WO1999000408A2 - Nouvelle proteine transmembranaire humaine - Google Patents

Nouvelle proteine transmembranaire humaine Download PDF

Info

Publication number
WO1999000408A2
WO1999000408A2 PCT/US1998/009878 US9809878W WO9900408A2 WO 1999000408 A2 WO1999000408 A2 WO 1999000408A2 US 9809878 W US9809878 W US 9809878W WO 9900408 A2 WO9900408 A2 WO 9900408A2
Authority
WO
WIPO (PCT)
Prior art keywords
ht4p
sequence
protein
sequences
expression
Prior art date
Application number
PCT/US1998/009878
Other languages
English (en)
Other versions
WO1999000408A3 (fr
Inventor
Jennifer L. Hillman
Neil C. Corley
Purvi Shah
Original Assignee
Incyte 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 Incyte Pharmaceuticals, Inc. filed Critical Incyte Pharmaceuticals, Inc.
Priority to AU75723/98A priority Critical patent/AU7572398A/en
Publication of WO1999000408A2 publication Critical patent/WO1999000408A2/fr
Publication of WO1999000408A3 publication Critical patent/WO1999000408A3/fr

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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • This invention relates to nucleic acid and amino acid sequences of a new transmembrane protein and to the use of these sequences in the diagnosis, prevention, and treatment of cancer, neurological disorders, and smooth muscle disorders.
  • Membrane proteins are divided into two groups based upon the ease with which the proteins can be removed from the membrane. Extrinsic or peripheral membrane proteins can be removed using extremes of ionic strength or pH, or other disruptors of protein interactions. Intrinsic or integral membrane proteins are released only when the lipid bilayer of the membrane is dissolved by detergent.
  • Transmembrane proteins The majority of known integral membrane proteins are transmembrane proteins which contain extracellular, transmembrane, and intracellular domains. Transmembrane proteins are typically embedded in a cell membrane by one or more regions comprising 15 to 25 hydrophobic amino acids that adopt an ⁇ -helical conformation. Transmembrane proteins are classified as bitopic (Types I and IT) and polytopic (Types IQ and IV). While bitopic proteins span the membrane only once, polytopic proteins span the membrane multiple times and type m proteins have multiple stretches of hydrophobic residues (Singer, S.J. (1990) Annu. Rev. Cell Biol. 6:247- 96). Transmembrane proteins carry out a variety of important cellular functions including signal transduction (e.g., growth factor receptors) and transport of ions or metabolites (e.g., ion channels).
  • signal transduction e.g., growth factor receptors
  • ions or metabolites e.g., ion channels
  • TM4SF transmembrane 4 superfamily
  • TM4SF proteins are found predominantly in cells of hematopoietic origin and in tumors and include a number of platelet and endothelial cell membrane proteins, CD9 (lung adenocarcinoma antigen MRP-1), the platelet and melanoma-associated antigen CD63, leukocyte surface glycoproteins, CD53, CD37, CD63, and R2, the tumor associated antigen TAPA-1 (CD81), the colonal carcinoma antigen CO-029, mink lung epithelial protein TI-1, the tumor-associated antigens L6 and SAS ,and a gene amplified in human sarcomas (Wright and Tomlinson, supra; Jankowski, S.A. et al. (1994) Oncogene 9:1205-11).
  • the proteins share 25-30% amino acid sequence identity.
  • TM4SF proteins The predicted structure of the TM4SF proteins reveals a topology where the N- and C- termini are intracellular and the major hydrophilic domain, located between transmembrane domains 3 and 4, is extracellular. Most of these proteins have N-linked glycosylation sites within the hydrophilic domain. TM4SF proteins are most conserved in their transmembrane and cytoplasmic domains and most divergent in their hydrophilic extracellular domains. The high level of conservation seen in the transmembrane and cytoplasmic domains suggest an effector/signaling function. The divergence of the extracellular domains suggests that these hydrophilic domains provide functions specific to each protein such as ligand binding or protein- protein interaction (Wright and Tomlinson, supra).
  • TM4SF proteins have been implicated in signal transduction, control of cell adhesion, regulation of cell growth and proliferation, including development, oncogenesis, motility, and the ability to suppress metastatic potential. Expression of a number of TM4SF proteins is associated with a variety of tumors and is altered when cells are growing or activated. For example, CD9, CD53 and CD82 are upregulated when lymphocytes are activated and cell surface expression of CD37 is lost when B cells are activated. Other TM4SF proteins are implicated in cell growth due to their association with tumor cells.
  • CD9 is not expressed on resting B and T lymphocytes, it is a marker for 90% of non-T acute lymphoblastic leukemia cells and for 50% of acute myeloid and chronic lymphoid leukemias.
  • Anti-CD9 antibodies inhibit the motility of a variety of cancer cell lines and inhibit the metastatic potential of the mouse BL6 cell line (Miyake, M. and Hakomori, S. (1991) Biochem. 30:3328-34). Expression of CD9 in transfection experiments correlated with suppression of metastatic potential and cell motility (Dceyama, S. et al. (1993) J. Exp. Med. 177:1231-37).
  • CD63 is not expressed on normal tissue melanocytes, but it is expressed in early stage melanoma.
  • the gene encoding SAS (sarcoma amplified sequence) is amplified in a subset of human sarcomas.
  • the chromosome encoding this gene, 12ql3-14, has previously been found to contain a number of growth-related genes including the GLI proto-oncogene and MDM2, a modulator of the tumor suppressor gene, p53 (Jankowski et al, supra).
  • TM4SF transmembrane 4 protein
  • polynucleotides encoding it provides additional tools for studying the function of TM4SF proteins in signal transduction and cell growth control under normal and disease conditions.
  • a new TM4SF satisfies a need in the art by providing new diagnostic or therapeutic compositions useful in the treatment or prevention of cancer, neurological disorders, and smooth muscle disorders.
  • the present invention features a new human transmembrane 4 protein hereinafter designated HT4P and characterized as having similarity to other transmembrane 4 proteins.
  • the invention features a substantially purified HT4P having the amino acid sequence shown in SEQ ID NO:l.
  • polynucleotide that encode HT4P.
  • the polynucleotide is the nucleotide sequence of SEQ ID NO:2.
  • the invention also relates to a polynucleotide sequence comprising the complement of SEQ ID NO:2 or variants thereof.
  • the invention features polynucleotide sequences which hybridize under stringent conditions to SEQ ID NO:2.
  • the invention additionally features fragments of the polynuclceotides encoding HT4P, expression vectors and host cells comprising polynucleotides that encode HT4P and a method for producing HT4P using the vectors and host cells.
  • the present invention also features antibodies which bind specifically to HT4P, and pharmaceutical compositions comprising substantially purified HT4P.
  • the invention also features agonists and antagonists of HT4P.
  • the invention also provides methods for treating disorders associated with expression of HT4P by administration of HT4P and methods for detection of polynucleotides encoding a transmembrane 4 protein in a biological sample.
  • FIGS. 1A, IB and 1C show the amino acid sequence (SEQ ID NO:l) and nucleic acid sequence (SEQ ID NO:2) of HT4P.
  • the alignment was produced using MacDNASIS PROTM software (Hitachi Software Engineering Co., Ltd., San Bruno, CA).
  • Figure 2 shows the amino acid sequence alignments among HT4P (SEQ ID NO:l), a human SAS member of TM4SF (GI 457937; SEQ ID NO:3), and a pig SAS (GI 971980; SEQ ID NO:4).
  • the alignment was produced using the multisequence alignment program of DNASTARTM software (DNASTAR Inc, Madison WI).
  • Figures 3A, 3B, and 3C show the hydrophobicity plots (DNASTARTM) for HT4P, SEQ ID NO: 1; human SAS, SEQ ID NO:3; and pig SAS (SEQ ID NO:4).
  • the positive X axis reflects amino acid position, and the positive Y axis, hydrophobicity.
  • Nucleic acid sequence refers to an oligonucleotide, nucleotide, or polynucleotide, and fragments or portions thereof, and to DNA or RNA of genomic or synthetic origin which may be single- or double-stranded, and represent the sense or antisense strand.
  • amino acid sequence refers to an oligopeptide, peptide, polypeptide, or protein sequence, and fragments or portions thereof, and to naturally occurring or synthetic molecules.
  • amino acid sequence is recited herein to refer to an amino acid sequence of a naturally occurring protein molecule
  • amino acid sequence and like terms, such as “polypeptide” or “protein” are not meant to limit the amino acid sequence to the complete, native amino acid sequence associated with the recited protein molecule.
  • Peptide nucleic acid refers to a molecule which comprises an oligomer to which an amino acid residue, such as lysine, and an amino group have been added. These small molecules, also designated anti-gene agents, stop transcript elongation by binding to their complementary strand of nucleic acid (Nielsen, P.E. et al. (1993) Anticancer Drug Des. 8:53-63).
  • HT4P refers to the amino acid sequences of substantially purified HT4P obtained from any species, particularly mammalian, including bovine, ovine, porcine, murine, equine, and preferably human, from any source whether natural, synthetic, semi-synthetic, or recombinant.
  • Consensus refers to a nucleic acid sequence which has been resequenced to resolve uncalled bases, or which has been extended using XL-PCRTM (Perkin Elmer, Norwalk, CT) in the 5' and/or the 3' direction and resequenced, or which has been assembled from the overlapping sequences of more than one Incyte clone using the GELVIEWTM Fragment Assembly system (GCG, Madison, WI), or which has been both extended and assembled.
  • XL-PCRTM Perkin Elmer, Norwalk, CT
  • GELVIEWTM Fragment Assembly system GELVIEWTM Fragment Assembly system
  • a “variant" of HT4P refers to an amino acid sequence that is altered by one or more amino acids.
  • the variant may have "conservative" changes, wherein a substituted amino acid has similar structural or chemical properties, e.g., replacement of leucine with isoleucine. More rarely, a variant may have "nonconservative" changes, e.g., replacement of a glycine with a tryptophan. Similar minor variations may also include amino acid deletions or insertions, or both.
  • Guidance in determining which amino acid residues may be substituted, inserted, or deleted without abolishing biological or immunological activity may be found using computer programs well known in the art, for example, DNASTAR software.
  • a “deletion”, as used herein, refers to a change in either amino acid or nucleotide sequence in which one or more amino acid or nucleotide residues, respectively, are absent.
  • An “insertion” or “addition”, as used herein, refers to a change in an amino acid or nucleotide sequence resulting in the addition of one or more amino acid or nucleotide residues, respectively, as compared to the naturally occurring molecule.
  • substitution refers to the replacement of one or more amino acids or nucleotides by different amino acids or nucleotides, respectively.
