGGAGTTCGCTCACGTTCTTATTCTTGCTC GTCACCCAAAATTTCTTTAGGAAAAACTCGTTTGGTGCGTGAATTAACAGTATGCAGTTCAAGTGAA GAGCAAAAAGCTTACAGCTTATCGGAGCCACCAAGAGAAAACAGGATTCAGGAAGAAGAATGGGATA AATACATCATACCTGCCAAATCAGAGTCTGAAAAATATAAAGTGAGTCGAACTTTCAGTTTCCTCAT GAATAGGATGACTAGCCCTCGGAATAAATCAAAGACAAAAAGCAAGGATGCCAAAGATAAAGAGAAG CTGAATCGACATCAGTTTGCCCCAGGAACATTCTCTGGGGTTCTGCAGTGTTTGGTTTGTGATAAAA CACTCCTGGGGAAAGAGTCACTGCAGTGTTCTAGTTGTAATGCAAATGTGCACAAAGGTTGTAAAGA TGCTGCGCCTGCATGCACCAAGAAATTCCAAGAGAAATATAACAAGAACAAACCACAGACCATCCTT GGAAGTTCTTCATTTAGAGACATCCCACAGCCTGGTCTCTCCTTGCACCCTTCTTCCTCCGTGCCTG TTGGATTGCCGACTGGAAGGAGGGAGACTGTGGGACAGGTCCATCCATTGTCCAGAAGTGTTCCAGG TACCACCTTGGAAAGCTTCAGGAGGTCAGCCACATCCTTGGAGTCTGAGAGTGACAATAACAGCTGC AGAAGCAGGTCTCATTCTGATGAGCTGCTACAGTCCATGGGCTCTTCTCCCTCTACAGAGTCTTTCA TAATGGAAGATGTTGTGGATTCTTCTCTGTGGAGTGACCTCAGCAGTGATGCCCAGGAGTTTGAAGC AGAATCTTGGAGTCTTGTGGTGGATCCCTCATTTTGTAATAGGCAGGAGAAGGATGTCATCAAAAGA CAGGATGTCATTTTTGAGCTAATGCAAACAGAGATGCATCACATCCAGACCCTGTTCATCATGTCTG AGATCTTCAGGAAAGGCATGAAAGAGGAGCTGCAGCTGGACCACAGCACCGTGGATAAAATTTTCCC CTGTTTAGATGAGTTGCTTGAAATCCACAGGCATTTCTTCTACAGTATGAAGGAACGAAGGCAGGAA TCAAGTGCTGGCAGCGACAGGAATTTTGTGATCGACCGAATTGGAGATATTTTGGTACAACAGTTTT CAGAAGAAAATGCAAGTAAAATGAAGAAAATATATGGAGAATTCTGTTGCCATCATAAAGAAGCTGT TAACCTCTTTAAAGAACTCCAGCAGAATAAAAAGTTTCAGAATTTTATTAAGCTCCGAAATAGTAAT CTTTTGGCTCGACGCCGAGGAATTCCAGAATGCATTCTGTTGGTCACTCAGCGTATTACAAAATACC CTGTCTTGGTGGAAAGGATATTGCAGTACACAAAGGAAAGAACTGAGGAACATAAAGACTTACGCAA AGCCCTTTGCTTAATTAAAGACATGATTGCAACAGTGGATTTAAAAGTCAATGAATATGAGAAAAAC CAAAAATGGCTTGAGATCCTAAATAAGATTGAAAACAAAACATACACGAAGCTCAAAAATGGACATG TGTTTAGGAAGCAGGCACTGATGAGTGAAGAAAGGACTCTGTTATATGATGGCCTTGTTTACTGGAA AACTGCTACAGGTCGTTTCAAAGATATCCTAGCTCTACTTCTAACTGATGTGCTGCTCTTTTTACAA GAAAAAGACCAGAAATACATCTTTGCAGCCGTTGATCAGAAGCCATCAGTTATTTCCCTTCAAAAGC TTATTGCTAGAGAAGTTGCTAATGAGGAGAGAGGAATGTTTCTGATCAGTGCTTCATCTGCTGGTCC TGAGATGTATGAAATTCACACCAATTCCAAGGAGGAACGCAATAACTGGATGAGACGGATCCAGCAG GCTGTAGAAAGTTGTCCTGAAGAAAAAGGGGGAAGGACAAGTGAATCTGATGAAGACAAGAGGAAAG CTGAAGCCAGAGTGGCCAAAATTCAGCAATGTCAAGAAATACTCACTAACCAAGACCAACAAATTTG TGCGTATTTGGAGGAGAAGCTGCATATCTATGCTGAACTTGGAGAACTGAGCGGATTTGAGGACGTC CATCTAGAGCCCCACCTCCTTATTAAACCTGACCCAGGCGAGCCTCCCCAGGCAGCCTCATTACTGG CAGCAGCACTGAAAGAAGCTGAGAGCCTACAAGTTGCAGTGAAGGCCTCACAGATGGGCGCCGTGAG TCAATCATGTGAGGACAGTTGTGGAGACTCTGTCTTGGCGGACACACTCAGTTCTCATGATGTACCA GGATCACCGACTGCCTCATTAGTCACAGGAGGGAGAGAAGGAAGAGGCTGTTCGGATGTGGATCCCG GGATCCAGGGTGTGGTAACCGACTTGGCCGTCTCTGATGCAGGGGAGAAGGTGGAATGTAGAAATTT TCCAGGTTCTTCACAATCAGAGATTATACAAGCCATACAGAATTTAACCCGTCTCTTATACAGCCTT CAGGCCGCCTTGACCATTCAGGACAGCCACATTGAGATCCACAGGCTGGTTCTCCAGCAGCAGGAGG GCCTGTCTCTCGGCCACTCTATCCTCCGAGGCGGCCCCTTGCAGGACCAGAAGTCTCGCGACGCGGA CAGGCAGCATGAGGAGCTGGCCAATGTGCACCAGCTTCAGCACCAGCTCCAGCAGGAGCAGCGGCGC TGGCTGCGCAGGTGTGAGCAGCAGCAGCGGGCGCAGGCGACCAGGGAGAGCTGGCTGCAGGAGCGGG AGCGGGAGTGCCAGTCGCAGGAGGAGCTGCTGCTGCGGAGCCGGGGCGAGCTGGACCTCCAGCTCCA GGAGTACCAGCACAGCCTGGAGCGGCTGAGGGAGGGCCAGCGCCTGGTGGAGAGGGAGCAGGCGAGG ATGCGGGCCCAGCAGAGCCTGCTGGGCCACTGGAAGCACGGCCGGCAGAGGAGCCTGCCCGCGGTGC TCCTTCCGGGTGGCCCCGAGGTAATGGAACTTAATCGATCTGAGAGTTTATGTCATGAAAACTCATT CTTCATCAATGAAGCTTTAGTACAAATGTCATTTAACACTTTCAACAAACTGAATCCGTCAGTTATC CATCAGGATGCCACTTACCCTACAACTCAATCTCATTCTGACTTGGTGAGGACTAGTGAACATCAAG TAGACCTCAAGGTGGACCCTTCTCAGCCTTCGAATGTCAGTCACAAACTGTGGACAGCCGCTGGTTC CGGCCATCAGATACTTCCTTTCCAAGAAAGCAGCAAGGATTCTTGTAAAAATGATTTGGACACCTCC CACACTGAGTCCCCAACCCCCCATGACTCAAATTCACACCGCCCTCAACTGCAGGCGTTTATAACAG AAGCAAAGCTAAATCTACCGACAAGGACAATGACCAGACAAGATGGGGAAACTGGAGATGGAGCCAA AGAAAATATTGTTTACCTCTAATTGTGTTGTCATTTTTCCAAACAAAACAAAACACTGGCACTTTTG GGAGAAACTTTTTGTCTCCATTCCTTATGTATGTGTGATTGTCTGTGTCCAAATTGCTTTAAGAATA ATATTTAATATTTCCTGGAAGCTCATTTTTTTGGCATGAGTCTAATTAAATTATTGAAAGCCAAAAA AAAAAAAAAAAA
The NOV16 protein (SEQ ID NO:34) encoded by SEQ ID NO:33 is 1706 amino acid residues in length and is presented using the one-letter amino acid code in Table 16B. Psort analysis predicts the NOVl 6 protein of the invention to be localized in the cytoplasm with a certainty of 0.4500.
Table 16B. Encoded NOV16 protein sequence (SEQ ID NO:34)
MELSCSEAPLYQGQMMIYAKFDKNVYLPEDAEFYFTYDGSHQRHVMIAERIED VLQSSVPGHGL QETVTVSVCLCSEGYSPVTMGSGSVTYVD MACRLARLLVTQANRLTACSHQTLLTPFALTAGAL PALDEELVLALTHLELPLEWTVLGSSSLEVSSHRESLLHLAMR GLAK SQFFLCLPGGVQALAL PNEEGATPLDLALREGHSKLVEDVTSFQGR SPSFSRVQLSEEASLHYIHSSETLTLTLNHTAEH LLEADIKLFRKYFWDRAFLVKAFEQEARPEERTAMPSSGAETEEEI NSVSSRSAAEKEDIKRVK SLWQHHREHEDQHSLDSRSLLRYPKKSKPPSTLLAAGRLSDMLNGGDEVYANCMVIDQVGDLDIS Λ,__
YINIEGITATTSPESRGCTL PQSSKHTLPTETSPSVYPLSENVEGT ΓAAHHΠTEAQQSFMSPSSSCAS NLNLSFG HGFEKEQSHLK RSSSLDALDADSEGEGHSEPSHICYTPGSQSSSRTGIPSGDELDS FETNTEPDFNISRAESLPLSSNLQLKESLLSGVRSRSYSCSSPKISLGKTRLVRE TVCSSSEEQ KAYSLSEPPRENRIQEEEWDKYIIPAKSESEKYKVSRTFSFLMNRMTSPRNKSKTKSKDAKDKEK LNRHQFAPGTFSGVLQCLVCD TLLGKESLQCSSCNANVHKGCKDAAPACTKKFQEKYNKNKPQT ILGSSSFRDIPQPGLSLHPSSSVPVGLPTGRRETVGQVHPLSRSVPGTTLESFRRSATSLESESD NNSCRSRSHSDELLQSMGSSPSTESFIMEDWDSSL SD SSDAQEFEAES SLWDPSFCNRQE KDVIKRQDVIFELMQTEMHHIQTLFIMSEIFRKGMKEELQLDHSTVDKIFPCLDELLEIHRHFFY SMKERRQESSAGSDRNFVIDRIGDILVQQFSEENASKMKKIYGEFCCHHKEAVN FKELQQNKKF QNFIKLR SNLLARRRGIPECIL VTQRITKYPVLVERILQYTKERTEEHKDLRKALCLIKDMIA TVDLKVNEYEKNQK LEILNKIENKTYTKLK GHVFRKQALMSEERT LYDGLVYWKTATGRFKD ILALL TDVLLFLQEKDQKYIFAAVDQKPSVISLQKLIAREVANEERGMFLISASSAGPEMYEIH TNSKEERNN MRRIQQAVESCPEEKGGRTSESDEDKRKAEARVAKIQQCQEILTNQDQQICAYLE EKLHIYAE GELSGFEDVHLEPHLLIKPDPGEPPQAASLLAAALKEAESLQVAVKASQMGAVSQS CEDSCGDSVLADTLSSHDVPGSPTASLVTGGREGRGCSDVDPGIQGWTDLAVSDAGEKVECRNF PGSSQSEIIQAIQNLTRLLYSLQAA TIQDSHIEIHRLVLQQQEGLSLGHSILRGGPLQDQKSRD ADRQHEELANVHQLQHQLQQEQRR LRRCEQQQRAQATRESWLQΞRERECQSQEE LLRSRGELD LQLQEYQHSLERLREGQRLVEREQARMRAQQSLLGHWKHGRQRSLPAVLLPGGPEVMELNRSESL CHENSFFINEALVQMSFNTFNKLNPSVIHQDATYPTTQSHSDLVRTSEHQVDLKVDPSQPS VSH KLWTAAGSGHQILPFQESSKDSCKNDLDTSHTΞSPTPHDSNSHRPQLQAFITEAKLN PTRTMTR QDGΞTGDGAKENIVYL
A search against the Patp database, a proprietary database that contains sequences published in patents and patent publications, yielded several homologous proteins shown in Table 16C.
Table 16C. Patp results for NOVl 6
Smallest
Sum
Reading High Prob
Sequences producing High-scoring Segment Pairs: Frame Score P(N)
>patp:ABB44551 Human wound healing related polypeptide +1 1470 2.8e-150
>patp:AA 93941 Human brx protein +1 1436 1.5e-148
>patp:ABG05537 Novel human diagnostic protein #5528 +1 1436 1.5e-14£
>patp:ABG05537 Novel human diagnostic protein #5528 +1 1436 1.5e-14£
>patp:ABG15870 Novel human diagnostic protein #15861 +1 1447 7.5e-14t
In a BLAST search of public sequence databases, it was found, for example, that the nucleic acid sequence of this invention has 4339 of 5274 bases (82%) identical to a gb:GENBANK-rD:MMU73199|acc:U73199.1 mRNA ϊromMus musculus (Rho-guanine nucleotide exchange factor mRNA, complete eds). The full amino acid sequence of the protein of the mvention was found to have 1350 of 1670 amino acid residues (80%) identical to, and 1460 of 1670 amino acid residues (87%) similar to, the 1693 amino acid residue ptnr:SWISSPROT-ACC:P97433 protein from as musculus (RHO-GUANINE NUCLEOTIDE EXCHANGE FACTOR (RHOGEF) (RIP2)).
NOVl 6 also has homology to the proteins shown in the BLASTP data in Table 16D. Table 16D. BLAST results for NOV16
Gene Index/ Protein/ Organism Length Identity Positives Expect Identifier (aa) (%) (%) gi I 7106395 I ref |NP_0 Rho interacting 1693 1350/1674 1460/1674 0.0 36156.1] (NM 012026) protein 2; Rho (80%) (86%) specific exchange factor
[Mus inuscu2us] gi 118602674 I ref |XP_ hypothetical 669 668/669 668/669 0.0 016989.5] (XM 016989 protein FLJ21817 (99% ) (99%) similar to Rhoip2 [Homo sapiens] gi 1104384411 dbj | BAB unnamed protein 669 669/669 669/669 0.0 15243.11 (AK025816) product (100%) (100%) [ omo sapiens ] gi 1153417611 gb IAAHl hypothetical 615 609/613 609/613 0.0 2946.11AAH12946 (BC0 protein FLJ21817 (99%) (99%) 12946) similar to Rhoip2 [Homo sapiens] gi 117437752 I ref |XP__ similar to Rho 590 290/292 291/292 e-170 068710. ll (XM 068710 interacting (99%) (99%) protein 2; Rho specific exchange factor
[Homo sapiens]
A multiple sequence alignment is given in Table 16E, with the NOV16 protein being shown on line 1 in Table 16E in a ClustalW analysis, and comparing the NOV16 protein with the related protein sequences shown in Table 16D. This BLASTP data is displayed graphically in the ClustalW in Table 16E.
Table 16E. ClustalW Analysis of NOVl 6
1) > NOVl 6; SEQ ID N0:34
2) > gi]7106395]/ Rho interacting protein 2; Rho specific exchange factor [Mus musculus]; SEQ ID
NO: 107
3) > gi|18602674|/ hypothetical protein FLJ21817 similar to Rhoip2 [Homo sapiens]; SEQ ID NO:108
4) > gi]10438441|/ unnamed protein product [Homo sapiens]; SEQ ID NO:109
5) > gi|15341761|/ hypothetical protein FLJ21817 similar to Rhoiρ2 [Homo sapiens]; SEQ ID NO:l 10
6) > gi| 17437752]/ similar to Rho interacting protein 2; Rho specific exchange factor [Homo sapiens];
SEQ ID NO:lll
10 20 30 40 50 60 0 9
20 19
Figure imgf000123_0001
130 140 150 160 170 180
....|....|....|....|....]....|....|....|....|....|....|....]
NOV16 121 TPFALTAGALPALDEELVLALTHLELPLEWTVLGSSSLEVSSHRESLLHLAMRWGLAKLS 180 79
40 39
00 99
60 59
Figure imgf000124_0001
370 380 390 400 410 420
NOVl6 361 AGRLSDMLNGGDEVYANCMVIDQVGDLDISYINIEGITATTSPESRGCTLWPQSSKHTLP 420 gi| | 7106395 ] 360 AGQLSDVLNGGDEVYANCMVIDQVGDLDINYINLEGLSTHTSPESGRSMLGPQACMHTLP 419 gi | 18602674 X X g | 10438441 X X gi | 15341761 X X gi | 17437752 X X
430 440 450 460 470 480
N0V16 421 TETSPSVYPLSENVEGTAHTEAQQSFMSPSSSCASNLNLSFGWHGFEKEQSHLKKRSSSL 480 gi | 7106395 | 420 PDTSPCGRPLIENSEGTLDAAASQSFVTPSSSRTSNLNLSFGLHGFEKEQSHLKKRSSSL 479 gi | 18602674 X X gi | 10438441 X gi |15341761 X X gi | 17437752 X X
40 38
00 98
Figure imgf000124_0002
Figure imgf000125_0001
610 620 630 640 650 660 60 58
20 18
80 76
40 36
00 96
60 56
020 016
Figure imgf000125_0002
1030 1040 . 1050 1060 1070 1080
Figure imgf000125_0003
Figure imgf000126_0001
1090 1100 1110 1120 1130 1140
NOVl6 1075 KQALMSEERTLLYDGLVY KTATGRFKDILALLLTDVLLFLQEKDQKYIFAAVDQKPSV 1137 gi 7106395 I 1074 KQAL|sgERgLL|jDGLVY KTATGRFKDILALLLTDVLLFLQEKDQKYIFAAVDQKPSV 1133 gi 18602674| 41 KQALMSEERTLLYDGLVYWKTATGRFKDILALLLTDVLLFLQEKDQKYIFAAVDQKPSV 100 gi 10438441 j 41 KQALMSEERTLLYDGLVY KTATGRFKDILALLLTDVLLFLQEKDQKYIFAAVDQKPSV 100 gi 15341761 j 41 KQALMSEERTLLYDGLVY KTATGRFKDILALLLTDVLLFLQEKDQKYIFAAVDQKPSV 100 gi 17437752 39 HGVE^QRSTS2RRAgGQGPSCLDgL[ PSSGKQJ5JlBgV'SD3VALFWGHPAGgFFR- - - -
1150 1160 1170 1180 1190 1200
NOV16 1138 SLQKLIAREVANEERGMFLISASSAGPEMYEIHTNSKEERNNWMRRIQQAVESCPEEKGC 1197 gi|7106395] 1134 SLQKLIAREVANEERGMFLISASSAGPEMYEIHTNSKEERNN MRRIQQAVESCPEEgGt 1193 gij 18602674 I 101 SLQKLIAREVANEERGMFLISASSAGPEMYEIHTNSKEERNNWMRRIQQAVESCPEEKGC 160 gij 10438441 j 101 SLQKLIAREVANEERGMFLISASSAGPEMYEIHTNSKEERNNWMRRIQQAVESCPEEKGG 160 gijl534176lj 101 SLQKLIAREVANEERGMFLISASSAGPEMYEIHTNSKEERNNWMRRIQQAVESCPEEKGC 160 gij 17437752 j 95 PADSgWLLGPVEGNPVLQTLPFJsJLGg ragPPSS PPTTAARRSgDDRREEjIjjVVQQQQBBGG^^II I1 I1 FFSS gFFBg GgG 148
1210 1220 1230 1240 1250 1260
Figure imgf000126_0002
1270 1280 1290 1300 1310 1320
NOVl6 1257 PHLLIKPDPGEPPQAASLLAAALKEAESLQVAVKASQMGAVSQSCEDSCGDSVLADTLSS gi 7106395 I 1253 PHLLIKPDPGEPPQAASLLAAAL|EAESLQVAVKAS|MGj2vSQS E|sgGgJVLJΪJDTgsg gi 18602674| 220 PHLLIKPDPGEPPQAASLLAAALKEAESLQVAVKASQMGAVSQSCEDSCGDSVLADTLSS gi 10438441 j 220 PHLLIKPDPGEPPQAASLLAAALKEAESLQVAVKASQMGAVSQSCEDSCGDSVLADTLSS gi 15341761 j 220 PHLLIKPDPGEPPQAASLLAAALKEAESLQVAVKASQMGAVSQSCEDSCGDSVLADTLSS gi 17437752 203 GQMMgYAKFDKN- -VYJ5PgDAEFYFTYDG§HQRH αA- -ERIE[gNajQSSVPG 252
1330 1340 1350 1360 1370 1380
NOVl6 1317 IDVPGSPTASLVTGGREGRGCSDVDPGIQGWTDLAVSDAGEKVECRNBFPGSSQSEI IΖ gi 7106395 I 1313 UDVPΞSPTASLVT^G^EGRGCG|DVDPG JOGVVTDLAVSDAGEKVEBR^FGGSSQSEIIL gi 18602674 I 280 3DVPGSPTASLVTGGREGRGCSDVDPGIQGWTDLAVSDAGEKVECRN1FPGSSQSEII( gi 10438441 j 280 3DVPGSPTASLVTGGREGRGCSDVDPGIQGWTDLAVSDAGEKVECRNIFPGSSQSEII( gi 15341761 j 280 3DVPGSPTASLVTGGREGRGCSDVDPGIQGWTDLAVSDAGEKVECRNIFPGSSQSEIK gi 17437752 253 gGLQETVUVgVCLCS-j^YSPVTMGSgSVTYgDKtt^CRLgRLLgTQAjJiRLTACgHQTΪiIiT 311 1390 1400 1410 1420 1430 1440
NOVl6 1376 Al QNLTRLLYSLQAALT I QDSHI EIHRLVLQQQEGLSLGHS I LRGGPLQDQOKSRDADRζ 1434 gi I 7106395 I 1372 AIQNLTRLLYSLQAALTIQDSHIEIH|LVLQQ EgLg^HSPBRGGPLQDQ κSRg5g 1430 gijl8602674| 339 AIQNLTRLLYSLQAALTIQDSHIEIHRLVLQQQEGLSLGHSILRGGPLQDQIKSRDADRC 397 gijl043844lj 339 AIQNLTRLLYSLQAALTIQDSHIEIHRLVLQQQEGLSLGHSILRGGPLQDQIKSRDADRC 397 gij 15341761 j 339 MQNLTRLLYSLQAALTIQDSHIEIHRLVLQQQEGLSLGHSILRGGPLQDQIKSRDADRC 397 gij 17437752 j 312 PFALTAGAjjP^^EEgVj ALTJ^LJgljP- -jjE pVJjGSSSJJEVS - 351
1450 1460 1470 1480 1490 1500
NOVl6 1435 HEELANVHQLQHQLQQEQRRWLRRCEQQQRAQATRESWLQERERECQSQEELLLRSRGEL gi|7106395| 1431 jgEELAN@H|LQHQ@QQEQRRWJgR0Ci5QQQRgQ[^ ES LQgRERECQSQEELLLRgJR|EL gi|l8602674| 398 HEELANVHQLQHQLQQEQRRWLRRCEQQQRAQATRESWLQERERECQSQEELLLRSRGEL gi 110438441 j 398 HEELANVHQLQHQLQQEQRRWLRRCEQQQRAQATRESWLQERERECQSQEELLLRSRGEL gijl534176lj 398 HEELANVHQLQHQLQQEQRRWLRRCEQQQRAQATRESWLQERERECQSQEELLLRSRGEL gi j 17437752 j 351 -S|^-gSLJpLA J^G3AKLSgFFLCLPGGVQAj^PNE[gGA^PLpjjAJ5a 399
1510 1520 1530 1540 1550 1560
NOVl6 1495 DLQLQEYQHSLERLREGQRLVEREQARMRAQQSLLGHWKHGRQRSLPAVLLPGGPEVMEL 1554 is so
Figure imgf000127_0001
456
Figure imgf000127_0002
1630 1640 1650 1660 1670 1680
Figure imgf000127_0003
The presence of identifiable domains in the protein disclosed herein was determined by searches using algorithms such as PROSITE, Blocks, Pfam, ProDomain, Prints and then determining the Interpro number by crossing the domain match (or numbers) using the Interpro website (http:www.ebi.ac.uk/interpro/). Table 16F lists the domain description from DOMAIN analysis results against NOV16.
Figure imgf000127_0004
Consistent with other known members of the subunit family of proteins, NOV 16 has, for example, a RhoGEF signature sequence and homology to other members of the RhoGEF- like Protein Family. NOVl 6 nucleic acids, and the encoded polypeptides, according to the invention are useful in a variety of applications and contexts. For example, NOVl 6 nucleic acids and polypeptides can be used to identify proteins that are members of the RhoGEF-like Protein Family. The NOVl 6 nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOVl 6 activity or function. Specifically, the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., cellular activation, cellular replication, and signal transduction. These molecules can be used to treat, e.g. , cancer, trauma, regeneration (in vitro and in vivo), viral/bacterial/parasitic infections, diabetes, Von Hippel-Lindau (VHL) syndrome, pancreatitis, obesity, anemia , bleeding disorders, scleroderma, transplantation, hemophilia, hypercoagulation, idiopathic thrombocytopenic purpura, immunodeficiencies, graft versus host disease, Alzheimer's disease, stroke, tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, cerebral palsy, epilepsy, Lesch-Nyhan syndrome, multiple sclerosis, ataxia-telangiectasia, leukodystrophies, behavioral disorders, addiction, anxiety, pain, neurodegeneration, cirrhosis, transplantation, adrenoleukodystrophy , congenital adrenal hyperplasia, hemophilia, hypercoagulation, idiopathic thrombocytopenic purpura, autoimmune disease, allergies, immunodeficiencies, transplantation, graft versus host disease, lymphedema , allergies, immunodeficiencies, osteoporosis, hypercalceimia, arthritis, ankylosing spondylitis, scoliosis, tendinitis, systemic lupus erythematosus, autoimmune disease, asthma, emphysema, scleroderma, ARDS, endometriosis, fertility, hyperthyroidism, hypothyroidism, diabetes, autoimmune disease, renal artery stenosis, interstitial nephritis, glomerulonephritis, polycystic kidney disease, systemic lupus erythematosus, renal tubular acidosis, IgA nephropathy, hypercalceimia, psoriasis, actinic keratosis, tuberous sclerosis, acne, hair growth/loss, alopecia, pigmentation disorders, endocrine disorders as well as other diseases, disorders and conditions.
In addition, various NOV 16 nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. For example, the NOVl 6 nucleic acids and their encoded polypeptides include structural motifs that are characteristic of proteins belonging to the RhoGEF-like Protein Family.
GEF (Guanine nucleotide exchange factor) for Rho/Rac/Cdc42-like GTPases is also called Dbl-homologous (DH) domain. It appears that PH (pleckstrin homology) domains invariably occur C-terminal to RhoGEF/DH domains. Although the exact function of PH domains is unclear, several choices include binding to the beta/gamma subunit of heterotrimeric G proteins, binding to lipids, e.g. phosphatidylinositol-4,5-bisphosphate, binding to phosphorylated Ser/Thr residues, attachment to membranes by an unknown mechanism. The DAG_PE-binding domain binds two zinc ions; the ligands of these metal ions are probably the six cysteines and two histidines that are conserved in this domain and can regulate signal transduction by the PKC family of kinases.
NOVl 6 belongs to the guanine nucleotide exchange factor family of proteins which play a significant role in signal transduction. The guanine nucleotide exchange factor (GEF) domain that regulates GTP binding protein signaling. The GEF domain regulates positively the signaling cascades that utilize GTP-binding proteins (such as those of the ras superfamily) that function as molecular switches in fundamental events such as signal transduction, cytoskeleton dynamics and intracellular trafficking. An example of a protein containing GEF and PH domains is FGD1 (faciogenital dyplasia protein) Experiments have shown that the GEF and (PH) domains of FGD1 can bind specifically to the Rho family GTPase Cdc42Hs and stimulates the GDP-GTP exchange of the isoprenylated form of Cdc42Hs. The GEF domain of FGD1 has also been shown to activate 2 kinases involved in cell proliferation; the Jun NH2- terminal kinase and the p70 S6 kinase (Zheng et al; J. Biol. Chem 1996 Dec 27;271(52):33169-72). Thus, NOV16 polypeptide may play an important role in normal development as well as disease. This class of molecules (GEFs) is also being considered as a good drug target as the guanine nucleotide exchange factor RasGRP is a high -affinity target for diacylglycerol and phorbol esters and is bound by bryostatin 1, a compound currently in clinical trials (Lorenzo et al; Mol. Pharmacol 2000 May; 57(5):840-6). The homolog of RhoGEF, DRhoGEF2 fail to gastrulate due to a defect in cell shape changes required for tissue invagination and the mRNA is found throughout oogenesis and embryogenesis (Barrett et al. ; Cell 1997; 91(7):905-15; Werner et al; Gene 1997; 187(1):107-14). RhoGEF also interacts with c-Jun amino-terminal kinase (JNK) interacting protein- 1 (JIP-1). JEP-1 might function as a scaffold protein by complexing specific components of the JNK signaling pathway, namely JNK, mitogen-activated protein kinase kinase 7, and mixed lineage kinase 3 (Meyer et al; J Biol Chem 1999; 274(49):35113-8).
The NOV 16 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications in the mediation of blood and nerve physiology. As such, the NOV16 nucleic acids and polypeptides, antibodies and related compounds according to the invention may be used to treat blood and nervous system disorders, e.g., cancer, trauma, regeneration (in vitro and in vivo), viral/bacterial/parasitic infections, diabetes, Von Hippel-Lindau (VHL) syndrome, pancreatitis, obesity, anemia , bleeding disorders, scleroderma, transplantation, hemophilia, hypercoagulation, idiopathic thrombocytopenic purpura, immunodeficiencies, graft versus host disease, Alzheimer's disease, stroke, tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, cerebral palsy, epilepsy, Lesch-Nyhan syndrome, multiple sclerosis, ataxia-telangiectasia, leukodystrophies, behavioral disorders, addiction, anxiety, pain, neurodegeneration, cirrhosis, transplantation, adrenoleukodystrophy , congenital adrenal hyperplasia, hemophilia, hypercoagulation, idiopathic thrombocytopenic purpura, autoimmune disease, allergies, immunodeficiencies, transplantation, graft versus host disease, lymphedema , allergies, immunodeficiencies, osteoporosis, hypercalceimia, arthritis, ankylosing spondylitis, scoliosis, tendinitis, systemic lupus erythematosus, autoimmune disease, asthma, emphysema, scleroderma, ARDS, endometriosis, fertility, hyperthyroidism, hypothyroidism, diabetes, autoimmune disease, renal artery stenosis, interstitial nephritis, glomerulonephritis, polycystic kidney disease, systemic lupus erythematosus, renal tubular acidosis, IgA nephropathy, hypercalceimia, psoriasis, actinic keratosis, tuberous sclerosis, acne, hair growth loss, alopecia, pigmentation disorders, endocrine disorders as well as other diseases, disorders and conditions.
The NOVl 6 nucleic acids and polypeptides are useful for detecting specific cell types. For example, expression analysis has demonstrated that a NOVl 6 nucleic acid is expressed in Adipose, Umbilical Vein, Pancreas, Thymus, Brain, Lung, Kidney, Adrenal Gland/Suprarenal gland, Peripheral Blood, Lymph node, Cartilage, Mammary gland/Breast, Uterus, Prostate, Trachea, Cochlea, Dermis, Heart, Aorta, Coronary Artery, Thyroid, Liver, Bone, Bone Marrow, Spinal Cord, Cervix, and Retina. Additional utilities for NOVl 6 nucleic acids and polypeptides according to the invention are disclosed herein.
NOVX Nucleic Acids and Polypeptides
One aspect of the invention pertains to isolated nucleic acid molecules that encode NOVX polypeptides or biologically active portions thereof. Also included in the invention are nucleic acid fragments sufficient for use as hybridization probes to identify NOVX-encoding nucleic acids (e.g., NOVX mRNAs) and fragments for use as PCR primers for the amplification and/or mutation of NOVX nucleic acid molecules. As used herein, the term "nucleic acid molecule" is intended to include DNA molecules (e.g., cDNA or genomic DNA), RNA molecules (e.g., mRNA), analogs of the DNA or RNA generated using nucleotide analogs, and derivatives, fragments and homologs thereof. The nucleic acid molecule may be single-stranded or double-stranded, but preferably is comprised double- stranded DNA.
An NOVX nucleic acid can encode a mature NOVX polypeptide. As used herein, a "mature" form of a polypeptide or protein disclosed in the present invention is the product of a naturally occurring polypeptide or precursor form or proprotein. The naturally occurring polypeptide, precursor or proprotein includes, by way of nonlimiting example, the full-length gene product, encoded by the corresponding gene. Alternatively, it may be defined as the polypeptide, precursor or proprotein encoded by an ORF described herein. The product "mature" form arises, again by way of nonlimiting example, as a result of one or more naturally occurring processing steps as they may take place within the cell, or host cell, in which the gene product arises. Examples of such processing steps leading to a "mature" form of a polypeptide or protein include the cleavage of the N-terminal methionine residue encoded by the initiation codon of an ORF, or the proteolytic cleavage of a signal peptide or leader sequence. Thus a mature form arising from a precursor polypeptide or protein that has residues 1 to N, where residue 1 is the N-terminal methionine, would have residues 2 through N remaining after removal of the N-terminal methionine. Alternatively, a mature form arising from a precursor polypeptide or protein having residues 1 to N, in which an N-terminal signal sequence from residue 1 to residue M is cleaved, would have the residues from residue M+l to residue N remaining. Further as used herein, a "mature" form of a polypeptide or protein may arise from a step of post-translational modification other than a proteolytic cleavage event. Such additional processes include, byway of non-limiting example, glycosylation, myristoylation or phosphorylation. In general, a mature polypeptide or protein may result from the operation of only one of these processes, or a combination of any of them. The term "probes", as utilized herein, refers to nucleic acid sequences of variable length, preferably between at least about 10 nucleotides (nt), 100 nt, or as many as approximately, e.g., 6,000 nt, depending upon the specific use. Probes are used in the detection of identical, similar, or complementary nucleic acid sequences. Longer length probes are generally obtained from a natural or recombinant source, are highly specific, and much slower to hybridize than shorter-length oligomer probes. Probes may be single- or double-stranded and designed to have specificity in PCR, membrane-based hybridization technologies, or ELISA- like technologies. The term "isolated" nucleic acid molecule, as utilized herein, is one, which is separated from other nucleic acid molecules which are present in the natural source of the nucleic acid. Preferably, an "isolated" nucleic acid is free of sequences which naturally flank the nucleic acid (i.e., sequences located at the 5'- and 3'-termini of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived. For example, in various embodiments, the isolated NOVX nucleic acid molecules can contain less than about 5 kb, 4 kb, 3 kb, 2 kb, 1 kb, 0.5 kb or 0.1 kb of nucleotide sequences which naturally flank the nucleic acid molecule in genomic DNA of the cell/tissue from which the nucleic acid is derived (e.g., brain, heart, liver, spleen, etc.). Moreover, an "isolated" nucleic acid molecule, such as a cDNA molecule, can be substantially free of other cellular material or culture medium when produced by recombinant techniques, or of chemical precursors or other chemicals when chemically synthesized.
A nucleic acid molecule of the invention, e.g., a nucleic acid molecule having the nucleotide sequence SEQ ID NOS:l, 3, 5, 1, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, or a complement of this aforementioned nucleotide sequence, can be isolated using standard molecular biology techniques and the sequence information provided herein. Using all or a portion of the nucleic acid sequence of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33 as a hybridization probe, NOVX molecules can be isolated using standard hybridization and cloning techniques (e.g., as described in Sambrook, et al, (eds.), MOLECULAR CLONING: A LABORATORY MANUAL 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989; and Ausubel, et al, (eds.), CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York, NY, 1993.)
A nucleic acid of the invention can be amplified using cDNA, mRNA or alternatively, genomic DNA, as a template and appropriate oligonucleotide primers according to standard PCR amplification techniques. The nucleic acid so amplified can be cloned into an appropriate vector and characterized by DNA sequence analysis. Furthermore, oligonucleotides corresponding to NOVX nucleotide sequences can be prepared by standard synthetic techniques, e.g., using an automated DNA synthesizer.
As used herein, the term "oligonucleotide" refers to a series of linked nucleotide residues, which oligonucleotide has a sufficient number of nucleotide bases to be used in a PCR reaction. A short oligonucleotide sequence may be based on, or designed from, a genomic or cDNA sequence and is used to amplify, confirm, or reveal the presence of an identical, similar or complementary DNA or RNA in a particular cell or tissue. Oligonucleotides comprise portions of a nucleic acid sequence having about 10 nt, 50 nt, or 100 nt in length, preferably about 15 nt to 30 nt in length. In one embodiment of the invention, an oligonucleotide comprising a nucleic acid molecule less than 100 nt in length would further comprise at least 6 contiguous nucleotides SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, or a complement thereof. Oligonucleotides maybe chemically synthesized and may also be used as probes. h another embodiment, an isolated nucleic acid molecule of the invention comprises a nucleic acid molecule that is a complement of the nucleotide sequence shown in SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, or 33, or a portion of this nucleotide sequence (e.g., a fragment that can be used as a probe or primer or a fragment encoding a biologically-active portion of an NOVX polypeptide). A nucleic acid molecule that is complementary to the nucleotide sequence shown SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, or 33 is one that is sufficiently complementary to the nucleotide sequence shown SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, or 33 that it can hydrogen bond with little or no mismatches to the nucleotide sequence shown SEQ D NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, or 33, thereby forming a stable duplex.
As used herein, the term "complementary" refers to Watson-Crick or Hoogsteen base pairing between nucleotides units of a nucleic acid molecule, and the term "binding" means the physical or chemical interaction between two polypeptides or compounds or associated polypeptides or compounds or combinations thereof. Binding includes ionic, non-ionic, van der Waals, hydrophobic interactions, and the like. A physical interaction can be either direct or indirect. Indirect interactions may be through or due to the effects of another polypeptide or compound. Direct binding refers to interactions that do not take place through, or due to, the effect of another polypeptide or compound, but instead are without other substantial chemical intermediates.
Fragments provided herein are defined as sequences of at least 6 (contiguous) nucleic acids or at least 4 (contiguous) amino acids, a length sufficient to allow for specific hybridization in the case of nucleic acids or for specific recognition of an epitope in the case of amino acids, respectively, and are at most some portion less than a full length sequence. Fragments may be derived from any contiguous portion of a nucleic acid or amino acid sequence of choice. Derivatives are nucleic acid sequences or amino acid sequences formed from the native compounds either directly or by modification or partial substitution. Analogs are nucleic acid sequences or amino acid sequences that have a structure similar to, but not identical to, the native compound but differs from it in respect to certain components or side chains. Analogs may be synthetic or from a different evolutionary origin and may have a similar or opposite metabolic activity compared to wild type. Homologs are nucleic acid sequences or amino acid sequences of a particular gene that are derived from different species. Derivatives and analogs may be full length or other than full length, if the derivative or analog contains a modified nucleic acid or amino acid, as described below. Derivatives or analogs of the nucleic acids or proteins of the invention include, but are not limited to, molecules comprising regions that are substantially homologous to the nucleic acids or proteins of the invention, in various embodiments, by at least about 70%, 80%, or 95% identity (with a preferred identity of 80-95%) over a nucleic acid or amino acid sequence of identical size or when compared to an aligned sequence in which the alignment is done by a computer homology program known in the art, or whose encoding nucleic acid is capable of hybridizing to the complement of a sequence encoding the aforementioned proteins under stringent, moderately stringent, or low stringent conditions. See e.g. Ausubel, βt al, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York, NY, 1993, and below. A "homologous nucleic acid sequence" or "homologous amino acid sequence," or variations thereof, refer to sequences characterized by a homology at the nucleotide level or amino acid level as discussed above. Homologous nucleotide sequences encode those sequences coding for isoforms of NOVX polypeptides. Isoforms can be expressed in different tissues of the same organism as a result of, for example, alternative splicing of RNA. Alternatively, isoforms can be encoded by different genes. In the invention, homologous nucleotide sequences include nucleotide sequences encoding for an NOVX polypeptide of species other than humans, including, but not limited to: vertebrates, and thus can include, e.g., frog, mouse, rat, rabbit, dog, cat cow, horse, and other organisms. Homologous nucleotide sequences also include, but are not limited to, naturally occurring allelic variations and mutations of the nucleotide sequences set forth herein. A homologous nucleotide sequence does not, however, include the exact nucleotide sequence encoding human NOVX protein. Homologous nucleic acid sequences include those nucleic acid sequences that encode conservative amino acid substitutions (see below) in SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, as well as a polypeptide possessing NOVX biological activity. Various biological activities of the NOVX proteins are described below.
An NOVX polypeptide is encoded by the open reading frame ("ORF") of an NOVX nucleic acid. An ORF corresponds to a nucleotide sequence that could potentially be translated into a polypeptide. A stretch of nucleic acids comprising an ORF is uninterrupted by a stop codon. An ORF that represents the coding sequence for a full protein begins with an ATG "start" codon and terminates with one of the three "stop" codons, namely, TAA, TAG, or TGA. For the purposes of this invention, an ORF may be any part of a coding sequence, with or without a start codon, a stop codon, or both. For an ORF to be considered as a good candidate for coding for a bonaβde cellular protein, a mimmum size requirement is often set, e.g., a stretch of DNA that would encode a protein of 50 amino acids or more.
The nucleotide sequences determined from the cloning of the human NOVX genes allows for the generation of probes and primers designed for use in identifying and/or cloning NOVX homologues in other cell types, e.g. from other tissues, as well as NOVX homologues from other vertebrates. The probe/primer typically comprises substantially purified oligonucleotide. The oligonucleotide typically comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least about 12, 25, 50, 100, 150, 200, 250, 300, 350 or 400 consecutive sense strand nucleotide sequence SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, or 33; or an anti-sense strand nucleotide sequence of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, or 33; or of a naturally occurring mutant of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33. Probes based on the human NOVX nucleotide sequences can be used to detect transcripts or genomic sequences encoding the same or homologous proteins, hi various embodiments, the probe further comprises a label group attached thereto, e.g. the label group can be a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor. Such probes can be used as a part of a diagnostic test kit for identifying cells or tissues which mis- express an NOVX protein, such as by measuring a level of an NOVX-encoding nucleic acid in a sample of cells from a subject e.g., detecting NOVX mRNA levels or determining whether a genomic NOVX gene has been mutated or deleted.
"A polypeptide having a biologically-active portion of an NOVX polypeptide" refers to polypeptides exhibiting activity similar, but not necessarily identical to, an activity of a polypeptide of the invention, including mature forms, as measured in a particular biological assay, with or without dose dependency. A nucleic acid fragment encoding a "biologically- active portion of NOVX" can be prepared by isolating a portion SEQ ID NOS: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, or 33, that encodes a polypeptide having an NOVX biological activity (the biological activities of the NOVX proteins are described below), expressing the encoded portion of NOVX protein (e.g., by recombinant expression in vitro) and assessing the activity of the encoded portion of NOVX. NOVX Nucleic Acid and Polypeptide Variants
The invention further encompasses nucleic acid molecules that differ from the nucleotide sequences shown in SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33 due to degeneracy of the genetic code and thus encode the same NOVX proteins as that encoded by the nucleotide sequences shown in SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33. In another embodiment, an isolated nucleic acid molecule of the invention has a nucleotide sequence encoding a protein having an amino acid sequence shown in SEQ D NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, or 34. In addition to the human NOVX nucleotide sequences shown in SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, it will be appreciated by those skilled in the art that DNA sequence polymorphisms that lead to changes in the amino acid sequences of the NOVX polypeptides may exist within a population (e.g., the human population). Such genetic polymorphism in the NOVX genes may exist among individuals within a population due to natural allelic variation. As used herein, the terms "gene" and "recombinant gene" refer to nucleic acid molecules comprising an open reading frame (ORF) encoding an NOVX protein, preferably a vertebrate NOVX protein. Such natural allelic variations can typically result in 1-5% variance in the nucleotide sequence of the NOVX genes. Any and all such nucleotide variations and resulting amino acid polymorphisms in the NOVX polypeptides, which are the result of natural allelic variation and that do not alter the functional activity of the NOVX polypeptides, are intended to be within the scope of the invention.
Moreover, nucleic acid molecules encoding NOVX proteins from other species, and thus that have a nucleotide sequence that differs from the human SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33 are intended to be within the scope of the invention. Nucleic acid molecules corresponding to natural allelic variants and homologues of the NOVX cDNAs of the invention can be isolated based on their homology to the human NOVX nucleic acids disclosed herein using the human cDNAs, or a portion thereof, as a hybridization probe according to standard hybridization techniques under stringent hybridization conditions.
Accordingly, in another embodiment, an isolated nucleic acid molecule of the invention is at least 6 nucleotides in length and hybridizes under stringent conditions to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33. In another embodiment, the nucleic acid is at least 10, 25, 50, 100, 250, 500, 750, 1000, 1500, or 2000 or more nucleotides in length, hi yet another embodiment, an isolated nucleic acid molecule of the invention hybridizes to the coding region. As used herein, the term "hybridizes under stringent conditions" is intended to describe conditions for hybridization and washing under which nucleotide sequences at least 60% homologous to each other typically remain hybridized to each other.
Homologs (i.e., nucleic acids encoding NOVX proteins derived from species other than human) or other related sequences (e.g., paralogs) can be obtained by low, moderate or high stringency hybridization with all or a portion of the particular human sequence as a probe using methods well known in the art for nucleic acid hybridization and cloning.
As used herein, the phrase "stringent hybridization conditions" refers to conditions under which a probe, primer or oligonucleotide will hybridize to its target sequence, but to no other sequences. Stringent conditions are sequence-dependent and will be different in different circumstances. Longer sequences hybridize specifically at higher temperatures than shorter sequences. Generally, stringent conditions are selected to be about 5 °C lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH. The Tm is the temperature (under defined ionic strength, pH and nucleic acid concentration) at which 50% of the probes complementary to the target sequence hybridize to the target sequence at equilibrium. Since the target sequences are generally present at excess, at Tm, 50% of the probes are occupied at equilibrium. Typically, stringent conditions will be those in which the salt concentration is less than about 1.0 M sodium ion, typically about 0.01 to 1.0 M sodium ion (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 30°C for short probes, primers or oligonucleotides (e.g., 10 nt to 50 nt) and at least about 60°C for longer probes, primers and oligonucleotides. Stringent conditions may also be achieved with the addition of destabilizing agents, such as formamide.