  • biologically active refers to a protein having structural, regulatory, or biochemical functions of a naturally occurring molecule.
  • immunologically active refers to the capability of the natural, recombinant, or synthetic HT4P, or any oligopeptide thereof, to induce a specific immune response in appropriate animals or cells and to bind with specific antibodies.
  • agonist refers to a molecule which, when bound to HT4P, causes a change in HT4P which modulates the activity of HT4P.
  • Agonists may include proteins, nucleic acids, carbohydrates, or any other molecules which bind to HT4P.
  • Antagonist refers to a molecule which, when bound to HT4P, blocks or modulates the biological or immunological activity of HT4P.
  • Antagonists and inhibitors may include proteins, nucleic acids, carbohydrates, or any other molecules which bind to HT4P.
  • modulate refers to a change or an alteration in the biological activity of HT4P. Modulation may be an increase or a decrease in protein activity, a change in binding characteristics, or any other change in the biological, functional or immunological 5 properties of HT4P.
  • mimetic refers to a molecule, the structure of which is developed from knowledge of the structure of HT4P or portions thereof and, as such, is able to effect some or all of the actions of serine protease TM4SF-like molecules.
  • derivative refers to the chemical modification of a nucleic 10 acid encoding HT4P or the encoded HT4P. Illustrative of such modifications would be replacement of hydrogen by an alkyl, acyl, or amino group.
  • a nucleic acid derivative would encode a polypeptide which retains essential biological characteristics of the natural molecule.
  • substantially purified refers to nucleic or amino acid sequences that are removed from their natural environment, isolated or separated, and are at least 15 60% free, preferably 75% free, and most preferably 90% free from other components with which they are naturally associated.
  • PCR polymerase chain reaction
  • hybridization refers to any process by which a strand of nucleic acid binds with a complementary strand through base pairing.
  • hybridization complex refers to a complex formed between two nucleic acid sequences by virtue of the formation of hydrogen binds between complementary 25 G and C bases and between complementary A and T bases; these hydrogen bonds may be further stabilized by base stacking interactions.
  • the two complementary nucleic acid sequences hydrogen bond in an antiparallel configuration.
  • a hybridization complex may be formed in solution (e.g., C 0 t or P ⁇ t analysis) or between one nucleic acid sequence present in solution and another nucleic acid sequence immobilized on a solid support (e.g., membranes, filters, chips, 30 pins or glass slides to which cells have been fixed for in situ hybridization).
  • complementarity refers to the natural binding of polynucleotides under permissive salt and temperature conditions by base-pairing. For example, for the sequence "A-G-T” binds to the complementary sequence "T-C-A". Complementarity between two single-stranded molecules may be “partial”, in which only some of the nucleic acids bind, or it may be complete when total complementarity exists between the single stranded molecules. The degree of complementarity between nucleic acid strands has significant effects on the efficiency and strength of hybridization between nucleic acid strands. This is of particular importance in amplification reactions, which depend upon binding between nucleic acids strands.
  • a partially complementary sequence is one that at least partially inhibits an identical sequence from hybridizing to a target nucleic acid; it is referred to using the functional term "substantially homologous.”
  • the inhibition of hybridization of the completely complementary sequence to the target sequence may be examined using a hybridization assay (Southern or northern blot, solution hybridization and the like) under conditions of low stringency.
  • a substantially homologous sequence or probe will compete for and inhibit the binding (i.e., the hybridization) of a completely homologous sequence or probe to the target sequence under conditions of low stringency.
  • low stringency conditions are such that non-specific binding is permitted; low stringency conditions require that the binding of two sequences to one another be a specific (i.e., selective) interaction.
  • the absence of non-specific binding may be tested by the use of a second target sequence which lacks even a partial degree of complementarity (e.g., less than about 30% identity); in the absence of non-specific binding, the probe will not hybridize to the second non-complementary target sequence.
  • stringent conditions is the “stringency” which occurs within a range from about Tm-5°C (5°C below the melting temperature (Tm) of the probe) to about 20°C to 25 °C below Tm. As will be understood by those of skill in the art, the stringency of hybridization may be altered in order to identify or detect identical or related polynucleotide sequences.
  • antisense refers to nucleotide sequences which are complementary to a specific DNA or RNA sequence.
  • antisense strand is used in reference to a nucleic acid strand that is complementary to the "sense” strand.
  • Antisense molecules may be produced by any method, including synthesis by ligating the gene(s) of interest in a reverse orientation to a viral promoter which permits the synthesis of a complementary strand. Once introduced into a cell, this transcribed strand combines with natural sequences produced by the cell to form duplexes. These duplexes then block either the further transcription or translation. In this manner, mutant phenotypes may be generated.
  • the designation "negative” is sometimes used in reference to the antisense strand, and "positive” is sometimes used in reference to the sense strand.
  • portion refers to fragments of that protein.
  • the fragments may range in size from four amino acid residues to the entire amino acid sequence minus one amino acid.
  • a protein "comprising at least a portion of the amino acid sequence of SEQ ID NO: 1" encompasses the full-length human HT4P and fragments thereof.
  • Transformation describes a process by which exogenous DNA enters and changes a recipient cell. It may occur under natural or artificial conditions using various methods well known in the art. Transformation may rely on any known method for the insertion of foreign nucleic acid sequences into a prokaryotic or eukaryotic host cell. The method is selected based on the host cell being transformed and may include, but is not limited to, viral infection, electroporation, lipofection, and particle bombardment.
  • Such "transformed” cells include stably transformed cells in which the inserted DNA is capable of replication either as an autonomously replicating plasmid or as part of the host chromosome. They also include cells which transiently express the inserted DNA or RNA for limited periods of time.
  • antigenic determinant refers to that portion of a molecule that makes contact with a particular antibody (i.e., an epitope).
  • a protein or fragment of a protein is used to immunize a host animal, numerous regions of the protein may induce the production of antibodies which bind specifically to a given region or three-dimensional structure on the protein; these regions or structures are referred to as antigenic determinants.
  • An antigenic determinant may compete with the intact antigen (i.e., the immunogen used to elicit the immune response) for binding to an antibody.
  • telomere binding in reference to the interaction of an antibody and a protein or peptide, mean that the interaction is dependent upon the presence of a particular structure (i.e., the antigenic determinant or epitope) on the protein; in other words, the antibody is recognizing and binding to a specific protein structure rather than to proteins in general. For example, if an antibody is specific for epitope "A”, the presence of a protein containing epitope A (or free, unlabeled A) in a reaction containing labeled "A” and the antibody will reduce the amount of labeled A bound to the antibody.
  • a biological sample suspected of containing nucleic acid encoding HT4P or fragments thereof may comprise a cell, chromosomes isolated from a cell (e.g., a spread of metaphase chromosomes), genomic DNA (in solution or bound to a solid support such as for Southern analysis), RNA (in solution or bound to a solid support such as for northern analysis), cDNA (in solution or bound to a solid support), an extract from cells or a tissue, and the like.
  • "Alterations" in the polynucleotide of SEQ ID NO:2, as used herein, comprise any alteration in the sequence of polynucleotides encoding HT4P including deletions, insertions, and point mutations that may be detected using hybridization assays.
  • alterations to the genomic DNA sequence which encodes HT4P e.g., by alterations in the pattern of restriction fragment length polymorphisms capable of hybridizing to SEQ ID NO:2
  • the inability of a selected fragment of SEQ ID NO:2 to hybridize to a sample of genomic DNA e.g., using allele-specific oligonucleotide probes
  • improper or unexpected hybridization such as hybridization to a locus other than the normal chromosomal locus for the polynucleotide sequence encoding HT4P (e.g., using fluorescent in situ hybridization [FISH] to metaphase chromosomes spreads).
  • FISH fluorescent in situ hybridization
  • antibody refers to intact molecules as well as fragments thereof, such as Fa, F(ab') 2 , and Fv, which are capable of binding the epitopic determinant.
  • Antibodies that bind HT4P polypeptides can be prepared using intact polypeptides or fragments containing small peptides of interest as the immunizing antigen.
  • the polypeptide or peptide used to immunize an animal can be derived from the transition of RNA or synthesized chemically, and can be conjugated to a carrier protein, if desired. Commonly used carriers that are chemically coupled to peptides include bovine serum albumin and thyroglobulin.
  • the coupled peptide is then used to immunize the animal (e.g., a mouse, a rat, or a rabbit).
  • animal e.g., a mouse, a rat, or a rabbit.
  • humanized antibody refers to antibody molecules in which amino acids have been replaced in the non-antigen binding regions in order to more closely resemble a human antibody, while still retaining the original binding ability.
  • the invention is based on the discovery of a new human transmembrane 4 protein
  • H4P the polynucleotides encoding HT4P
  • use of these compositions for the diagnosis, prevention, or treatment of cancer, neurological disorders, and smooth muscle disorders.
  • Nucleic acids encoding the human HT4P of the present invention were first identified in Incyte Clone 2279874 from the normal prostate cDNA library (PROSNON01) through a computer search for amino acid sequence alignments.
  • a consensus sequence, SEQ ID NO:2 was derived from the following overlapping and/or extended nucleic acid sequences: Incyte Clones 638873/ BRSTNOT03, 1418260/ KIDNNOT09, 1444655/ THYRNOT03, 2228841/ PROSNOT16, 2243238/ PANCTUT02, 2279874/ PROSNON01, and 2726714/ OVARTUT05.
  • the invention encompasses a polypeptide comprising the amino acid sequence of SEQ ID NO: 1 as shown in Figures 1 A, IB and 1C.
  • HT4P is 204 amino acids in length and has potential N-linked glycosylation sites at Nl 13 and N137. Cysteine residues at C3, C8, C12, C14, C94, C96, C128, C129, C140, C144, C151, and C154 represent potential disulfide bridging sites.
  • a potential casein kinase II phosphorylation site is located at SI 77, and potential protein kinase C phosphorylation sites are located at S42, S 117, and T187.
  • HT4P has chemical and structural homology with human SAS (GI 457937; SEQ ID NO:3) and pig SAS (GI 971980; SEQ ID NO:4).
  • HT4P shares 61% and 68% identity with the human and pig SAS proteins, respectively.
  • HT4P shares several features characteristic of TM4FS proteins.
  • the four conserved hydrophobic, transmembrane domains are Tl (K10-G34), T2 (V48-Q72), T3 (V73-A98) and T4 (F168-R191).
  • Tl contains the highly conserved asparagine residue, N17, and T3 and T4 contain conserved residues at Q89 and El 80, respectively.
  • the major hydrophilic, extracellular domain in HT4P is located approximately between residues N100 and R167. This region of HT4P is the most divergent in terms of sequence similarity to the human SAS which suggests a different ligand. Several cysteine residues located in this region are conserved and are believed to be important for correct folding of this domain.
  • the two N-linked glycosylation sites of HT4P are located in the hydrophilic domain.
  • the potential casein kinase II phosphorylation site in HT4P at S 177 is shared by the human SAS. As illustrated by Figures 3 A, 3B, and 3C, HT4P and the human and pig SAS proteins have rather similar hydrophobicity plots.
  • the four hydrophobic, transmembrane domains of HT4P are centered approximately at residues 30, 60, 90, and 190.
  • the major hydrophilic, extracellular domain of HT4P is centered approximately at residue 140.
  • Northern analysis shows the expression of HT4P in various libraries, 41% of which are associated with cancer and immortalized cell lines, 33% of which are associated with smooth muscle and the sympathetic nervous system, and 18% of which are associated with the brain and neural tissues.
  • the invention also encompasses HT4P variants.
  • a preferred HT4P variant is one having at least 80%, and more preferably 90%, amino acid sequence identity to the HT4P amino acid sequence (SEQ ED NO: 1).
  • a most preferred HT4P variant is one having at least 95% amino acid sequence identity to SEQ ED NO: 1.
  • the invention also encompasses polynucleotide which encode HT4P. Accordingly, any nucleic acid sequence which encodes the amino acid sequence of HT4P can be used to generate recombinant molecules which express HT4P.
  • the invention encompasses the polynucleotide comprising the nucleic acid sequence of SEQ ED NO:2 as shown in Figures 1A, IB and lC.
  • nucleotide sequences encoding HT4P may be produced.
  • the invention contemplates each and every possible variation of nucleotide sequence that could be made by selecting combinations based on possible codon choices. These combinations are made in accordance with the standard triplet genetic code as applied to the nucleotide sequence of naturally occurring HT4P, and all such variations are to be considered as being specifically disclosed.
  • nucleotide sequences which encode HT4P and its variants are preferably capable of hybridizing to the nucleotide sequence of the naturally occurring HT4P under appropriately selected conditions of stringency, it may be advantageous to produce nucleotide sequences encoding HT4P or its derivatives possessing a substantially different codon usage. Codons may be selected to increase the rate at which expression of the peptide occurs in a particular prokaryotic or eukaryotic host in accordance with the frequency with which particular codons are utilized by the host.
  • RNA transcripts having more desirable properties such as a greater half-life, than transcripts produced from the naturally occurring sequence.
  • the invention also encompasses production of DNA sequences, or portions thereof, which encode HT4P and its derivatives, entirely by synthetic chemistry.
  • the synthetic sequence may be inserted into any of the many available expression vectors and cell systems using reagents that are well known in the art at the time of the filing of this application.
  • synthetic chemistry may be used to introduce mutations into a sequence encoding HT4P or any portion thereof.
  • polynucleotide sequences that are capable of hybridizing to the claimed nucleotide sequences, and in particular, those shown in SEQ ED NO:2, under various conditions of stringency.
  • Hybridization conditions are based on the melting temperature (Tm) of the nucleic acid binding complex or probe, as taught in Wahl, G.M. and S.L. Berger (1987; Methods Enzymol. 152:399-407) and Kimmel, A.R. (1987; Methods Enzymol. 152:507-511), and may be used at a defined stringency.
  • Altered nucleic acid sequences encoding HT4P which are encompassed by the invention include deletions, insertions, or substitutions of different nucleotides resulting in a polynucleotide that encodes the same or a functionally equivalent HT4P.
  • the encoded protein may also contain deletions, insertions, or substitutions of amino acid residues which produce a silent change and result in a functionally equivalent HT4P. Deliberate amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues as long as the biological activity of HT4P is retained.
  • negatively charged amino acids may include aspartic acid and glutamic acid; positively charged amino acids may include lysine and arginine; and amino acids with uncharged polar head groups having similar hydrophilicity values may include leucine, isoleucine, and valine; glycine and alanine; asparagine and glutamine; serine and threonine; phenylalanine and tyrosine.
  • alleles of the genes encoding HT4P are also included within the scope of the present invention.
  • an "allele” or “allelic sequence” is an alternative form of the gene which may result from at least one mutation in the nucleic acid sequence. Alleles may result in altered mRNAs or polypeptides whose structure or function may or may not be altered. Any given gene may have none, one, or many allelic forms. Common mutational changes which give rise to alleles are generally ascribed to natural deletions, additions, or substitutions of nucleotides. Each of these types of changes may occur alone, or in combination with the others, one or more times in a given sequence.
  • Methods for DNA sequencing which are well known and generally available in the art may be used to practice any embodiments of the invention.
  • the methods may employ such enzymes as the Klenow fragment of DNA polymerase I, Sequenase® (US Biochemical Corp, Cleveland, OH), Taq polymerase (Perkin Elmer), thermostable T7 polymerase (Amersham, Chicago, EL), or combinations of recombinant polymerases and proofreading exonucleases such as the ELONGASE Amplification System marketed by Gibco BRL (Gaithersburg, MD).
  • the process is automated with machines such as the Hamilton Micro Lab 2200
  • nucleic acid sequences encoding HT4P may be extended utilizing a partial nucleotide sequence and employing various methods known in the art to detect upstream sequences such as promoters and regulatory elements. For example, one method which may be employed,
  • PCR "restriction-site" PCR, uses universal primers to retrieve unknown sequence adjacent to a known locus (Sarkar, G. (1993) PCR Methods Applic. 2:318-322).
  • genomic DNA is first amplified in the presence of primer to linker sequence and a primer specific to the known region.
  • the amplified sequences are then subjected to a second round of PCR with the same linker primer and another specific primer internal to the first one. Products of each round of PCR are
  • RNA polymerase 15 transcribed with an appropriate RNA polymerase and sequenced using reverse transcriptase.
  • Inverse PCR may also be used to amplify or extend sequences using divergent primers based on a known region (Triglia, T. et al. (1988) Nucleic Acids Res. 16:8186).
  • the primers may be designed using OLIGO 4.06 Primer Analysis software (National Biosciences Inc., Madison, MN), or another appropriate program, to be 22-30 nucleotides in length, to have a GC content of
  • the method uses several restriction enzymes to generate a suitable fragment in the known region of a gene. The fragment is then circularized by intramolecular ligation and used as a PCR template.
  • capture PCR involves PCR amplification of DNA fragments adjacent to a known sequence in human and yeast artificial chromosome DNA
  • Genomic libraries may be useful for extension of sequence into the 5' and 3' non-transcribed regulatory regions.
  • Capillary electrophoresis systems which are commercially available may be used to analyze the size or confirm the nucleotide sequence of sequencing or PCR products.
  • capillary sequencing may employ flowable polymers for electrophoretic separation, four different fluorescent dyes (one for each nucleotide) which are laser activated, and detection of the emitted wavelengths by a charge coupled devise camera.
  • Output light intensity may be converted to electrical signal using appropriate software (e.g. GenotyperTM and Sequence NavigatorTM, Perkin Elmer) and the entire process from loading of samples to computer analysis and electronic data display may be computer controlled.
  • Capillary electrophoresis is especially preferable for the sequencing of small pieces of DNA which might be present in limited amounts in a particular sample.
  • polynucleotide sequences or fragments thereof which encode HT4P, or fusion proteins or functional equivalents thereof may be used in recombinant DNA molecules to direct expression of HT4P in appropriate host cells. Due to the inherent degeneracy of the genetic code, other DNA sequences which encode substantially the same or a functionally equivalent amino acid sequence may be produced and these sequences may be used to clone and express HT4P.
  • codons preferred by a particular prokaryotic or eukaryotic host can be selected to increase the rate of protein expression or to produce a recombinant RNA transcript having desirable properties, such as a half-life which is longer than that of a transcript generated from the naturally occurring sequence.
  • nucleotide sequences of the present invention can be engineered using methods generally known in the art in order to alter HT4P encoding sequences for a variety of reasons, including but not limited to, alterations which modify the cloning, processing, and/or expression of the gene product.
  • DNA shuffling by random fragmentation and PCR reassembly of gene fragments and synthetic oligonucleotides may be used to engineer the nucleotide sequences.
  • site-directed mutagenesis may be used to insert new restriction sites, alter glycosylation patterns, change codon preference, produce splice variants, or introduce mutations, and so forth.
  • natural, modified, or recombinant nucleic acid sequences encoding HT4P may be ligated to a heterologous sequence to encode a fusion protein.
  • a heterologous sequence to encode a fusion protein.
  • a fusion protein may also be engineered to contain a cleavage site located between the HT4P encoding sequence and the heterologous protein sequence, so that HT4P may be cleaved and purified away from the heterologous moiety.
  • sequences encoding HT4P may be synthesized, in whole or in part, using chemical methods well known in the art (see Caruthers, M.H. et al. (1980) Nucl. Acids Res. Symp. Ser. 215-223, Horn, T. et al. (1980) Nucl. Acids Res. Symp. Ser. 225-232).
  • the protein itself may be produced using chemical methods to synthesize the amino acid sequence of HT4P, or a portion thereof.
  • peptide synthesis can be performed using various solid-phase techniques (Roberge, J.Y. et al. (1995) Science 269:202-204) and automated synthesis may be achieved, for example, using the ABI 431 A Peptide Synthesizer (Perkin Elmer).
  • the newly synthesized peptide may be substantially purified by preparative high performance liquid chromatography (e.g., Creighton, T. (1983) Proteins. Structures and Molecular Principles. WH Freeman and Co., New York, NY).
  • the composition of the synthetic peptides may be confirmed by amino acid analysis or sequencing (e.g., the Edman degradation procedure; Creighton, supra).
  • the amino acid sequence of HT4P, or any part thereof may be altered during direct synthesis and/or combined using chemical methods with sequences from other proteins, or any part thereof, to produce a variant polypeptide.
  • nucleotide sequences encoding HT4P or functional equivalents may be inserted into appropriate expression vector, i.e., a vector which contains the necessary elements for the transcription and translation of the inserted coding sequence.
  • a variety of expression vector/host systems may be utilized to contain and express sequences encoding HT4P. These include, but are not limited to, microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with virus expression vectors (e.g., baculovirus); plant cell systems transformed with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or with bacterial expression vectors (e.g., Ti or pBR322 plasmids); or animal cell systems.
  • microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors
  • yeast transformed with yeast expression vectors e.g., insect cell systems infected with virus expression vectors (e.g., baculovirus)
  • plant cell systems transformed with virus expression vectors e.g., cauliflower mosaic virus
  • control elements are those non-translated regions of the vector— enhancers, promoters, 5' and 3' untranslated regions—which interact with host cellular proteins to carry out transcription and translation. Such elements may vary in their strength and specificity. Depending on the vector system and host utilized, any number of suitable transcription and translation elements, including constitutive and inducible promoters, may be used. For example, when cloning in bacterial systems, inducible promoters such as the hybrid lacZ promoter of the Bluescript® phagemid (Stratagene, LaJolla, CA) or pSportlTM plasmid (Gibco BRL) and the like may be used.
  • inducible promoters such as the hybrid lacZ promoter of the Bluescript® phagemid (Stratagene, LaJolla, CA) or pSportlTM plasmid (Gibco BRL) and the like may be used.
  • the baculovirus polyhedrin promoter may be used in insect cells. Promoters or enhancers derived from the genomes of plant cells (e.g., heat shock, RUBISCO; and storage protein genes) or from plant viruses (e.g., viral promoters or leader sequences) may be cloned into the vector. In mammalian cell systems, promoters from mammalian genes or from mammalian viruses are preferable. If it is necessary to generate a cell line that contains multiple copies of the sequence encoding HT4P, vectors based on SV40 or EBV may be used with an appropriate selectable marker.
  • Promoters or enhancers derived from the genomes of plant cells e.g., heat shock, RUBISCO; and storage protein genes
  • plant viruses e.g., viral promoters or leader sequences
  • a number of expression vectors may be selected depending upon the use intended for HT4P.
  • vectors which direct high level expression of fusion proteins that are readily purified may be used.
  • Such vectors include, but are not limited to, the multifunctional E. coli cloning and expression vectors such as Bluescript® (Stratagene), in which the sequence encoding HT4P may be ligated into the vector in frame with sequences for the amino-terminal Met and the subsequent 7 residues of ⁇ -galactosidase so that a hybrid protein is produced; pEN vectors (Van Heeke, G. and S.M. Schuster (1989) J. Biol. Chem.
  • pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST).
  • GST glutathione S-transferase
  • fusion proteins are soluble and can easily be purified from lysed cells by adsorption to glutathione-agarose beads followed by elution in the presence of free glutathione.
  • Proteins made in such systems may be designed to include heparin, thrombin, or factor XA protease cleavage sites so that the cloned polypeptide of interest can be released from the GST moiety at will.
  • Saccharomyces cerevisiae a number of vectors containing constitutive or inducible promoters such as alpha factor, alcohol oxidase, and PGH may be used.
  • constitutive or inducible promoters such as alpha factor, alcohol oxidase, and PGH.
  • sequences encoding HT4P may be driven by any of a number of promoters.
  • viral promoters such as the 35S and 19S promoters of CaMV may be used alone or in combination with the omega leader sequence from TMV (Takamatsu, N. (1987) EMBO J. 3:1271-1680; Brogue, R. et al. (1984) Science 224:838-843; and Winter, J. et al. (1991) Results Probl. Cell Differ.
  • constructs can be introduced into plant cells by direct DNA transformation or pathogen-mediated transfection.
  • pathogen-mediated transfection Such techniques are described in a number of generally available reviews (see, for example, Hobbs, S. or Murry, L.E. in McGraw Hill Yearbook of Science and Technology (1992) McGraw Hill, New York, NY; pp. 191-196.
  • An insect system may also be used to express HT4P.
  • Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes in Spodoptera frugiperda cells or in Trichoplusia larvae.
  • the sequences encoding HT4P may be cloned into a non-essential region of the virus, such as the polyhedrin gene, and placed under control of the polyhedrin promoter. Successful insertion of HT4P will render the polyhedrin gene inactive and produce recombinant virus lacking coat protein.
  • the recombinant viruses may then be used to infect, for example, S. frugiperda cells or Trichoplusia larvae in which HT4P may be expressed (Engelhard, E.K. et al. (1994) Proc. Nat. Acad. Sci. 91:3224-3227).
  • a number of viral-based expression systems may be utilized.
  • sequences encoding HT4P may be ligated into an adenovirus transcription/translation complex consisting of the late promoter and tripartite leader sequence. Insertion in a non-essential El or E3 region of the viral genome may be used to obtain a viable virus which is capable of expressing HT4P in infected host cells (Logan, J. and Shenk, T. (1984) Proc. Natl. Acad. Sci. 81:3655-3659).
  • transcription enhancers such as the Rous sarcoma virus (RSV) enhancer, may be used to increase expression in mammalian host cells.
  • RSV Rous sarcoma virus
  • Specific initiation signals may also be used to achieve more efficient translation of sequences encoding HT4P. Such signals include the ATG initiation codon and adjacent sequences. In cases where sequences encoding HT4P, its initiation codon, and upstream sequences are inserted into the appropriate expression vector, no additional transcriptional or translational control signals may be needed. However, in cases where only coding sequence, or a portion thereof, is inserted, exogenous translational control signals including the ATG initiation codon should be provided. Furthermore, the initiation codon should be in the correct reading frame to ensure translation of the entire insert. Exogenous translational elements and initiation codons may be of various origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of enhancers which are appropriate for the particular cell system which is used, such as those described in the literature (Scharf, D. et al. (1994) Results Probl. Cell Differ. 20:125-162).
  • a host cell strain may be chosen for its ability to modulate the expression of the inserted sequences or to process the expressed protein in the desired fashion.
  • modifications of the polypeptide include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation, and acylation.
  • Post-translational processing which cleaves a "prepro" form of the protein may also be used to facilitate correct insertion, folding and/or function.
  • Different host cells such as CHO, HeLa, MDCK, HEK293, and WI38, which have specific cellular machinery and characteristic mechanisms for such post-translational activities, may be chosen to ensure the correct modification and processing of the foreign protein.
  • cell lines which stably express HT4P may be transformed using expression vectors which may contain viral origins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate vector. Following the introduction of the vector, cells may be allowed to grow for 1-2 days in an enriched media before they are switched to selective media.
  • the purpose of the selectable marker is to confer resistance to selection, and its presence allows growth and recovery of cells which successfully express the introduced sequences.
  • Resistant clones of stably transformed cells may be proliferated using tissue culture techniques appropriate to the cell type.
  • any number of selection systems may be used to recover transformed cell lines. These include, but are not limited to, the herpes simplex virus thymidine kinase (Wigler, M. et al. (1977) Cell 11:223-32) and adenine phosphoribosyltransferase (Lowy, I. et al. (1980) Cell 22:817-23) genes which can be employed in tk " or aprt " cells, respectively. Also, antimetabolite, antibiotic or herbicide resistance can be used as the basis for selection; for example, dhfr which confers resistance to methotrexate (Wigler, M. et al. (1980) Proc. Natl. Acad. Sci.
  • npt which confers resistance to the aminoglycosides neomycin and G-418 (Colbere-Garapin, F. et al (1981) J. Mol. Biol. 150:1-14) and als or pat, which confer resistance to chlorsulfuron and phosphinotricin acetyltransferase, respectively (Murry, supra). Additional selectable genes have been described, for example, trpB, which allows cells to utilize indole in place of tryptophan, or hisD, which allows cells to utilize histinol in place of histidine (Hartman, S.C. and R.C. Mulligan (1988) Proc. Natl. Acad. Sci.
  • marker gene expression suggests that the gene of interest is also present, its presence and expression may need to be confirmed.
  • sequence encoding HT4P is inserted within a marker gene sequence
  • recombinant cells containing sequences encoding HT4P can be identified by the absence of marker gene function.
  • a marker gene can be placed in tandem with a sequence encoding HT4P under the control of a single promoter. Expression of the marker gene in response to induction or selection usually indicates expression of the tandem gene as well.
  • host cells which contain the nucleic acid sequence encoding HT4P and express HT4P may be identified by a variety of procedures known to those of skill in the art. These procedures include, but are not limited to, DNA-DNA or DNA-RNA hybridizations and protein bioassay or immunoassay techniques which include membrane, solution, or chip based technologies for the detection and/or quantification of nucleic acid or protein.
  • polynucleotide sequences encoding HT4P can be detected by DNA-DNA or DNA-RNA hybridization or amplification using probes or portions or fragments of polynucleotides encoding HT4P.
  • Nucleic acid amplification based assays involve the use of oligonucleotides or oligomers based on the sequences encoding HT4P to detect transformants containing DNA or RNA encoding HT4P.
  • oligonucleotides or “oligomers” refer to a nucleic acid sequence of at least about 10 nucleotides and as many as about 60 nucleotides, preferably about 15 to 30 nucleotides, and more preferably about 20-25 nucleotides, which can be used as a probe or amplimer.
  • ELISA enzyme-linked immunosorbent assay
  • RIA radioimmunoassay
  • FACS fluorescence activated cell sorting
  • a two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering epitopes on HT4P is preferred, but a competitive binding assay may be employed.
  • a competitive binding assay may be employed.
  • Means for producing labeled hybridization or PCR probes for detecting sequences related to polynucleotides encoding HT4P include oligolabeling, nick translation, end-labeling or PCR amplification using a labeled nucleotide.
  • sequences encoding HT4P, or any portions thereof may be cloned into a vector for the production of an mRNA probe.
  • RNA polymerase such as T7, T3, or SP6 and labeled nucleotides.
  • T7, T3, or SP6 RNA polymerase
  • Suitable reporter molecules or labels include radionuclides, enzymes, fluorescent, chemiluminescent, or chromogenic agents as well as substrates, cofactors, inhibitors, magnetic particles, and the like.
  • Host cells transformed with nucleotide sequences encoding HT4P may be cultured under conditions suitable for the expression and recovery of the protein from cell culture.
  • the protein produced by a recombinant cell may be secreted or contained intracellularly depending on the sequence and/or the vector used.
  • expression vectors containing polynucleotides which encode HT4P may be designed to contain signal sequences which direct secretion of HT4P through a prokaryotic or eukaryotic cell membrane.
  • Other recombinant constructions may be used to join sequences encoding HT4P to nucleotide sequence encoding a polypeptide domain which will facilitate purification of soluble proteins.
  • Such purification facilitating domains include, but are not limited to, metal chelating peptides such as histidine-tryptophan modules that allow purification on immobilized metals, protein A domains that allow purification on immobilized immunoglobulin, and the domain utilized in the FLAGS extension/affinity purification system (Ernmunex Corp., Seattle, WA).
  • metal chelating peptides such as histidine-tryptophan modules that allow purification on immobilized metals
  • protein A domains that allow purification on immobilized immunoglobulin
  • the domain utilized in the FLAGS extension/affinity purification system Ernmunex Corp., Seattle, WA.