Stringent conditions are known to those skilled in the art and can be found in Ausubel, et al, (eds.), CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6. Preferably, the conditions are such that sequences at least about 65%, 70%, 15%, 85%, 90%, 95%, 98%, or 99% homologous to each other typically remain hybridized to each other. A non-limiting example of stringent hybridization conditions are hybridization in a high salt buffer comprising 6X SSC, 50 mM Tris-HCl (pH 7.5), 1 mM EDTA, 0.02% PVP, 0.02% Ficoll, 0.02% BSA, and 500 mg ml denatured salmon sperm DNA at 65°C, followed by one or more washes in 0.2X SSC, 0.01% BSA at 50°C. An isolated nucleic acid molecule of the invention that hybridizes under stringent conditions to the sequences SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, corresponds to a naturally-occurring nucleic acid molecule. As used herein, a "naturally-occurring" nucleic acid molecule refers to an RNA or DNA molecule having a nucleotide sequence that occurs in nature (e.g., encodes a natural protein).
In a second embodiment, a nucleic acid sequence that is hybridizable to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, or fragments, analogs or derivatives thereof, under conditions of moderate stringency is provided. A non-limiting example of moderate stringency hybridization conditions are hybridization in 6X SSC, 5X Denhardt's solution, 0.5% SDS and 100 mg/ml denatured salmon sperm DNA at 55°C, followed by one or more washes in IX SSC, 0.1% SDS at 37°C. Other conditions of moderate stringency that may be used are well-known within the art. See, e.g., Ausubel, et al. (eds.), 1993, CURRENT
PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, NY, and Kriegler, 1990; GENE TRANSFER AND EXPRESSION, A LABORATORY MANUAL, Stockton Press, NY.
In a third embodiment, a nucleic acid that is hybridizable to the nucleic acid molecule comprising the nucleotide sequences SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, or fragments, analogs or derivatives thereof, under conditions of low stringency, is provided. A non-limiting example of low stringency hybridization conditions are hybridization in 35% formamide, 5X SSC, 50 mM Tris-HCl (pH 7.5), 5 mM EDTA, 0.02% PVP, 0.02% Ficoll, 0.2% BSA, 100 mg/ml denatured salmon sperm DNA, 10% (wt/vol) dextran sulfate at 40°C, followed by one or more washes in 2X SSC, 25 mM Tris-HCl (pH 7.4), 5 mM EDTA, and 0.1 % SDS at 50°C. Other conditions of low stringency that may be used are well known in the art (e.g., as employed for cross-species hybridizations). See, e.g., Ausubel, et al. (eds.), 1993, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, NY, and Kriegler, 1990, GENE TRANSFER AND EXPRESSION, A LABORATORY MANUAL, Stockton Press, NY; Shilo and Weinberg, 1981. Proc Natl Acad Sci USA 78: 6789-6792.
Conservative Mutations
In addition to naturally-occurring allelic variants of NOVX sequences that may exist in the population, the skilled artisan will further appreciate that changes can be introduced by mutation into the nucleotide sequences SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23,
25, 27, 29, 31, and 33, thereby leading to changes in the amino acid sequences of the encoded
NOVX proteins, without altering the functional ability of said NOVX proteins. For example, nucleotide substitutions leading to amino acid substitutions at "non-essential" amino acid residues can be made in the sequence SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, or 34. A "non-essential" amino acid residue is a residue that can be altered from the wild-type sequences of the NOVX proteins without altering their biological activity, whereas an "essential" amino acid residue is required for such biological activity. For example, amino acid residues that are conserved among the NOVX proteins of the invention are predicted to be particularly non-amenable to alteration. Amino acids for which conservative substitutions can be made are well-known within the art.
Another aspect of the invention pertains to nucleic acid molecules encoding NOVX proteins that contain changes in amino acid residues that are not essential for activity. Such NOVX proteins differ in amino acid sequence from SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34 yet retain biological activity. In one embodiment, the isolated nucleic acid molecule comprises a nucleotide sequence encoding a protein, wherein the protein comprises an amino acid sequence at least about 45% homologous to the amino acid sequences SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34. Preferably, the protein encoded by the nucleic acid molecule is at least about 60% homologous to SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34; more preferably at least about 70% homologous SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, or 34; still more preferably at least about 80% homologous to SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, or 34; even more preferably at least about 90% homologous to SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, or 34; and most preferably at least about 95% homologous to SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, or 34.
An isolated nucleic acid molecule encoding an NOVX protein homologous to the protein of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, or 34 can be created by introducing one or more nucleotide substitutions, additions or deletions into the nucleotide sequence of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, such that one or more amino acid substitutions, additions or deletions are introduced into the encoded protein.
Mutations can be introduced into SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33 by standard techniques, such as site-directed mutagenesis and PCR-mediated mutagenesis. Preferably, conservative amino acid substitutions are made at one or more predicted, non-essential amino acid residues. A "conservative amino acid substitution" is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined within the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Thus, a predicted non-essential amino acid residue in the NOVX protein is replaced with another amino acid residue from the same side chain family. Alternatively, in another embodiment, mutations can be introduced randomly along all or part of an NOVX coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for NOVX biological activity to identify mutants that retain activity. Following mutagenesis of SEQ ID NOS : 1 , 3 , 5, 7, 9, 11 , 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, the encoded protein can be expressed by any recombinant technology known in the art and the activity of the protein can be determined. The relatedness of amino acid families may also be determined based on side chain interactions. Substituted amino acids may be fully conserved "strong" residues or fully conserved "weak" residues. The "strong" group of conserved amino acid residues may be any one of the following groups: STA, NEQK, NHQK, NDEQ, QHRK, MILV, MILF, HY, FYW, wherein the single letter amino acid codes are grouped by those amino acids that may be substituted for each other. Likewise, the "weak" group of conserved residues may be any one of the following: CSA, ATV, SAG, STNK, STPA, SGND, SNDEQK, NDEQHK, NEQHRK, HFY, wherein the letters within each group represent the single letter amino acid code. In one embodiment, a mutant NOVX protein can be assayed for (i) the ability to form proteimprotein interactions with other NOVX proteins, other cell-surface proteins, or biologically-active portions thereof, (ii) complex formation between a mutant NOVX protein and an NOVX ligand; or (iii) the ability of a mutant NOVX protein to bind to an intracellular target protein or biologically-active portion thereof; (e.g. avidin proteins). In yet another embodiment, a mutant NOVX protein can be assayed for the ability to regulate a specific biological function (e.g., regulation of insulin release).
Antisense Nucleic Acids
Another aspect of the invention pertains to isolated antisense nucleic acid molecules that are hybridizable to or complementary to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, or fragments, analogs or derivatives thereof. An "antisense" nucleic acid comprises a nucleotide sequence that is complementary to a "sense" nucleic acid encoding a protein (e.g., complementary to the coding strand of a double-stranded cDNA molecule or complementary to an mRNA sequence). In specific aspects, antisense nucleic acid molecules are provided that comprise a sequence complementary to at least about 10, 25, 50, 100, 250 or 500 nucleotides or an entire NOVX coding strand, or to only a portion thereof. Nucleic acid molecules encoding fragments, homologs, derivatives and analogs of an NOVX protein of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, or 34, or antisense nucleic acids complementary to an NOVX nucleic acid sequence of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, are additionally provided.
In one embodiment, an antisense nucleic acid molecule is antisense to a "coding region" of the coding strand of a nucleotide sequence encoding an NOVX protein. The term "coding region" refers to the region of the nucleotide sequence comprising codons which are translated into amino acid residues, hi another embodiment, the antisense nucleic acid molecule is antisense to a "noncoding region" of the coding strand of a nucleotide sequence encoding the NOVX protein. The term "noncoding region" refers to 5' and 3' sequences which flank the coding region that are not translated into amino acids (i.e., also referred to as 5' and 3 ' untranslated regions) .
Given the coding strand sequences encoding the NOVX protein disclosed herein, antisense nucleic acids of the invention can be designed according to the rules of Watson and Crick or Hoogsteen base pairing. The antisense nucleic acid molecule can be complementary to the entire coding region of NOVX mRNA, but more preferably is an oligonucleotide that is antisense to only a portion of the coding or noncoding region of NOVX mRNA. For example, the antisense oligonucleotide can be complementary to the region surrounding the translation start site of NOVX mRNA. An antisense oligonucleotide can be, for example, about 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 nucleotides in length. An antisense nucleic acid of the invention can be constructed using chemical synthesis or enzymatic ligation reactions using procedures known in the art. For example, an antisense nucleic acid (e.g., an antisense oligonucleotide) can be chemically synthesized using naturally-occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids (e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used). Examples of modified nucleotides that can be used to generate the antisense nucleic acid include: 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxyhnethyl) uracil, 5-carboxymethylaminomethyl- 2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5'-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5 -methyluracil, uracil-5-oxyacetic acid methylester, uracil-5-oxyacetic acid (v), 5-methyl-2-thiouracil, 3-(3-amino-3-N-2-carboxypropyl) uracil, (acp3)w, and 2,6-diaminopurine. Alternatively, the antisense nucleic acid can be produced biologically using an expression vector into which a nucleic acid has been subcloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest, described further in the following subsection).
The antisense nucleic acid molecules of the invention are typically administered to a subject or generated in situ such that they hybridize with or bind to cellular mRNA and/or genomic DNA encoding an NOVX protein to thereby inhibit expression of the protein (e.g., by inhibiting transcription and/or translation). The hybridization can be by conventional nucleotide complementarity to form a stable duplex, or, for example, in the case of an antisense nucleic acid molecule that binds to DNA duplexes, through specific interactions in the maj or groove of the double helix. An example of a route of administration of antisense nucleic acid molecules of the invention includes direct injection at a tissue site. Alternatively, antisense nucleic acid molecules can be modified to target selected cells and then administered systemically. For example, for systemic administration, antisense molecules can be modified such that they specifically bind to receptors or antigens expressed on a selected cell surface (e.g., by linking the antisense nucleic acid molecules to peptides or antibodies that bind to cell surface receptors or antigens). The antisense nucleic acid molecules can also be delivered to cells using the vectors described herein. To achieve sufficient nucleic acid molecules, vector constructs in which the antisense nucleic acid molecule is placed under the control of a strong pol IT or pol III promoter are preferred.
In yet another embodiment, the antisense nucleic acid molecule of the invention is an α-anomeric nucleic acid molecule. An α-anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual β -units, the strands run parallel to each other. See, e.g., Gaultier, et al, 1987. Nucl. Acids Res. 15: 6625-6641. The antisense nucleic acid molecule can also comprise a 2'-o-methylribonucleotide (See, e.g., Inoue, et al. 1987. Nucl. Acids Res. 15: 6131-6148) or a chimeric RNA-DNA analogue (See, e.g., Inoue, et al, 1987. FEBSLett. 215: 327-330. Ribozymes and PNA Moieties
Nucleic acid modifications include, by way of non-limiting example, modified bases, and nucleic acids whose sugar phosphate backbones are modified or derivatized. These modifications are carried out at least in part to enhance the chemical stability of the modified nucleic acid, such that they may be used, for example, as antisense binding nucleic acids in therapeutic applications in a subject.
In one embodiment, an antisense nucleic acid of the invention is a ribozyme. Ribozymes are catalytic RNA molecules with ribonuclease activity that are capable of cleaving a single-stranded nucleic acid, such as an mRNA, to which they have a complementary region. Thus, ribozymes (e.g., hammerhead ribozymes as described in Haselhoff and Gerlach 1988. Nature 334: 585-591) can be used to catalytically cleave NOVX mRNA transcripts to thereby inhibit translation of NOVX mRNA. A ribozyme having specificity for an NOVX-encoding nucleic acid can be designed based upon the nucleotide sequence of an NOVX cDNA disclosed herein (i.e., SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33). For example, a derivative of a Tetrahymena L-19 TVS RNA can be constructed in which the nucleotide sequence of the active site is complementary to the nucleotide sequence to be cleaved in an NOVX-encoding mRNA. See, e.g., U.S. Patent 4,987,071 to Cech, et al. and U.S. Patent 5,116,742 to Cech, et al. NOVX mRNA can also be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules. See, e.g., Bartel et al, (1993) Science 261:1411-1418.
Alternatively, NOVX gene expression can be inhibited by targeting nucleotide sequences complementary to the regulatory region of the NOVX nucleic acid (e.g., the NOVX promoter and/or enhancers) to form triple helical structures that prevent transcription of the NOVX gene in target cells. See, e.g., Helene, 1991. Anticancer Drug Des. 6: 569-84; Helene, et al. 1992. Ann. N.Y. Acad. Sci. 660: 27-36; Maher, 1992. Bioassays 14: 807-15.
In various embodiments, the NOVX nucleic acids can be modified at the base moiety, sugar moiety or phosphate backbone to improve, e.g., the stability, hybridization, or solubility of the molecule. For example, the deoxyribose phosphate backbone of the nucleic acids can be modified to generate peptide nucleic acids. See, e.g., Hyrup, et al, 1996. BioorgMed
Chem 4: 5-23. As used herein, the terms "peptide nucleic acids" or "PNAs" refer to nucleic acid mimics (e.g., DNA mimics) in which the deoxyribose phosphate backbone is replaced by a pseudopeptide backbone and only the four natural nucleobases are retained. The neutral backbone of PNAs has been shown to allow for specific hybridization to DNA and RNA under conditions of low ionic strength. The synthesis of PNA oligomers can be performed using standard solid phase peptide synthesis protocols as described in Hyrup, et al, 1996. supra; Perry-O'Keefe, et al, 1996. Proc. Natl. Acad. Sci. USA 93: 14670-14675.
PNAs of NOVX can be used in therapeutic and diagnostic applications. For example, PNAs can be used as antisense or antigene agents for sequence-specific modulation of gene expression by, e.g., inducing transcription or translation arrest or inhibiting replication. PNAs of NOVX can also be used, for example, in the analysis of single base pair mutations in a gene (e.g. , PNA directed PCR clamping; as artificial restriction enzymes when used in combination with other enzymes, e.g., S\ nucleases (See, Hyrup, et al, I996.supra); or as probes or primers for DNA sequence and hybridization (See, Hyrup, et al, 1996, supra; Perry-O'Keefe, et al, 1996. supra).
In another embodiment, PNAs of NOVX can be modified, e.g., to enhance their stability or cellular uptake, by attaching lipophilic or other helper groups to PNA, by the formation of PNA-DNA chimeras, or by the use of liposomes or other techniques of drug delivery known in the art. For example, PNA-DNA chimeras of NOVX can be generated that may combine the advantageous properties of PNA and DNA. Such chimeras allow DNA recognition enzymes (e.g., RNase H and DNA polymerases) to interact with the DNA portion while the PNA portion would provide high binding affinity and specificity. PNA-DNA chimeras can be linked using linkers of appropriate lengths selected in terms of base stacking, number of bonds between the nucleobases, and orientation (see, Hyrup, et al, 1996. supra). The synthesis of PNA-DNA chimeras can be performed as described in Hyrup, et al, 1996. supra and Finn, et al, 1996. Nucl Acids Res 24: 3357-3363. For example, a DNA chain can be synthesized on a solid support using standard phosphoramidite coupling chemistry, and modified nucleoside analogs, e.g., 5'-(4-methoxytrityl)amino-5'-deoxy-thymidine phosphoramidite, can be used between the PNA and the 5' end of DNA. See, e.g., Mag, et al, 1989. Nucl Acid Res 17: 5973-5988. PNA monomers are then coupled in a stepwise manner to produce a chimeric molecule with a 5' PNA segment and a 3' DNA segment. See, e.g., Finn, et al, 1996. supra. Alternatively, chimeric molecules can be synthesized with a 5' DNA segment and a 3' PNA segment. See, e.g., Petersen, et al, 1975. Bioorg. Med. Chem. Lett. 5: 1119-11124. hi other embodiments, the oligonucleotide may include other appended groups such as peptides (e.g., for targeting host cell receptors in vivo), or agents facilitating transport across the cell membrane (see, e.g., Letsinger, et al, 1989. Proc. Natl. Acad. Sci. U.S.A. 86: 6553-6556; Lemaifre, et al, 1987. Proc. Natl. Acad. Sci. 84: 648-652; PCT Publication No. WO88/09810) or the blood-brain barrier (see, e.g., PCT Publication No. WO 89/10134). In addition, oligonucleotides can be modified with hybridization triggered cleavage agents (see, e.g., Krol, et al, 1988. BioTechniques 6:958-976) or intercalating agents (.see, e.g., Zon, 1988. Pharm. Res. 5: 539-549). To this end, the oligonucleotide may be conjugated to another molecule, e.g., a peptide, a hybridization triggered cross-linking agent, a transport agent, a hybridization-triggered cleavage agent, and the like.
NOVX Polypeptides
A polypeptide according to the invention includes a polypeptide including the amino acid sequence of NOVX polypeptides whose sequences are provided in SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, or 34. The invention also includes a mutant or variant protein any of whose residues may be changed from the corresponding residues shown in SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, or 34 while still encoding a protein that maintains its NOVX activities and physiological functions, or a functional fragment thereof. hi general, an NOVX variant that preserves NOVX-like function includes any variant in which residues at a particular position in the sequence have been substituted by other amino acids, and further include the possibility of inserting an additional residue or residues between two residues of the parent protein as well as the possibility of deleting one or more residues from the parent sequence. Any amino acid substitution, insertion, or deletion is encompassed by the invention. In favorable circumstances, the substitution is a conservative substitution as defined above.
One aspect of the invention pertains to isolated NOVX proteins, and biologically- active portions thereof, or derivatives, fragments, analogs or homologs thereof. Also provided are polypeptide fragments suitable for use as immunogens to raise anti-NOVX antibodies. Tn one embodiment, native NOVX proteins can be isolated from cells or tissue sources by an appropriate purification scheme using standard protein purification techniques. In another embodiment, NOVX proteins are produced by recombinant DNA techniques. Alternative to recombinant expression, an NOVX protein or polypeptide can be synthesized chemically using standard peptide synthesis techniques.
An "isolated" or "purified" polypeptide or protein or biologically-active portion thereof is substantially free of cellular material or other contaminating proteins from the cell or tissue source from which the NOVX protein is derived, or substantially free from chemical precursors or other chemicals when chemically synthesized. The language "substantially free of cellular material" includes preparations of NOVX proteins in which the protein is separated from cellular components of the cells from which it is isolated or recombinantly-produced. In one embodiment, the language "substantially free of cellular material" includes preparations of NOVX proteins having less than about 30% (by dry weight) of non-NOVX proteins (also referred to herein as a "contaminating protein"), more preferably less than about 20% of non-NOVX proteins, still more preferably less than about 10% of non-NOVX proteins, and most preferably less than about 5% of non-NOVX proteins. When the NOVX protein or biologically-active portion thereof is recombinantly-produced, it is also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%, more preferably less than about 10%, and most preferably less than about 5% of the volume of the NOVX protein preparation.
The language "substantially free of chemical precursors or other chemicals" includes preparations of NOVX proteins in which the protein is separated from chemical precursors or other chemicals that are involved in the synthesis of the protein. In one embodiment, the language "substantially free of chemical precursors or other chemicals" includes preparations of NOVX proteins having less than about 30% (by dry weight) of chemical precursors or non-NOVX chemicals, more preferably less than about 20% chemical precursors or non-NOVX chemicals, still more preferably less than about 10% chemical precursors or non-NOVX chemicals, and most preferably less than about 5% chemical precursors or non-NOVX chemicals. Biologically-active portions of NOVX proteins include peptides comprising amino acid sequences sufficiently homologous to or derived from the amino acid sequences of the NOVX proteins (e.g., the amino acid sequence shown in SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, or 34) that include fewer amino acids than the full-length NOVX proteins, and exhibit at least one activity of an NOVX protein. Typically, biologically- active portions comprise a domain or motif with at least one activity of the NOVX protein. A biologically-active portion of an NOVX protein can be a polypeptide which is, for example, 10, 25, 50, 100 or more amino acid residues in length.
Moreover, other biologically-active portions, in which other regions of the protein are deleted, can be prepared by recombinant techniques and evaluated for one or more of the functional activities of a native NOVX protein.
In an embodiment, the NOVX protein has an amino acid sequence shown SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, or 34. In other embodiments, the NOVX protein is substantially homologous to SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, or 34, and retains the functional activity of the protein of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, or 34, yet differs in amino acid sequence due to natural allelic variation or mutagenesis, as described in detail, below.
Accordingly, in another embodiment, the NOVX protein is a protein that comprises an amino acid sequence at least about 45% homologous to the amino acid sequence SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, or 34, and retains the functional activity of the NOVX proteins of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, or 34.
Determining Homology Between Two or More Sequences To determine the percent homology of two amino acid sequences or of two nucleic acids, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino or nucleic acid sequence). The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are homologous at that position (i.e., as used herein amino acid or nucleic acid "homology" is equivalent to amino acid or nucleic acid "identity").
The nucleic acid sequence homology may be determined as the degree of identity between two sequences. The homology may be determined using computer programs known in the art, such as GAP software provided in the GCG program package. See, Needleman and Wunsch, 1970. JMol Biol 48: 443-453. Using GCG GAP software with the following settings for nucleic acid sequence comparison: GAP creation penalty of 5.0 and GAP extension penalty of 0.3, the coding region of the analogous nucleic acid sequences referred to above exhibits a degree of identity preferably of at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99%, with the CDS (encoding) part of the DNA sequence shown in SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33.
The term "sequence identity" refers to the degree to which two polynucleotide or polypeptide sequences are identical on a residue-by-residue basis over a particular region of comparison. The term "percentage of sequence identity" is calculated by comparing two optimally aligned sequences over that region of comparison, determining the number of positions at which the identical nucleic acid base (e.g., A, T, C, G, U, or I, in the case of nucleic acids) occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the region of comparison (i.e., the window size), and multiplying the result by 100 to yield the percentage of sequence identity. The term "substantial identity" as used herein denotes a characteristic of a polynucleotide sequence, wherein the polynucleotide comprises a sequence that has at least 80 percent sequence identity, preferably at least 85 percent identity and often 90 to 95 percent sequence identity, more usually at least 99 percent sequence identity as compared to a reference sequence over a comparison region.
Chimeric and Fusion Proteins
The invention also provides NOVX chimeric or fusion proteins. As used herein, an
NOVX "chimeric protein" or "fusion protein" comprises an NOVX polypeptide operatively- linked to a non-NOVX polypeptide. An "NOVX polypeptide" refers to a polypeptide having an amino acid sequence corresponding to an NOVX protein SEQ TD NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, or 34), whereas a "non-NOVX polypeptide" refers to a polypeptide having an amino acid sequence corresponding to a protein that is not substantially homologous to the NOVX protein, e.g., a protein that is different from the NOVX protein and that is derived from the same or a different organism. Within an NOVX fusion protein the NOVX polypeptide can correspond to all or a portion of an NOVX protein. In one embodiment, an NOVX fusion protein comprises at least one biologically-active portion of an NOVX protein. In another embodiment, an NOVX fusion protein comprises at least two biologically-active portions of an NOVX protein. In yet another embodiment, an NOVX fusion protein comprises at least three biologically-active portions of an NOVX protein.
Within the fusion protein, the term "operatively-linked" is intended to indicate that the NOVX polypeptide and the non-NOVX polypeptide are fused in-frame with one another. The non-NOVX polypeptide can be fused to the N-terminus or C-terminus of the NOVX polypeptide. h one embodiment, the fusion protein is a GST-NO VX fusion protein in which the
NOVX sequences are fused to the C-terminus of the GST (glutathione S-transferase) sequences. Such fusion proteins can facilitate the purification of recombinant NOVX polypeptides. hi another embodiment, the fusion protein is an NOVX protein containing a heterologous signal sequence at its N-terminus. In certain host cells (e.g., mammalian host cells), expression and/or secretion of NOVX can be increased through use of a heterologous signal sequence.
In yet another embodiment, the fusion protein is an NOVX-immunoglobulin fusion protein in which the NOVX sequences are fused to sequences derived from a member of the immunoglobulin protein family. The NONX-immunoglobulin fusion proteins of the invention can be incorporated into pharmaceutical compositions and administered to a subject to inhibit an interaction between an NOVX ligand and an NOVX protein on the surface of a cell, to thereby suppress NOVX-mediated signal transduction in vivo. The NOVX-immunoglobulin fusion proteins can be used to affect the bioavailability of an NOVX cognate ligand. Inhibition of the NOVX ligand/NOVX interaction may be useful therapeutically for both the treatment of proliferative and differentiative disorders, as well as modulating (e.g. promoting or inhibiting) cell survival. Moreover, the NOVX-immunoglobulin fusion proteins of the invention can be used as immunogens to produce anti-NOVX antibodies in a subject, to purify NOVX ligands, and in screening assays to identify molecules that inhibit the interaction of NOVX with an NOVX ligand.
An NOVX chimeric or fusion protein of the invention can be produced by standard recombinant DNA teclmiques. For example, DNA fragments coding for the different polypeptide sequences are ligated together in-frame in accordance with conventional techniques, e.g., by employing blunt-ended or stagger-ended termini for ligation, restriction enzyme digestion to provide for appropriate termini, filling-in of cohesive ends as appropriate, alkaline phosphatase treatment to avoid undesirable joining, and enzymatic ligation. In another embodiment, the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers. Alternatively, PCR amplification of gene fragments can be carried out using anchor primers that give rise to complementary overhangs between two consecutive gene fragments that can subsequently be annealed and reamplified to generate a chimeric gene sequence (see, e.g., Ausubel, et al. (eds.) CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, 1992). Moreover, many expression vectors are commercially available that already encode a fusion moiety (e.g., a GST polypeptide). An NOVX-encoding nucleic acid can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the NOVX protein.
NOVX Agonists and Antagonists
The invention also pertains to variants of the NOVX proteins that function as either
NOVX agonists (i.e., mimetics) or as NOVX antagonists. Variants of the NOVX protein can be generated by mutagenesis (e.g., discrete point mutation or truncation of the NOVX protein). An agonist of the NOVX protein can retain substantially the same, or a subset of, the biological activities of the naturally occurring form of the NOVX protein. An antagonist of the NOVX protein can inhibit one or more of the activities of the naturally occurring form of the NOVX protein by, for example, competitively binding to a downstream or upstream member of a cellular signaling cascade which includes the NOVX protein. Thus, specific biological effects can be elicited by treatment with a variant of limited function. In one embodiment, treatment of a subject with a variant having a subset of the biological activities of the naturally occurring form of the protein has fewer side effects in a subject relative to treatment with the naturally occurring form of the NOVX proteins. Variants of the NOVX proteins that function as either NOVX agonists (i.e., mimetics) or as NOVX antagonists can be identified by screening combinatorial libraries of mutants (e.g., truncation mutants) of the NOVX proteins for NOVX protein agonist or antagonist activity. In one embodiment, a variegated library of NOVX variants is generated by combinatorial mutagenesis at the nucleic acid level and is encoded by a variegated gene library. A variegated library of NOVX variants can be produced by, for example, enzymatically ligating a mixture of synthetic oligonucleotides into gene sequences such that a degenerate set of potential NOVX sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g., for phage display) containing the set of NOVX sequences therein. There are a variety of methods which can be used to produce libraries of potential NOVX variants from a degenerate oligonucleotide sequence. Chemical synthesis of a degenerate gene sequence can be performed in an automatic DNA synthesizer, and the synthetic gene then ligated into an appropriate expression vector. Use of a degenerate set of genes allows for the provision, in one mixture, of all of the sequences encoding the desired set of potential NOVX sequences. Methods for synthesizing degenerate oligonucleotides are well-known within the art. See, e.g., Narang, 1983. Tetrahedron 39: 3; Itakura, et al, 1984. Ann . Rev. Biochem. 53: 323; Itakura, et al, 1984. Science 198: 1056; He, et al, 1983. Nucl. Acids Res. 11 : 477.
Polypeptide Libraries In addition, libraries of fragments of the NOVX protein coding sequences can be used to generate a variegated population of NOVX fragments for screening and subsequent selection of variants of an NOVX protein, hi one embodiment, a library of coding sequence fragments can be generated by treating a double stranded PCR fragment of an NOVX coding sequence with a nuclease under conditions wherein nicking occurs only about once per molecule, denaturing the double stranded DNA, renaturing the DNA to form double-stranded DNA that can include sense/antisense pairs from different nicked products, removing single stranded portions from reformed duplexes by treatment with Si nuclease, and ligating the resulting fragment library into an expression vector. By this method, expression libraries can be derived which encodes N-terminal and internal fragments of various sizes of the NOVX proteins. Various techniques are known in the art for screening gene products of combinatorial libraries made by point mutations or truncation, and for screening cDNA libraries for gene products having a selected property. Such techniques are adaptable for rapid screening of the gene libraries generated by the combinatorial mutagenesis of NOVX proteins. The most widely used techniques, which are amenable to high throughput analysis, for screening large gene libraries typically include cloning the gene library into replicable expression vectors, transforming appropriate cells with the resulting library of vectors, and expressing the combinatorial genes under conditions in which detection of a desired activity facilitates isolation of the vector encoding the gene whose product was detected. Recursive ensemble mutagenesis (REM), a new technique that enhances the frequency of functional mutants in the libraries, can be used in combination with the screening assays to identify NOVX variants. See, e.g., Arkin and Yourvan, 1992. Proc. Natl. Acad. Sci. USA 89: 7811-7815; Delgrave, et al, 1993. Protein Engineering 6:327-331.
Anti-NOVX Antibodies
Also included in the invention are antibodies to NOVX proteins, or fragments of NOVX proteins. The term "antibody" as used herein refers to immunoglobulin molecules and immunologically active portions of immunoglobulin (Ig) molecules, i.e., molecules that contain an antigen binding site that specifically binds (immunoreacts with) an antigen. Such antibodies include, but are not limited to, polyclonal, monoclonal, chimeric, single chain, Fab, Fab' and F(ay)2 fragments, and an Fa expression library. In general, an antibody molecule obtained from humans relates to any of the classes IgG, IgM, IgA, IgE and IgD, which differ from one another by the nature of the heavy chain present in the molecule. Certain classes have subclasses as well, such as IgGu IgG2, and others. Furthermore, in humans, the light chain may be a kappa chain or a lambda chain. Reference herein to antibodies includes a reference to all such classes, subclasses and types of human antibody species.
An isolated NOVX-related protein of the invention may be intended to serve as an antigen, or a portion or fragment thereof, and additionally can be used as an immunogen to generate antibodies that immunospecifically bind the antigen, using standard techniques for polyclonal and monoclonal antibody preparation. The full-length protein can be used or, alternatively, the invention provides antigenic peptide fragments of the antigen for use as immunogens. An antigenic peptide fragment comprises at least 6 amino acid residues of the amino acid sequence of the full length protein and encompasses an epitope thereof such that an antibody raised against the peptide forms a specific immune complex with the full length protein or with any fragment that contains the epitope. Preferably, the antigenic peptide comprises at least 10 amino acid residues, or at least 15 amino acid residues, or at least 20 amino acid residues, or at least 30 amino acid residues. Preferred epitopes encompassed by the antigenic peptide are regions of the protein that are located on its surface; commonly these are hydrophilic regions.
In certain embodiments of the invention, at least one epitope encompassed by the antigenic peptide is a region of NOVX-related protein that is located on the surface of the protein, e.g., a hydrophilic region. A hydrophobicity analysis of the human NOVX-related protein sequence will indicate which regions of a NOVX-related protein are particularly hydrophilic and, therefore, are likely to encode surface residues useful for targeting antibody production. As a means for targeting antibody production, hydropathy plots showing regions of hydrophilicity and hydrophobicity may be generated by any method well known in the art, including, for example, the Kyte Doolittle or the Hopp Woods methods, either with or without Fourier transformation. See, e.g., Hopp and Woods, 1981, Proc. Nat. Acad. Sci. USA 78:
3824-3828; Kyte and Doolittle 1982, J. Mol. Biol. 157: 105-142, each of which is incorporated herein by reference in its entirety. Antibodies that are specific for one or more domains within an antigenic protein, or derivatives, fragments, analogs or homologs thereof, are also provided herein. A protein of the invention, or a derivative, fragment, analog, homolog or ortholog thereof, may be utilized as an immunogen in the generation of antibodies that immunospecifically bind these protein components.
Various procedures known within the art may be used for the production of polyclonal or monoclonal antibodies directed against a protein of the invention, or against derivatives, fragments, analogs homologs or orthologs thereof (see, for example, Antibodies: A Laboratory Manual, Harlow and Lane, 1988, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, incorporated herein by reference). Some of these antibodies are discussed below.