  • cleavable linker sequences such as those specific for Factor XA or enterokinase (Invitrogen, San Diego, CA) between the purification domain and HT4P may be used to facilitate purification.
  • One such expression vector provides for expression of a fusion protein containing HT4P and a nucleic acid encoding 6 histidine residues preceding a thioredoxin or an enterokinase cleavage site.
  • the histidine residues facilitate purification on EMIAC (immobilized metal ion affinity chromatography as described in Porath, J. et al. (1992, Prot. Exp. Purif. 3:263-281) while the enterokinase cleavage site provides a means for purifying HT4P from the fusion protein.
  • EMIAC immobilized metal ion affinity chromatography as described in Porath, J. et al. (1992, Prot. Exp. Purif. 3:263-281
  • the enterokinase cleavage site provides a means for purifying HT4P from the fusion protein.
  • fragments of HT4P may be produced by direct peptide synthesis using solid-phase techniques Merrifield J. (1963) J. Am. Chem. Soc. 85:2149-2154). Protein synthesis may be performed using manual techniques or by automation. Automated synthesis may be achieved, for example, using Applied Biosystems 431 A Peptide Synthesizer (Perkin Elmer). Various fragments of HT4P may be chemically synthesized separately and combined using chemical methods to produce the full length molecule. THERAPEUTICS
  • HT4P HT4P
  • human SAS pig SAS
  • northern analysis shows the expression of HT4P in tissues associated with cancer, smooth muscle, and neural tissues. HT4P therefore appears to be associated with the development of cancer, smooth muscle disorders, and neurological disorders.
  • HT4P or a fragment or derivative thereof may be administered to a subject to treat or prevent a neurological disorder.
  • disorders may include, but are not limited to, akathesia, Alzheimer's disease, amnesia, amyotrophic lateral sclerosis, bipolar disorder, catatonia, cerebral neoplasms, dementia, depression, Down's syndrome, tardive dyskinesia, dystonias, epilepsy, Huntington's disease, multiple sclerosis, neurofibromatosis, Parkinson's disease, paranoid psychoses, schizophrenia, and Tourette's disorder.
  • a vector capable of expressing HT4P, or a fragment or a derivative thereof may also be administered to a subject to treat any of the neurological disorders listed above.
  • HT4P or a fragment or derivative thereof may be administered to a subject to treat or prevent a smooth muscle disorder.
  • a smooth muscle disorder is defined as any impairment or alteration in the normal action of smooth muscle and may include, but is not limited to, angina, anaphylactic shock, arrhythmias, asthma, cardiovascular shock, Cushing's syndrome, hypertension, hypoglycemia, myocardial infarction, migraine, and pheochromocytoma, and myopathies including cardiomyopathy, encephalopathy, epilepsy, Kearns-Sayre syndrome, lactic acidosis, myoclonic disorder, and ophthalmoplegia.
  • Smooth muscle includes, but is not limited to, that of the blood vessels, gastrointestinal tract, heart, and uterus.
  • a vector capable of expressing HT4P, or a fragment or a derivative thereof may also be administered to a subject to treat any of the smooth muscle disorders listed above.
  • an antagonist or inhibitor of HT4P may be administered to a subject to treat or prevent cancer, including astrocytoma, glioma, ganglioneuroma, neurocytoma, neuroblastoma. adenocarcinoma, sarcoma, melanoma, lymphoma, leukemia, and myeloma.
  • types of cancer may include, but are not limited to, cancer of the adrenal gland, bladder, bone, brain, breast, gastrointestinal tract, heart, kidney, liver, lung, ovary, pancreas, paraganglia, parathyroid, prostate, salivary glands, skin, spleen, testis, thyroid, and uterus.
  • antibodies which are specific for HT4P may be used directly as an antagonist, or indirectly as a targeting or delivery mechanism for bringing a pharmaceutical agent to cells or tissues which express HT4P.
  • the complement of the polynucleotide encoding HT4P or an antisense molecule may be administered to a subject to treat or prevent any of the types of cancer listed above.
  • any of the therapeutic proteins, antagonists, antibodies, agonists, antisense sequences or vectors of this invention may be administered in combination with other appropriate therapeutic agents. Selection of the appropriate agents for use in combination therapy may be made by one of ordinary skill in the art, according to conventional pharmaceutical principles.
  • the combination of therapeutic agents may act synergistically to effect the treatment or prevention of the various disorders described above. Using this approach, one may be able to achieve therapeutic efficacy with lower dosages of each agent, thus reducing the potential for adverse side effects.
  • Antagonists or inhibitors of HT4P may be produced using methods which are generally known in the art.
  • purified HT4P may be used to produce antibodies or to screen libraries of pharmaceutical agents to identify those which specifically bind HT4P.
  • the antibodies may be generated using methods that are well known in the art. Such antibodies may include, but are not limited to, polyclonal, monoclonal, chimeric, single chain, Fab fragments, and fragments produced by a Fab expression library. Neutralizing antibodies, (i.e., those which inhibit dimer formation) are especially preferred for therapeutic use.
  • various hosts including goats, rabbits, rats, mice, humans, and others, may be immunized by injection with HT4P or any fragment or oligopeptide thereof which has immunogenic properties.
  • various adjuvants may be used to increase immunological response.
  • adjuvants include, but are not limited to, Freund's, mineral gels such as aluminum hydroxide, and surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol.
  • BCG Bacilli Calmette-Guerin
  • Corynebacterium parvum are especially preferable. It is preferred that the peptides, fragments, or oligopeptides used to induce antibodies to
  • HT4P have an amino acid sequence consisting of at least five amino acids, and more preferably at least 10 amino acids. It is also preferable that they are identical to a portion of the amino acid sequence of the natural protein, and they may contain the entire amino acid sequence of a small, naturally occurring molecule. Short stretches of HT4P amino acids may be fused with those of another protein such as keyhole limpet hemocyanin and antibody produced against the chimeric molecule.
  • Monoclonal antibodies to HT4P may be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique, the human B-cell hybridoma technique, and the EBV-hybridoma technique (Kohler, G. et al. (1975) Nature 256:495-497; Kozbor, D. et al. (1985) J. Immunol. Methods 81:31-42; Cote, R.J. et al. (1983) Proc. Natl. Acad. Sci. 80:2026-2030; Cole, S.P. et al. (1984) Mol. Cell Biol. 62:109-120).
  • chimeric antibodies the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity can be used (Morrison, S.L. et al. (1984) Proc. Natl. Acad. Sci. 81:6851-6855; Neuberger, M.S. et al. (1984) Nature 312:604-608; Takeda, S. et al. (1985) Nature 314:452-454).
  • techniques described for the production of single chain antibodies may be adapted, using methods known in the art, to produce HT4P-specific single chain antibodies.
  • Antibodies with related specificity, but of distinct idiotypic composition may be generated by chain shuffling from random combinatorial immunoglobin libraries (Burton D.R. (1991) Proc. Natl. Acad. Sci. 88: 11120-3).
  • Antibodies may also be produced by inducing in vivo production in the lymphocyte population or by screening recombinant immunoglobulin libraries or panels of highly specific binding reagents as disclosed in the literature (Orlandi, R. et al. (1989) Proc. Natl. Acad. Sci. 86: 3833-3837; Winter, G. et al. (1991) Nature 349:293-299).
  • Antibody fragments which contain specific binding sites for HT4P may also be generated.
  • fragments include, but are not limited to, the F(ab')2 fragments which can be produced by pepsin digestion of the antibody molecule and the Fab fragments which can be generated by reducing the disulfide bridges of the F(ab ⁇ )2 fragments.
  • Fab expression libraries may be constructed to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity (Huse, W.D. et al. (1989) Science 254:1275-1281).
  • Various immunoassays may be used for screening to identify antibodies having the desired specificity.
  • the polynucleotides encoding HT4P, or any fragment thereof, or antisense molecules may be used for therapeutic purposes.
  • antisense to the polynucleotide encoding HT4P may be used in situations in which it would be desirable to block the transcription of the mRNA.
  • cells may be transformed with sequences complementary to polynucleotides encoding HT4P.
  • antisense molecules may be used to modulate HT4P activity, or to achieve regulation of gene function.
  • sense or antisense oligomers or larger fragments can be designed from various locations along the coding or control regions of sequences encoding HT4P.
  • Expression vectors derived from retro viruses, adenovirus, herpes or vaccinia viruses, or from various bacterial plasmids may be used for delivery of nucleotide sequences to the targeted organ, tissue or cell population. Methods which are well known to those skilled in the art can be used to construct recombinant vectors which will express antisense molecules complementary to the polynucleotides of the gene encoding HT4P. These techniques are described both in Sambrook et al. (supra) and in Ausubel et al. (supra).
  • Genes encoding HT4P can be turned off by transforming a cell or tissue with expression vectors which express high levels of a polynucleotide or fragment thereof which encodes HT4P. Such constructs may be used to introduce untranslatable sense or antisense sequences into a cell. Even in the absence of integration into the DNA, such vectors may continue to transcribe RNA molecules until they are disabled by endogenous nucleases. Transient expression may last for a month or more with a non-replicating vector and even longer if appropriate replication elements are part of the vector system.
  • modifications of gene expression can be obtained by designing antisense molecules, DNA, RNA, or PNA, to the control regions of the gene encoding HT4P, i.e., the promoters, enhancers, and introns. Oligonucleotides derived from the transcription initiation site, e.g., between positions -10 and +10 from the start site, are preferred. Similarly, inhibition can be achieved using "triple helix" base-pairing methodology. Triple helix pairing is useful because it causes inhibition of the ability of the double helix to open sufficiently for the binding of polymerases, transcription factors, or regulatory molecules. Recent therapeutic advances using triplex DNA have been described in the literature (Gee, J.E. et al.
  • the antisense molecules may also be designed to block translation of mRNA by preventing the transcript from binding to ribosomes.
  • Ribozymes enzymatic RNA molecules, may also be used to catalyze the specific cleavage of RNA. The mechanism of ribozyme action involves sequence-specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage.
  • Examples which may be used include engineered hammerhead motif ribozyme molecules that can specifically and efficiently catalyze endonucleolytic cleavage of sequences encoding HT4P.
  • Specific ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites which include the following sequences: GUA, GUU, and GUC. Once identified, short RNA sequences of between 15 and 20 ribonucleotides corresponding to the region of the target gene containing the cleavage site may be evaluated for secondary structural features which may render the oligonucleotide inoperable.
  • the suitability of candidate targets may also be evaluated by testing accessibility to hybridization with complementary oligonucleotides using ribonuclease protection assays.