Polyclonal Antibodies For the production of polyclonal antibodies, various suitable host animals (e.g., rabbit, goat, mouse or other mammal) may be immunized by one or more injections with the native protein, a synthetic variant thereof, or a derivative of the foregoing. An appropriate immunogenic preparation can contain, for example, the naturally occurring immunogenic protein, a chemically synthesized polypeptide representing the immunogenic protein, or a recombinantly expressed immunogenic protein. Furthermore, the protein may be conjugated to a second protein known to be immunogenic in the mammal being immunized. Examples of such immunogenic proteins include but are not limited to keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, and soybean trypsin inhibitor. The preparation can further include an adjuvant. Various adjuvants used to increase the immunological response include, but are not limited to, Freund's (complete and incomplete), mineral gels (e.g., aluminum hydroxide), surface active substances (e.g., lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, dinitrophenol, etc.), adjuvants usable in humans such as Bacille Calmette-Guerin and Corynebacterium parvum, or similar imm iostimulatory agents. Additional examples of adjuvants which can be employed include MPL-TDM adjuvant (monophosphoiyl Lipid A, synthetic trehalose dicorynomycolate).
The polyclonal antibody molecules directed against the immunogenic protein can be isolated from the mammal (e.g., from the blood) and further purified by well known techniques, such as affinity chromatography using protein A or protein G, which provide primarily the IgG fraction of immune serum. Subsequently, or alternatively, the specific antigen which is the target of the immunoglobulin sought, or an epitope thereof, maybe immobilized on a column to purify the immune specific antibody by immunoaffmity chromatography. Purification of immunoglobulins is discussed, for example, by D. Wilkinson (The Scientist, published by The Scientist, Inc., Philadelphia PA, Vol. 14, No. 8 (April 17, 2000), pp. 25-28).
Monoclonal Antibodies
The term "monoclonal antibody" (MAb) or "monoclonal antibody composition", as used herein, refers to a population of antibody molecules that contain only one molecular species of antibody molecule consisting of a unique light chain gene product and a unique heavy chain gene product, hi particular, the complementarity determining regions (CDRs) of the monoclonal antibody are identical in all the molecules of the population. MAbs thus contain an antigen binding site capable of immunoreacting with a particular epitope of the antigen characterized by a unique binding affinity for it. Monoclonal antibodies can be prepared using hybridoma methods, such as those described by Kohler and Milstein, Nature, 256:495 (1975). In a hybridoma method, a mouse, hamster, or other appropriate host animal, is typically immunized with an immunizing agent to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the immunizing agent. Alternatively, the lymphocytes can be immunized in vitro. The immunizing agent will typically include the protein antigen, a fragment thereof or a fusion protein thereof. Generally, either peripheral blood lymphocytes are used if cells of human origin are desired, or spleen cells or lymph node cells are used if non-human mammalian sources are desired. The lymphocytes are then fused with an immortalized cell line using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, MONOCLONAL ANTIBODIES: PRINCIPLES AND PRACTICE, Academic Press, (1986) pp. 59-103). Immortalized cell lines are usually transformed mammalian cells, particularly myeloma cells of rodent, bovine and human origin. Usually, rat or mouse myeloma cell lines are employed. The hybridoma cells can be cultured in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, immortalized cells. For example, if the parental cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT), the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine ("HAT medium"), which substances prevent the growth of HGPRT-deficient cells. Preferred immortalized cell lines are those that fuse efficiently, support stable high level expression of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium. More preferred immortalized cell lines are murine myeloma lines, which can be obtained, for instance, from the Salk Institute Cell Distribution Center, San Diego, California and the American Type Culture Collection, Manassas, Virginia. Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies (Kozbor, J Immunol, 133:3001 (1984); Brodeur et al. , MONOCLONAL ANTIBODY PRODUCTION TECHNIQUES AND APPLICATIONS, Marcel Dekker, Inc., New York, (1987) pp. 51-63).
The culture medium in which the hybridoma cells are cultured can then be assayed for the presence of monoclonal antibodies directed against the antigen. Preferably, the binding specificity of monoclonal antibodies produced by the hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA). Such techniques and assays are known in the art. The binding affinity of the monoclonal antibody can, for example, be determined by the Scatchard analysis of Munson and Pollard, Anal. Biochem., 107:220 (1980). Preferably, antibodies having a high degree of specificity and a high binding affinity for the target antigen are isolated.
After the desired hybridoma cells are identified, the clones can be subcloned by limiting dilution procedures and grown by standard methods. Suitable culture media for this purpose include, for example, Dulbecco's Modified Eagle's Medium and RPMI- 1640 medium. Alternatively, the hybridoma cells can be grown in vivo as ascites in a mammal. The monoclonal antibodies secreted by the subclones can be isolated or purified from the culture medium or ascites fluid by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
The monoclonal antibodies can also be made by recombinant DNA methods, such as those described in U.S. Patent No. 4,816,567. DNA encoding the monoclonal antibodies of the invention can be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies). The hybridoma cells of the invention serve as a preferred source of such DNA. Once isolated, the DNA can be placed into expression vectors, which are then transfected into host cells such as simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. The DNA also can be modified, for example, by substituting the coding sequence for human heavy and light chain constant domains in place of the homologous murine sequences (U.S. Patent No. 4,816,567; Morrison, Nature 368, 812-13 (1994)) or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide. Such a non-immunoglobulin polypeptide can be substituted for the constant domains of an antibody of the invention, or can be substituted for the variable domains of one antigen-combining site of an antibody of the invention to create a chimeric bivalent antibody.
Humanized Antibodies The antibodies directed against the protein antigens of the invention can further comprise humanized antibodies or human antibodies. These antibodies are suitable for administration to humans without engendering an immune response by the human against the administered immunoglobulin. Humanized forms of antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab')2 or other antigen- binding subsequences of antibodies) that are principally comprised of the sequence of a human immunoglobulin, and contain minimal sequence derived from a non-human immunoglobulin. Humanization can be performed following the method of Winter and co-workers (Jones et al, Nature, 321:522-525 (1986); Riechmann et al, Nature, 332:323-327 (1988); Verhoeyen et al, Science, 239:1534-1536 (1988)), by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. (See also U.S. Patent No. 5,225,539.) h some instances, Fv framework residues of the human immunoglobulin are replaced by corresponding non-human residues. Humanized antibodies can also comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the framework regions are those of a human immunoglobulin consensus sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin (Jones et al, 1986; Rieclimann et al, 1988; and Presta, Curr. Op. Struct. Biol, 2:593-596 (1992)).
Human Antibodies
Fully human antibodies relate to antibody molecules in which essentially the entire sequences of both the light chain and the heavy chain, including the CDRs, arise from human genes. Such antibodies are termed "human antibodies", or "fully human antibodies" herein. Human monoclonal antibodies can be prepared by the trioma technique; the human B-cell hybridoma technique (see Kozbor, et al, 1983 Immunol Today 4: 72) and the EBV hybridoma technique to produce human monoclonal antibodies (see Cole, et al, 1985 In: MONOCLONAL ANTIBODIES AND CANCER THERAPY, Alan R. Liss, Inc., pp. 77-96). Human monoclonal antibodies may be utilized in the practice of the present invention and may be produced by using human hybridomas (see Cote, et al, 1983. Proc Natl Acad Sci USA 80: 2026-2030) or by transforming human B-cells with Epstein Barr Virus in vitro (see Cole, et al, 1985 In: MONOCLONAL ANTIBODIES AND CANCER THERAPY, Alan R. Liss, Inc., pp. 77-96). In addition, human antibodies can also be produced using additional techniques, including phage display libraries (Hoogenboom and Winter, J. Mol. Biol, 227:381 (1991); Marks et al, J. Mol. Biol, 222:581 (1991)). Similarly, human antibodies can be made by introducing human immunoglobulin loci into transgenic animals, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. Upon challenge, human antibody production is observed, which closely resembles that seen in humans in all respects, including gene rearrangement, assembly, and antibody repertoire. This approach is described, for example, in U.S. Patent Nos. 5,545,807; 5,545,806; 5,569,825; 5,625,126; 5,633,425; 5,661,016, and in Marks et al. (Bio/Technology 10, 779-783 (1992)); Lonberg et al. (Nature 368 856-859 (1994)); Morrison ( Nature 368, 812-13 (1994)); Fishwild et al,( Nature Biotechnology 14, 845-51 (1996)); Neuberger (Nature Biotechnology 14, 826 (1996)); and Lonberg and Huszar (Intern. Rev. Immunol. 13 65-93 (1995)).
Human antibodies may additionally be produced using transgenic nonhuman animals which are modified so as to produce fully human antibodies rather than the animal's endogenous antibodies in response to challenge by an antigen. (See PCT publication
WO94/02602). The endogenous genes encoding the heavy and light immunoglobulin chains in the nonhuman host have been incapacitated, and active loci encoding human heavy and light chain immunoglobulins are inserted into the host's genome. The human genes are incorporated, for example, using yeast artificial chromosomes containing the requisite human DNA segments. An animal which provides all the desired modifications is then obtained as progeny by crossbreeding intermediate transgenic animals containing fewer than the full complement of the modifications. The preferred embodiment of such a nonhuman animal is a mouse, and is termed the Xenomouse™ as disclosed in PCT publications WO 96/33735 and WO 96/34096. This animal produces B cells which secrete fully human immunoglobulins. The antibodies can be obtained directly from the animal after immunization with an immunogen of interest, as, for example, a preparation of a polyclonal antibody, or alternatively from immortalized B cells derived from the animal, such as hybridomas producing monoclonal antibodies. Additionally, the genes encoding the immunoglobulins with human variable regions can be recovered and expressed to obtain the antibodies directly, or can be further modified to obtain analogs of antibodies such as, for example, single chain Fv molecules.
An example of a method of producing a nonhuman host, exemplified as a mouse, lacking expression of an endogenous immunoglobulin heavy chain is disclosed in U.S. Patent No. 5,939,598. It can be obtained by a method including deleting the J segment genes from at least one endogenous heavy chain locus in an embryonic stem cell to prevent rearrangement of the locus and to prevent formation of a transcript of a rearranged immunoglobulin heavy chain locus, the deletion being effected by a targeting vector containing a gene encoding a selectable marker; and producing from the embryonic stem cell a transgenic mouse whose somatic and germ cells contain the gene encoding the selectable marker. A method for producing an antibody of interest, such as a human antibody, is disclosed in U.S. Patent No. 5,916,771. It includes introducing an expression vector that contains a nucleotide sequence encoding a heavy chain into one mammalian host cell in culture, introducing an expression vector containing a nucleotide sequence encoding a light chain into another mammalian host cell, and fusing the two cells to form a hybrid cell. The hybrid cell expresses an antibody containing the heavy chain and the light chain.
In a further improvement on this procedure, a method for identifying a clinically relevant epitope on an immunogen, and a correlative method for selecting an antibody that binds immunospecifically to the relevant epitope with high affinity, are disclosed in PCT publication WO 99/53049.
Fab Fragments and Single Chain Antibodies
According to the invention, techniques can be adapted for the production of single-chain antibodies specific to an antigenic protein of the invention (see e.g., U.S. Patent No. 4,946,778). In addition, methods can be adapted for the construction of Fab expression libraries (see e.g., Huse, et al, 1989 Science 246: 1275-1281) to allow rapid and effective identification of monoclonal Fab fragments with the desired specificity for a protein or derivatives, fragments, analogs or homologs thereof. Antibody fragments that contain the idiotypes to a protein antigen may be produced by techniques known in the art including, but not limited to: (i) an F(ab')2 fragment produced by pepsin digestion of an antibody molecule; (ii) an Fab fragment generated by reducing the disulfide bridges of an F(a ')2 fragment; (iii) an Fab fragment generated by the treatment of the antibody molecule with papain and a reducing agent and (iv) Fv fragments.
Bispecific Antibodies
Bispecific antibodies are monoclonal, preferably human or humanized, antibodies that have binding specificities for at least two different antigens, hi the present case, one of the binding specificities is for an antigenic protein of the invention. The second binding target is any other antigen, and advantageously is a cell-surface protein or receptor or receptor subunit. Methods for making bispecific antibodies are known in the art. Traditionally, the recombinant production of bispecific antibodies is based on the co-expression of two immunoglobulin heavy-chain/light-chain pairs, where the two heavy chains have different specificities (Milstein and Cuello, Nature, 305:537-539 (1983)). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture often different antibody molecules, of which only one has the correct bispecific structure. The purification of the correct molecule is usually accomplished by affinity chromatography steps. Similar procedures are disclosed in WO 93/08829, published 13 May 1993, and in Traunecker et al, 1991 EMBOJ., 10:3655-3659. Antibody variable domains with the desired binding specificities (antibody-antigen combining sites) can be fused to immunoglobulin constant domain sequences. The fusion preferably is with an immunoglobulin heavy-chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is preferred to have the first heavy-chain constant region (CHI) containing the site necessary for light-chain binding present in at least one of the fusions. DNAs encoding the immunoglobulin heavy-chain fusions and, if desired, the immunoglobulin light chain, are inserted into separate expression vectors, and are co- transfected into a suitable host organism. For further details of generating bispecific antibodies see, for example, Suresh et al, Methods in Enzymology, 121:210 (1986). According to another approach described in WO 96/27011 , the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers which are recovered from recombinant cell culture. The preferred interface comprises at least a part of the CH3 region of an antibody constant domain, h this method, one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g. tyrosine or tryptophan). Compensatory "cavities" of identical or similar size to the large side chain(s) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine). This provides a mechanism for increasing the yield of the heterodimer over other unwanted end-products such as homodimers. Bispecific antibodies can be prepared as full length antibodies or antibody fragments
(e.g. F(ab')2 bispecific antibodies). Techniques for generating bispecific antibodies from antibody fragments have been described in the literature. For example, bispecific antibodies • can be prepared using chemical linkage. Brennan et al, Science 229:81 (1985) describe a procedure wherein intact antibodies are proteolytically cleaved to generate F(ab') fragments. These fragments are reduced in the presence of the dithiol complexing agent sodium arsenite to stabilize vicinal dithiols and prevent intermolecular disulfide formation. The Fab' fragments generated are then converted to thionitrobenzoate (TNB) derivatives. One of the Fab'-TNB derivatives is then reconverted to the Fab'-thiol by reduction with mercaptoethylamine and is mixed with an equimolar amount of the other Fab'-TNB derivative to form the bispecific antibody. The bispecific antibodies produced can be used as agents for the selective iinmobilization of enzymes.
Additionally, Fab' fragments can be directly recovered from E. coli and chemically coupled to form bispecific antibodies. Shalaby et al, J. Exp. Med. 175:217-225 (1992) describe the production of a fully humanized bispecific antibody F(ab') molecule. Each Fab' fragment was separately secreted from E. coli and subjected to directed chemical coupling in vitro to form the bispecific antibody. The bispecific antibody thus formed was able to bind to cells overexpressing the ErbB2 receptor and normal human T cells, as well as trigger the lytic activity of human cytotoxic lymphocytes against human breast tumor targets. Various techniques for making and isolating bispecific antibody fragments directly from recombinant cell culture have also been described. For example, bispecific antibodies have been produced using leucine zippers. Kostelny et al, J. Immunol. 148(5):1547-1553 (1992). The leucine zipper peptides from the Fos and Jun proteins were linked to the Fab' portions of two different antibodies by gene fusion. The antibody homodimers were reduced at the hinge region to form monomers and then re-oxidized to form the antibody heterodimers. This method can also be utilized for the production of antibody homodimers. The "diabody" technology described by Hollinger et al, Proc. Natl. Acad. Sci. USA 90:6444-6448 (1993) has provided an alternative mechanism for making bispecific antibody fragments. The fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (VL) by a linker which is too short to allow pairing between the two domains on the same chain. Accordingly, the VH and VL domains of one fragment are forced to pair with the complementary VL and VH domains of another fragment, thereby forming two antigen-binding sites. Another strategy for making bispecific antibody fragments by the use of single-chain Fv (sFv) dimers has also been reported. See, Gruber et al, J. Immunol. 152:5368 (1994). Antibodies with more than two valencies are contemplated. For example, trispecific antibodies can be prepared. Tutt et al, J. Immunol. 147:60 (1991).
Exemplary bispecific antibodies can bind to two different epitopes, at least one of which originates in the protein antigen of the invention. Alternatively, an anti-antigenic arm of an immunoglobulin molecule can be combined with an arm which binds to a triggering molecule on a leukocyte such as a T-cell receptor molecule (e.g. CD2, CD3, CD28, or B7), or Fc receptors for IgG (FcγR), such as FcγRI (CD64), FcγRII (CD32) and FcγRIII (CD 16) so as to focus cellular defense mechanisms to the cell expressing the particular antigen. Bispecific antibodies can also be used to direct cytotoxic agents to cells which express a particular antigen. These antibodies possess an antigen-binding arm and an arm which binds a cytotoxic agent or a radionuclide chelator, such as EOTTJBE, DPT A, DOT A, or TETA. Another bispecific antibody of interest binds the protein antigen described herein and further binds tissue factor (TF). Heteroconjugate Antibodies
Heteroconjugate antibodies are also within the scope of the present invention. Heteroconjugate antibodies are composed of two covalently joined antibodies. Such antibodies have, for example, been proposed to target immune system cells to unwanted cells (U.S. Patent No. 4,676,980), and for treatment of HIV infection (WO 91/00360; WO
92/200373; EP 03089). It is contemplated that the antibodies can be prepared in vitro using known methods in synthetic protein chemistry, including those involving crosslinking agents. For example, immunotoxins can be constructed using a disulfide exchange reaction or by forming a thioether bond. Examples of suitable reagents for this purpose include iminothiolate and methyl-4-mercaptobutyrimidate and those disclosed, for example, in U.S. Patent No. 4,676,980.
Effector Function Engineering
It can be desirable to modify the antibody of the invention with respect to effector function, so as to enhance, e.g., the effectiveness of the antibody in treating cancer. For example, cysteine residue(s) can be introduced into the Fc region, thereby allowing interchain disulfide bond formation in this region. The homodimeric antibody thus generated can have improved internalization capability and/or increased complement-mediated cell killing and antibody-dependent cellular cytotoxicity (ADCC). See Caron et al, J. Exp Med., 176: 1191- 1195 (1992) and Shopes, J. Immunol., 148: 2918-2922 (1992). Homodimeric antibodies with enhanced anti-tumor activity can also be prepared using heterobifunctional cross-linkers as described in Wolff et al. Cancer Research, 53: 2560-2565 (1993). Alternatively, an antibody can be engineered that has dual Fc regions and can thereby have enhanced complement lysis and ADCC capabilities. See Stevenson et al, Anti-Cancer Drug Design, 3: 219-230 (1989).
Immunoconjugates
The invention also pertains to immunoconjugates comprising an antibody conjugated to a cytotoxic agent such as a chemotherapeutic agent, toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).
Chemotherapeutic agents useful in the generation of such immunoconjugates have been described above. Enzymatically active toxins and fragments thereof that can be used include diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes. A variety of radionuclides are available for the production of radioconjugated antibodies. Examples include 212Bi, 1311, 131hι, 90Y, and 186Re.
Conjugates of the antibody and cytotoxic agent are made using a variety of bifunctional protein-coupling agents such as N-succinimidyl-3-(2-pyridyldithiol) propionate (SPDP), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HCL), active esters (such as disuccinimidyl suberate), aldehydes (such as glutareldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis- diazonium derivatives (such as bis-(p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as tolyene 2,6-diisocyanate), and bis-active fluorine compounds (such as 1,5-difmoro- 2,4-dinitrobenzene). For example, a ricin immunotoxin can be prepared as described in Vitetta et al, Science, 238: 1098 (1987). Carbon- 14-labeled l-isothiocyanatobenzyl-3- methyldiethylene triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody. See WO94/11026.
In another embodiment, the antibody can be conjugated to a "receptor" (such streptavidin) for utilization in tumor pretargeting wherein the antibody-receptor conjugate is administered to the patient, followed by removal of unbound conjugate from the circulation using a clearing agent and then administration of a "ligand" (e.g., avidin) that is in turn conjugated to a cytotoxic agent.
In one embodiment, methods for the screening of antibodies that possess the desired specificity include, but are not limited to, enzyme-linked immunosorbent assay (ELISA) and other immunologically-mediated techniques known within the art. In a specific embodiment, selection of antibodies that are specific to a particular domain of an NOVX protein is facilitated by generation of hybridomas that bind to the fragment of an NOVX protein possessing such a domain. Thus, antibodies that are specific for a desired domain within an NOVX protein, or derivatives, fragments, analogs or homologs thereof, are also provided herein. Anti-NOVX antibodies may be used in methods known within the art relating to the localization and/or quantitation of an NOVX protein (e.g., for use in measuring levels of the NOVX protein within appropriate physiological samples, for use in diagnostic methods, for use in imaging the protein, and the like). In a given embodiment, antibodies for NOVX proteins, or derivatives, fragments, analogs or homologs thereof, that contain the antibody derived binding domain, are utilized as pharmacologically-active compounds (hereinafter "Therapeutics").
An anti-NOVX antibody (e.g., monoclonal antibody) can be used to isolate an NOVX polypeptide by standard techniques, such as affinity chromatography or immunoprecipitation. An anti-NOVX antibody can facilitate the purification of natural NOVX polypeptide from cells and of recombinantly-produced NOVX polypeptide expressed in host cells. Moreover, an anti-NOVX antibody can be used to detect NOVX protein (e.g., in a cellular lysate or cell supernatant) in order to evaluate the abundance and pattern of expression of the NOVX protein. Anti-NOVX antibodies can be used diagnostically to monitor protein levels in tissue as part of a clinical testing procedure, e.g., to, for example, determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling (i.e., physically linking) the antibody to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, β-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive material include 125I, 131I, 35S or 3H.
NOVX Recombinant Expression Vectors and Host Cells
Another aspect of the invention pertains to vectors, preferably expression vectors, containing a nucleic acid encoding an NOVX protein, or derivatives, fragments, analogs or homologs thereof. As used herein, the term "vector" refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a "plasmid", which refers to a circular double stranded DNA loop into which additional DNA segments can be ligated. Another type of vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operatively-linked. Such vectors are referred to herein as "expression vectors", hi general, expression vectors of utility in recombinant DNA techniques are often in the form of plasmids. In the present specification, "plasmid" and "vector" can be used interchangeably as the plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions. The recombinant expression vectors of the invention comprise a nucleic acid of the mvention in a form suitable for expression of the nucleic acid in a host cell, which means that the recombinant expression vectors include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, that is operatively-linked to the nucleic acid sequence to be expressed. Within a recombinant expression vector, "operably-linked" is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner that allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell).
The term "regulatory sequence" is intended to includes promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990). Regulatory sequences include those that direct constitutive expression of a nucleotide sequence in many types of host cell and those that direct expression of the nucleotide sequence only in certain host cells (e.g., tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc. The expression vectors of the invention can be introduced into host cells to thereby produce proteins or peptides, including fusion proteins or peptides, encoded by nucleic acids as described herein (e.g., NOVX proteins, mutant forms of NOVX proteins, fusion proteins, etc.).
The recombinant expression vectors of the invention can be designed for expression of NOVX proteins in prokaryotic or eukaryotic cells. For example, NOVX proteins can be expressed in bacterial cells such as Escherichia coli, insect cells (using baculovirus expression vectors) yeast cells or mammalian cells. Suitable host cells are discussed further in Goeddel, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990). Alternatively, the recombinant expression vector can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase. Expression of proteins in prokaryotes is most often carried out in Escherichia coli with vectors containing constitutive or inducible promoters directing the expression of either fusion or non-fusion proteins. Fusion vectors add a number of amino acids to a protein encoded therein, usually to the amino terminus of the recombinant protein. Such fusion vectors typically serve three purposes: (i) to increase expression of recombinant protein; (ii) to increase the solubility of the recombinant protein; and (iii) to aid in the purification of the recombinant protein by acting as a ligand in affinity purification. Often, in fusion expression vectors, a proteolytic cleavage site is introduced at the junction of the fusion moiety and the recombinant protein to enable separation of the recombinant protein from the fusion moiety subsequent to purification of the fusion protein. Such enzymes, and their cognate recognition sequences, include Factor Xa, thrombin and enterokinase. Typical fusion expression vectors include pGEX (Pharmacia Biotech Inc; Smith and Johnson, 1988. Gene 67: 31-40), pMAL (New England Biolabs, Beverly, Mass.) and pRIT5 (Pharmacia, Piscataway, N.J.) that fuse glutathione S-transferase (GST), maltose E binding protein, or protein A, respectively, to the target recombinant protein. Examples of suitable inducible non-fusion E. coli expression vectors include pTrc (Amrann et al, (1988) Gene 69:301-315) and pET l id (Sn dier et al, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990) 60-89). One strategy to maximize recombinant protein expression in E. coli is to express the protein in a host bacteria with an impaired capacity to proteolytically cleave the recombinant protein. See, e.g., Gottesman, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990) 119-128. Another strategy is to alter the nucleic acid sequence of the nucleic acid to be inserted into an expression vector so that the individual codons for each amino acid are those preferentially utilized in E. coli (see, e.g., Wada, et al, 1992. Nucl. Acids Res. 20: 2111-2118). Such alteration of nucleic acid sequences of the invention can be carried out by standard DNA synthesis techniques. hi another embodiment, the NOVX expression vector is a yeast expression vector. Examples of vectors for expression in yeast Saccharomyces cerivisae include pYepSecl (Baldari, et al, 1987. EMBO J. 6: 229-234), pMFa (Kurjan and Herskowitz, 1982. Cell 30: 933-943), pJRY88 (Schultz et al, 1987. Gene 54: 113-123), pYES2 (Invitrogen Corporation, San Diego, Calif), and picZ (InVitrogen Corp, San Diego, Calif).
Alternatively, NOVX can be expressed in insect cells using baculovirus expression vectors. Baculovirus vectors available for expression of proteins in cultured insect cells (e.g., SF9 cells) include the pAc series (Smith, et al, 1983. Mol. Cell. Biol. 3: 2156-2165) and the pVL series (Lucklow and Summers, 1989. Virology 170: 31-39).
In yet another embodiment, a nucleic acid of the invention is expressed in mammalian cells using a mammalian expression vector. Examples of mammalian expression vectors include pCDM8 (Seed, 1987. Nature 329: 840) and ρMT2PC (Kaufman, et al, 1987. EMBO J. 6: 187-195). When used in mammalian cells, the expression vector's control functions are often provided by viral regulatory elements. For example, commonly used promoters are derived from polyoma, adenovirus 2, cytomegalovirus, and simian virus 40. For other suitable expression systems for both prokaryotic and eukaryotic cells see, e.g., Chapters 16 and 17 of Sambrook, et al, MOLECULAR CLONING: A LABORATORY MANUAL. 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989. In another embodiment, the recombinant mammalian expression vector is capable of directing expression of the nucleic acid preferentially in a particular cell type (e.g., tissue-specific regulatory elements are used to express the nucleic acid). Tissue-specific regulatory elements are known in the art. Non-limiting examples of suitable tissue-specific promoters include the albumin promoter (liver-specific; Pinkert, et al, 1987. Genes Dev. 1: 268-277), lymphoid-specific promoters (Calame and Eaton, 1988. Adv. Immunol. 43: 235-275), in particular promoters of T cell receptors (Winoto and Baltimore, 1989. EMBO J. 8: 729-733) and immunoglobulins (Banerji, et al, 1983. Cell 33: 729-740; Queen and Baltimore, 1983. Cell 33: 741-748), neuron-specific promoters (e.g., the neurofilament promoter; Byrne and Ruddle, 1989. Proc. Natl. Acad. Sci. USA 86: 5473-5477), pancreas-specific promoters (Edlund, et al, 1985. Science 230: 912-916), and mammary gland-specific promoters (e.g., milk whey promoter; U.S. Pat. No. 4,873,316 and European Application Publication No. 264,166). Developmentally-regulated promoters are also encompassed, e.g., the murine hox promoters (Kessel and Grass, 1990. Science 249: 374-379) and the D-fetoprotein promoter (Campes and Tilghman, 1989. Genes Dev. 3: 537-546).
The invention further provides a recombinant expression vector comprising a DNA molecule of the invention cloned into the expression vector in an antisense orientation. That is, the DNA molecule is operatively-linked to a regulatory sequence in a manner that allows for expression (by transcription of the DNA molecule) of an RNA molecule that is antisense to NOVX mRNA. Regulatory sequences operatively linked to a nucleic acid cloned in the antisense orientation can be chosen that direct the continuous expression of the antisense RNA molecule in a variety of cell types, for instance viral promoters and/or enhancers, or regulatory sequences can be chosen that direct constitutive, tissue specific or cell type specific expression of antisense RNA. The antisense expression vector can be in the form of a recombinant plasmid, phagemid or attenuated virus in which antisense nucleic acids are produced under the control of a high efficiency regulatory region, the activity of which can be determined by the cell type into which the vector is introduced. For a discussion of the regulation of gene expression using antisense genes see, e.g., Weintraub, et al, "Antisense RNA as a molecular tool for genetic analysis," Reviews-Trends in Genetics, Vol. 1(1) 1986.
Another aspect of the invention pertains to host cells into which a recombinant expression vector of the invention has been introduced. The terms "host cell" and "recombinant host cell" are used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but also to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
A host cell can be any prokaryotic or eukaryotic cell. For example, NOVX protein can be expressed in bacterial cells such as E. coli, insect cells, yeast or mammalian cells (such as Chinese hamster ovary cells (CHO) or COS cells). Other suitable host cells are known to those skilled in the art.
Vector DNA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques. As used herein, the terms "transformation" and "transfection" are intended to refer to a variety of art-recognized techniques for introducing foreign nucleic acid (e.g., DNA) into a host cell, including calcium phosphate or calcium chloride co-precipitation, DEAE-dextran-mediated transfection, lipofection, or electroporation. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. (MOLECULAR CLONING: A LABORATORY MANUAL. 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989), and other laboratory manuals.
For stable transfection of mammalian cells, it is known that, depending upon the expression vector and transfection technique used, only a small fraction of cells may integrate the foreign DNA into their genome. In order to identify and select these integrants, a gene that encodes a selectable marker (e.g., resistance to antibiotics) is generally introduced into the host cells along with the gene of interest. Various selectable markers include those that confer resistance to drugs, such as G418, hygromycin and methotrexate. Nucleic acid encoding a selectable marker can be introduced into a host cell on the same vector as that encoding NOVX or can be introduced on a separate vector. Cells stably transfected with the introduced nucleic acid can be identified by drag selection (e.g., cells that have incoφorated the selectable marker gene will survive, while the other cells die).
A host cell of the invention, such as a prokaryotic or eukaryotic host cell in culture, can be used to produce (i.e., express) NOVX protein. Accordingly, the invention further provides methods for producing NOVX protein using the host cells of the invention, hi one embodiment, the method comprises culturing the host cell of invention (into which a recombinant expression vector encoding NOVX protein has been introduced) in a suitable medium such that NOVX protein is produced. In another embodiment, the method further comprises isolating NOVX protein from the medium or the host cell.
Transgenic NO X Animals
The host cells of the invention can also be used to produce non-human transgenic animals. For example, in one embodiment, a host cell of the invention is a fertilized oocyte or an embryonic stem cell into which NOVX protein-coding sequences have been introduced. Such host cells can then be used to create non-human transgenic animals in which exogenous NOVX sequences have been introduced into their genome or homologous recombinant animals in which endogenous NOVX sequences have been altered. Such animals are useful for studying the function and/or activity of NOVX protein and for identifying and/or evaluating modulators of NOVX protein activity. As used herein, a "transgenic animal" is a non-human animal, preferably a mammal, more preferably a rodent such as a rat or mouse, in which one or more of the cells of the animal includes a transgene. Other examples of transgenic animals include non-human primates, sheep, dogs, cows, goats, chickens, amphibians, etc. A transgene is exogenous DNA that is integrated into the genome of a cell from which a transgenic animal develops and that remains in the genome of the mature animal, thereby directing the expression of an encoded gene product in one or more cell types or tissues of the transgenic animal. As used herein, a "homologous recombinant animal" is a non-human animal, preferably a mammal, more preferably a mouse, in which an endogenous NOVX gene has been altered by homologous recombination between the endogenous gene and an exogenous DNA molecule introduced into a cell of the animal, e.g., an embryonic cell of the animal, prior to development of the animal. A transgenic animal of the invention can be created by introducing NOVX-encoding nucleic acid into the male pronuclei of a fertilized oocyte (e.g., by microinjection, retroviral infection) and allowing the oocyte to develop in a pseudopregnant female foster animal. The human NOVX cDNA sequences SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33 can be introduced as a transgene into the genome of a non-human animal. Alternatively, a non-human homologue of the human NOVX gene, such as a mouse NOVX gene, can be isolated based on hybridization to the human NOVX cDNA (described further supra) and used as a transgene. Intronic sequences and polyadenylation signals can also be included in the transgene to increase the efficiency of expression of the transgene. A tissue-specific regulatory sequence(s) can be operably-linked to the NOVX transgene to direct expression of NOVX protein to particular cells. Methods for generating transgenic animals via embryo manipulation and microinjection, particularly animals such as mice, have become conventional in the art and are described, for example, in U.S. Patent Nos. 4,736,866; 4,870,009; and 4,873,191; and Hogan, 1986. In: MANIPULATING THE MOUSE EMBRYO, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N. Y. Similar methods are used for production of other transgenic animals. A transgenic founder animal can be identified based upon the presence of the NOVX transgene in its genome and/or expression of NOVX mRNA in tissues or cells of the animals. A transgenic founder animal can then be used to breed additional animals carrying the transgene. Moreover, transgenic animals carrying a transgene- encoding NOVX protein can further be bred to other transgenic animals carrying other transgenes.