  • Antisense molecules and ribozymes of the invention may be prepared by any method known in the art for the synthesis of nucleic acid molecules. These include techniques for chemically synthesizing oligonucleotides such as solid phase phosphoramidite chemical synthesis. Alternatively, RNA molecules may be generated by in vitro and in vivo transcription of DNA sequences encoding HT4P. Such DNA sequences may be incorporated into a wide variety of vectors with suitable RNA polymerase promoters such as T7 or SP6. Alternatively, these cDNA constructs that synthesize antisense RNA constitutively or inducibly can be introduced into cell lines, cells, or tissues.
  • RNA molecules may be modified to increase intracellular stability and half-life. Possible modifications include, but are not limited to, the addition of flanking sequences at the 5' and/or 3' ends of the molecule or the use of phosphorothioate or 2' O-methyl rather than phosphodiesterase linkages within the backbone of the molecule.
  • Delivery by transfection and by liposome injections may be achieved using methods which are well known in the art. Any of the therapeutic methods described above may be applied to any subject in need of such therapy, including, for example, mammals such as dogs, cats, cows, horses, rabbits, monkeys, and most preferably, humans.
  • An additional embodiment of the invention relates to the administration of a pharmaceutical composition, in conjunction with a pharmaceutically acceptable carrier, for any of the therapeutic effects discussed above.
  • Such pharmaceutical compositions may consist of HT4P, antibodies to HT4P, mimetics, agonists, antagonists, or inhibitors of HT4P.
  • the compositions may be administered alone or in combination with at least one other agent, such as stabilizing compound, which may be administered in any sterile, biocompatible pharmaceutical carrier, including, but not limited to, saline, buffered saline, dextrose, and water.
  • the compositions may be administered to a patient alone, or in combination with other agents, drugs or hormones.
  • compositions utilized in this invention may be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, intraventricular, transdermal, subcutaneous, intraperitoneal, intranasal, enteral, topical, sublingual, or rectal means.
  • these pharmaceutical compositions may contain suitable pharmaceutically-acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Further details on techniques for formulation and administration may be found in the latest edition of Remington's Pharmaceutical Sciences (Maack Publishing Co., Easton,
  • compositions for oral administration can be formulated using pharmaceutically acceptable carriers well known in the art in dosages suitable for oral administration.
  • Such carriers enable the pharmaceutical compositions to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for ingestion by the patient.
  • compositions for oral use can be obtained through combination of active compounds with solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients are carbohydrate or protein fillers, such as sugars, including lactose, sucrose, mannitol, or sorbitol; starch from corn, wheat, rice, potato, or other plants; cellulose, such as methyl cellulose, hydroxypropylmethyl-cellulose, or sodium carboxymethylcellulose; gums including arabic and tragacanth; and proteins such as gelatin and collagen.
  • disintegrating or solubilizing agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, alginic acid, or a salt thereof, such as sodium alginate.
  • Dragee cores may be used in conjunction with suitable coatings, such as concentrated sugar solutions, which may also contain gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • suitable coatings such as concentrated sugar solutions, which may also contain gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for product identification or to characterize the quantity of active compound, i.e., dosage.
  • compositions which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a coating, such as glycerol or sorbitol.
  • Push-fit capsules can contain active ingredients mixed with a filler or binders, such as lactose or starches, lubricants, such as talc or magnesium stearate, and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid, or liquid polyethylene glycol with or without stabilizers.
  • compositions suitable for parenteral administration may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiologically buffered saline.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • suspensions of the active compounds may be prepared as appropriate oily injection suspensions.
  • Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • penetrants appropriate to the particular barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art.
  • compositions of the present invention may be manufactured in a manner that is known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, or lyophilizing processes.
  • the pharmaceutical composition may be provided as a salt and can be formed with many acids, including but not limited to, hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents than are the corresponding free base forms.
  • the preferred preparation may be a lyophilized powder which may contain any or all of the following: 1-50 mM histidine, 0.1 %-2% sucrose, and 2-7% mannitol, at a pH range of 4.5 to 5.5, that is combined with buffer prior to use.
  • compositions suitable for use in the invention include compositions wherein the active ingredients are contained in an effective amount to achieve the intended purpose. The determination of an effective dose is well within the capability of those skilled in the art.
  • the therapeutically effective dose can be estimated initially either in cell culture assays, e.g., of neoplastic cells, or in animal models, usually mice, rabbits, dogs, or pigs.
  • the animal model may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans.
  • a therapeutically effective dose refers to that amount of active ingredient, for example HT4P or fragments thereof, antibodies of HT4P, agonists, antagonists or inhibitors of HT4P, which ameliorates the symptoms or condition.
  • Therapeutic efficacy and toxicity may be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., ED50 (the dose therapeutically effective in 50% of the population) and LD50 (the dose lethal to 50% of the population).
  • the dose ratio between therapeutic and toxic effects is the therapeutic index, and it can be expressed as the ratio, LD50/ED50.
  • Pharmaceutical compositions which exhibit large therapeutic indices are preferred.
  • the data obtained from cell culture assays and animal studies is used in formulating a range of dosage for human use.
  • the dosage contained in such compositions is preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage varies within this range depending upon the dosage form employed, sensitivity of the patient, and the route of administration.
  • the exact dosage will be determined by the practitioner, in light of factors related to the subject that requires treatment. Dosage and administration are adjusted to provide sufficient levels of the active moiety or to maintain the desired effect. Factors which may be taken into account include the severity of the disease state, general health of the subject, age, weight, and gender of the subject, diet, time and frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy. Long-acting pharmaceutical compositions may be administered every 3 to 4 days, every week, or once every two weeks depending on half-life and clearance rate of the particular formulation.
  • Normal dosage amounts may vary from 0.1 to 100,000 micrograms, up to a total dose of about 1 g, depending upon the route of administration.
  • Guidance as to particular dosages and methods of delivery is provided in the literature and generally available to practitioners in the art. Those skilled in the art will employ different formulations for nucleotides than for proteins or their inhibitors. Similarly, delivery of polynucleotides or polypeptides will be specific to particular cells, conditions, locations, etc. DIAGNOSTICS
  • antibodies which specifically bind HT4P may be used for the diagnosis of conditions or diseases characterized by expression of HT4P, or in assays to monitor patients being treated with HT4P, agonists, antagonists or inhibitors.
  • the antibodies useful for diagnostic purposes may be prepared in the same manner as those described above for therapeutics. Diagnostic assays for HT4P include methods which utilize the antibody and a label to detect HT4P in human body fluids or extracts of cells or tissues.
  • the antibodies may be used with or without modification, and may be labeled by joining them, either covalently or non-covalently, with a reporter molecule.
  • a wide variety of reporter molecules which are known in the art may be used, several of which are described above.
  • ELIS A, RIA, and FACS for measuring HT4P are known in the art and provide a basis for diagnosing altered or abnormal levels of HT4P expression.
  • Normal or standard values for HT4P expression are established by combining body fluids or cell extracts taken from normal mammalian subjects, preferably human, with antibody to HT4P under conditions suitable for complex formation. The amount of standard complex formation may be quantified by various methods, but preferably by photometric, means. Quantities of HT4P expressed in subject, control and disease, samples from biopsied tissues are compared with the standard values. Deviation between standard and subject values establishes the parameters for diagnosing disease.
  • the polynucleotides encoding HT4P may be used for diagnostic purposes.
  • the polynucleotides which may be used include oligonucleotide sequences, antisense RNA and DNA molecules, and PNAs.
  • the polynucleotides may be used to detect and quantitate gene expression in biopsied tissues in which expression of HT4P may be correlated with disease.
  • the diagnostic assay may be used to distinguish between absence, presence, and excess expression of HT4P, and to monitor regulation of HT4P levels during therapeutic intervention.
  • hybridization with PCR probes which are capable of detecting polynucleotide sequences, including genomic sequences, encoding HT4P or closely related molecules, may be used to identify nucleic acid sequences which encode HT4P.
  • the specificity of the probe whether it is made from a highly specific region, e.g., 10 unique nucleotides in the 5' regulatory region, or a less specific region, e.g., especially in the 3' coding region, and the stringency of the hybridization or amplification (maximal, high, intermediate, or low) will determine whether the probe identifies only naturally occurring sequences encoding HT4P, alleles, or related sequences.
  • Probes may also be used for the detection of related sequences, and should preferably contain at least 50% of the nucleotides from any of the HT4P encoding sequences.
  • the hybridization probes of the subject invention may be DNA or RNA and derived from the nucleotide sequence of SEQ ED NO:2 or from genomic sequence including promoter, enhancer elements, and introns of the naturally occurring HT4P.
  • Means for producing specific hybridization probes for DNAs encoding HT4P include the cloning of nucleic acid sequences encoding HT4P or HT4P derivatives into vectors for the production of mRNA probes.
  • Such vectors are known in the art, commercially available, and may be used to synthesize RNA probes in vitro by means of the addition of the appropriate RNA polymerases and the appropriate labeled nucleotides.
  • Hybridization probes may be labeled by a variety of reporter groups, for example, radionuclides such as 32P or 35S, or enzymatic labels, such as alkaline phosphatase coupled to the probe via avidin/biotin coupling systems, and the like.
  • Polynucleotide sequences encoding HT4P may be used for the diagnosis of conditions or diseases which are associated with expression of HT4P.
  • conditions or diseases include neurological disorders such as akathesia, Alzheimer's disease, amnesia, amyotrophic lateral sclerosis, bipolar disorder, catatonia, cerebral neoplasms, dementia, depression, Down's syndrome, tardive dyskinesia, dystonias, epilepsy, Huntington's disease, multiple sclerosis, neurofibromatosis, Parkinson's disease, paranoid psychoses, schizophrenia, and Tourette's disorder; smooth muscle disorders such as angina, anaphylactic shock, arrhythmias, asthma, cardiovascular shock, Cushing's syndrome, hypertension, hypoglycemia, myocardial infarction, migraine, and pheochromocytoma, and myopathies including cardiomyopathy, encephalopathy, epilepsy, Kearns-Sayre syndrome, lactic acidosis, myo
  • the polynucleotide sequences encoding HT4P may be used in Southern or northern analysis, dot blot, or other membrane-based technologies; in PCR technologies; or in dip stick, pin, ELIS A or chip assays utilizing fluids or tissues from patient biopsies to detect altered HT4P expression. Such qualitative or quantitative methods are well known in the art.
  • the nucleotide sequences encoding HT4P may be useful in assays that detect activation or induction of various cancers, particularly those mentioned above.