To create a homologous recombinant animal, a vector is prepared which contains at least a portion of an NOVX gene into which a deletion, addition or substitution has been introduced to thereby alter, e.g., functionally disrupt, the NOVX gene. The NOVX gene can be a human gene (e.g., the cDNA of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33), but more preferably, is a non-human homologue of a human NOVX gene. For example, a mouse homologue of human NOVX gene of SEQ ID NOS: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33 can be used to construct a homologous recombination vector suitable for altering an endogenous NOVX gene in the mouse genome. In one embodiment, the vector is designed such that, upon homologous recombination, the endogenous NOVX gene is functionally disrupted (i.e., no longer encodes a functional protein; also referred to as a "knock out" vector).
Alternatively, the vector can be designed such that, upon homologous recombination, the endogenous NOVX gene is mutated or otherwise altered but still encodes functional protein (e.g., the upstream regulatory region can be altered to thereby alter the expression of the endogenous NOVX protein). In the homologous recombination vector, the altered portion of the NOVX gene is flanked at its 5'- and 3'-termini by additional nucleic acid of the NOVX gene to allow for homologous recombination to occur between the exogenous NOVX gene carried by the vector and an endogenous NOVX gene in an embryonic stem cell. The additional flanking NOVX nucleic acid is of sufficient length for successful homologous recombination with the endogenous gene. Typically, several kilobases of flanking DNA (both at the 5'- and 3'-termini) are included in the vector. See, e.g., Thomas, et al, 1987. Cell 51: 503 for a description of homologous recombination vectors. The vector is ten introduced into an embryonic stem cell line (e.g., by electroporation) and cells in which the introduced NOVX gene has homologously-recombined with the endogenous NOVX gene are selected. See, e.g., Li, et al, 1992. Cell 69: 915.
The selected cells are then injected into a blastocyst of an animal (e.g., a mouse) to form aggregation chimeras. See, e.g., Bradley, 1987. In: TERATOCARCINOMAS AND
EMBRYONIC STEM CELLS: A PRACTICAL APPROACH, Robertson, ed. IRL, Oxford, pp. 113-152. A chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal and the embryo brought to term. Progeny harboring the homologously-recombined DNA in their germ cells can be used to breed animals in which all cells of the animal contain the homologously-recombined DNA by germline transmission of the transgene. Methods for constructing homologous recombination vectors and homologous recombinant animals are described further in Bradley, 1991. Curr. Opin. Biotechnol 2: 823-829; PCT International Publication Nos.: WO 90/11354; WO 91/01140; WO 92/0968; and WO 93/04169. i another embodiment, transgenic non-humans animals can be produced that contain selected systems that allow for regulated expression of the transgene. One example of such a system is the cre/loxP recombinase system of bacteriophage PI. For a description of the cre/loxP recombinase system, See, e.g., Lakso, et al, 1992. Proc. Natl. Acad. Sci. USA 89: 6232-6236. Another example of a recombinase system is the FLP recombinase system of Saccharomyces cerevisiae. See, O'Gorman, et al, 1991. Science 251:1351-1355. If a cre/loxP recombinase system is used to regulate expression of the transgene, animals containing transgenes encoding both the Cre recombinase and a selected protein are required. Such animals can be provided through the construction of "double" transgenic animals, e.g., by mating two transgenic animals, one containing a transgene encoding a selected protein and the other containing a transgene encoding a recombinase. Clones of the non-human transgenic animals described herein can also be produced according to the methods described in Wilmut, et al, 1997. Nature 385: 810-813. In brief, a cell (e.g., a somatic cell) from the transgenic animal can be isolated and induced to exit the growth cycle and enter G0 phase. The quiescent cell can then be fused, e.g., through the use of electrical pulses, to an enucleated oocyte from an animal of the same species from which the quiescent cell is isolated. The reconstructed oocyte is then cultured such that it develops to morula or blastocyte and then transferred to pseudopregnant female foster animal. The offspring borne of this female foster animal will be a clone of the animal from which the cell (e.g., the somatic cell) is isolated.
Pharmaceutical Compositions
The NOVX nucleic acid molecules, NOVX proteins, and anti-NOVX antibodies (also referred to herein as "active compounds") of the invention, and derivatives, fragments, analogs and homologs thereof, can be incorporated into pharmaceutical compositions suitable for administration. Such compositions typically comprise the nucleic acid molecule, protein, or antibody and a pharmaceutically acceptable carrier. As used herein, "pharmaceutically acceptable carrier" is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. Suitable carriers are described in the most recent edition of Remington's Pharmaceutical Sciences, a standard reference text in the field, which is incoφorated herein by reference. Preferred examples of such carriers or diluents include, but are not limited to, water, saline, finger's solutions, dextrose solution, and 5% human serum albumin. Liposomes and non-aqueous vehicles such as fixed oils may also be used. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incoφorated into the compositions.
A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, infradermal, subcutaneous, oral (e.g., inhalation), transdermal (i.e., topical), transmucosal, and rectal administration. Solutions or suspensions used for parenteral, infradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetefraacetic acid (EDTA); buffers such as acetates, citrates or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose. The pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, tbimerosal, and the like, hi many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as manitol, sorbitol, sodium chloride in the composition. Prolonged absoφtion of the injectable compositions can be brought about by including in the composition an agent which delays absoφtion, for example, aluminum monostearate and gelatin.
Sterile injectable solutions can be prepared by incoφorating the active compound (e.g., an NOVX protein or anti-NOVX antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incoφorating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the puφose of oral therapeutic administration, the active compound can be incoφorated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring. For administration by inhalation, the compounds are delivered in the form of an aerosol spray from pressured container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art. The compounds can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.
In one embodiment, the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Coφoration and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Patent No. 4,522,811. It is especially advantageous to formulate oral or parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals. The nucleic acid molecules of the invention can be inserted into vectors and used as gene therapy vectors. Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (.see, e.g., U.S. Patent No. 5,328,470) or by stereotactic injection (see, e.g., Chen, et al, 1994. Proc. Natl. Acad. Sci. USA 91: 3054-3057). The pharmaceutical preparation of the gene therapy vector can include the gene therapy vector in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded. Alternatively, where the complete gene delivery vector can be produced intact from recombinant cells, e.g., retroviral vectors, the pharmaceutical preparation can include one or more cells that produce the gene delivery system.
The pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration.
Screening and Detection Methods
The isolated nucleic acid molecules of the invention can be used to express NOVX protein (e.g., via a recombinant expression vector in a host cell in gene therapy applications), to detect NOVX mRNA (e.g., in a biological sample) or a genetic lesion in an NOVX gene, and to modulate NOVX activity, as described further, below. In addition, the NOVX proteins can be used to screen drugs or compounds that modulate the NOVX protein activity or expression as well as to treat disorders characterized by insufficient or excessive production of NOVX protein or production of NOVX protein forms that have decreased or aberrant activity compared to NOVX wild-type protein (e.g.; diabetes (regulates insulin release); obesity (binds and fransport lipids); metabolic disturbances associated with obesity, the metabolic syndrome X as well as anorexia and wasting disorders associated with chronic diseases and various cancers, and infectious diseaseψossesses anti-microbial activity) and the various dyslipidemias. fri addition, the anti-NOVX antibodies of the invention can be used to detect and isolate NOVX proteins and modulate NOVX activity. In yet a further aspect, the invention can be used in methods to influence appetite, absoφtion of nutrients and the disposition of metabolic substrates in both a positive and negative fashion.
The invention further pertains to novel agents identified by the screening assays described herein and uses thereof for treatments as described, supra.
Screening Assays
The invention provides a method (also referred to herein as a "screening assay") for identifying modulators, i.e., candidate or test compounds or agents (e.g., peptides, peptidomimetics, small molecules or other drags) that bind to NOVX proteins or have a stimulatory or inhibitory effect on, e.g., NOVX protein expression or NOVX protein activity.
The invention also includes compounds identified in the screening assays described herein.
In one embodiment, the invention provides assays for screening candidate or test compounds which bind to or modulate the activity of the membrane-bound form of an NOVX protein or polypeptide or biologically-active portion thereof. The test compounds of the invention can be obtained using any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the "one-bead one-compound" library method; and synthetic library methods using affinity chromatography selection. The biological library approach is limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds. See, e.g., Lam, 1997 '. Anticancer Drug Design 12: 145.
A "small molecule" as used herein, is meant to refer to a composition that has a molecular weight of less than about 5 kD and most preferably less than about 4 kD. Small molecules can be, e.g., nucleic acids, peptides, polypeptides, peptidomimetics, carbohydrates, lipids or other organic or inorganic molecules. Libraries of chemical and/or biological mixtures, such as fungal, bacterial, or algal extracts, are known in the art and can be screened with any of the assays of the invention.
Examples of methods for the synthesis of molecular libraries can be found in the art, for example in: DeWitt, et al, 1993. Proc. Natl. Acad. Sci. U.S.A. 90: 6909; Erb, et al, 1994.
Proc. Natl. Acad. Sci. U.S.A. 91: 11422; Zuckermann, et al, 1994. J. Med. Chem. 37: 2678;
Cho, et al, 1993. Science 261: 1303; Carrell, et al, 1994. Angew. Chem. Int. Ed. Engl. 33:
2059; Carell, et al, 1994. Angew. Chem. Int. Ed. Engl. 33: 2061; and Gallop, et al, 1994. J.
Med. Chem. 37: 1233. Libraries of compounds may be presented in solution (e.g., Houghten, 1992. Biotechniques 13: 412-421), or on beads (Lam, 1991. Nature 354: 82-84), on chips (Fodor, 1993. Nature 364: 555-556), bacteria (Ladner, U.S. Patent No. 5,223,409), spores (Ladner, U.S. Patent 5,233,409), plasmids (Cull, et al, 1992. Proc. Natl. Acad. Sci. USA 89: 1865-1869) or on phage (Scott and Smith, 1990. Science 249: 386-390; Devlin, 1990. Science 249: 404-406; Cwirla, et al, 1990. Proc. Natl. Acad. Sci. U.S.A. 87: 6378-6382; Felici, 1991. J. Mol. Biol. 222: 301-310; Ladner, U.S. Patent No. 5,233,409.).
In one embodiment, an assay is a cell-based assay in which a cell which expresses a membrane-bound form of NOVX protein, or a biologically-active portion thereof, on the cell surface is contacted with a test compound and the ability of the test compound to bind to an NOVX protein determined. The cell, for example, can of mammalian origin or a yeast cell. Determining the ability of the test compound to bind to the NOVX protein can be accomplished, for example, by coupling the test compound with a radioisotope or enzymatic label such that binding of the test compound to the NOVX protein or biologically-active portion thereof can be determined by detecting the labeled compound in a complex. For example, test compounds can be labeled with 1251, 35S, 14C, or 3H, either directly or indirectly, and the radioisotope detected by direct counting of radioemission or by scintillation counting. Alternatively, test compounds can be enzymatically-labeled with, for example, horseradish peroxidase, alkaline phosphatase, or luciferase, and the enzymatic label detected by determination of conversion of an appropriate substrate to product. In one embodiment, the assay comprises contacting a cell which expresses a membrane-bound form of NOVX protein, or a biologically-active portion thereof, on the cell surface with a known compound which binds NOVX to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with an NOVX protein, wherein determining the ability of the test compound to interact with an NOVX protein comprises determining the ability of the test compound to preferentially bind to NOVX protein or a biologically-active portion thereof as compared to the known compound. hi another embodiment, an assay is a cell-based assay comprising contacting a cell expressing a membrane-bound form of NOVX protein, or a biologically-active portion thereof, on the cell surface with a test compound and determining the ability of the test compound to modulate (e.g., stimulate or inhibit) the activity of the NOVX protein or biologically-active portion thereof. Determining the ability of the test compound to modulate the activity of NOVX or a biologically-active portion thereof can be accomplished, for example, by determining the ability of the NOVX protein to bind to or interact with an NOVX target molecule. As used herein, a "target molecule" is a molecule with which an NOVX protein binds or interacts in nature, for example, a molecule on the surface of a cell which expresses an NOVX interacting protein, a molecule on the surface of a second cell, a molecule in the extracellular milieu, a molecule associated with the internal surface of a cell membrane or a cytoplasmic molecule. An NOVX target molecule can be a non-NOVX molecule or an NOVX protein or polypeptide of the invention. In one embodiment, an NOVX target molecule is a component of a signal transduction pathway that facilitates transduction of an extracellular signal (e.g. a signal generated by binding of a compound to a membrane-bound NOVX molecule) through the cell membrane and into the cell. The target, for example, can be a second intercellular protein that has catalytic activity or a protein that facilitates the association of downstream signaling molecules with NOVX.
Determimng the ability of the NOVX protein to bind to or interact with an NOVX target molecule can be accomplished by one of the methods described above for determining direct binding. In one embodiment, determining the ability of the NOVX protein to bind to or interact with an NOVX target molecule can be accomplished by determining the activity of the target molecule. For example, the activity of the target molecule can be determined by detecting induction of a cellular second messenger of the target (i.e. intracellular Ca2+, diacylglycerol, TP3, etc.), detecting catalytic/enzymatic activity of the target an appropriate substrate, detecting the induction of a reporter gene (comprising an NOVX-responsive regulatory element operatively linked to a nucleic acid encoding a detectable marker, e.g., luciferase), or detecting a cellular response, for example, cell survival, cellular differentiation, or cell proliferation.
In yet another embodiment, an assay of the invention is a cell-free assay comprising contacting an NOVX protein or biologically-active portion thereof with a test compound and determining the ability of the test compound to bind to the NOVX protein or biologically- active portion thereof. Binding of the test compound to the NOVX protein can be determined either directly or indirectly as described above. In one such embodiment, the assay comprises contacting the NOVX protein or biologically-active portion thereof with a known compound which binds NOVX to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with an NOVX protein, wherein determining the ability of the test compound to interact with an NOVX protein comprises determining the ability of the test compound to preferentially bind to NOVX or biologically-active portion thereof as compared to the known compound. In still another embodiment, an assay is a cell-free assay comprising contacting NOVX protein or biologically-active portion thereof with a test compound and determining the ability of the test compound to modulate (e.g. stimulate or inhibit) the activity of the NOVX protein or biologically-active portion thereof. Determimng the ability of the test compound to modulate the activity of NOVX can be accomplished, for example, by determining the ability of the NOVX protein to bind to an NOVX target molecule by one of the methods described above for determining direct binding, hi an alternative embodiment, determining the ability of the test compound to modulate the activity of NOVX protein can be accomplished by determining the ability of the NOVX protein further modulate an NOVX target molecule. For example, the catalytic/enzymatic activity of the target molecule on an appropriate subsfrate can be determined as described, supra.
In yet another embodiment, the cell-free assay comprises contacting the NOVX protein or biologically-active portion thereof with a known compound which binds NOVX protein to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with an NOVX protein, wherein determining the ability of the test compound to interact with an NOVX protein comprises determining the ability of the NOVX protein to preferentially bind to or modulate the activity of an NOVX target molecule.
The cell-free assays of the invention are amenable to use of both the soluble form or the membrane-bound form of NOVX protein. In the case of cell-free assays comprising the membrane-bound form of NOVX protein, it may be desirable to utilize a solubilizing agent such that the membrane-bound form of NOVX protein is maintained in solution. Examples of such solubilizing agents include non-ionic detergents such as n-octylglucoside, n-dodecylglucoside, n-dodecyhnaltoside, octanoyl-N-methylglucamide, decanoyl-N-methylglucamide, Triton® X-100, Triton® X-114, Thesit®,
Isotridecypoly(ethylene glycol ether)n, N-dodecyl— N,N-dimethyl-3-ammonio-l -propane sulfonate, 3-(3-cholamidopropyι) dimethylamminiol-1 -propane sulfonate (CHAPS), or
3-(3-cholamidopropyl)dimethylamminiol-2-hydroxy-l-propane sulfonate (CHAPSO).
In more than one embodiment of the above assay methods of the invention, it may be desirable to immobilize either NOVX protein or its target molecule to facilitate separation of complexed from uncomplexed forms of one or both of the proteins, as well as to accommodate automation of the assay. Binding of a test compound to NOVX protein, or interaction of NOVX protein with a target molecule in the presence and absence of a candidate compound, can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtiter plates, test tubes, and micro-centrifuge tubes. In one embodiment, a fusion protein can be provided that adds a domain that allows one or both of the proteins to be bound to a matrix. For example, GST-NONX fusion proteins or GST-target fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, MO) or glutathione derivatized microtiter plates, that are then combined with the. test compound or the test compound and either the non-adsorbed target protein or ΝOVX protein, and the mixture is incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH). Following incubation, the beads or microtiter plate wells are washed to remove any unbound components, the matrix immobilized in the case of beads, complex determined either directly or indirectly, for example, as described, supra. Alternatively, the complexes can be dissociated from the matrix, and the level of ΝOVX protein binding or activity determined using standard techniques.
Other techniques for immobilizing proteins on matrices can also be used in the screening assays of the invention. For example, either the ΝOVX protein or its target molecule can be immobilized utilizing conjugation of biotin and streptavidin. Biotinylated ΝOVX protein or target molecules can be prepared from biotin-ΝHS (Ν-hydroxy-succinimide) using techniques well-known within the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, 111.), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical). Alternatively, antibodies reactive with ΝOVX protein or target molecules, but which do not interfere with binding of the ΝOVX protein to its target molecule, can be derivatized to the wells of the plate, and unbound target or ΝOVX protein trapped in the wells by antibody conjugation. Methods for detecting such complexes, in addition to those described above for the GST-immobilized complexes, include immunodetection of complexes using antibodies reactive with the ΝOVX protein or target molecule, as well as enzyme-linked assays that rely on detecting an enzymatic activity associated with the ΝONX protein or target molecule.
In another embodiment, modulators of ΝONX protein expression are identified in a method wherein a cell is contacted with a candidate compound and the expression of ΝONX rnRΝA or protein in the cell is determined. The level of expression of ΝONX mRΝA or protein in the presence of the candidate compound is compared to the level of expression of ΝONX mRΝA or protein in the absence of the candidate compound. The candidate compound can then be identified as a modulator of ΝONX mRΝA or protein expression based upon this comparison. For example, when expression of ΝONX mRΝA or protein is greater (i.e., statistically significantly greater) in the presence of the candidate compound than in its absence, the candidate compound is identified as a stimulator of NONX mRΝA or protein expression. Alternatively, when expression of ΝONX mRΝA or protein is less (statistically significantly less) in the presence of the candidate compound than in its absence, the candidate compound is identified as an inhibitor of ΝONX mRΝA or protein expression. The level of ΝONX mRΝA or protein expression in the cells can be determined by methods described herein for detecting ΝONX mRΝA or protein.
In yet another aspect of the invention, the ΝONX proteins can be used as "bait proteins" in a two-hybrid assay or three hybrid assay (.see, e.g., U.S. Patent No. 5,283,317; Zervos, et al, 1993. Cell 72: 223-232; Madura, et al, 1993. J. Biol. Chem. 268: 12046-12054; Bartel, et al, 1993. Biotechniques 14: 920-924; Iwabuchi, et al, 1993. Oncogene 8:
1693-1696; and Brent WO 94/10300), to identify other proteins that bind to or interact with NOVX ("NOVX-binding proteins" or "NOVX-bp") and modulate NONX activity. Such ΝONX-binding proteins are also likely to be involved in the propagation of signals by the ΝOVX proteins as, for example, upstream or downstream elements of the ΝOVX pathway. The two-hybrid system is based on the modular nature of most transcription factors, which consist of separable DΝA-binding and activation domains. Briefly, the assay utilizes two different DΝA constructs. In one construct, the gene that codes for ΝOVX is fused to a gene encoding the DΝA binding domain of a known transcription factor (e.g. , GAL-4). In the other construct, a DΝA sequence, from a library of DΝA sequences, that encodes an unidentified protein ("prey" or "sample") is fused to a gene that codes for the activation domain of the known transcription factor. If the "bait" and the "prey" proteins are able to interact, in vivo, forming an ΝOVX-dependent complex, the DΝA-binding and activation domains of the transcription factor are brought into close proximity. This proximity allows transcription of a reporter gene (e.g., LacZ) that is operably linked to a transcriptional regulatory site responsive to the transcription factor. Expression of the reporter gene can be detected and cell colonies containing the functional transcription factor can be isolated and used to obtain the cloned gene that encodes the protein which interacts with ΝOVX.
The invention further pertains to novel agents identified by the aforementioned screening assays and uses thereof for treatments as described herein.
Detection Assays
Portions or fragments of the cDΝA sequences identified herein (and the corresponding complete gene sequences) can be used in numerous ways as polynucleotide reagents. By way of example, and not of limitation, these sequences can be used to: (i) map their respective genes on a chromosome; and, thus, locate gene regions associated with genetic disease; (ii) identify an individual from a minute biological sample (tissue typing); and (iii) aid in forensic identification of a biological sample. Some of these applications are described in the subsections, below.
Chromosome Mapping
Once the sequence (or a portion of the sequence) of a gene has been isolated, this sequence can be used to map the location of the gene on a chromosome. This process is called chromosome mapping. Accordingly, portions or fragments of the NOVX sequences, SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, or fragments or derivatives thereof, can be used to map the location of the NOVX genes, respectively, on a chromosome. The mapping of the NOVX sequences to chromosomes is an important first step in correlating these sequences with genes associated with disease.
Briefly, NOVX genes can be mapped to chromosomes by preparing PCR primers (preferably 15-25 bp in length) from the NOVX sequences. Computer analysis of the NOVX, sequences can be used to rapidly select primers that do not span more than one exon in the genomic DNA, thus complicating the amplification process. These primers can then be used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene coπesponding to the NOVX sequences will yield an amplified fragment.
Somatic cell hybrids are prepared by fusing somatic cells from different mammals (e.g., human and mouse cells). As hybrids of human and mouse cells grow and divide, they gradually lose human chromosomes in random order, but retain the mouse chromosomes. By using media in which mouse cells cannot grow, because they lack a particular enzyme, but in which human cells can, the one human chromosome that contains the gene encoding the needed enzyme will be retained. By using various media, panels of hybrid cell lines can be established. Each cell line in a panel contains either a single human chromosome or a small number of human chromosomes, and a full set of mouse chromosomes, allowing easy mapping of individual genes to specific human chromosomes. See, e.g., D'Eustachio, et al, 1983. Science 220: 919-924. Somatic cell hybrids containing only fragments of human chromosomes can also be produced by using human chromosomes with translocations and deletions.
PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular sequence to a particular chromosome. Three or more sequences can be assigned per day using a single thermal cycler. Using the NOVX sequences to design oligonucleotide primers, sub- localization can be achieved with panels of fragments from specific chromosomes.
Fluorescence in situ hybridization (FISH) of a DNA sequence to a metaphase chromosomal spread can further be used to provide a precise chromosomal location in one step. Chromosome spreads can be made using cells whose division has been blocked in metaphase by a chemical like colcemid that disrupts the mitotic spindle. The chromosomes can be treated briefly with trypsin, and then stained with Giemsa. A pattern of light and dark bands develops on each chromosome, so that the chromosomes can be identified individually. The FISH technique can be used with a DNA sequence as short as 500 or 600 bases. However, clones larger than 1,000 bases have a higher likelihood of binding to a unique chromosomal location with sufficient signal intensity for simple detection. Preferably 1,000 bases, and more preferably 2,000 bases, will suffice to get good results at a reasonable amount of time. For a review of this technique, .see, Verma, et al, HUMAN CHROMOSOMES: A MANUAL OF BASIC TECHNIQUES (Pergamon Press, New York 1988). Reagents for chromosome mapping can be used individually to mark a single chromosome or a single site on that chromosome, or panels of reagents can be used for marking multiple sites and/or multiple chromosomes. Reagents coπesponding to noncoding regions of the genes actually are preferred for mapping puφoses. Coding sequences are more likely to be conserved within gene families, thus increasing the chance of cross hybridizations during chromosomal mapping.
Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be correlated with genetic map data. Such data are found, e.g., in McKusick, MENDELIAN INHERITANCE IN MAN, available on-line through Johns Hopkins University Welch Medical Library). The relationship between genes and disease, mapped to the same chromosomal region, can then be identified through linkage analysis (co-inheritance of physically adjacent genes), described in, e.g., Egeland, et al, 1987. Nature, 325: 783-787.
Moreover, differences in the DNA sequences between individuals affected and unaffected with a disease associated with the NOVX gene, can be determined. If a mutation is observed in some or all of the affected individuals but not in any unaffected individuals, then the mutation is likely to be the causative agent of the particular disease. Comparison of affected and unaffected individuals generally involves first looking for structural alterations in the chromosomes, such as deletions or translocations that are visible from chromosome spreads or detectable using PCR based on that DNA sequence. Ultimately, complete sequencing of genes from several individuals can be performed to confirm the presence of a mutation and to distinguish mutations from polymoφhisms.
Tissue Typing
The NOVX sequences of the invention can also be used to identify individuals from minute biological samples. In this technique, an individual's genomic DNA is digested with one or more restriction enzymes, and probed on a Southern blot to yield unique bands for identification. The sequences of the invention are useful as additional DNA markers for RFLP ("restriction fragment length polymoφhisms," described in U.S. Patent No. 5,272,057). Furthermore, the sequences of the invention can be used to provide an alternative technique that determines the actual base-by-base DNA sequence of selected portions of an individual's genome. Thus, the NOVX sequences described herein can be used to prepare two PCR primers from the 5'- and 3'-termini of the sequences. These primers can then be used to amplify an individual's DNA and subsequently sequence it.
Panels of coπesponding DNA sequences from individuals, prepared in this manner, can provide unique individual identifications, as each individual will have a unique set of such DNA sequences due to allelic differences. The sequences of the invention can be used to obtain such identification sequences from individuals "and from tissue. The NOVX sequences of the invention uniquely represent portions of the human genome. Allelic variation occurs to some degree in the coding regions of these sequences, and to a greater degree in the noncoding regions. It is estimated that allelic variation between individual humans occurs with a frequency of about once per each 500 bases. Much of the allelic variation is due to single nucleotide polymoφhisms (SNPs), which include restriction fragment length polymoφhisms (RFLPs).
Each of the sequences described herein can, to some degree, be used as a standard against which DNA from an individual can be compared for identification pmposes. Because greater numbers of polymoφbisms occur in the noncoding regions, fewer sequences are necessary to differentiate individuals. The noncoding sequences can comfortably provide positive individual identification with a panel of perhaps 10 to 1 ,000 primers that each yield a noncoding amplified sequence of 100 bases. If predicted coding sequences, such as those in SEQ JD NOSrl, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33 are used, amore appropriate number of primers for positive individual identification would be 500-2,000.
Predictive Medicine The invention also pertains to the field of predictive medicine in which diagnostic assays, prognostic assays, pharmacogenomics, and monitoring clinical trials are used for prognostic (predictive) pmposes to thereby treat an individual prophylactically. Accordingly, one aspect of the invention relates to diagnostic assays for determining NOVX protein and/or nucleic acid expression as well as NOVX activity, in the context of a biological sample (e.g. , blood, serum, cells, tissue) to thereby determine whether an individual is afflicted with a disease or disorder, or is at risk of developing a disorder, associated with aberrant NOVX expression or activity. The disorders include metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders, and the various dyslipidemias, metabolic disturbances associated with obesity, the metabolic syndrome X and wasting disorders associated with chronic diseases and various cancers. The invention also provides for prognostic (or predictive) assays for determining whether an individual is at risk of developing a disorder associated with NOVX protein, nucleic acid expression or activity. For example, mutations in an NOVX gene can be assayed in a biological sample. Such assays can be used for prognostic or predictive pmpose to thereby prophylactically treat an individual prior to the onset of a disorder characterized by or associated with NOVX protein, nucleic acid expression, or biological activity.
Another aspect of the invention provides methods for determining NOVX protein, nucleic acid expression or activity in an individual to thereby select appropriate therapeutic or prophylactic agents for that individual (referred to herein as "pharmacogenomics"). Pharmacogenomics allows for the selection of agents (e.g., drugs) for therapeutic or prophylactic treatment of an individual based on the genotype of the individual (e.g., the genotype of the individual examined to determine the ability of the individual to respond to a particular agent.)
Yet another aspect of the invention pertains to monitoring the influence of agents (e.g., drugs, compounds) on the expression or activity of NOVX in clinical trials. These and other agents are described in further detail in the following sections.
Diagnostic Assays
An exemplary method for detecting the presence or absence of NOVX in a biological sample involves obtaining a biological sample from a test subject and contacting the biological sample with a compound or an agent capable of detecting NOVX protein or nucleic acid (e.g., mRNA, genomic DNA) that encodes NOVX protein such that the presence of NOVX is detected in the biological sample. An agent for detecting NOVX mRNA or genomic DNA is a labeled nucleic acid probe capable of hybridizing to NOVX mRNA or genomic DNA. The nucleic acid probe can be, for example, a full-length NOVX nucleic acid, such as the nucleic acid of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, or a portion thereof, such as an oligonucleotide of at least 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to NOVX mRNA or genomic DNA. Other suitable probes for use in the diagnostic assays of the invention are described herein.
An agent for detecting NOVX protein is an antibody capable of binding to NOVX protein, preferably an antibody with a detectable label. Antibodies can be polyclonal, or more preferably, monoclonal. An intact antibody, or a fragment thereof (e.g., Fab or F(ab')2) can be used. The term "labeled", with regard to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled. Examples of indirect labeling include detection of a primary antibody using a fluorescently-labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently- labeled streptavidin. The term "biological sample" is intended to include tissues, cells and biological fluids isolated from a subject, as well as tissues, cells and fluids present within a subject. That is, the detection method of the invention can be used to detect NOVX mRNA, protein, or genomic DNA in a biological sample in vitro as well as in vivo. For example, in vitro techniques for detection of NOVX mRNA include Northern hybridizations and in situ hybridizations. In vitro techniques for detection of NOVX protein include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations, and immunofluorescence. In vitro techniques for detection of NOVX genomic DNA include Southern hybridizations. Furthermore, in vivo techniques for detection of NOVX protein include introducing into a subject a labeled anti-NOVX antibody. For example, the antibody can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques.
In one embodiment, the biological sample contains protein molecules from the test subject. Alternatively, the biological sample can contain mRNA molecules from the test subject or genomic DNA molecules from the test subject. A prefeπed biological sample is a peripheral blood leukocyte sample isolated by conventional means from a subject.
In another embodiment, the methods further involve obtaining a confrol biological sample from a control subject, contacting the control sample with a compound or agent capable of detecting NOVX protem, mRNA, or genomic DNA, such that the presence of NOVX protein, mRNA or genomic DNA is detected in the biological sample, and comparing the presence of NOVX protein, mRNA or genomic DNA in the control sample with the presence of NOVX protein, mRNA or genomic DNA in the test sample. The invention also encompasses kits for detecting the presence of NOVX in a biological sample. For example, the kit can comprise: a labeled compound or agent capable of detecting NOVX protein or mRNA in a biological sample; means for determining the amount of NOVX in the sample; and means for comparing the amount of NOVX in the sample with a standard., The compound or agent can be packaged in a suitable container. The kit can further comprise instructions for using the kit to detect NOVX protein or nucleic acid.