  • the nucleotide sequences encoding HT4P may be labeled by standard methods, and added to a fluid or tissue sample from a patient under conditions suitable for the formation of hybridization complexes.
  • the sample is washed and the signal is quantitated and compared with a standard value. If the amount of signal in the biopsied or extracted sample is significantly altered from that of a comparable control sample, the nucleotide sequences have hybridized with nucleotide sequences in the sample, and the presence of altered levels of nucleotide sequences encoding HT4P in the sample indicates the presence of the associated disease.
  • Such assays may also be used to evaluate the efficacy of a particular therapeutic treatment regimen in animal studies, in clinical trials, or in monitoring the treatment of an individual patient.
  • a normal or standard profile for expression is established. This may be accomplished by combining body fluids or cell extracts taken from normal subjects, either animal or human, with a sequence, or a fragment thereof, which encodes HT4P, under conditions suitable for hybridization or amplification. Standard hybridization may be quantified by comparing the values obtained from normal subjects with those from an experiment where a known amount of a substantially purified polynucleotide is used. Standard values obtained from normal samples may be compared with values obtained from samples from patients who are symptomatic for disease. Deviation between standard and subject values is used to establish the presence of disease.
  • hybridization assays may be repeated on a regular basis to evaluate whether the level of expression in the patient begins to approximate that which is observed in the normal patient.
  • the results obtained from successive assays may be used to show the efficacy of treatment over a period ranging from several days to months.
  • the presence of a relatively high amount of transcript in biopsied tissue from an individual may indicate a predisposition for the development of the disease, or may provide a means for detecting the disease prior to the appearance of actual clinical symptoms.
  • a more definitive diagnosis of this type may allow health professionals to employ preventative measures or aggressive treatment earlier thereby preventing the development or further progression of the cancer.
  • oligonucleotides designed from the sequences encoding HT4P may involve the use of PCR.
  • Such oligomers may be chemically synthesized, generated enzymatically, or produced from a recombinant source.
  • Oligomers will preferably consist of two nucleotide sequences, one with sense orientation (5'->3') and another with antisense (3' ⁇ -5'), employed under optimized conditions for identification of a specific gene or condition.
  • the same two oligomers, nested sets of oligomers, or even a degenerate pool of oligomers may be employed under less stringent conditions for detection and/or quantitation of closely related DNA or RNA sequences.
  • Methods which may also be used to quantitate the expression of HT4P include radiolabeling or biotinylating nucleotides, coamplification of a control nucleic acid, and standard curves onto which the experimental results are interpolated (Melby, P.C. et al. (1993) J. Immunol. Methods, 159:235-244; Duplaa, C. et al. (1993) Anal. Biochem. 229-236).
  • the speed of quantitation of multiple samples may be accelerated by running the assay in an ELISA format where the oligomer of interest is presented in various dilutions and a spectrophotometric or colorimetric response gives rapid quantitation.
  • the nucleic acid sequences which encode HT4P may also be used to generate hybridization probes which are useful for mapping the naturally occurring genomic sequence.
  • the sequences may be mapped to a particular chromosome or to a specific region of the chromosome using well known techniques.
  • Such techniques include FISH, FACS, or artificial chromosome constructions, such as yeast artificial chromosomes, bacterial artificial chromosomes, bacterial PI constructions or single chromosome cDNA libraries as reviewed in Price, CM. (1993) Blood Rev. 7: 127-134, and Trask, B.J. (1991) Trends Genet. 7: 149-154.
  • FISH FISH (as described in Verma et al. (1988) Human Chromosomes: A Manual of Basic Techniques. Pergamon Press, New York, NY) may be correlated with other physical chromosome mapping techniques and genetic map data. Examples of genetic map data can be found in the 1994 Genome Issue of Science (265: 198 If). Correlation between the location of the gene encoding HT4P on a physical chromosomal map and a specific disease , or predisposition to a specific disease, may help delimit the region of DNA associated with that genetic disease.
  • the nucleotide sequences of the subject invention may be used to detect differences in gene sequences between normal, carrier, or affected individuals.
  • En situ hybridization of chromosomal preparations and physical mapping techniques such as linkage analysis using established chromosomal markers may be used for extending genetic maps. Often the placement of a gene on the chromosome of another mammalian species, such as mouse, may reveal associated markers even if the number or arm of a particular human chromosome is not known. New sequences can be assigned to chromosomal arms, or parts thereof, by physical mapping. This provides valuable information to investigators searching for disease genes using positional cloning or other gene discovery techniques. Once the disease or syndrome has been crudely localized by genetic linkage to a particular genomic region, for example, AT to 1 lq22-23 (Gatti, R.A. et al.
  • HT4P its catalytic or immunogenic fragments or oligopeptides thereof, can be used for screening libraries of compounds in any of a variety of drug screening techniques.
  • the fragment employed in such screening may be free in solution, affixed to a solid support, borne on a cell surface, or located intracellularly. The formation of binding complexes, between HT4P and the agent being tested, may be measured.
  • Another technique for drug screening provides for high throughput screening of compounds having suitable binding affinity to the protein of interest as described in published PCT application WO84/03564.
  • HT4P large numbers of different small test compounds are synthesized on a solid substrate, such as plastic pins or some other surface.
  • the test compounds are reacted with HT4P, or fragments thereof, and washed.
  • Bound HT4P is then detected by methods well known in the art.
  • Purified HT4P can also be coated directly onto plates for use in the aforementioned drug screening techniques.
  • non-neutralizing antibodies can be used to capture the peptide and immobilize it on a solid support.
  • nucleotide sequences which encode HT4P may be used in any molecular biology techniques that have yet to be developed, provided the new techniques rely on properties of nucleotide sequences that are currently known, including, but not limited to, such properties as the triplet genetic code and specific base pair interactions.
  • PROSNON01 is a normalized library constructed from 4.4 million independent clones from the PROSNOT11 cDNA library.
  • the PROSNOT11 cDNA library was constructed from microscopically normal prostate tissue obtained from a 28-year-old Caucasian male (specimen #RA95-09-0677; International Institute of Advanced Medicine, Exton, PA) who died from a self inflicted gun shot wound.
  • the frozen tissue was homogenized and lysed using a Brinkmann Homogenizer Polytron PT-3000 (Brinkmann Instruments, Westbury NJ) in guanidinium isothiocyanate solution.
  • the lysate was centrifuged over a 5.7 M CsCl cushion using an Beckman SW28 rotor in a Beckman L8-70M Ultracentrifuge (Beckman Instruments) for 18 hours at 25,000 rpm at ambient temperature.
  • the RNA was extracted with acid phenol pH 4.7, precipitated using 0.3 M sodium acetate and 2.5 volumes of ethanol, resuspended in RNAse-free water and DNase treated at 37 °C RNA extraction and precipitation were repeated as before.
  • the mRNA was then isolated with the Qiagen Oligotex kit (QIAGEN, Chatsworth, CA) and used to construct the cDNA library.
  • the normalization and hybridization conditions were adapted from Soares et al., PNAS 91 (1994):9228, except that a longer (19-hour) reannealing hybridization was used.
  • mRNA was handled according to the recommended protocols in the Superscript Plasmid System for cDNA Synthesis and Plasmid Cloning (Cat. #18248-013, Gibco/BRL, Gaithersburg, MD). cDNAs were fractionated on a Sepharose CL4B column (Cat. #275105-01, Pharmacia), and those cDNAs exceeding 400 bp were ligated into pSport I. The plasmid pSport I was subsequently transformed into DH5aTM competent cells (Cat. #18258-012, Gibco/BRL).
  • Plasmid DNA was released from the cells and purified using the REAL Prep 96 Plasmid
  • Kit (Catalog #26173, QEAGEN, Inc.). This kit enabled the simultaneous purification of 96 samples in a 96-well block using multi-channel reagent dispensers.
  • the recommended protocol was employed except for the following changes: 1) the bacteria were cultured in 1 ml of sterile Terrific Broth (Catalog #22711, Gibco/BRL) with carbenicillin at 25 mg/L and glycerol at 0.4%; 2) after inoculation, the cultures were incubated for 19 hours and at the end of incubation, the cells were lysed with 0.3 ml of lysis buffer; and 3) following isopropanol precipitation, the plasmid DNA pellet was resuspended in 0.1 ml of distilled water. After the last step in the protocol, samples were transferred to a 96-well block for storage at 4° C.
  • the cDNAs were sequenced by the method of Sanger et al. (1975, J. Mol. Biol. 94:441f), using a Hamilton Micro Lab 2200 (Hamilton, Reno, NV) in combination with Peltier Thermal Cyclers (PTC200 from MJ Research, Watertown, MA) and Applied Biosystems 377 DNA Sequencing Systems; and the reading frame was determined.
  • nucleotide sequences of the Sequence Listing or amino acid sequences deduced from them were used as query sequences against databases such as GenBank, SwissProt, BLOCKS, and Pima EL These databases which contain previously identified and annotated sequences were searched for regions of homology (similarity) using BLAST, which stands for Basic Local Alignment Search Tool (Altschul, S.F. (1993) J. Mol. Evol. 36:290-300; Altschul et al. (1990) J. Mol. Biol. 215:403-410).
  • BLAST produces alignments of both nucleotide and amino acid sequences to determine sequence similarity. Because of the local nature of the alignments, BLAST is especially useful in determining exact matches or in identifying homologs which may be of prokaryotic (bacterial) or eukaryotic (animal, fungal or plant) origin. Other algorithms such as the one described in Smith RF and TF Smith (1992; Protein Engineering 5:35-51), incorporated herein by reference, can be used when dealing with primary sequence patterns and secondary structure gap penalties. As disclosed in this application, the sequences have lengths of at least 49 nucleotides, and no more than 12% uncalled bases (where N is recorded rather than A, C, G, or T). The BLAST approach, as detailed in Karlin, S. and S.F. Atschul (1993; Proc. Nat. Acad.
  • Northern analysis is a laboratory technique used to detect the presence of a transcript of a gene and involves the hybridization of a labeled nucleotide sequence to a membrane on which RNAs from a particular cell type or tissue have been bound (Sambrook et al., supra).
  • the product score takes into account both the degree of similarity between two sequences and the length of the sequence match. For example, with a product score of 40, the match will be exact within a 1-2% error; and at 70, the match will be exact. Homologous molecules are usually identified by selecting those which show product scores between 15 and 40, although lower scores may identify related molecules.
  • One primer is synthesized to initiate extension in the antisense direction (XLR) and the other is synthesized to extend sequence in the sense direction (XLF).
  • Primers are used to facilitate the extension of the known sequence "outward" generating amplicons containing new, unknown nucleotide sequence for the region of interest.