Prognostic Assays
The diagnostic methods described herein can furthermore be utilized to identify subjects having or at risk of developing a disease or disorder associated with abeπant NOVX expression or activity. For example, the assays described herein, such as the preceding diagnostic assays or the following assays, can be utilized to identify a subject having or at risk of developing a disorder associated with NOVX protein, nucleic acid expression or activity. Alternatively, the prognostic assays can be utilized to identify a subject having or at risk for developing a disease or disorder. Thus, the invention provides a method for identifying a disease or disorder associated with aberrant NOVX expression or activity in which a test sample is obtained from a subject and NOVX protein or nucleic acid (e.g., mRNA, genomic DNA) is detected, wherein the presence of NOVX protein or nucleic acid is diagnostic for a subject having or at risk of developing a disease or disorder associated with abeπant NOVX expression or activity. As used herein, a "test sample" refers to a biological sample obtained from a subject of interest. For example, a test sample can be a biological fluid (e.g., serum), cell sample, or tissue.
Furthermore, the prognostic assays described herein can be used to determine whether a subject can be administered an agent (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drug candidate) to treat a disease or disorder associated with abeπant NOVX expression or activity. For example, such methods can be used to determine whether a subject can be effectively treated with an agent for a disorder. Thus, the invention provides methods for determining whether a subject can be effectively treated with an agent for a disorder associated with abeπant NOVX expression or activity in which a test sample is obtained and NOVX protein or nucleic acid is detected (e.g., wherein the presence of NOVX protein or nucleic acid is diagnostic for a subject that can be administered the agent to treat a disorder associated with abeπant NOVX expression or activity).
The methods of the invention can also be used to detect genetic lesions in an NOVX gene, thereby determining if a subject with the lesioned gene is at risk for a disorder characterized by abeπant cell proliferation and/or differentiation. In various embodiments, the methods include detecting, in a sample of cells from the subject, the presence or absence of a genetic lesion characterized by at least one of an alteration affecting the integrity of a gene encoding an NOVX-protein, or the misexpression of the NOVX gene. For example, such genetic lesions can be detected by ascertaining the existence of at least one of: (i) a deletion of one or more nucleotides from an NOVX gene; (ii) an addition of one or more nucleotides to an NOVX gene; (iii) a substitution of one or more nucleotides of an NOVX gene, (iv) a chromosomal rearrangement of an NOVX gene; (v) an alteration in the level of a messenger RNA transcript of an NOVX gene, (vi) abeπant modification of an NOVX gene, such as of the methylation pattern of the genomic DNA, (vii) the presence of a non-wild-type splicing pattern of a messenger RNA transcript of an NOVX gene, (viii) a non-wild-type level of an NOVX protein, (ix) allelic loss of an NOVX gene, and (x) inappropriate post-translational modification of an NOVX protein. As described herein, there are a large number of assay techniques known in the art which can be used for detecting lesions in an NOVX gene. A prefeπed biological sample is a peripheral blood leukocyte sample isolated by conventional means from a subject. However, any biological sample containing nucleated cells may be used, including, for example, buccal mucosal cells.
In certain embodiments, detection of the lesion involves the use of a probe/primer in a polymerase chain reaction (PCR) (see, e.g., U.S. Patent Nos.4,683,195 and 4,683,202), such as anchor PCR or RACE PCR, or, alternatively, in a ligation chain reaction (LCR) (see, e.g., Landegran, et al, 1988. Sezewce 241: 1077-1080; and Nakazawa, et al, 1994. Proc. Natl
Acad. Sci. USA 91 : 360-364), the latter of which can be particularly useful for detecting point mutations in the NOVX-gene (see, Abravaya, et al, 1995. Nucl Acids Res. 23: 675-682). This method can include the steps of collecting a sample of cells from a patient, isolating nucleic acid (e.g., genomic, mRNA or both) from the cells of the sample, contacting the nucleic acid sample with one or more primers that specifically hybridize to an NOVX gene under conditions such that hybridization and amplification of the NOVX gene (if present) occurs, and detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample. It is anticipated that PCR and/or LCR may be desirable to use as a preliminary amplification step in conjunction with any of the techniques used for detecting mutations described herein. Alternative amplification methods include: self sustained sequence replication (see, Guatelli, et al, 1990. Proc. Natl. Acad. Sci. USA 87: 1874-1878), transcriptional amplification system (see, Kwoh, et al, 1989. Proc. Natl. Acad. Sci. USA 86: 1173-1177); Qβ Replicase (see,
Lizardi, et al, 1988. BioTechnology 6: 1197), or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers. In an alternative embodiment, mutations in an NOVX gene from a sample cell can be identified by alterations in restriction enzyme cleavage patterns. For example, sample and control DNA is isolated, amplified (optionally), digested with one or more restriction endonucleases, and fragment length sizes are determined by gel electrophoresis and compared. Differences in fragment length sizes between sample and control DNA indicates mutations in the sample DNA. Moreover, the use of sequence specific ribozymes (see, e.g., U.S. Patent No. 5,493,531) can be used to score for the presence of specific mutations by development or loss of a ribozyme cleavage site. hi other embodiments, genetic mutations in NOVX can be identified by hybridizing a sample and confrol nucleic acids, e.g., DNA or RNA, to high-density arrays containing hundreds or thousands of oligonucleotides probes. See, e.g., Cronin, et al, 1996. Human Mutation 1: 244-255; Kozal, et al, 1996. Nat. Med. 2: 753-759. For example, genetic mutations in ΝOVX can be identified in two dimensional arrays containing light-generated DΝA probes as described in Cronin, et al, supra. Briefly, a first hybridization array of probes can be used to scan through long stretches of DΝA in a sample and control to identify base changes between the sequences by making linear arrays of sequential overlapping probes. This step allows the identification of point mutations. This is followed by a second hybridization array that allows the characterization of specific mutations by using smaller, specialized probe arrays complementary to all variants or mutations detected. Each mutation array is composed of parallel probe sets, one complementary to the wild-type gene and the other complementary to the mutant gene.
In yet another embodiment, any of a variety of sequencing reactions known in the art can be used to directly sequence the ΝOVX gene and detect mutations by comparing the sequence of the sample ΝOVX with the coπesponding wild-type (control) sequence. Examples of sequencing reactions include those based on techniques developed by Maxim and Gilbert, 1977. Proc. Natl. Acad. Sci. USA 74: 560 or Sanger, 1977. Proc. Natl Acad. Sci. USA 74: 5463. It is also contemplated that any of a variety of automated sequencing procedures can be utilized when performing the diagnostic assays (see, e.g., Naeve, et al, 1995. Biotechniques 19: 448), including sequencing by mass spectrometry (see, e.g., PCT International Publication No. WO 94/16101; Cohen, et al, 1996. Adv. Chromatography 36: 127-162; and Griffin, et al, 1993. Appl. Biochem. Biotechnol 38: 147-159).
Other methods for detecting mutations in the NOVX gene include methods in which protection from cleavage agents is used to detect mismatched bases in RNA/RNA or RNA/DNA heteroduplexes. See, e.g., Myers, et al, 1985. Science 230: 1242. In general, the art technique of "mismatch cleavage" starts by providing heteroduplexes of formed by hybridizing (labeled) RNA or DNA containing the wild-type NOVX sequence with potentially mutant RNA or DNA obtained from a tissue sample. The double-stranded duplexes are treated with an agent that cleaves single-stranded regions of the duplex such as which will exist due to basepair mismatches between the confrol and sample strands. For instance, RNA/DNA duplexes can be treated with RNase and DNA/DNA hybrids treated with Si nuclease to enzymatically digesting the mismatched regions. In other embodiments, either DNA/DNA or RNA/DNA duplexes can be treated with hydroxylamine or osmium tetroxide and with piperidine in order to digest mismatched regions. After digestion of the mismatched regions, the resulting material is then separated by size on denaturing polyacrylamide gels to determine the site of mutation. See, e.g., Cotton, etal, 1988. Proc. Natl Acad. Sci. USA 85: 4397; Saleeba, et al, 1992. Methods Enzymol 217: 286-295. In an embodiment, the confrol DNA or RNA can be labeled for detection.
In still another embodiment, the mismatch cleavage reaction employs one or more proteins that recognize mismatched base pairs in double-stranded DNA (so called "DNA mismatch repair" enzymes) in defined systems for detecting and mapping point mutations in NOVX cDNAs obtained from samples of cells. For example, the mutY enzyme of E. coli cleaves A at G/A mismatches and the thymidine DNA glycosylase from HeLa cells cleaves T at G/T mismatches. See, e.g., Hsu, et al, 1994. Carcinogenesis 15: 1657-1662. According to an exemplary embodiment, a probe based on an NOVX sequence, e.g., a wild-type NOVX sequence, is hybridized to a cDNA or other DNA product from a test cell(s). The duplex is treated with a DNA mismatch repair enzyme, and the cleavage products, if any, can be detected from electrophoresis protocols or the like. See, e.g., U.S. Patent No. 5,459,039. In other embodiments, alterations in electrophoretic mobility will be used to identify mutations in NOVX genes. For example, single strand conformation polymoφhism (SSCP) may be used to detect differences in electrophoretic mobility between mutant and wild type nucleic acids. See, e.g., Orita, et al, 1989. Proc. Natl. Acad. Sci. USA: 86: 2766; Cotton, 1993. Mutat. Res. 285: 125-144; Hayashi, 1992. Genet. Anal. Tech. Appl. 9: 73-79. Single-stranded DNA fragments of sample and control NOVX nucleic acids will be denatured and allowed to renature. The secondary structure of single-stranded nucleic acids varies according to sequence, the resulting alteration in electrophoretic mobility enables the detection of even a single base change. The DNA fragments may be labeled or detected with labeled probes. The sensitivity of the assay may be enhanced by using RNA (rather than DNA), in which the secondary structure is more sensitive to a change in sequence. In one embodiment, the subject method utilizes heteroduplex analysis to separate double stranded heteroduplex molecules on the basis of changes in electrophoretic mobility. See, e.g., Keen, et al, 1991. Trends Genet. 1: 5.
In yet another embodiment, the movement of mutant or wild-type fragments in polyacrylamide gels containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (DGGE). See, e.g., Myers, et al, 1985. Nature 313: 495. When DGGE is used as the method of analysis, DNA will be modified to insure that it does not completely denature, for example by adding a GC clamp ofapproximately 40 bp of high-melting GC-rich DNA by PCR. In a further embodiment, a temperature gradient is used in place of a denaturing gradient to identify differences in the mobility of control and sample DNA. See, e.g., Rosenbaum and Reissner, 1987. Biophys. Chem. 265: 12753.
Examples of other techniques for detecting point mutations include, but are not limited to, selective oligonucleotide hybridization, selective amplification, or selective primer extension. For example, oligonucleotide primers may be prepared in which the known mutation is placed centrally and then hybridized to target DNA under conditions that permit hybridization only if a perfect match is found. See, e.g., Saiki, et al, 1986. Nature 324: 163; Saiki, et al, 1989. Proc. Natl. Acad. Sci. USA 86: 6230. Such allele specific oligonucleotides are hybridized to PCR amplified target DNA or a number of different mutations when the oligonucleotides are attached to the hybridizing membrane and hybridized with labeled target DNA. Alternatively, allele specific amplification technology that depends on selective PCR amplification may be used in conjunction with the instant invention. Oligonucleotides used as primers for specific amplification may carry the mutation of interest in the center of the molecule (so that amplification depends on differential hybridization; .see, e.g., Gibbs, et al, 1989. Nucl. Acids Res. 17: 2437-2448) or at the extreme 3'-terminus of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerase extension (see, e.g., Prossner, 1993. Tibtech. 11 : 238). In addition it may be desirable to introduce a novel restriction site in the region of the mutation to create cleavage-based detection. See, e.g., Gasparini, et al, 1992. Mol. Cell Probes 6: 1. It is anticipated that in certain embodiments amplification may also be performed using Taq ligase for amplification. See, e.g., Barany, 1991. Proc. Natl. Acad. Sci. USA 88: 189. In such cases, ligation will occur only if there is a perfect match, at the 3'-terminus of the 5' sequence, making it possible to detect the presence of a known mutation at a specific site by looking for the presence or absence of amplification. The methods described herein may be performed, for example, by utilizing pre-packaged diagnostic kits comprising at least one probe nucleic acid or antibody reagent described herein, which may be conveniently used, e.g., in clinical settings to diagnose patients exhibiting symptoms or family history of a disease or illness involving an NOVX gene.
Furthermore, any cell type or tissue, preferably peripheral blood leukocytes, in which NOVX is expressed may be utilized in the prognostic assays described herein. However, any biological sample containing nucleated cells may be used, including, for example, buccal mucosal cells.
Pharmacogenomics
Agents, or modulators that have a stimulatory or inhibitory effect on NOVX activity (e.g., NOVX gene expression), as identified by a screening assay described herein can be administered to individuals to treat (prophylactically or therapeutically) disorders (The disorders include metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer- associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders, and the various dyslipidemias, metabolic disturbances associated with obesity, the metabolic syndrome X and wasting disorders associated with chronic diseases and various cancers.) In conjunction with such treatment, the pharmacogenomics (i.e., the study of the relationship between an individual's genotype and that individual's response to a foreign compound or drug) of the individual may be considered. Differences in metabolism of therapeutics can lead to severe toxicity or therapeutic failure by altering the relation between dose and blood concentration of the pharmacologically active drag. Thus, the pharmacogenomics of the individual permits the selection of effective agents (e.g., drugs) for prophylactic or therapeutic treatments based on a consideration of the individual's genotype. Such pharmacogenomics can further be used to determine appropriate dosages and therapeutic regimens. Accordingly, the activity of NOVX protein, expression of NOVX nucleic acid, or mutation content of NOVX genes in an individual can be determined to thereby select appropriate agent(s) for therapeutic or prophylactic treatment of the individual.
Pharmacogenomics deals with clinically significant hereditary variations in the response to drags due to altered drug disposition and abnormal action in affected persons. See e.g., Eichelbaum, 1996. Clin. Exp. Pharmacol Physiol, 23: 983-985; Linder, 1997. Clin. Chem., 43: 254-266. In general, two types of pharmacogenetic conditions can be differentiated. Genetic conditions transmitted as a single factor altering the way drugs act on the body (altered drug action) or genetic conditions transmitted as single factors altering the way the body acts on drags (altered drug metabolism). These pharmacogenetic conditions can occur either as rare defects or as polymoφhisms. For example, glucose-6-phosphate dehydrogenase (G6PD) deficiency is a common inherited enzymopathy in which the main clinical complication is hemolysis after ingestion of oxidant drugs (anti-malarials, sulfonamides, analgesics, nitrofurans) and consumption of fava beans. As an illustrative embodiment, the activity of drug metabolizing enzymes is a major determinant of both the intensity and duration of drug action. The discovery of genetic polymoφhisms of drag metabolizing enzymes (e.g., N-acetyltransferase 2 (NAT 2) and cytochrome P450 enzymes CYP2D6 and CYP2C19) has provided an explanation as to why some patients do not obtain the expected drug effects or show exaggerated drag response and serious toxicity after taking the standard and safe dose of a drag. These polymoφhisms are expressed in two phenotypes in the population, the extensive metabolizer (EM) and poor metabolizer (PM). The prevalence of PM is different among different populations. For example, the gene coding for CYP2D6 is highly polymoφhic and several mutations have been identified in PM, which all lead to the absence of functional CYP2D6. Poor metabolizers of CYP2D6 and CYP2C19 quite frequently experience exaggerated drag response and side effects when they receive standard doses. If a metabolite is the active therapeutic moiety, PM show no therapeutic response, as demonstrated for the analgesic effect of codeine mediated by its CYP2D6-formed metabolite moφhine. At the other extreme are the so called ultra-rapid metabolizers who do not respond to standard doses. Recently, the molecular basis of ultra-rapid metabolism has been identified to be due to CYP2D6 gene amplification.
Thus, the activity of NOVX protein, expression of NOVX nucleic acid, or mutation content of NOVX genes in an individual can be determined to thereby select appropriate agent(s) for therapeutic or prophylactic treatment of the individual. In addition, pharmacogenetic studies can be used to apply genotyping of polymoφhic alleles encoding drag-metabolizing enzymes to the identification of an individual's drag responsiveness phenotype. This knowledge, when applied to dosing or drug selection, can avoid adverse reactions or therapeutic failure and thus enhance therapeutic or prophylactic efficiency when treating a subject with an NOVX modulator, such as a modulator identified by one of the exemplary screening assays described herein.
Monitoring of Effects During Clinical Trials
Monitoring the influence of agents (e.g., drags, compounds) on the expression or activity of NOVX (e.g., the ability to modulate abeπant cell proliferation and/or differentiation) can be applied not only in basic drug screening, but also in clinical trials. For example, the effectiveness of an agent determined by a screening assay as described herein to increase NOVX gene expression, protein levels, or upregulate NOVX activity, can be monitored in clinical trails of subjects exhibiting decreased NOVX gene expression, protein levels, or downregulated NOVX activity. Alternatively, the effectiveness of an agent deteπnined by a screening assay to decrease NOVX gene expression, protein levels, or downregulate NOVX activity, can be monitored in clinical trails of subjects exhibiting increased NOVX gene expression, protein levels, or upregulated NOVX activity. In such clinical trials, the expression or activity of NOVX and, preferably, other genes that have been implicated in, for example, a cellular proliferation or immune disorder can be used as a "read out" or markers of the immune responsiveness of a particular cell.
Byway of example, and not of limitation, genes, including NOVX, that are modulated in cells by treatment with an agent (e.g., compound, drug or small molecule) that modulates NOVX activity (e.g., identified in a screening assay as described herein) can be identified. Thus, to study the effect of agents on cellular proliferation disorders, for example, in a clinical trial, cells can be isolated and RNA prepared and analyzed for the levels of expression of
NOVX and other genes implicated in the disorder. The levels of gene expression (i.e., a gene expression pattern) can be quantified by Northern blot analysis or RT-PCR, as described herein, or alternatively by measuring the amount of protein produced, by one of the methods as described herein, or by measuring the levels of activity of NOVX or other genes. In this manner, the gene expression pattern can serve as a marker, indicative of the physiological response of the cells to the agent. Accordingly, this response state may be determined before, and at various points during, treatment of the individual with the agent.
In one embodiment, the invention provides a method for monitoring the effectiveness of treatment of a subject with an agent (e.g., an agonist, antagonist, protein, peptide, peptidomimetic, nucleic acid, small molecule, or other drag candidate identified by the screening assays described herein) comprising the steps of (i) obtaining a pre-administration sample from a subject prior to administration of the agent; (ii) detecting the level of expression of an NOVX protein, mRNA, or genomic DNA in the preadministration sample; (iii) obtaining one or more post-administration samples from the subject; (iv) detecting the level of expression or activity of the NOVX protein, mRNA, or genomic DNA in the post-administration samples; (v) comparing the level of expression or activity of the NOVX protein, mRNA, or genomic DNA in the pre-administration sample with the NOVX protein, mRNA, or genomic DNA in the post admimsfration sample or samples; and (vi) altering the administration of the agent to the subject accordingly. For example, increased administration of the agent may be desirable to increase the expression or activity of NOVX to higher levels than detected, i.e., to increase the effectiveness of the agent. Alternatively, decreased administration of the agent may be desirable to decrease expression or activity of NOVX to lower levels than detected, i.e., to decrease the effectiveness of the agent.
Methods of Treatment The invention provides for both prophylactic and therapeutic methods of treating a subject at risk of (or susceptible to) a disorder or having a disorder associated with aberrant NOVX expression or activity. The disorders include cardiomyopathy, atherosclerosis, hypertension, congenital heart defects, aortic stenosis, atrial septal defect (ASD), atrioventricular (A-V) canal defect, ductus arteriosus, pulmonary stenosis, subaortic stenosis, ventricular septal defect (VSD), valve diseases, tuberous sclerosis, scleroderma, obesity, transplantation, adrenoleukodystrophy, congenital adrenal hypeφlasia, prostate cancer, neoplasm; adenocarcinoma, lymphoma, uterus cancer, fertility, hemophilia, hypercoagulation, idiopathic thrombocytopenic puφura, immunodeficiencies, graft versus host disease, AIDS, bronchial asthma, Crohn's disease; multiple sclerosis, treatment of Albright Hereditary Ostoeodystrophy, and other diseases, disorders and conditions of the like. These methods of treatment will be discussed more fully, below.
Disease and Disorders
Diseases and disorders that are characterized by increased (relative to a subject not suffering from the disease or disorder) levels or biological activity may be treated with
Therapeutics that antagonize (i.e., reduce or inhibit) activity. Therapeutics that antagonize activity may be administered in a therapeutic or prophylactic manner. Therapeutics that may be utilized include, but are not limited to: (i) an aforementioned peptide, or analogs, derivatives, fragments or homologs thereof; (ii) antibodies to an aforementioned peptide; (iii) nucleic acids encoding an aforementioned peptide; (iv) administration of antisense nucleic acid and nucleic acids that are "dysfunctional" (i.e., due to a heterologous insertion within the coding sequences of coding sequences to an aforementioned peptide) that are utilized to "knockout" endogenous function of an aforementioned peptide by homologous recombination (see, e.g., Capecchi, 1989. Science 244: 1288-1292); or (v) modulators ( i.e., inhibitors, agonists and antagonists, including additional peptide mimetic of the invention or antibodies specific to a peptide of the invention) that alter the interaction between an aforementioned peptide and its binding partner.
Diseases and disorders that are characterized by decreased (relative to a subject not suffering from the disease or disorder) levels or biological activity may be treated with
Therapeutics that increase (i.e., are agonists to) activity. Therapeutics that upregulate activity may be administered in a therapeutic or prophylactic manner. Therapeutics that may be utilized include, but are not limited to, an aforementioned peptide, or analogs, derivatives, fragments or homologs thereof; or an agonist that increases bioavailability. Increased or decreased levels can be readily detected by quantifying peptide and/or
RNA, by obtaining a patient tissue sample (e.g., from biopsy tissue) and assaying it in vitro for RNA or peptide levels, structure and/or activity of the expressed peptides (or mRNAs of an aforementioned peptide). Methods that are well-known within the art include, but are not limited to, immunoassays (e.g., by Western blot analysis, immunoprecipitation followed by sodium dodecyl sulfate (SDS) polyacrylamide gel electrophoresis, immunocytochemistry, etc.) and/or hybridization assays to detect expression of mRNAs (e.g., Northern assays, dot blots, in situ hybridization, and the like).
Prophylactic Methods
In one aspect, the invention provides a method for preventing, in a subject, a disease or condition associated with an abeπant NOVX expression or activity, by adπiinistering to the subject an agent that modulates NOVX expression or at least one NOVX activity. Subjects at risk for a disease that is caused or contributed to by aberrant NOVX expression or activity can be identified by, for example, any or a combination of diagnostic or prognostic assays as described herein. Administration of a prophylactic agent can occur prior to the manifestation of symptoms characteristic of the NOVX abeπancy, such that a disease or disorder is prevented or, alternatively, delayed in its progression. Depending upon the type of NOVX abeπancy, for example, an NOVX agonist or NOVX antagonist agent can be used for treating the subject. The appropriate agent can be determined based on screening assays described herein. The prophylactic methods of the invention are further discussed in the following subsections. Therapeutic Methods
Another aspect of the invention pertains to methods of modulating NOVX expression or activity for therapeutic pmposes. The modulatory method of the invention involves contacting a cell with an agent that modulates one or more of the activities of NOVX protein activity associated with the cell. An agent that modulates NOVX protein activity can be an agent as described herein, such as a nucleic acid or a protein, a naturally-occurring cognate ligand of an NOVX protein, a peptide, an NOVX peptidomimetic, or other small molecule. In one embodiment, the agent stimulates one or more NOVX protein activity. Examples of such stimulatory agents include active NOVX protein and a nucleic acid molecule encoding NOVX that has been introduced into the cell. In another embodiment, the agent inhibits one or more NOVX protein activity. Examples of such inhibitory agents include antisense NOVX nucleic acid molecules and anti-NOVX antibodies. These modulatory methods can be performed in vitro (e.g., by culturing the cell with the agent) or, alternatively, in vivo (e.g., by administering the agent to a subject). As such, the invention provides methods of treating an individual afflicted with a disease or disorder characterized by abeπant expression or activity of an NOVX protein or nucleic acid molecule. In one embodiment, the method involves administering an agent (e.g., an agent identified by a screening assay described herein), or combination of agents that modulates (e.g., up-regulates or down-regulates) NOVX expression or activity. In another embodiment, the method involves administering an NOVX protein or nucleic acid molecule as therapy to compensate for reduced or aberrant NOVX expression or activity.
Stimulation of NOVX activity is desirable in .sttwations in which NOVX is abnormally downregulated and/or in which increased NOVX activity is likely to have a beneficial effect. One example of such a situation is where a subject has a disorder characterized by abeπant cell proliferation and/or differentiation (e.g., cancer or immune associated disorders). Another example of such a situation is where the subject has a gestational disease (e.g., preclampsia).
Determination of the Biological Effect of the Therapeutic
In various embodiments of the invention, suitable in vitro or in vivo assays are performed to determine the effect of a specific Therapeutic and whether its administration is indicated for treatment of the affected tissue.
In various specific embodiments, in vitro assays may be performed with representative cells of the type(s) involved in the patient's disorder, to determine if a given Therapeutic exerts the desired effect upon the cell type(s). Compounds for use in therapy may be tested in suitable animal model systems including, but not limited to rats, mice, chicken, cows, monkeys, rabbits, and the like, prior to testing in human subjects. Similarly, for in vivo testing, any of the animal model system known in the art may be used prior to administration to human subjects.
Prophylactic and Therapeutic Uses of the Compositions of the Invention
The NOVX nucleic acids and proteins of the invention are useful in potential prophylactic and therapeutic applications implicated in a variety of disorders including, but not limited to: metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer- associated cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, hematopoietic disorders, and the various dyslipidemias, metabolic disturbances associated with obesity, the metabolic syndrome X and wasting disorders associated with chronic diseases and various cancers.
As an example, a cDNA encoding the NOVX protein of the invention may be useful in gene therapy, and the protein may be useful when administered to a subject in need thereof. By way of non-limiting example, the compositions of the invention will have efficacy for treatment of patients suffering from: metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, hematopoietic disorders, and the various dyslipidemias. Both the novel nucleic acid encoding the NOVX protein, and the NOVX protein of the invention, or fragments thereof, may also be useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed. A further use could be as an anti-bacterial molecule (i.e., some peptides have been found to possess anti-bacterial properties). These materials are further useful in the generation of antibodies, which immunospecifically-bind to the novel substances of the invention for use in therapeutic or diagnostic methods.
The invention will be further described in the following examples, which do not limit the scope of the invention described in the claims.
EXAMPLES Example 1. Identification of NOVX clones
The novel NOVX target sequences identified in the present invention were subjected to the exon linking process to confirm the sequence. As shown in Table 17, PCR primers were designed by starting at the most upstream sequence available, for the forward primer, and at the most downstream sequence available for the reverse primer In each case, the sequence was examined, walking inward from the respective termini toward the coding sequence, until a suitable sequence that is either unique or highly selective was encountered, or, in the case of the reverse primer, until the stop codon was reached. Such primers were designed based on in silico predictions for the full length cDNA, part (one or more exons) of the DNA or protein sequence of the target sequence, or by translated homology of the predicted exons to closely related human sequences from other species. These primers were then employed in PCR amplification based on the following pool of human cDNAs: adrenal gland, bone marrow, brain - amygdala, brain - cerebellum, brain - hippocampus, brain - substantia nigra, brain - thalamus, brain -whole, fetal brain, fetal kidney, fetal liver, fetal lung, heart, kidney, lymphoma - Raji, mammary gland, pancreas, pituitary gland, placenta, prostate, salivary gland, skeletal muscle, small intestine, spinal cord, spleen, stomach, testis, thyroid, trachea, uterus. Usually the resulting amplicons were gel purified, cloned and sequenced to high redundancy. The PCR product derived from exon linking was cloned into the pCR2.1 vector from Invitrogen. The resulting bacterial clone has an insert covering the entire open reading frame cloned into the pCR2.1 vector. The resulting sequences from all clones were assembled with themselves, with other fragments in CuraGen Coφoration' s database and with public ESTs. Fragments and ESTs were included as components for an assembly when the extent of their identity with another component of the assembly was at least 95% over 50 bp. In addition, sequence traces were evaluated manually and edited for corrections if appropriate. These procedures provide the sequence reported herein.
Table 17. PCR Primers for Exon Linking
Figure imgf000198_0001
Example 2. Quantitative expression analysis of clones in various cells and tissues
The quantitative expression of various clones was assessed using microtiter plates containing RNA samples from a variety of normal and pathology-derived cells, cell lines and tissues using real time quantitative PCR (RTQ PCR). RTQ PCR was performed on an Applied Biosystems ABI PRISM® 7700 or an ABI PRISM® 7900 HT Sequence Detection System. Various collections of samples are assembled on the plates, and referred to as Panel 1 (containing normal tissues and cancer cell lines), Panel 2 (containing samples derived from tissues from normal and cancer sources), Panel 3 (containing cancer cell lines), Panel 4 (containing cells and cell lines from normal tissues and cells related to inflammatory conditions), Panel 5D/5I (containing human tissues and cell lines with an emphasis on metabolic diseases), AI_comprehensive__panel (containing normal tissue and samples from autoimmune diseases), Panel CNSD.01 (containing central nervous system samples from normal and diseased brains) and CNS_neurodegeneration_panel (containing samples from normal and Alzheimer's diseased brains).
RNA integrity from all samples is controlled for quality by visual assessment of agarose gel electropherograms using 28S and 18S ribosomal RNA staining intensity ratio as a guide (2:1 to 2.5:1 28s: 18s) and the absence of low molecular weight RNAs that would be indicative of degradation products. Samples are controlled against genomic DNA contamination by RTQ PCR reactions run in the absence of reverse transcriptase using probe and primer sets designed to amplify across the span of a single exon.
First, the RNA samples were normalized to reference nucleic acids such as constitutively expressed genes (for example, β-actin and GAPDH). Normalized RNA (5 ul) was converted to cDNA and analyzed by RTQ-PCR using One Step RT-PCR Master Mix
Reagents (Applied Biosystems; Catalog No.4309169) and gene-specific primers according to the manufacturer's instructions.
In other cases, non-normalized RNA samples were converted to single strand cDNA (sscDNA) using Superscript JJ (Invitrogen Coφoration; Catalog No. 18064-147) and random hexamers according to the manufacturer's instractions. Reactions containing up to 10 μg of total RNA were performed in a volume of 20 μl and incubated for 60 minutes at 42°C. This reaction can be scaled up to 50 μg of total RNA in a final volume of 100 μl. sscDNA samples are then normalized to reference nucleic acids as described previously, using IX TaqMan® Universal Master mix (Applied Biosystems; catalog No. 4324020), following the manufacturer's instractions.
Probes and primers were designed for each assay according to Applied Biosystems
Primer Express Software package (version I for Apple Computer's Macintosh Power PC) or a similar algorithm using the target sequence as input. Default settings were used for reaction conditions and the following parameters were set before selecting primers: primer concentration = 250 nM, primer melting temperature (Tm) range = 58°-60°C, primer optimal Tm = 59°C, maximum primer difference = 2°C, probe does not have 5'G, probe Tm must be 10°C greater than primer Tm, amplicon size 75bp to lOObp. The probes and primers selected (see below) were synthesized by Synthegen (Houston, TX, USA). Probes were double purified by HPLC to remove uncoupled dye and evaluated by mass spectroscopy to verify coupling of reporter and quencher dyes to the 5' and 3' ends of the probe, respectively. Their final concentrations were: forward and reverse primers, 900nM each, and probe, 200nM.
PCR conditions: When working with RNA samples, normalized RNA from each tissue and each cell line was spotted in each well of either a 96 well or a 384- well PCR plate (Applied Biosystems). PCR cocktails included either a single gene specific probe and primers set, or two multiplexed probe and primers sets (a set specific for the target clone and another gene-specific set multiplexed with the target probe). PCR reactions were set up using TaqMan® One-Step RT-PCR Master Mix (Applied Biosystems, Catalog No.4313803) following manufacturer's instructions. Reverse transcription was performed at 48°C for 30 minutes followed by amplification/PCR cycles as follows: 95°C 10 min, then 40 cycles of 95°C for 15 seconds, 60°C for 1 minute. Results were recorded as CT values (cycle at which a given sample crosses a threshold level of fluorescence) using a log scale, with the difference in RNA concentration between a given sample and the sample with the lowest CT value being represented as 2 to the power of delta CT. The percent relative expression is then obtained by taking the reciprocal of this RNA difference and multiplying by 100.
When working with sscDNA samples, normalized sscDNA was used as described previously for RNA samples. PCR reactions containing one or two sets of probe and primers were set up as described previously, using IX TaqMan® Universal Master mix (Applied Biosystems; catalog No. 4324020), following the manufacturer's instractions. PCR amplification was performed as follows: 95°C 10 min, then 40 cycles of 95°C for 15 seconds, 60°C for 1 minute. Results were analyzed and processed as described previously.
Panels 1, 1.1, 1.2, and 1.3D
The plates for Panels 1, 1.1, 1.2 and 1.3D include 2 control wells (genomic DNA control and chemistry control) and 94 wells containing cDNA from various samples. The samples in these panels are broken into 2 classes: samples derived from cultured cell lines and samples derived from primary normal tissues. The cell lines are derived from cancers of the following types: lung cancer, breast cancer, melanoma, colon cancer, prostate cancer, CNS cancer, squamous cell carcinoma, ovarian cancer, liver cancer, renal cancer, gastric cancer and pancreatic cancer. Cell lines used in these panels are widely available through the American Type Culture Collection (ATCC), a repository for cultured cell lines, and were cultured using the conditions recommended by the ATCC. The normal tissues found on these panels are comprised of samples derived from all major organ systems from single adult individuals or fetuses. These samples are derived from the following organs: adult skeletal muscle, fetal skeletal muscle, adult heart, fetal heart, adult kidney, fetal kidney, adult liver, fetal liver, adult lung, fetal lung, various regions of the brain, the spleen, bone marrow, lymph node, pancreas, salivary gland, pituitary gland, adrenal gland, spinal cord, thymus, stomach, small intestine, colon, bladder, trachea, breast, ovary, uterus, placenta, prostate, testis and adipose.
In the results for Panels 1, 1.1, 1.2 and 1.3D, the following abbreviations are used:
ca. = carcinoma, k * = established from metastasis, met = metastasis, s cell var = small cell variant, non-s = non-sm = non-small, squam = squamous, pi. eff = pi effusion = pleural effusion, glio = glioma, astro = astrocytoma, and neuro = neuroblastoma.
General_screening panel vl .4
The plates for Panel 1.4 include 2 control wells (genomic DNA control and chemistry control) and 94 wells containing cDNA from various samples. The samples in Panel 1.4 are broken into 2 classes: samples derived from cultured cell lines and samples derived from primary normal tissues. The cell lines are derived from cancers of the following types: lung cancer, breast cancer, melanoma, colon cancer, prostate cancer, CNS cancer, squamous cell carcinoma, ovarian cancer, liver cancer, renal cancer, gastric cancer and pancreatic cancer.
Cell lines used in Panel 1.4 are widely available through the American Type Culture
Collection (ATCC), a repository for cultured cell lines, and were cultured using the conditions recommended by the ATCC. The normal tissues found on Panel 1.4 are comprised of pools of samples derived from all major organ systems from 2 to 5 different adult individuals or fetuses. These samples are derived from the following organs: adult skeletal muscle, fetal skeletal muscle, adult heart, fetal heart, adult kidney, fetal kidney, adult liver, fetal liver, adult lung, fetal lung, various regions of the brain, the spleen, bone marrow, lymph node, pancreas, salivary gland, pituitary gland, adrenal gland, spinal cord, thymus, stomach, small intestine, colon, bladder, trachea, breast, ovary, uterus, placenta, prostate, testis and adipose. Abbreviations are as described for Panels 1, 1.1, 1.2, and 1.3D.
Panels 2D and 2.2
The plates for Panels 2D and 2.2 generally include 2 control wells and 94 test samples composed of RNA or cDNA isolated from human tissue procured by surgeons working in close cooperation with the National Cancer Institute's Cooperative Human Tissue Network (CHTN) or the National Disease Research Initiative (NDRI). The tissues are derived from human malignancies and in cases where indicated many malignant tissues have "matched margins" obtained from noncancerous tissue just adjacent to the tumor. These are termed normal adjacent tissues and are denoted "NAT" in the results below. The tumor tissue and the "matched margins" are evaluated by two independent pathologists (the surgical pathologists and again by a pathologist at NDRI or CHTN). This analysis provides a gross histopathological assessment of tumor differentiation grade. Moreover, most samples include the original surgical pathology report that provides information regarding the clinical stage of the patient. These matched margins are taken from the tissue suπounding (i.e. immediately proximal) to the zone of surgery (designated "NAT", for normal adjacent tissue, in Table RR). In addition, RNA and cDNA samples were obtained from various human tissues derived from autopsies performed on elderly people or sudden death victims (accidents, etc.). These tissues were ascertained to be free of disease and were purchased from various commercial sources such as Clontech (Palo Alto, CA), Research Genetics, and mvitrogen.
Panel 3D
The plates of Panel 3D are comprised of 94 cDNA samples and two control samples.
Specifically, 92 of these samples are derived from cultured human cancer cell lines, 2 samples of human primary cerebellar tissue and 2 controls. The human cell lines are generally obtained from ATCC (American Type Culture Collection), NCI or the German tumor cell bank and fall into the following tissue groups: Squamous cell carcinoma of the tongue, breast cancer, prostate cancer, melanoma, epidermoid carcinoma, sarcomas, bladder carcinomas, pancreatic cancers, kidney cancers, leukemias/lymphomas, ovarian/uterine/cervical, gastric, colon, lung and CNS cancer cell lines, m addition, there are two independent samples of cerebellum. These cells are all cultured under standard recommended conditions and RNA extracted using the standard procedures. The cell lines in panel 3D and 1.3D are of the most common cell lines used in the scientific literature.
Panels 4D, 4R, and 4.1D
Panel 4 includes samples on a 96 well plate (2 control wells, 94 test samples) composed of RNA (Panel 4R) or cDNA (Panels 4D/4.1D) isolated from various human cell lines or tissues related to inflammatory conditions. Total RNA from control normal tissues such as colon and lung (Stratagene, La Jolla, CA) and thymus and kidney (Clontech) was employed. Total RNA from liver tissue from cirrhosis patients and kidney from lupus patients was obtained from BioChain (Biochain Institute, hie, Hayward, CA). Intestinal tissue for RNA preparation from patients diagnosed as having Crohn's disease and ulcerative colitis was obtained from the National Disease Research Interchange (NDRI) (Philadelphia, PA).
Astrocytes, lung fibroblasts, dermal fibroblasts, coronary artery smooth muscle cells, small airway epithelium, bronchial epithelium, microvascular dermal endothelial cells, microvascular lung endothelial cells, human pulmonary aortic endothelial cells, human umbilical vein endothelial cells were all purchased from Clonetics (Walkersville, MD) and grown in the media supplied for these cell types by Clonetics. These primary cell types were activated with various cytokines or combinations of cytokines for 6 and/or 12-14 hours, as indicated. The following cytokines were used; IL-1 beta at approximately l-5ng ml, TNF alpha at approximately 5-10ng/ml, TFN gamma at approximately 20-50ng/ml, IL-4 at approximately 5-lOng/ml, IL-9 at approximately 5-lOng/ml, IL-13 at approximately 5- lOng/ml. Endothelial cells were sometimes starved for various times by culture in the basal media from Clonetics with 0.1% serum.
Mononuclear cells were prepared from blood of employees at CuraGen Coφoration, using Ficoll. LAK cells were prepared from these cells by culture in DMEM 5% FCS
(Hyclone), lOOμM non essential amino acids (Gibco/Life Technologies, Rockville, MD), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5xl0"5M (Gibco), and lOmM Hepes
(Gibco) and Interleukin 2 for 4-6 days. Cells were then either activated with 10-20ng/ml PMA and l-2μg/ml ionomycin, IL-12 at 5-lOng/ml, IFN gamma at 20-50ng/ml and IL-18 at 5- 10ng/ml for 6 hours. In some cases, mononuclear cells were cultured for 4-5 days in DMEM 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5x10"5M (Gibco), and lOmM Hepes (Gibco) with PHA (phytohemagglutinin) or PWM (pokeweed mitogen) at approximately 5μg/ml. Samples were taken at 24, 48 and 72 hours for RNA preparation. MLR (mixed lymphocyte reaction) samples were obtained by taking blood from two donors, isolating the mononuclear cells using Ficoll and mixing the isolated mononuclear cells 1:1 at a final concentration ofapproximately 2xl06cells/ml in DMEM 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol (5.5x10"5M) (Gibco), and lOmM Hepes (Gibco). The MLR was cultured and samples taken at various time points ranging from 1- 7 days for RNA preparation.
Monocytes were isolated from mononuclear cells using CD 14 Miltenyi Beads, +ve VS selection columns and a Vario Magnet according to the manufacturer's instructions. Monocytes were differentiated into dendritic cells by culture in DMEM 5% fetal calf serum (FCS) (Hyclone, Logan, UT), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5xl0"5M (Gibco), and lOmM Hepes (Gibco), 50ng/ml GMCSF and 5ng/ml IL-4 for 5-7 days. Macrophages were prepared by culture of monocytes for 5-7 days in DMEM 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5xlO"5M (Gibco), lOmM Hepes (Gibco) and 10% AB Human Serum or MCSF at approximately 50ng/ml. Monocytes, macrophages and dendritic cells were stimulated for 6 and 12-14 hours with lipopolysaccharide (LPS) at lOOng/ml. Dendritic cells were also stimulated with anti-CD40 monoclonal antibody (Pharmingen) at lOμg/ml for 6 and 12-14 hours.
CD4 lymphocytes, CD8 lymphocytes and NK cells were also isolated from mononuclear cells using CD4, CD8 and CD56 Miltenyi beads, positive VS selection columns and a Vario Magnet according to the manufacturer's instractions. CD45RA and CD45RO CD4 lymphocytes were isolated by depleting mononuclear cells of CD8, CD56, CD14 and CD19 cells using CD8, CD56, CD14 and CD19 Miltenyi beads and positive selection. CD45RO beads were then used to isolate the CD45RO CD4 lymphocytes with the remaining cells being CD45RA CD4 lymphocytes. CD45RA CD4, CD45RO CD4 and CD8 lymphocytes were placed in DMEM 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5xlO"5M (Gibco), and lOmM Hepes (Gibco) and plated at 106cells/ml onto Falcon 6 well tissue culture plates that had been coated overnight with 0.5μg/ml anti-CD28 (Pharmingen) and 3ug/ml anti-CD3 (OKT3, ATCC) in PBS. After 6 and 24 hours, the cells were harvested for RNA preparation. To prepare chronically activated CD8 lymphocytes, we activated the isolated CD8 lymphocytes for 4 days on anti-CD28 and anti-CD3 coated plates and then harvested the cells and expanded them in DMEM 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5xlO"5M (Gibco), and lOmM Hepes (Gibco) and IL-2. The expanded CD8 cells were then activated again with plate bound anti-CD3 and anti-CD28 for 4 days and expanded as before. RNA was isolated 6 and 24 hours after the second activation and after 4 days of the second expansion culture. The isolated NK cells were cultured in DMEM 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5xlO"5M (Gibco), and lOmM Hepes (Gibco) and IL-2 for 4-6 days before RNA was prepared.
To obtain B cells, tonsils were procured from NDRI. The tonsil was cut up with sterile dissecting scissors and then passed through a sieve. Tonsil cells were then spun down and resupended at 106cells/ml in DMEM 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5xlO"5M (Gibco), and lOmM Hepes (Gibco). To activate the cells, we used PWM at 5μg/ml or anti-CD40 (Pharmingen) at approximately lOμg/ml and IL-4 at 5-lOng/ml. Cells were harvested for RNA preparation at 24,48 and 72 hours.
To prepare the primary and secondary Thl/Th2 and Tri cells, six-well Falcon plates were coated overnight with lOμg/ml anti-CD28 (Pharmingen) and 2μg/ml OKT3 (ATCC), and then washed twice with PBS. Umbilical cord blood CD4 lymphocytes (Poietic Systems, German Town, MD) were cultured at 105-106cells/ml in DMEM 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5x10" 5M (Gibco), lOmM Hepes (Gibco) and IL-2 (4ng/ml). IL-12 (5ng/ml) and anti-IL4 (1 μg/ml) were used to direct to Thl, while IL-4 (5ng ml) and anti-IFN gamma (1 μg/ml) were used to direct to Th2 and IL-10 at 5ng/ml was used to direct to Tri. After 4-5 days, the activated Thl, Th2 and Tri lymphocytes were washed once in DMEM and expanded for 4-7 days in DMEM 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5xl0"5M (Gibco), lOmM Hepes (Gibco) and JL-2 (lng/ml). Following this, the activated Thl, Th2 and Tri lymphocytes were re-stimulated for 5 days with anti-CD28/OKT3 and cytokines as described above, but with the addition of anti-CD95L (1 μg/ml) to prevent apoptosis. After 4-5 days, the Thl, Th2 and Tri lymphocytes were washed and then expanded again with IL-2 for 4-7 days. Activated Thl and Th2 lymphocytes were maintained in this way for a maximum of three cycles. RNA was prepared from primary and secondary Thl, Th2 and Tri after 6 and 24 hours following the second and third activations with plate bound anti-CD3 and anti-CD28 mAbs and 4 days into the second and third expansion cultures in Interleukin 2.
The following leukocyte cells lines were obtained from the ATCC: Ramos, EOL-1, KU-812. EOL cells were further differentiated by culture in O.lmM dbcAMP at 5xl05cells/ml for 8 days, changing the media every 3 days and adjusting the cell concentration to 5xl05cells/ml. For the culture of these cells, we used DMEM or RPMI (as recommended by the ATCC), with the addition of 5% FCS (Hyclone), 1 OOμM non essential amino acids
(Gibco), ImM sodium pyravate (Gibco), mercaptoethanol 5.5xl0"5M (Gibco), lOmM Hepes (Gibco). RNA was either prepared from resting cells or cells activated with PMA at lOng/ml and ionomycin at 1 μg ml for 6 and 14 hours. Keratinocyte line CCD106 and an airway epithelial tumor line NCI-H292 were also obtained from the ATCC. Both were cultured in DMEM 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyravate (Gibco), mercaptoethanol 5.5xlO"5M (Gibco), and lOmM Hepes (Gibco). CCD1106 cells were activated for 6 and 14 hours with approximately 5 ng/ml TNF alpha and 1 ng/ml IL-1 beta, while NCI-H292 cells were activated for 6 and 14 hours with the following cytokines: 5ng/ml IL-4, 5ng/ml IL-9, 5ng/ml IL-13 and 25ng/ml TEN gamma.
For these cell lines and blood cells, RNA was prepared by lysing approximately
107cells/ml using Trizol (Gibco BRL). Briefly, 1/10 volume of bromochloropropane (Molecular Research Coφoration) was added to the RNA sample, vortexed and after 10 minutes at room temperature, the tubes were spun at 14,000 φm in a Sorvall SS34 rotor. The aqueous phase was removed and placed in a 15ml Falcon Tube. An equal volume of isopropanol was added and left at -20°C overnight. The precipitated RNA was spun down at 9,000 rpm for 15 min in a Sorvall SS34 rotor and washed in 70% ethanol. The pellet was redissolved in 300μl of RNAse-free water and 35μl buffer (Promega) 5μl DTT, 7μl RNAsin and 8μl DNAse were added. The tube was incubated at 37°C for 30 minutes to remove contaminating genomic DNA, extracted once with phenol chloroform and re-precipitated with 1/10 volume of 3M sodium acetate and 2 volumes of 100% ethanol. The RNA was spun down and placed in RNAse free water. RNA was stored at -80°C.
AI_comprehensive panel_vl.O The plates for AI_comprehensive panel_vl.O include two control wells and 89 test samples comprised of cDNA isolated from surgical and postmortem human tissues obtained from the Backus Hospital and Clinomics (Frederick, MD). Total RNA was extracted from tissue samples from the Backus Hospital in the Facility at CuraGen. Total RNA from other tissues was obtained from Clinomics.
Joint tissues including synovial fluid, synovium, bone and cartilage were obtained from patients undergoing total knee or hip replacement surgery at the Backus Hospital. Tissue samples were immediately snap frozen in liquid nitrogen to ensure that isolated RNA was of optimal quality and not degraded. Additional samples of osteoarthritis and rheumatoid arthritis joint tissues were obtained from Clinomics. Normal control tissues were supplied by Clinomics and were obtained during autopsy of trauma victims.
Surgical specimens of psoriatic tissues and adjacent matched tissues were provided as total RNA by Clinomics. Two male and two female patients were selected between the ages of 25 and 47. None of the patients were taking prescription drugs at the time samples were isolated.
Surgical specimens of diseased colon from patients with ulcerative colitis and Crohns disease and adjacent matched tissues were obtained from Clinomics. Bowel tissue from three female and three male Crohn's patients between the ages of 41-69 were used. Two patients were not on prescription medication while the others were taking dexamethasone, phenobarbital, or tylenol. Ulcerative colitis tissue was from three male and four female patients. Four of the patients were taking lebvid and two were on phenobarbital.
Total RNA from post mortem lung tissue from trauma victims with no disease or with emphysema, asthma or COPD was purchased from Clinomics. Emphysema patients ranged in age from 40-70 and all were smokers, this age range was chosen to focus on patients with cigarette-linked emphysema and to avoid those patients with alpha-1 anti-trypsin deficiencies. Asthma patients ranged in age from 36-75, and excluded smokers to prevent those patients that could also have COPD. COPD patients ranged in age from 35-80 and included both smokers and non-smokers. Most patients were taking corticosteroids, and bronchodilators.
In the labels employed to identify tissues in the AI_comprehensive panel_vl.0 panel, the following abbreviations are used: Al = Autoimmunity Syn = Synovial Normal = No apparent disease Rep22 /Rep20 = individual patients RA = Rheumatoid arthritis
Backus = From Backus Hospital OA = Osteoarthritis (SS) (BA) (MF) = Individual patients Adj = Adjacent tissue Match control = adjacent tissues
-M = Male -F = Female COPD = Chronic obstructive pulmonary disease
Panels 5D and 51
The plates for Panel 5D and 51 include two control wells and a variety of cDNAs isolated from human tissues and cell lines with an emphasis on metabolic diseases. Metabolic tissues were obtained from patients enrolled in the Gestational Diabetes study. Cells were obtained during different stages in the differentiation of adipocytes from human mesenchymal stem cells. Human pancreatic islets were also obtained.
In the Gestational Diabetes study subjects are young (18 - 40 years), otherwise healthy women with and without gestational diabetes undergoing routine (elective) Caesarean section. After delivery of the infant, when the surgical incisions were being repaired/closed, the obstetrician removed a small sample (<1 cc) of the exposed metabolic tissues during the closure of each surgical level. The biopsy material was rinsed in sterile saline, blotted and fast frozen within 5 minutes from the time of removal. The tissue was then flash frozen in liquid nitrogen and stored, individually, in sterile screw-top tubes and kept on dry ice for shipment to or to be picked up by CuraGen. The metabolic tissues of interest include uterine wall (smooth muscle), visceral adipose, skeletal muscle (rectus) and subcutaneous adipose. Patient descriptions are as follows:
Patient 2: Diabetic Hispanic, overweight, not on insulin
Patient 7-9: Nondiabetic Caucasian and obese (BMI>30)
Patient 10: Diabetic Hispanic, overweight, on insulin Patient 11: Nondiabetic African American and overweight Patient 12: Diabetic Hispanic on insulin
Adipocyte differentiation was induced in donor progenitor cells obtained from Osiras (a division of Clonetics/BioWhittaker) in triplicate, except for Donor 3U which had only two replicates. Scientists at Clonetics isolated, grew and differentiated human mesenchymal stem cells (HuMSCs) for CuraGen based on the published protocol found in Mark F. Pittenger, et al, Multilineage Potential of Adult Human Mesenchymal Stem Cells Science Apr 2 1999: 143-147. Clonetics provided Trizol lysates or frozen pellets suitable for mRNA isolation and ds cDNA production. A general description of each donor is as follows:
Donor 2 and 3 U: Mesenchymal Stem cells, Undifferentiated Adipose
Donor 2 and 3 AM: Adipose, AdiposeMidway Differentiated Donor 2 and 3 AD: Adipose, Adipose Differentiated
Human cell lines were generally obtained from ATCC (American Type Culture Collection), NCI or the German tumor cell bank and fall into the following tissue groups: kidney proximal convoluted tubule, uterine smooth muscle cells, small intestine, liver HepG2 cancer cells, heart primary stromal cells, and adrenal cortical adenoma cells. These cells are all cultured under standard recommended conditions and RNA extracted using the standard procedures. All samples were processed at CuraGen to produce single stranded cDNA.
Panel 51 contains all samples previously described with the addition of pancreatic islets from a 58 year old female patient obtained from the Diabetes Research Institute at the University of Miami School of Medicine. Islet tissue was processed to total RNA at an outside source and delivered to CuraGen for addition to panel 51.
In the labels employed to identify tissues in the 5D and 51 panels, the following abbreviations are used:
GO Adipose = Greater Omentum Adipose
SK = Skeletal Muscle UT = Uterus PL = Placenta AD = Adipose Differentiated AM = Adipose Midway Differentiated U = Undifferentiated Stem Cells
Panel CNSD.01
The plates for Panel CNSD.01 include two control wells and 94 test samples comprised of cDNA isolated from postmortem human brain tissue obtained from the Harvard Brain Tissue Resource Center. Brains are removed from calvaria of donors between 4 and 24 hours after death, sectioned by neuroanatomists, and frozen at -80°C in liquid nitrogen vapor. All brains are sectioned and examined by neuropathologists to confirm diagnoses with clear associated neuropathology.
Disease diagnoses are taken from patient records. The panel contains two brains from each of the following diagnoses: Alzheimer's disease, Parkinson's disease, Huntington's disease, Progressive Supernuclear Palsy, Depression, and "Normal controls". Within each of these brains, the following regions are represented: cingulate gyrus, temporal pole, globus palladus, substantia nigra, Brodman Area 4 (primary motor strip), Brodman Area 7 (parietal cortex), Brodman Area 9 (prefrontal cortex), and Brodman area 17 (occipital cortex). Not all brain regions are represented in all cases; e.g., Huntington's disease is characterized in part by neurodegeneration in the globus palladus, thus this region is impossible to obtain from confirmed Huntington's cases. Likewise Parkinson's disease is characterized by degeneration of the substantia nigra making this region more difficult to obtain. Normal control brains were examined for neuropathology and found to be free of any pathology consistent with neurodegeneration.
In the labels employed to identify tissues in the CNS panel, the following abbreviations are used:
PSP = Progressive supranuclear palsy Sub Nigra = Substantia nigra
Glob Palladus= Globus palladus Temp Pole = Temporal pole Cing Gyr = Cingulate gyrus BA 4 = Brodman Area 4
Panel CNS_Neurodegeneration_V1.0 The plates for Panel CNS_Neurodegeneration_V1.0 include two control wells and 47 test samples comprised of cDNA isolated from postmortem human brain tissue obtained from the Harvard Brain Tissue Resource Center (McLean Hospital) and the Human Brain and Spinal Fluid Resource Center (VA Greater Los Angeles Healthcare System). Brains are removed from calvaria of donors between 4 and 24 hours after death, sectioned by neuroanatomists, and frozen at -80°C in liquid nitrogen vapor. All brains are sectioned and examined by neuropathologists to confirm diagnoses with clear associated neuropathology.
Disease diagnoses are taken from patient records. The panel contains six brains from Alzheimer's disease (AD) patients, and eight brains from "Normal controls" who showed no evidence of dementia prior to death. The eight normal confrol brains are divided into two categories: Controls with no dementia and no Alzheimer's like pathology (Controls) and controls with no dementia but evidence of severe Alzheimer's like pathology, (specifically senile plaque load rated as level 3 on a scale of 0-3; 0 = no evidence of plaques, 3 = severe AD senile plaque load). Within each of these brains, the following regions are represented: hippocampus, temporal cortex (Brodman Area 21), parietal cortex (Brodman area 7), and occipital cortex (Brodman area 17). These regions were chosen to encompass all levels of neurodegeneration in AD. The hippocampus is a region of early and severe neuronal loss in AD; the temporal cortex is known to show neurodegeneration in AD after the hippocampus; the parietal cortex shows moderate neuronal death in the late stages of the disease; the occipital cortex is spared in AD and therefore acts as a "control" region within AD patients. Not all brain regions are represented in all cases.
In the labels employed to identify tissues in the CNS_Neurodegeneration_V1.0 panel, the following abbreviations are used:
AD = Alzheimer's disease brain; patient was demented and showed AD-like pathology upon autopsy
Control = Control brains; patient not demented, showing no neuropathology
Control (Path) = Control brains; pateint not demented but showing sever AD-like pathology
SupTemporal Ctx = Superior Temporal Cortex frif Temporal Ctx = Inferior Temporal Cortex
A. NOV2 (CG93210-01: Plasma membrane ring finger protein ) Expression of NON2 gene (CG93210-01) was assessed using the primer-probe set Ag3845, described in Table AA. Results of the RTQ-PCR runs are shown in Tables AB, AC, AD and AE.
Table AA. Probe Name Ag3845
Figure imgf000212_0001
Table AB. CNS_neurodegeneration_vl.O
Figure imgf000212_0002
Figure imgf000213_0001
Table AC. General_screenmgjpanel_vl.4
Figure imgf000213_0002
Figure imgf000214_0001
Table AD. Panel 2.1
Tissue Name ReI.Exp.(%) I Tissue Name I Rel.Exp.(%)
Figure imgf000215_0001
Figure imgf000216_0001
Table AE. Panel 4. ID
Figure imgf000216_0002
Figure imgf000217_0001
Figure imgf000218_0001
CNS_neurodegeneration_vl.0 Summary: Ag3845 This panel does not show differential expression of the CG93210-01 gene in Alzheimer's disease. However, this expression profile confirms the presence of this gene in the brain. Please see Panel 1.4 for discussion of utility of this gene in the central nervous system.
General_screening_panel_vl.4 Summary: Ag3845 Highest expression of the CG93210-01 gene is seen in abreast cancer cell line-(CT=26.1). In addition, significant levels of expression are seen in a cluster of samples derived from brain, ovarian, breast and lung cancer cell lines. Thus, expression of this gene could be used to differentiate between these samples and other samples on this panel and as a marker to detect the presence of these cancers. This protein contains a domain that is homologous to a ring finger domain that is though to have intrinsic function as a ubiquituin ligase. Ubiquituin ligase activity of BRCA1 is thought to be important in the prevention of breast and ovarian cancers. Therefore, therapeutic modulation of the expression or function of this gene may be effective in the treatment of ovarian, brain, breast and lung cancers.
Among tissues with metabolic function, this gene is expressed at moderate to low levels in pituitary, adipose, adrenal gland, pancreas, thyroid, and adult and fetal skeletal muscle, heart, and liver. This widespread expression among these tissues suggests that this gene product may play a role in normal neuroendocrine and metabolic and that disregulated expression of this gene may contribute to neuroendocrine disorders or metabolic diseases, such as obesity and diabetes. In addition, this gene is expressed at much higher levels in fetal liver (CT=29) when compared to expression in adult liver (CT=34). Thus, expression of this gene may be used to differentiate between the fetal and adult source of these tissue.
This molecule is also expressed at moderate levels in the CNS, including the hippocampus, thalamus, substantia nigra and cerebral cortex. Therefore, therapeutic modulation of the expression or function of this gene may be useful in the treatment of neurologic disorders, such as Alzheimer's disease, Parkinson's disease, schizophrenia, multiple sclerosis, stroke and epilepsy.
Overall, the ubiquitous expression of this gene suggests a wider role for the coπesponding protein product in cell function.
References:
Hashizume R, Fukuda M, Maeda I, Nishikawa H, Oyake D, Yabuki Y, Ogata H, Ohta T. The RING heterodimer BRCAl-BARDl is a ubiquitin ligase inactivated by a breast cancer- derived mutation. J Biol Chem 2001 May 4;276(18): 14537-40
BRCAl-BARDl constitutes a heterodimeric RING finger complex associated through its N- terminal regions. Here we demonstrate that the BRCAl-BARDl heterodimeric RING finger complex contains sigmficant ubiquitin ligase activity that can be disrupted by a breast cancer- derived RING finger mutation in BRCA1. Whereas individually BRCA1 and BARDl have very low ubiquitin ligase activities in vitro, BRCA1 combined with BARDl exhibits dramatically higher activity. Bacterially purified RING finger domains comprising residues 1- 304 of BRCA1 and residues 25-189 of BARDl are capable of polymerizing ubiquitin. The steady-state level of transfected BRCA1 in vivo was increased by co-transfection of BARDl, and reciprocally that of transfected BARDl was increased by BRCA1 in a dose-dependent manner. The breast cancer-derived BARDl -interaction-deficient mutant, BRCA1(C61G), does not exhibit ubiquitin ligase activity in vitro. These results suggest that the BRCAl-BARDl complex contains a ubiquitin ligase activity that is important in prevention of breast and ovarian cancer development. PMID: 11278247
Panel 2.1 Summary: Ag3845 Highest expression of the CG93210-01 gene is seen in normal kidney (CT=30.3). There is also higher expression in a kidney tumor when compared to the coπesponding matched normal tissue. Overall, this gene is widely expressed in this panel, consistent with expression in the previous panels. Thus, expression of this gene could be used to differentiate these samples from other samples on this panel. Furthermore, therapeutic modulation of the expression or function of this gene may be effective in the treatment of kidney cancer.
Panel 4.1D Summary: Ag3845 This gene is expressed in most of the samples on this panel, with highest expression of the CG93210-10 gene in untreated lung microvascular endothelial cells. This gene is also expressed at moderate to low levels in a wide range of cell types of significance in the immune response in health and disease. These cells include members of the T-cell, B-cell, endothelial cell, macrophage/monocyte, and peripheral blood mononuclear cell family, as well as epithelial and fibroblast cell types from lung and skin, and normal tissues represented by colon, lung, thymus and kidney. This ubiquitous pattern of expression suggests that this gene product may be involved in homeostatic processes for these and other cell types and tissues. This pattern is in agreement with the expression profile in General_screening_panel_vl.4 and also suggests a role for the gene product in cell survival and proliferation. Therefore, modulation of the gene product with a functional therapeutic may lead to the alteration of functions associated with these cell types and lead to improvement of the symptoms of patients suffering from autoimmune and inflammatory diseases such as asthma, allergies, inflammatory bowel disease, lupus erythematosus, psoriasis, rheumatoid arthritis, and osteoarthritis.
B. NOV4 (CG93187-01): PROTOCADHERIN ALPHA C2 SHORT FORM PROTEIN
Expression of gene CG93187-01 was assessed using the primer-probe set Ag3844, described in Table BA. Results of the RTQ-PCR runs are shown in Tables BB, BC and BD.
Table BA. Probe Name Ag3844
Figure imgf000220_0001
Reverse 5'-ctgtcccactggttctcagtat-3' (Seq ID NO: 119) 22 2357
Table BB. CNS_neurodegeneration_vl.O
Figure imgf000221_0001
Temporal Ctx Parietal Ctx
Table BC. Panel 2.1
Figure imgf000222_0001
Figure imgf000223_0001
Table BD. Panel 4. ID
Figure imgf000223_0002
Figure imgf000224_0001
Figure imgf000225_0001
CΝS_neurodegeneration_vl.O Summary: Ag3844 The CG93187-01 gene, a protocadherin homolog, is detected at low levels in the CNS, with highest expression in the hippocampus of an Alzheimer's patient. While this gene shows no differential expression between the brains of Alzheimer's patients and controls, this expression profile suggests a role for this gene in the CNS. The cadherins have been shown to be critical for CNS development, specifically for the guidance of axons, dendrites and/or growth cones in general. Therapeutic modulation of the levels of this protein, or possible signaling via this protein may be of utility in enhancing/directing compensatory synaptogenesis and fiber growth in the CNS in response to neuronal death (stroke, head trauma), axon lesion (spinal cord injury), or neurodegeneration (Alzheimer's, Parkinson's, Huntington's, vascular dementia or any neurodegenerative disease). Since protocadherins play an important role in synaptogenesis this gene product may also be involved in depression, schizophrenia, which also involve synaptogeneisis.
References: Hilschmann N, Barnikol HU, Barnikol-Watanabe S, Gotz H, Kratzin H, Thinnes FP. The immunoglobulin-like genetic predetermination of the brain: the protocadherins, blueprint of the neuronal network. Naturwissenschaften 2001 Jan;88(l):2-12
The morphogenesis of the brain is governed by synaptogenesis. Synaptogenesis in turn is determined by cell adhesion molecules, which bridge the synaptic cleft and, by homophilic contact, decide which neurons are connected and which are not. Because of their enormous diversification in specificities, protocadherins (pcdh alpha, pcdh beta, pcdh gamma), a new class of cadherins, play a decisive role. Surprisingly, the genetic confrol of the protocadherins is very similar to that of the immunoglobulins. There are three sets of variable (V) genes followed by a coπesponding constant (C) gene. Applying the rules of the immunoglobulin genes to the protocadherin genes leads, despite of this similarity, to quite different results in the central nervous system. The lymphocyte expresses one single receptor molecule specifically directed against an outside stimulus. In contrast, there are three specific recognition sites in each neuron, each expressing a different protocadherin. In this way, 4,950 different neurons arising from one stem cell form a neuronal network, in which homophilic contacts can be formed in 52 layers, permitting an enormous number of different connections and restraints between neurons. This network is one module of the central computer of the brain. Since the V-genes are generated during evolution and V-gene translocation during embryogenesis, outside stimuli have no influence on this network. The network is an inborn property of the protocadherin genes. Every circuit produced, as well as learning and memory, has to be based on this genetically predetermined network. This network is so universal that it can cope with everything, even the unexpected. In this respect the neuronal network resembles the recognition sites of the immunoglobulins.
General_screening_panel_vl.4 Summary: Ag3844 Results from one experiment with the CG93187-01 gene are not included. The amp plot indicates that there were experimental difficulties with this run.
Panel 2.1 Summary: Ag3844 Significant expression of the CG93187-01 gene is restricted to the lung in this panel (CTs=34.5-35). Thus, expression of this gene could be used to differentiate between lung derived tissue and other samples on this panel and as a marker to detect the presence of lung cancer. Furthermore, therapeutic modulation of the expression or function of this gene may be effective in the treatment of lung cancer. Panel 4.1D Summary: Ag3844 Highest expression of the CG93187-01 gene, a protocadherin alpha homolog, is seen in secondary Thl/TH2/Trl cells treated with anti-CD95 (CT=30.5). Overall, expression appears to be higher in hematopoietically derived samples when compared to expression in fibroblasts and endothelial cells. Detection in LAK cells suggests that modulation of the function of this gene product may also lead to improvement of symptoms associated with tumor immunology and tumor cell clearance, as well as removal of virally and bacterial infected cells. In addition, the gene product could also potentially be used therapeutically in the treatment of asthma, emphysema, IBD, lupus or arthritis and in other diseases in which T cells and B cells are activated.
C. NOV5 (CG95083-01): Novel Nuclear Protein
Expression of gene CG95083-01 was assessed using the primer-probe set Ag3918, described in Table CA. Results of the RTQ-PCR runs are shown in Tables CB, CC and CD.
Table CA. Probe Name Ag3918
Figure imgf000227_0001
Table CB. CNS_neurodegeneration_vl .0
Figure imgf000227_0002
Figure imgf000228_0001
Table CC. General_screening_panel_vl.4
Figure imgf000228_0002
Figure imgf000229_0001
Figure imgf000230_0001
Table CD. Panel 4. ID
Figure imgf000230_0002
Figure imgf000231_0001
CNS_neurodegeneration_vl.O Summary: Ag3918 This panel does not show differential expression of the CG95083-01 gene in Alzheimer's disease. However, this expression profile confirms the presence of this gene in the brain. Please see Panel 1.4 for discussion of utility of this gene in the central nervous system.
General_screening_panel_vl.4 Summary: Ag3918 Highest expression of the CG95083-01 gene is seen in the fetal lung (CT=26.5). In addition, this gene is expressed at much higher levels in fetal lung when compared to expression in the adult counterpart (CT=31.3). Thus, expression of this gene may be used to differentiate between the fetal and adult source of this tissue.
hi addition, significant levels of expression are seen in a samples derived from colon and lung cancer cell lines. Thus, expression of this gene could be used to differentiate between these samples and other samples on this panel and as a marker to detect the presence of these cancers. Furthermore, therapeutic modulation of the expression or function of this gene may be effective in the treatment of colon and lung cancers.
Among tissues with metabolic function, this gene is expressed at moderate to low levels in pituitary, adipose, adrenal gland, pancreas, thyroid, and adult and fetal skeletal muscle, heart, and liver. This widespread expression among these tissues suggests that this gene product may play a role in normal neuroendocrine and metabolic and that disregulated expression of this gene may contribute to neuroendocrine disorders or metabolic diseases, such as obesity and diabetes.
This molecule is also expressed at low levels in the CNS, including the hippocampus, amygdala, cerebellum, thalamus, substantia nigra and cerebral cortex. Therefore, therapeutic modulation of the expression or function of this gene may be useful in the treatment of neurologic disorders, such as Alzheimer's disease, Parkinson's disease, schizophrenia, multiple sclerosis, stroke and epilepsy.
Panel 4.1D Summary: Ag3918 Highest expression of the CG95083-01 gene is seen in untreated lung microvascular endothelial cells (CT=26.6). This gene is expressed consistently in endothelium samples including HPAEC, HUVEC and lung and dermal microvascular EC.
Therefore, expression of this gene could be used as a marker of endothelial cells. Furthermore, therapies designed with the protein encoded by this transcript could be important in the regulating endothelium function including leukocyte extravasation, a major component of inflammation during asthma, IBD, and psoriasis.
D. NOV6 (CG94989-01): Novel Secretory Protein
Expression of gene CG94989-01 was assessed using the primer-probe set Ag3980, described in Table DA. Results of the RTQ-PCR runs are shown in Tables DB, DC, DD and DE.
Table DA. Probe Name Ag3980
Figure imgf000233_0001
Table DB. CNS_neurodegeneration_vl.O
Figure imgf000233_0002
Figure imgf000234_0001
Table DC. General_screening_panel_vl.4
Figure imgf000234_0002
Figure imgf000235_0001
Table DD. Panel 2.1
Figure imgf000236_0001
Figure imgf000237_0001
Table DE. Panel 4. ID
Figure imgf000237_0002
Figure imgf000238_0001
Figure imgf000239_0001
CNS_neurodegeneration_vl.O Summary: Ag3980 This panel does not show differential expression of the CG94989-01 gene in Alzheimer's disease. However, this expression profile confirms the presence of this gene in the brain. Please see Panel 1.4 for discussion of utility of this gene in the central nervous system.
General_screening_panel_vl.4 Summary: Ag3980 Highest expression of the CG94989-01 gene is seen in the cerebellum (CT=25.8). In addition, expression of this gene appears to be highly brain preferential, with high to moderate levels of expression in all regions of the CNS examined. Therefore, therapeutic modulation of the expression or function of this gene may be useful in the treatment of neurologic disorders, such as Alzheimer's disease, Parkinson's disease, schizophrenia, multiple sclerosis, stroke and epilepsy.
Among tissues with metabolic function, this gene is expressed at moderate to low levels in pituitary, adipose, adrenal gland, pancreas, thyroid, and adult and fetal skeletal muscle and heart, and fetal liver. This widespread expression among these tissues suggests that this gene product may play a role in normal neuroendocrine and metabolic and that disregulated expression of this gene may contribute to neuroendocrine disorders or metabolic diseases, such as obesity and diabetes.
In addition, this gene is expressed at much higher levels in fetal liver (CT=31) when compared to expression in the adult counterpart (CT=35). Thus, expression of this gene may be used to differentiate between the fetal and adult source of these tissue. Furthermore, the higher levels of expression in fetal liver suggest a role for this protein product in the development of the organ. Therefore, therapeutic modulation of the expression or function of this gene may help in the regeneration of the liver in the adult and in the treatment of diseases that affect the liver such as Von Hippel-Lindau (VHL) syndrome and ciπhosis.
In addition, there is significant expression in a sample derived from a prostate cancer cell line (CT=26.5) and a cluster of renal cancer cell lines. Thus, expression of this gene could be used to differentiate between the prostate cancer cell line sample and other samples on this panel and as a marker to detect the presence of prostate cancer. Furthermore, therapeutic modulation of the expression or function of this gene may be effective in the treatment of prostate and kidney cancers.
Panel 2.1 Summary: Ag3980 Highest expression of the CG94989-01 gene is seen in a kidney cancer (CT=30.7). Futhermore, expression is higher in kidney, lung and liver cancers when compared to expression in normal adjacent tissue. Conversely, expression is higher in colon tissue than in the corresponding tumor samples. Thus, expression of this gene could be used to differentiate between these samples and other samples on this panel and as a marker to detect the presence of these cancers. Furthermore, therapeutic modulation of the expression or function of this gene may be effective in the treatment of colon, kidney, lung and liver cancers.
Panel 4.1D Summary: Ag3980 Highest expression of the CG94989-01 gene is seen in untreated pulmonary aortic endothelial cells (CT=30.6). Significant expression is also seen in a cluster of samples derived from HUVEC endothelial cells. Thus, expression of this gene could be used to differentiate these endothelial cells from other samples on this panel. Furthermore, this expression profile suggests that this gene product may be involved in endothelial cell function. Therefore, therapeutic modulation of the gene product may reduce or eliminate the symptoms in patients with autoimmune and inflammatory diseases in which endothelial cells are involved, such as lupus erythematosus, asthma, emphysema, Crohn's disease, ulcerative colitis, rheumatoid arthritis, osteoarthritis, and psoriasis. E. NOV7 (CG94978-01): TRANSMISSION- BLOCKING TARGET ANTIGEN S230 PRECURSOR
Expression of gene CG94978-01 was assessed using the primer-probe set Ag3977, described in Table AA. Results of the RTQ-PCR runs are shown in Tables EB, EC and ED.
Table EA. Probe Name Ag3977
Figure imgf000241_0001
Table EB. CNS_neurodegeneration_vl.0
Figure imgf000241_0002
Figure imgf000242_0001
Table EC. General_screening_panel_vl.4
Figure imgf000242_0002
Figure imgf000243_0001
Table ED. Panel 4. ID
Figure imgf000244_0001
Figure imgf000245_0001
CNS_neurodegeneration_vl.O Summary: Ag3977 This panel does not show differential expression of the CG94978-01 gene in Alzheimer's disease. However, this expression profile confirms the presence of this gene in the brain. Please see Panel 1.4 for discussion of utility of this gene in the central nervous system.
General_screening_panel_vl.4 Summary: Ag3977 Highest expression of the CG94978-01 gene is seen in a gastric cancer cell line (CT=25.3). Overall, this gene appears to show a moderate association with cancer cell lines, when compared to expression in normal tissue samples. Thus, expression of this gene could be used as a marker for the presence of cancer. Furthermore, therapeutic modulation of the expression or function of this gene may be useful in the treatment of cancer. Among tissues with metabolic function, this gene is expressed at high to moderate levels in pituitary, adipose, adrenal gland, pancreas, thyroid, and adult and fetal skeletal muscle, heart, and liver. This widespread expression among these tissues suggests that this gene product may play a role in normal neuroendocrine and metabolic and that disregulated expression of this gene may contribute to neuroendocrine disorders or metabolic diseases, such as obesity and diabetes.
This molecule is also expressed at high to moderate levels in the CNS, including the hippocampus, amygdala, cerebellum, thalamus, substantia nigra and cerebral cortex. Therefore, therapeutic modulation of the expression or function of this gene maybe useful in the treatment of neurologic disorders, such as Alzheimer's disease, Parkinson's disease, schizophrenia, multiple sclerosis, stroke and epilepsy.
Panel 4.1D Summary: Ag3977 Highest expression of the CG94978-01 gene is seen in LPS stimulated monocytes (CT=29.7). In addition, this gene is expressed at high to moderate levels in a wide range of cell types of significance in the immune response in health and disease. These cells include members of the T-cell, B-cell, endothelial cell, macrophage/monocyte, and peripheral blood mononuclear cell family, as well as epithelial and fibroblast cell types from lung and skin, and normal tissues represented by colon, lung, thymus and kidney. This ubiquitous pattern of expression suggests that this gene product may be involved in homeostatic processes for these and other cell types and tissues. This pattern is in agreement with the expression profile in General_screening_panel_vl .5 and also suggests a role for the gene product in cell survival and proliferation. Therefore, modulation of the gene product with a functional therapeutic may lead to the alteration of functions associated with these cell types and lead to improvement of the symptoms of patients suffering from autoimmune and inflammatory diseases such as asthma, allergies, inflammatory bowel disease, lupus erythematosus, psoriasis, rheumatoid arthritis, and osteoarthritis.
F. NOV8 (CG94713-01): nuclear protein
Expression of gene CG94713-01 was assessed using the primer-probe sets Ag3945 and Ag4790, described in Tables FA and FB. Results of the RTQ-PCR runs are shown in Tables FC, FD, FE and FF.
Table FA. Probe Name Ag3945
Figure imgf000247_0001
Table FB. Probe Name Ag4790
Primers Sequences Length Start Position
Forward 5'-ttatcaaaagctggcaccat-3' (Seq ID NO: 132) 20 1039
Probe fTE-T-S'-aagggcattcatcatcaataagacaa-S'-TAMRA (Seq ID NO: 133) 26 1011
Reverse 5'-aaggaaatgggccaaaatc-3' (Seq ID NO: 134) 19 973
Table FC. CNS_neurodegeneration_vl.0
Figure imgf000247_0002
Figure imgf000248_0001
Table FD. General_screening_panel_vl.4
Figure imgf000248_0002
Figure imgf000249_0001
Figure imgf000250_0001
Table FE. Panel 2.1
Figure imgf000250_0002
Figure imgf000251_0001
Figure imgf000252_0001
Table FF. Panel 4. ID
Figure imgf000252_0002
Figure imgf000253_0001
Figure imgf000254_0001
CNS_neurodegeneration_vl.0 Summary: Ag3945 This panel does not show differential expression of the CG94713-01 gene in Alzheimer's disease. However, this expression profile confirms the presence of this gene in the brain. Please see Panel 1.4 for discussion of utility of this gene in the central nervous system.
General_screening_panel_vl.4 Summary: Ag3945/Ag4790 Two experiments with two different probe and primer sets produce results that are in excellent agreement, with highest expression of the CG94713-01 gene in a lung cancer cell line (CTs=24-30). In addition, significant levels of expression are seen in a cluster of samples derived from brain, colon, gastric, ovarian, and melanoma cancer cell lines. Thus, expression of this gene could be used to differentiate between these samples and other samples on this panel and as a marker to detect the presence of these cancers. Furthermore, therapeutic modulation of the expression or function of this gene may be effective in the treatment of brain, colon, gastric, ovarian, melanoma, and lung cancers.
Among tissues with metabolic function, the region of the gene corresponding to the Ag4890 probe and primer set is seen at high to moderate in pituitary, adipose, adrenal gland, pancreas, thyroid, and adult and fetal skeletal muscle, heart, and liver. This widespread expression among these tissues suggests that this gene product may play a role in normal neuroendocrine and metabolic and that disregulated expression of this gene may contribute to neuroendocrine disorders or metabolic diseases, such as obesity and diabetes. In contrast, the Ag3945 probe and primer set showed lower levels of expression of this gene in pancreas, fetal heart and fetal liver (CTs=33-35). Expression in the other metabolic samples is low/undetectable.
The gene is also expressed at moderate to low levels in all regions of the CNS examined, including the hippocampus, amygdala, cerebellum, thalamus, substantia nigra and cerebral cortex. Therefore, therapeutic modulation of the expression or function of this gene may be useful in the treatment of neurologic disorders, such as Alzheimer's disease, Parkinson's disease, schizophrenia, multiple sclerosis, stroke and epilepsy.
Panel 2.1 Summary: Ag3945 Highest expression of the CG94713-01 gene is seen in a kidney cancer sample (CT=32.1). Significant expression is also seen in normal uterine, breast, stomach and bladder. Thus, expression of this gene could be used to as a marker to detect the presence of these cancers. Furthermore, therapeutic modulation of the expression or function of this gene may be effective in the treatment of uterine, breast, stomach and bladder cancers.
Panel 4.1D Summary: Ag3945/Ag4790 Two experiments with two different probe and primer sets produce results that are in excellent agreement, with highest expression of the CG94713-01 gene in IL-9 and IFN-gamma treated NCI-H292 cells (CTs=28-32). The gene is also expressed in a cluster of treated and untreated samples derived from the NCI-H292 cell line, a human airway epithelial cell line that produces mucins. Mucus overproduction is an important feature of bronchial asthma and chronic obstructive pulmonary disease samples. The transcript is also expressed at lower but still significant levels in small airway epithelium treated with IL-1 beta and TNF-alpha. The expression of the transcript in this mucoepidermoid cell line that is often used as a model for airway epithelium (NCI-H292 cells) suggests that this transcript may be important in the proliferation or activation of airway epithelium.
Therefore, therapeutics designed with the protein encoded by the transcript may reduce or eliminate symptoms caused by inflammation in lung epithelia in chronic obstractive pulmonary disease, asthma, allergy, and emphysema.
G. NOV9 (CG94702-01): Hemicentin precursor
Expression of gene CG94702-01 was assessed using the primer-probe sets Ag3944 and Ag986, described in Tables GA and GB. Results of the RTQ-PCR runs are shown in Tables GC, GD, GE and GF.
Table GA. Probe Name Ag3944
Figure imgf000256_0001
Table GB. Probe Name Ag986
Figure imgf000256_0002
Table GC. General_screening_panel_vl.4
Figure imgf000256_0003
Figure imgf000257_0001
Figure imgf000258_0001
Table GD. Panel 2.1
Figure imgf000258_0002
Figure imgf000259_0001
Figure imgf000260_0001
Table GE. Panel 3D
Figure imgf000260_0002
Figure imgf000261_0001
Figure imgf000262_0001
Table GF. Panel 4. ID
Figure imgf000262_0002
Figure imgf000263_0001
Figure imgf000264_0001
Figure imgf000265_0001
CNS_neurodegeneration_vl.O Summary: Ag986/Ag3944 Expression of the CG94702-01 gene is low/undetectable in all samples on this panel (CTs>35). (Data not shown.)
General_screening_panel_vl.4 Summary: Ag986/Ag3944 Two experiments with two different probe and primer sets produce results that are in excellent agreement, with highest expression of the CG94702-01 gene in the kidney (CTs=32-33). Significant expression appears to be generally associated with normal tisuse and is also seen in samples derived from pancreas, stomach, small intestine, fetal and adult skeletal muscle and colon. Thus, expression of this gene could be used to differentiate between kidney and fetal kidney (CTs=40) and between these samples and the other samples on this panel.
Panel 2.1 Summary: Ag3944 Highest expression of the CG947-02 gene is seen in the uterus (CT=32.8). Overall, expression appears to be higher in normal tissues when compared to expression in cancer as seen in the previous panel. Thus, expression of this gene could be used as a marker of uterine tissue.
Panel 3D Summary: Ag986 Expression of the CG94702-01 gene is restricted to two samples derived from lung cancer cell lines (CTs=32-33). Thus, expression of this gene could be used to differentiate between these samples and other samples on this panel and as a marker to detect the presence of lung cancer. Furthermore, therapeutic modulation of the expression or function of this gene may be effective in the treatment of lung cancer.
Panel 4.1D Summary: Ag986/Ag3944 Two experiments with two different probe and primer sets produce results that are in very good agreement, with expression of the CG94702-01 gene restricted to normal tissue samples derived from colon and kidney (CTs=33.5-34.5). This preferential expression in normal tissue is consistent with expression seen in previous panels and suggests that expression of this gene could be used to differentiate between these samples and other samples on this panel and as a marker for these tissues. Furthermore, expression of this gene is decreased in colon samples from patients with IBD colitis and Crohn's disease relative to normal colon. Therefore, therapeutic modulation of the activity of the protein encoded by this gene may be useful in the treatment of inflammatory bowel disease. Furthermore, therapies designed with the protein encoded by this gene may modulate kidney function and be important in the treatment of inflammatory or autoimmune diseases that affect the kidney, including lupus and glomerulonephritis.
H. NOVlOa (COR_CG94661-01): selectin like
Expression of gene COR_CG94661-01 was assessed using the primer-probe set Ag3954, described in Table HA. Results of the RTQ-PCR runs are shown in Tables HB, HC and HD.
Table HA. Probe Name Ag3954
Figure imgf000266_0001
Table HB. CNS_neurodegeneration_vl.O
Figure imgf000266_0002
Figure imgf000267_0001
Table HC. General _screening_panel_v 1.4
Figure imgf000267_0002
Figure imgf000268_0001
Table HD. Panel 4.1D
Figure imgf000269_0001
Figure imgf000270_0001
Figure imgf000271_0001
CNS_neurodegeneration_vl.O Summary: Ag3954 This panel confirms the expression of the CG94661-01 gene at low levels in the brain in an independent group of individuals. However, no differential expression of this gene was detected between Alzheimer's diseased postmortem brains and those of non-demented controls in this experiment. Please see Panel 1.4 for a discussion of the potential utility of this gene in treatment of central nervous system disorders.
General_screening_panel_vl.4 Summary: Ag3954 Highest expression of the CG94661-01 gene is seen in a breast cancer cell line (CT=27.6). Significant expression is also seen in a cluster of ovarian, breast and colon cancer cell lines. Thus, expression of this gene could be used to differentiate these samples from other samples on this panel and as a marker for the presence of these cancers. Furthermore, therapeutic modulation of the expression or function of this gene may be effective in the treatment of these cancers.
In addition, this gene is expressed at higher levels in the adult kidney (Ct=29.7) and heart (CT=32) when compared to expression in fetal kidney (CT=32.2) and heart (CT=37), respectively. Thus, expression of this gene could be used to differentiate between these two sources of tissues.
Among tissues with metabolic fimction, this gene is expressed in thyroid, pancreas, fetal skeletal, heart, adult and fetal liver, pituitary and adipose. This widespread expression suggests that this gene product may play a role in normal metabolic and neuroendocrine function and that disregulated expression of this gene may contribute to metabolic diseases (such as obesity and diabetes) or neuroendocrine disorders.
This gene is also expressed at moderate levels in all regions of the central nervous system examined, including amygdala, hippocampus, substantia nigra, thalamus, cerebellum, cerebral cortex, and spinal cord. This gene contains a region homologous to a sushi domain, which is a common motif of protein-protein interactions. These domains are found in receptors with important neuronal function, such as IL-15R and GABARs. Therefore, this gene may have utility as a small molecule or antibody target to modulate CNS processes involved in CNS disorders.
References:
Wei Xq, Orchardson M, Gracie JA, Leung BP, Gao Bm, Guan H, Niedbala W, Paterson GK, Mclhnes IB, Liew FY. (2001) The Sushi domain of soluble IL-15 receptor alpha is essential for binding IL-15 and inhibiting inflammatory and allogenic responses in vitro and in vivo. J Immunol 2001 Jul l;167(l):277-82
IL- 15 is a pleiotropic cytokine that plays important roles in both innate and adaptive immunity. It is associated with a range of immunopathology, including rheumatoid arthritis and allograft rejection. IL-15 functions through the trimeric IL-15R complex, which consists of a high affinity binding alpha-chain and the common IL-2R beta- and gamma-chains. Characterization of 1L-15/IL-15R interactions may facilitate the development of improved IL- 15 antagonists for therapeutic interventions. We previously constructed soluble murine IL- 15Ralpha (sIL-15Ralpha) by deleting the cytoplasmic and transmembrane domains. To localize the functional domain of IL-15Ralpha, we have now constructed various truncated versions of slL-15Raipha. The shortest region retaining IL-15 binding activity is a 65-aa sequence spanning the Sushi domain of IL-15Ralpha. Sushi domains, common motifs in protein-protein interactions, contain four cysteines forming two disulfide bonds in a 1-3 and 2- 4 pattern. Amino acid substitution of the first or fourth cysteine in sIL-15Ralpha completely abolished its IL-15 binding activity. This also abrogated the ability of sIL-15Ralpha to neutralize D -15-induced proinflammatory cytokine production and anti-apoptotic response in vitro. Furthermore, the mutant sIL-15Ralpha lost its ability to inhibit carrageenan-induced local inflammation and allogenic cell-induced T cell proliferation and cytokine production in vivo. Thus, the Sushi domain is critical for the functional activity of sIL-15Ralpha. PMJD: 11418660
Panel 4.1D Summary: Ag3954 Two experiments with the same probe and primer set produce results that are in excellent agreement, with highest expression of the CG94661-01 gene in the ionomycin treated B cell line Ramos (CTs=27-29). Overall, this transcript is expressed in hematopietic cells, preferentially on B and T cells. The protein encoded by this transcript includes a sushi domain which is important in proteimprotein interactions (see reference in panel 1.4). This protein appears to be similar to selectin and complement activation proteins and thus may function as a receptor or adhesion molecule. Therefore, therapeutic modulation of the protein encoded by this transcript could be important in the freament of inflammation including asthma, emphysema, arthritis, psoriasis, and inflammatory bowel disease.
I. NOVll (CG94325-01): Novel nuclear protein
Expression of gene CG94325-01 was assessed using the primer-probe set Ag3913, described in Table IA. Results of the RTQ-PCR runs are shown in Table IB.
Table IA. Probe Name Ag3913
Figure imgf000273_0001
Table IB. General_screening_panel_vl.4
Figure imgf000273_0002
Figure imgf000274_0001
Figure imgf000275_0001
General_screening_panel_vl.4 Summary: Ag3913 Highest expression of the CG94325-01 gene is seen in a gastric cancer cell line (CT=27.9). In addition, significant levels of expression are seen in a cluster of samples derived from ovarian, breast, colon, melanoma and lung cancer cell lines. Thus, expression of this gene could be used to differentiate between these samples and other samples on this panel and as a marker to detect the presence of these cancers. In addition, this gene is expressed at much higher levels in all fetal tissues on this panel (CTs=29-31) when compared to expression in the adult counterpart (CTs=35-40). Thus, expression of this gene may be used to differentiate between the fetal and adult sources of brain, liver, lung, skeletal muscle, kidney and heart. Furthermore, this predominant expression in fetal tissues and cancer cell lines further reinforces the suggestion that this gene product maybe involved in cellular growth and proliferation. Therefore, therapeutic modulation of the expression or function of this gene may be effective in the treatment of ovarian, breast, colon, melanoma and lung cancers.
Among tissues with metabolic function, this gene is expressed at low levels in adipose and pancreas. This expression among these tissues suggests that this gene product may play a role in normal neuroendocrine and metabolic and that disregulated expression of this gene may contribute to obesity and diabetes.
J. NOV12 (CG94282-01): Novel Notch Domain Containing Protein
Expression of gene CG94282-01 was assessed using the primer-probe set Ag3910, described in Table JA.
Table JA. Probe Name Ag3910
Figure imgf000276_0002
CNS_neurodegeneration_vl.O Summary: Ag3910 Expression of the CG94282-01 gene is low/undetectable in all samples on this panel (CTs>35). (Data not shown.) The amp plot indicates that there is a high probability of a probe failure.
General_screening_panel_vl.4 Summary: Ag3910 Expression of the CG94282-01 gene is low/undetectable in all samples on this panel (CTs>35). (Data not shown.) The amp plot indicates that there is a high probability of a probe failure.
Panel 2.1 Summary: Ag3910 Expression of the CG94282-01 gene is low/undetectable in all samples on this panel (CTs>35). (Data not shown.) The amp plot indicates that there is a high probability of a probe failure.
Panel 4.1D Summary: Ag3910 Expression of the CG94282-01 gene is low/undetectable in all samples on this panel (CTs>35). (Data not shown.) The amp plot indicates that there is a high probability of a probe failure.
K. NOV13 (CG94399-01): BHLH FACTOR MATH6
Expression of gene CG94399-01 was assessed using the primer-probe set Ag3919, described in Table KA. Results of the RTQ-PCR runs are shown in Tables KB, KC and KD.
Table KA. Probe Name Ag39i9
Figure imgf000276_0001
Table KB. CNS_neurodegeneration_vl.O
Figure imgf000276_0003
Figure imgf000277_0001
Table KC. General_screening_panel_vl.4
Figure imgf000277_0002
Figure imgf000278_0001
Figure imgf000279_0001
Table KD. Panel 4. ID
Figure imgf000279_0002
Figure imgf000280_0001
Figure imgf000281_0001
CNS_neurodegeneration_vl.O Summary: Ag3919 This panel does not show differential expression of the CG94399-01 gene in Alzheimer's disease. However, this expression profile confirms the presence of this gene in the brain. Please see Panel 1.4 for discussion of utility of this gene in the central nervous system.
General_screening_panel_vl.4 Summary: Ag3919 Highest expression of the CG94399-01 gene is seen in a brain cancer cell line (CT=28.4). In addition, significant levels of expression are seen in a cluster of samples derived from ovarian, melanoma and lung cancer cell lines. Thus, expression of this gene could be used to differentiate between these samples and other samples on this panel and as a marker to detect the presence of these cancers. Furthermore, therapeutic modulation of the expression or function of this gene maybe effective in the treatment of ovarian, melanoma, brain and lung cancers.
Among tissues with metabolic function, this gene is expressed at moderate to low levels in pituitary, adipose, adrenal gland, pancreas, thyroid, liver, and adult and fetal skeletal muscle, and heart. This widespread expression among these tissues suggests that this gene product may play a role in normal neuroendocrine and metabolic and that disregulated expression of this gene may contribute to neuroendocrine disorders or metabolic diseases, such as obesity and diabetes.
This molecule is also expressed at low levels in the CNS, including the hippocampus, thalamus, substantia nigra, amygdala, cerebellum and cerebral cortex. Therefore, therapeutic modulation of the expression or function of this gene may be useful in the treatment of neurologic disorders, such as Alzheimer's disease, Parkinson's disease, schizophrenia, multiple sclerosis, stroke and epilepsy.
In addition, this gene is expressed at much higher levels in fetal lung (CT=30) when compared to expression in the adult lung (CT=34), and at much higher levels in adult liver (CT=31) when compared to expression in fetal liver (CT=35). Thus, expression of this gene maybe used to differentiate between the fetal and adult source of these tissue.
Panel 4.1D Summary: Ag3919 Ag3919 Highest expression of the CG94399-01 gene is seen in resting dermal fibroblasts (CT=29.2). Moderate levels of expression are also seen in treated and untreated lung fibroblasts, dermal fibroblasts, lung microvascular endothelium and the mucoepidermoid cell line, NCI-H292. The expression of this gene in cells derived from or within the lung and skin suggests that this gene may be involved in normal conditions as well as pathological and inflammatory lung disorders that include chronic obstructive pulmonary disease, asthma, allergy, psoriasis and emphysema.
L. NOV15 (CG95387-02): LRR protein
Expression of gene CG95387-02 was assessed using the primer-probe set Ag4112, described in Table LA. Results of the RTQ-PCR runs are shown in Tables LB and LC.
Table LA. Probe Name Ag4112
Figure imgf000282_0001
Table LB. General_screening_panel_vl.4
Figure imgf000282_0002
Figure imgf000283_0001
Figure imgf000284_0001
Table LC. Panel 4. ID
Figure imgf000284_0002
Figure imgf000285_0001
CNS_neurodegeneration_vl.O Summary: Ag4112 Expression of the CG95387-01 gene is low/undetectable in all samples on this panel (CTs>35). (Data not shown.)
General_screening_panel_vl.4 Summary: Ag4112 Expression of the CG95387-01 gene is highest in the T47D breast cancer cell line (CT=31.7), an estrogen receptor positive cell line. In addition, low but significant levels of expression are seen in a cluster of samples derived from prostate, ovarian, gastric, brain, colon, and lung cancer cell lines. Thus, expression of this gene could be used to differentiate between these samples and other samples on this panel and as a marker to detect the presence of these cancers. Overall, expression of this gene appears to be associated with the cancer cell lines when compared to expression in the normal samples. This gene encodes a protein with homology to a leucine rich repeat protein. This motif is believed to participate in protein-protein interactions. A novel member of the leucine rich repeat family has been shown to be upregulated in esrogen positive breast cancers. Therefore based on the published literature and the expression of this gene, therapeutic modulation of the expression or function of this gene may be effective in the treatment of breast, prostate, ovarian, gastric, brain, colon, and lung cancers.
References:
Charpentier AH, Bednarek AK, Daniel RL, Hawkins KA, Laflin KJ, Gaddis S, MacLeod MC, Aldaz CM. Effects of estrogen on global gene expression: identification of novel targets of esfrogen action. Cancer Res 2000 Nov l;60(21):5977-83
The important role played by the sex hormone estrogen in disease and physiological processes has been well documented. However, the mechanisms by which this hormone elicits many of its normal as well as pathological effects are unclear. To identify both known and unknown genes that are regulated by or associated with estrogen action, we performed serial analysis of gene expression on estrogen-responsive breast cancer cells after exposure to this hormone. We examined approximately 190,000 mRNA transcripts and momtored the expression behavior of 12,550 genes. Expression levels for the vast majority of those transcripts were observed to remain constant upon 17beta estradiol (E2) treatment. Only approximately 0.4% of the genes showed an increase in expression of > or =3-fold by 3 h post-E2 treatment. We cloned five novel genes (E2IG1-5), which were observed up-regulated by the hormonal treatment. Of these the most highly induced transcript, E2IG1, appears to be a novel member of the family of small heat shock proteins. The E2IG4 gene is a new member of the large family of leucine- rich repeat-containing proteins. On the basis of architectural and domain homology, this gene appears to be a good candidate for secretion in the extracellular environment and, therefore, may play a role in breast tissue remodeling and/or epithelium-stroma interactions. Several interesting genes with a potential role in the regulation of cell cycle progression were also identified to increase in expression, including Pescadillo and chaperonin CCT2. Two putative paracrine/autocrine factors of potential importance in the regulation of the growth of breast cancer cells were identified to be highly up-regulated by E2: stanniocalcin 2, a calcium/phosphate homeostatic hormone; and inhibin-beta B, a TGF-beta-like factor. Interestingly, we also determined that E2IG1 and stanniocalcin 2 were exclusively overexpressed in estrogen-receptor-positive breast cancer lines, and thus they have the potential to serve as breast cancer biomarkers. This data provides a comprehensive view of the changes induced by E2 on the transcriptional program of human E2-responsive cells, and it also identifies novel and previously unsuspected gene targets whose expression is affected by this hormone.
PMID: 11085516
Panel 4.1D Summary: Ag4112 Expression of the CG95387-01 gene is restricted to the kidney (CT=33.7). The putative protein encoded by this gene could allow cells within the kidney to respond to specific microenvironmental signals. Therefore, therapies designed with the protein encoded for by this gene could modulate kidney function and be important in the treatment of inflammatory or autoimmune diseases that affect the kidney, including lupus and glomerulonephritis.
Example 3. Identification of Single Nucleotide Polymorphisms in NOVX nucleic acid sequences
Variant sequences are also included in this application. A variant sequence can include a single nucleotide polymorphism (SNP). A SNP can, in some instances, be referred to as a "cSNP" to denote that the nucleotide sequence containing the SNP originates as a cDNA. A SNP can arise in several ways. For example, a SNP may be due to a substitution of one nucleotide for another at the polymorphic site. Such a substitution can be either a transition or a transversion. A SNP can also arise from a deletion of a nucleotide or an insertion of a nucleotide, relative to a reference allele. In this case, the polymorphic site is a site at which one allele bears a gap with respect to a particular nucleotide in another allele. SNPs occurring within genes may result in an alteration of the amino acid encoded by the gene at the position of the SNP. Intragenic SNPs may also be silent, when a codon including a SNP encodes the same amino acid as a result of the redundancy of the genetic code. SNPs occurring outside the region of a gene, or in an intron within a gene, do not result in changes in any amino acid sequence of a protein but may result in altered regulation of the expression pattern. Examples include alteration in temporal expression, physiological response regulation, cell type expression regulation, intensity of expression, and stability of transcribed message.
SeqCalling assemblies produced by the exon linking process were selected and extended using the following criteria. Genomic clones having regions with 98% identity to all or part of the initial or extended sequence were identified by BLASTN searches using the relevant sequence to query human genomic databases. The genomic clones that resulted were selected for further analysis because this identity indicates that these clones contain the genomic locus for these SeqCalling assemblies. These sequences were analyzed for putative coding regions as well as for similarity to the known DNA and protein sequences. Programs used for these analyses include Grail, Genscan, BLAST, HMMER, FASTA, Hybrid and other relevant programs. Some additional genomic regions may have also been identified because selected
SeqCalling assemblies map to those regions. Such SeqCalling sequences may have overlapped with regions defined by homology or exon prediction. They may also be included because the location of the fragment was in the vicinity of genomic regions identified by similarity or exon prediction that had been included in the original predicted sequence. The sequence so identified was manually assembled and then may have been extended using one or more additional sequences taken from CuraGen Corporation's human SeqCalling database. SeqCalling fragments suitable for inclusion were identified by the CuraTools™ program SeqExtend or by identifying SeqCalling fragments mapping to the appropriate regions of the genomic clones analyzed. The regions defined by the procedures described above were then manually integrated and corrected for apparent inconsistencies that may have arisen, for example, from miscalled bases in the original fragments or from discrepancies between predicted exon junctions, EST locations and regions of sequence similarity, to derive the final sequence disclosed herein. When necessary, the process to identify and analyze SeqCalling assemblies and genomic clones was reiterated to derive the full length sequence (Alderborn et al, Determination of Single Nucleotide Polymorphisms by Real-time Pyrophosphate DNA Sequencing. Genome Research. 10 (8): 1249-1265, 2000).
Variants are reported individually but any combination of all or a select subset of variants are also included as contemplated NOVX embodiments of the invention. NOVl SNP data:
In the following positions, one or more consensus positions (Cons. Pos.) of the nucleotide sequence have been identified as SNPs. NOVl has one SNP variant (Variant 13377181), whose variant positions for its nucleotide and amino acid sequences is numbered according to SEQ ID NOS:l and 2, respectively. The nucleotide sequence of the NOVl variant differs as shown in Table 18.
Table 18. cSNP and Coding Variants for NOVl
Figure imgf000289_0001
NOV5 SNP data:
In the following positions, one or more consensus positions (Cons. Pos.) of the nucleotide sequence have been identified as SNPs. NOV5 has single SNP variant (Variant 13377186), whose variant positions for its nucleotide and amino acid sequences is numbered according to SEQ ID NOS: 9 and 10, respectively. The nucleotide sequence of the NOV5 variant differs as shown in Table 19.
Table 19. cSNP and Coding Variants for NOV5
Figure imgf000289_0002
NOV8 SNP data: ha the following positions, one or more consensus positions (Cons. Pos.) of the nucleotide sequence have been identified as SNPs. NOV8 has two SNP variants (Variant 1377197 and Variant 13377196), whose variant positions for its nucleotide and amino acid sequences is numbered according to SEQ ID NOS: 15 and 16, respectively. The nucleotide sequence of the NOV8 variant differs as shown in Table 20.
Table 20. cSNP and Coding Variants for NOV8
Figure imgf000290_0001
NOV9 SNP data:
In the following positions, one or more consensus positions (Cons. Pos.) of the nucleotide sequence have been identified as SNPs. NOV9 has a single SNP variant (Variant 13377850), whose variant positions for its nucleotide and amino acid sequences is numbered according to SEQ ID NOS: 17 and 18, respectively. The nucleotide sequence of the NOV9 variant differs as shown in Table 21.
Table 21. cSNP and Coding Variants for NOV9
Figure imgf000290_0002
NOVlOb SNP data:
In the following positions, one or more consensus positions (Cons. Pos.) of the nucleotide sequence have been identified as SNPs. NOVlOb has seven SNP variants, whose variant positions for its nucleotide and amino acid sequences is numbered according to SEQ ID NOS:21 and 22, respectively. The nucleotide sequence of the NOVlOb variant differs as shown in Table 22.
Figure imgf000290_0003
NOVll SNP data:
In the following positions, one or more consensus positions (Cons. Pos.) of the nucleotide sequence have been identified as SNPs. NOVl 1 has two SNP variants (Variant 13377199 and Variant 13377200), whose variant positions for its nucleotide and amino acid sequences is numbered according to SEQ ID NOS:23 and 24, respectively. The nucleotide sequence of the NOVl 1 variant differs as shown in Table 23.
Table 23. cSNP and Coding Variants for NOVll
Figure imgf000291_0001
NOV12 SNP data:
In the following positions, one or more consensus positions (Cons. Pos.) of the nucleotide sequence have been identified as SNPs. NOVl 2 has one SNP variant (Variant 13377201), whose variant positions for its nucleotide and amino acid sequences is numbered according to SEQ ID NOS:25 and 26, respectively. The nucleotide sequence of the NOVl 2 variant differs as shown in Table 24.
Table 24. cSNP and Coding Variants for NOV12
Figure imgf000291_0002
NOV13 SNP data:
In the following positions, one or more consensus positions (Cons. Pos.) of the nucleotide sequence have been identified as SNPs. NOVl 3 has two SNP variants (Variant 13377852 and Variant 13377851), whose variant positions for its nucleotide and amino acid sequences is numbered according to SEQ ID NOS:27 and 28, respectively. The nucleotide sequence of the NOV13 variant differs as shown in Table 25.
Table 25. cSNP and Coding Variants for NOV13
Figure imgf000292_0001
NOV15 SNP data:
In the following positions, one or more consensus positions (Cons. Pos.) of the nucleotide sequence have been identified as SNPs. NOV 15 has seven SNP variants, whose variant positions for its nucleotide and amino acid sequences is numbered according to SEQ ID NOS:31 and 32, respectively. The nucleotide sequence of the NOVl 5 variant differs as shown in Table 26.
Table 26. cSNP and Coding Variants for NOV15
Figure imgf000292_0002
OTHER EMBODIMENTS
Although particular embodiments have been disclosed herein in detail, this has been done by way of example for purposes of illustration only, and is not intended to be limiting with respect to the scope of the appended claims, which follow, hi particular, it is contemplated by the inventors that various substitutions, alterations, and modifications may be made to the invention without departing from the spirit and scope of the invention as defined by the claims. The choice of nucleic acid starting material, clone of interest, or library type is believed to be a matter of routine for a person of ordinary skill in the art with knowledge of the embodiments described herein. Other aspects, advantages, and modifications considered to be within the scope of the following claims.

Claims

WHAT IS CLAIMED IS:
1. An isolated polypeptide comprising an amino acid sequence selected from the group consisting of:
(a) a mature form of an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34;
(b) a variant of a mature form of an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34, wherein one or more amino acid residues in said variant differs from the amino acid sequence of said mature form, provided that said variant differs in no more than 15% of the amino acid residues from the amino acid sequence of said mature form;
(c) an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34; and
(d) a variant of an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34 wherein one or more amino acid residues in said variant differs from the amino acid sequence of said mature form, provided that said variant differs in no more than 15% of amino acid residues from said amino acid sequence.
2. The polypeptide of claim 1, wherein said polypeptide comprises the amino acid sequence of a natixrally-occurring allehc variant of an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34.
3. The polypeptide of claim 2, wherein said allelic variant comprises an amino acid sequence that is the translation of a nucleic acid sequence differing by a single nucleotide from a nucleic acid sequence selected from the group consisting of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33.
4. The polypeptide of claim 1, wherein the amino acid sequence of said variant comprises a conservative amino acid substitution.
5. An isolated nucleic acid molecule comprising a nucleic acid sequence encoding a polypeptide comprising an amino acid sequence selected from the group consisting of:
(a) a mature form of an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34;
(b) a variant of a mature form of an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34, wherein one or more amino acid residues in said variant differs from the amino acid sequence of said mature form, provided that said variant differs in no more than 15% of the amino acid residues from the amino acid sequence of said mature form;
(c) an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34;
(d) a variant of an amino acid sequence selected from the group consisting of SEQ D NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34, wherein one or more amino acid residues in said variant differs from the amino acid sequence of said mature form, provided that said variant differs in no more than 15% of amino acid residues from said amino acid sequence;
(e) a nucleic acid fragment encoding at least a portion of a polypeptide comprising an amino acid sequence chosen from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34, or a variant of said polypeptide, wherein one or more amino acid residues in said variant differs from the amino acid sequence of said mature form, provided that said variant differs in no more than 15% of amino acid residues from said amino acid sequence; and
(f) a nucleic acid molecule comprising the complement of (a), (b), (c), (d) or (e).
6. The nucleic acid molecule of claim 5, wherein the nucleic acid molecule comprises the nucleotide sequence of a naturally-occurring allehc nucleic acid variant.
7. The nucleic acid molecule of claim 5, wherein the nucleic acid molecule encodes a polypeptide comprising the amino acid sequence of a naturally-occurring polypeptide variant.
8. The nucleic acid molecule of claim 5, wherein the nucleic acid molecule differs by a single nucleotide from a nucleic acid sequence selected from the group consisting of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33.
9. The nucleic acid molecule of claim 5, wherein said nucleic acid molecule comprises a nucleotide sequence selected from the group consisting of
(a) a nucleotide sequence selected from the group consisting of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33;
(b) a nucleotide sequence differing by one or more nucleotides from a nucleotide sequence selected from the group consisting of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, provided that no more than 20% of the nucleotides differ from said nucleotide sequence;
(c) a nucleic acid fragment of (a); and
(d) a nucleic acid fragment of (b).
10. The nucleic acid molecule of claim 5, wherein said nucleic acid molecule hybridizes under stringent conditions to a nucleotide sequence chosen from the group consisting of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, or a complement of said nucleotide sequence.
11. The nucleic acid molecule of claim 5 , wherein the nucleic acid molecule comprises a nucleotide sequence selected from the group consisting of
(a) a first nucleotide sequence comprising a coding sequence differing by one or more nucleotide sequences from a coding sequence encoding said amino acid sequence, provided that no more than 20% of the nucleotides in the coding sequence in said first nucleotide sequence differ from said coding sequence;
(b) an isolated second polynucleotide that is a complement of the first polynucleotide; and
(c) a nucleic acid fragment of (a) or (b).
12. A vector comprising the nucleic acid molecule of claim 11.
13. The vector of claim 12, further comprising a promoter operably-linked to said nucleic acid molecule.
14. A cell comprising the vector of claim 12.
15. An antibody that immunospecifically-binds to the polypeptide of claim 1.
16. The antibody of claim 15, wherein said antibody is a monoclonal antibody.
17. The antibody of claim 15, wherein the antibody is a humanized antibody.
18. A method for determining the presence or amount of the polypeptide of claim 1 in a sample, the method comprising:
(a) providing the sample;
(b) contacting the sample with an antibody that binds immunospecifically to the polypeptide; and
(c) deteimining the presence or amount of antibody bound to said polypeptide, thereby detennining the presence or amount of polypeptide in said sample.
19. A method for deterniining the presence or amount of the nucleic acid molecule of claim 5 in a sample, the method comprising:
(a) providing the sample;
(b) contacting the sample with a probe that binds to said nucleic acid molecule; and
(c) determining the presence or amount of the probe bound to said nucleic acid molecule, thereby determining the presence or amount of the nucleic acid molecule in said sample.
20. A method of identifying an agent that binds to a polypeptide of claim 1 , the method comprising:
(a) contacting said polypeptide with said agent; and
(b) determining whether said agent binds to said polypeptide.
21. A method for identifying an agent that modulates the expression or activity of the polypeptide of claim 1, the method comprising:
(a) providing a cell expressing said polypeptide;
(b) contacting the cell with said agent; and
(c) determining whether the agent modulates expression or activity of said polypeptide, whereby an alteration in expression or activity of said peptide indicates said agent modulates expression or activity of said polypeptide.
22. A method for modulating the activity of the polypeptide of claim 1, the method comprising contacting a cell sample expressing the polypeptide of said claim with a compound that binds to said polypeptide in an amount sufficient to modulate the activity of the polypeptide.
23. A method of treating or preventing a NOVX-associated disorder, said method comprising administering to a subject in which such treatment or prevention is desired the polypeptide of claim 1 in an amount sufficient to treat or prevent said NOVX-associated disorder in said subject.
24. The method of claim 23, wherein said subject is a human.
25. A method of treating or preventing a NOVX-associated disorder, said method comprising administering to a subject in which such treatment or prevention is desired the nucleic acid of claim 5 in an amount sufficient to treat or prevent said NOVX-associated disorder in said subject.
26. The method of claim 25, wherein said subject is a human.
27. A method of treating or preventing a NOVX-associated disorder, said method comprising administering to a subject in which such treatment or prevention is desired the antibody of claim 15 in an amount sufficient to treat or prevent said NOVX-associated disorder in said subject.
28. The method of claim 27, wherein the subject is a human.
29. A pharmaceutical composition comprising the polypeptide of claim 1 and a pharmaceutically-acceptable carrier.
30. A pharmaceutical composition comprising the nucleic acid molecule of claim 5 and a pharmaceutically-acceptable carrier.
31. A pharmaceutical composition comprising the antibody of claim 15 and a pharmaceutically-acceptable carrier.
32. A kit comprising in one or more containers, the pharmaceutical composition of claim 29.
33. A kit comprising in one or more containers, the pharmaceutical composition of claim 30.
34. A kit comprising in one or more containers, the pharmaceutical composition of claim 31.
35. The use of a therapeutic in the manufacture of a medicament for treating a syndrome associated with a human disease, the disease selected from a NOVX-associated disorder, wherein said therapeutic is selected from the group consisting of a NOVX polypeptide, a NOVX nucleic acid, and a NOVX antibody.
36. A method for screening for a modulator of activity or of latency or predisposition to a NOVX-associated disorder, said method comprising:
(a) administering a test compound to a test animal at increased risk for a NOVX-associated disorder, wherein said test animal recombinantly expresses the polypeptide of claim 1;
(b) measuring the activity of said polypeptide in said test animal after administering the compound of step (a);
(c) comparing the activity of said protein in said test animal with the activity of said polypeptide in a control animal not administered said polypeptide, wherein a change in the activity of said polypeptide in said test animal relative to said control animal indicates the test compound is a modulator of latency of or predisposition to a NOVX- associated disorder.
37. The method of claim 36, wherein said test animal is a recombinant test animal that expresses a test protein transgene or expresses said fransgene under the control of a promoter at an increased level relative to a wild-type test animal, and wherein said promoter is not the native gene promoter of said transgene.
38. A method for determining the presence of or predisposition to a disease associated with altered levels of the polypeptide of claim 1 in a first mammalian subject, the method comprising:
(a) measuring the level of expression of the polypeptide in a sample from the first mammalian subject; and
(b) comparing the amount of said polypeptide in the sample of step (a) to the amount of the polypeptide present in a control sample from a second mammalian subject known not to have, or not to be predisposed to, said disease, wherein an alteration in the expression level of the polypeptide in the first subject as compared to the confrol sample indicates the presence of or predisposition to said disease.
39. A method for determining the presence of or predisposition to a disease associated with altered levels of the nucleic acid molecule of claim 5 in a first mammalian subject, the method comprising:
(a) measuring the amount of the nucleic acid in a sample from the first mammalian subject; and
(b) comparing the amount of said nucleic acid in the sample of step (a) to the amount of the nucleic acid present in a confrol sample from a second mammalian subject known not to have or not be predisposed to, the disease; wherein an alteration in the level of the nucleic acid in the first subject as compared to the control sample indicates the presence of or predisposition to the disease.
40. A method of treating a pathological state in a mammal, the method comprising administering to the mammal a polypeptide in an amount that is sufficient to alleviate the pathological state, wherein the polypeptide is a polypeptide having an amino acid sequence at least 95% identical to a polypeptide comprising an amino acid sequence of at least one of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34, or a biologically active fragment thereof.
41. A method of treating a pathological state in a mammal, the method comprising administering to the mammal the antibody of claim 15 in an amount sufficient to alleviate the pathological state.
PCT/US2002/011634 2001-04-23 2002-04-11 Proteins and nucleic acids encoding same WO2002085922A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP02723842A EP1383533A4 (en) 2001-04-23 2002-04-11 Proteins and nucleic acids encoding same
CA002443770A CA2443770A1 (en) 2001-04-23 2002-04-11 Proteins and nucleic acids encoding same

Applications Claiming Priority (12)

Application Number Priority Date Filing Date Title
US28574801P 2001-04-23 2001-04-23
US60/285,748 2001-04-23
US28606801P 2001-04-24 2001-04-24
US60/286,068 2001-04-24
US28629201P 2001-04-25 2001-04-25
US60/286,292 2001-04-25
US28833401P 2001-05-03 2001-05-03
US60/288,334 2001-05-03
US29124101P 2001-05-16 2001-05-16
US60/291,241 2001-05-16
US32228401P 2001-09-14 2001-09-14
US60/322,284 2001-09-14

Publications (2)

Publication Number Publication Date
WO2002085922A2 true WO2002085922A2 (en) 2002-10-31
WO2002085922A3 WO2002085922A3 (en) 2003-05-30

Family

ID=27559584

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/011634 WO2002085922A2 (en) 2001-04-23 2002-04-11 Proteins and nucleic acids encoding same

Country Status (3)

Country Link
EP (1) EP1383533A4 (en)
CA (1) CA2443770A1 (en)
WO (1) WO2002085922A2 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1316560A1 (en) * 2000-09-08 2003-06-04 Japan Science and Technology Corporation Novel human cancer/testis antigen and gene thereof
US8148093B2 (en) 2003-08-15 2012-04-03 Diadexus, Inc. Pro108 antibody compositions and methods of use and use of Pro108 to assess cancer risk
US9062101B2 (en) 2010-08-14 2015-06-23 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US9822171B2 (en) 2010-04-15 2017-11-21 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US9951125B2 (en) 2006-11-30 2018-04-24 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
US10208109B2 (en) 2005-11-30 2019-02-19 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US10464976B2 (en) 2003-01-31 2019-11-05 AbbVie Deutschland GmbH & Co. KG Amyloid β(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
US10538581B2 (en) 2005-11-30 2020-01-21 Abbvie Inc. Anti-Aβ globulomer 4D10 antibodies

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100311767A1 (en) 2007-02-27 2010-12-09 Abbott Gmbh & Co. Kg Method for the treatment of amyloidoses

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000058473A2 (en) * 1999-03-31 2000-10-05 Curagen Corporation Nucleic acids including open reading frames encoding polypeptides; 'orfx'

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2387371A1 (en) * 1999-06-14 2000-12-21 Zymogenetics, Inc. Helical cytokine zalpha33
AU2001273151A1 (en) * 2000-06-30 2002-01-14 Incyte Genomics, Inc. Human extracellular matrix and cell adhesion polypeptides

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000058473A2 (en) * 1999-03-31 2000-10-05 Curagen Corporation Nucleic acids including open reading frames encoding polypeptides; 'orfx'

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE PROTEIN [Online] NAGASE ET AL.: 'Prediction of the coding sequences of unidentified human genes', XP002959446 Retrieved from EMBL Database accession no. (BAA86588) & DNA RES vol. 6, no. 5, 11 November 1999, pages 337 - 345 *
See also references of EP1383533A2 *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1316560A1 (en) * 2000-09-08 2003-06-04 Japan Science and Technology Corporation Novel human cancer/testis antigen and gene thereof
EP1316560A4 (en) * 2000-09-08 2005-01-05 Japan Science & Tech Agency Novel human cancer/testis antigen and gene thereof
US10464976B2 (en) 2003-01-31 2019-11-05 AbbVie Deutschland GmbH & Co. KG Amyloid β(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
US8148093B2 (en) 2003-08-15 2012-04-03 Diadexus, Inc. Pro108 antibody compositions and methods of use and use of Pro108 to assess cancer risk
US10208109B2 (en) 2005-11-30 2019-02-19 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US10323084B2 (en) 2005-11-30 2019-06-18 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US10538581B2 (en) 2005-11-30 2020-01-21 Abbvie Inc. Anti-Aβ globulomer 4D10 antibodies
US9951125B2 (en) 2006-11-30 2018-04-24 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
US9822171B2 (en) 2010-04-15 2017-11-21 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US9062101B2 (en) 2010-08-14 2015-06-23 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US10047121B2 (en) 2010-08-14 2018-08-14 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins

Also Published As

Publication number Publication date
WO2002085922A3 (en) 2003-05-30
EP1383533A4 (en) 2006-11-02
CA2443770A1 (en) 2002-10-31
EP1383533A2 (en) 2004-01-28

Similar Documents

Publication Publication Date Title
WO2002057450A2 (en) Proteins and nucleic acids encoding same
WO2002046409A2 (en) Proteins and nucleic acids encoding same
EP1487864A2 (en) Therapeutic polypeptides nucleic acids encoding same, and methods of use
WO2002085922A2 (en) Proteins and nucleic acids encoding same
WO2002079398A2 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
WO2002099116A2 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
WO2003078572A2 (en) Therapeutic polypeptides, nucleic acids encoding same and methods of use
WO2003031571A2 (en) Novel human proteins, polynucleotides encoding them and methods of using the same
EP1356047A2 (en) Human proteins, polynucleotides encoding them and methods of using the same
EP1399537A2 (en) Therapeutic polypeptides, nucleic acids encoding same and methods of use
WO2002090500A2 (en) Novel human proteins, polynucleotides encoding them and methods of using the same
EP1463747A2 (en) Novel antibodies that bind to antigenic polypeptides,nucleic acids encodings the antigens, and methodes of use
EP1443913A2 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20030203843A1 (en) Proteins and nucleic acids encoding same
WO2002081629A2 (en) Novel human proteins, polynucleotides encoding them and methods of using the same
EP1622634A2 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
WO2003064589A2 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
WO2003052061A2 (en) Novel human proteins, polynucleotides encoding them and methods of using the same
AU2002254605A1 (en) Proteins and nucleic acids encoding same
WO2003004617A2 (en) Novel human proteins, polynucleotides encoding them and methods of using the same
WO2002072770A2 (en) Novel human proteins, polynucleotides encoding them and methods of using the same
WO2002072771A2 (en) Novel antibodies that bind to antigenic polypeptides, nucleic acids encoding the antigens, and methods of use
EP1401486A2 (en) Novel human proteins, polynucleotides encoding them and methods of using the same
JP2005509400A (en) Protein and nucleic acid encoding it
AU2002257115A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US US US US US US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2002254605

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2443770

Country of ref document: CA

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2002723842

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2002723842

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

NENP Non-entry into the national phase in:

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP

WWW Wipo information: withdrawn in national office

Ref document number: 2002723842

Country of ref document: EP