  • the initial primers are designed from the cDNA using OLIGO 4.06 (National Biosciences), or another appropriate program, to be 22-30 nucleotides in length, to have a GC content of 50% or more, and to anneal to the target sequence at temperatures about 68°-72° C Any stretch of nucleotides which would result in hairpin structures and primer-primer dimerizations is avoided.
  • the original, selected cDNA libraries, or a human genomic library are used to extend the sequence; the latter is most useful to obtain 5' upstream regions. If more extension is necessary or desired, additional sets of primers are designed to further extend the known region.
  • PCR is performed using the Peltier Thermal Cycler (PTC200; M.J. Research, Watertown, MA) and the following parameters:
  • Step 1 94° C for 1 min (initial denaturation)
  • Step 2 65° C for 1 min
  • Step 3 68° C for 6 min
  • Step 4 94° C for 15 sec
  • Step 6 68° C for 7 min Step 7 Repeat step 4-6 for 15 additional cycles Step 8 94° C for 15 sec
  • Step 10 68° C for 7: 15 min
  • Step 11 Repeat step 8-10 for 12 cycles Step 12 72° C for 8 min
  • a 5-10 ⁇ l aliquot of the reaction mixture is analyzed by electrophoresis on a low concentration (about 0.6-0.8%) agarose mini-gel to determine which reactions were successful in extending the sequence. Bands thought to contain the largest products are selected and removed from the gel. Further purification involves using a commercial gel extraction method such as QIAQuickTM (QIAGEN Inc., Chatsworth, CA). After recovery of the DNA, Klenow enzyme is used to trim single-stranded, nucleotide overhangs creating blunt ends which facilitate religation and cloning. After ethanol precipitation, the products are redissolved in 13 ⁇ l of ligation buffer, l ⁇ l
  • T4-DNA ligase (15 units) and l ⁇ l T4 polynucleotide kinase are added, and the mixture is incubated at room temperature for 2-3 hours or overnight at 16° C.
  • Competent E. coli cells (in 40 ⁇ l of appropriate media) are transformed with 3 ⁇ l of ligation mixture and cultured in 80 ⁇ l of SOC medium (Sambrook et al., supra). After incubation for one hour at 37° C, the whole transformation mixture is plated on Luria Bertani (LB)-agar (Sambrook et al., supra) containing 2x Carb.
  • PCR amplification For PCR amplification, 18 ⁇ l of concentrated PCR reaction mix (3.3x) containing 4 units of rTth DNA polymerase, a vector primer, and one or both of the gene specific primers used for the extension reaction are added to each well. Amplification is performed using the following conditions: Step 1 94° C for 60 sec
  • Step 2 94° C for 20 sec
  • Step 5 Repeat steps 2-4 for an additional 29 cycles Step 6 72° C for 180 sec
  • Step 7 4° C (and holding) Aliquots of the PCR reactions are run on agarose gels together with molecular weight markers. The sizes of the PCR products are compared to the original partial cDNAs, and appropriate clones are selected, ligated into plasmid, and sequenced.
  • Hybridization probes derived from SEQ ID NO:2 are employed to screen cDNAs, genomic DNAs, or mRNAs. Although the labeling of oligonucleotides, consisting of about 20 base-pairs, is specifically described, essentially the same procedure is used with larger cDNA fragments.
  • Oligonucleotides are designed using state-of-the-art software such as OLIGO 4.06 (National Biosciences), labeled by combining 50 pmol of each oligomer and 250 ⁇ Ci of [ ⁇ - 2 P] adenosine triphosphate (Amersham) and T4 polynucleotide kinase (DuPont NEN ® , Boston, MA).
  • the labeled oligonucleotides are substantially purified with Sephadex G-25 superfine resin column (Pharmacia & Upjohn).
  • a portion containing 10 7 counts per minute of each of the sense and antisense oligonucleotides is used in a typical membrane based hybridization analysis of human genomic DNA digested with one of the following endonucleases (Ase I, Bgl II, Eco RI, Pst I, Xba 1, or Pvu El; DuPont NEN ® ).
  • the DNA from each digest is fractionated on a 0.7 percent agarose gel and transferred to nylon membranes (Nytran Plus, Schleicher & Schuell, Durham, NH). Hybridization is carried out for 16 hours at 40°C To remove nonspecific signals, blots are sequentially washed at room temperature under increasingly stringent conditions up to 0.1 x saline sodium citrate and 0.5% sodium dodecyl sulfate. After XOMAT ARTM film (Kodak, Rochester, NY) is exposed to the blots in a Phosphoimager cassette (Molecular Dynamics, Sunnyvale, CA) for several hours, hybridization patterns are compared visually.
  • Polynucleotide complementary to the HT4P-encoding sequence, or any part thereof, or an antisense molecule is used to inhibit in vivo expression of naturally occurring HT4P.
  • antisense oligonucleotides comprising about 20 base-pairs, is specifically described, essentially the same procedure is used with larger cDNA fragments.
  • the complementary oligonucleotide is designed from the most unique 5' sequence as shown in Figures 1A, IB and IC and used either to inhibit transcription by preventing promoter binding to the upstream nontranslated sequence or translation of an HT4P-encoding transcript by preventing the ribosome from binding.
  • an effective antisense oligonucleotide includes any 15-20 nucleotides spanning the region which translates into the signal or 5' coding sequence of the polypeptide as shown in Figures 1A, IB and IC
  • HT4P expression of HT4P is accomplished by subcloning the cDNAs into appropriate vectors and transforming the vectors into host cells.
  • the cloning vector is used to express HT4P in E. coli. Upstream of the cloning site, this vector contains a promoter for ⁇ -galactosidase, followed by sequence containing the amino-terminal Met, and the subsequent seven residues of ⁇ -galactosidase. Immediately following these eight residues is a bacteriophage promoter useful for transcription and a linker containing a number of unique restriction sites.
  • Induction of an isolated, transformed bacterial strain with EPTG using standard methods produces a fusion protein which consists of the first eight residues of ⁇ -galactosidase, about 5 to 15 residues of linker, and the full length protein.
  • the signal residues direct the secretion of HT4P into the bacterial growth media which can be used directly in the following assay for activity.
  • human HT4P is identified as a member of the TM4SF and is presumed to be involved in the regulation of cell growth.
  • HT4P is expressed in various human carcinomas (prostate, lung, and breast), malignant growths that consist of epithelial cells that tend to infiltrate surrounding tissues and give rise to metastases.
  • HT4P is also expressed in lung and brain tumor metastases and thus, expression of HT4P is presumed to be related to metastatic potential.
  • expression vectors encoding HT4P are transfected (e.g., electroporated) into non-motile or poorly motile cell lines (the majority of cell lines including cancer cell lines are non- or poorly motile), and the motility of the transfected and untransfected (control) cells are compared.
  • Methods for the design and construction of an expression vector capable of expressing HT4P in the desired mammalian cell line(s) chosen are well known to the art.
  • Assays for examining the motility of cells in culture are known to the art (Miyake, M. et al. (1991) J. Exp. Med.
  • the correlation between HT4P and metastatic potential can be measured using an animal model in which the invasiveness of a non-metastatic cell line such as the mouse B16B1 melanoma, transfected with HT4P is compared with non- transfected B16B1 and with the highly metastatic BL6 cell line (Ekeyama, et al., supra).
  • Metastatic deposition in the lung is measured following intravenous injections of cells in Balb/c mice. An increase in lung deposition in the HT4P-transfected B 16B 1 cells relative to the non- transfected B 16B 1 that approaches the metastasis produced by BL6 cells is evidence that levels of HT4P in tumor cells correlates with metastatic potential.
  • HT4P that is substantially purified using PAGE electrophoresis (Sambrook, supra), or other purification techniques, is used to immunize rabbits and to produce antibodies using standard protocols.
  • the amino acid sequence deduced from SEQ ED NO:2 is analyzed using DNASTAR software (DNASTAR Lnc) to determine regions of high immunogenicity and a corresponding oligopolypeptide is synthesized and used to raise antibodies by means known to those of skill in the art. Selection of appropriate epitopes, such as those near the C-terminus or in hydrophilic regions, is described by Ausubel et al. (supra), and others. Typically, the oligopeptides are 15 residues in length, synthesized using an Applied
  • N-maleimidobenzoyl-N-hydroxysuccinimide ester (MBS; Ausubel et al., supra). Rabbits are immunized with the oligopeptide-KLH complex in complete Freund's adjuvant. The resulting antisera are tested for antipeptide activity, for example, by binding the peptide to plastic, blocking with 1% BSA, reacting with rabbit antisera, washing, and reacting with radioiodinated, goat anti-rabbit IgG.
  • Naturally occurring or recombinant HT4P is substantially purified by immunoaffinity chromatography using antibodies specific for HT4P.
  • An immunoaffinity column is constructed by covalently coupling HT4P antibody to an activated chromatographic resin, such as CnBr-activated Sepharose (Pharmacia & Upjohn). After the coupling, the resin is blocked and washed according to the manufacturer's instructions.
  • HT4P Media containing HT4P is passed over the immunoaffinity column, and the column is washed under conditions that allow the preferential absorbance of HT4P (e.g., high ionic strength buffers in the presence of detergent).
  • the column is eluted under conditions that disrupt antibody/HT4P binding (eg, a buffer of pH 2-3 or a high concentration of a chaotrope, such as urea or thiocyanate ion), and HT4P is collected.
  • a chaotrope such as urea or thiocyanate ion
  • HT4P or biologically active fragments thereof are labeled with 125 I Bolton-Hunter reagent (Bolton et al. (1973) Biochem. J. 133:529).
  • Candidate molecules previously arrayed in the wells of a multi-well plate are incubated with the labeled HT4P, washed and any wells with labeled HT4P complex are assayed. Data obtained using different concentrations of HT4P are used to calculate values for the number, affinity, and association of HT4P with the candidate molecules.
  • CTTGGTTAGT CTGCTGCTAA TTGGAATTGC TGCGTGGGGC ATTGGCTTCG GGCTGATTTC 240

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Zoology (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cell Biology (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

L'invention concerne une protéine 4 transmembranaire humaine (HT4P) et des polynucléotides qui codent HTP4. Elle porte aussi sur des vecteurs d'expression, des cellules hôtes, des molécules antisens, des anticorps ou des antagonistes, ainsi que sur des procédés de traitement de troubles associés à l'expression de HT4P.
PCT/US1998/009878 1997-05-13 1998-05-13 Nouvelle proteine transmembranaire humaine WO1999000408A2 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU75723/98A AU7572398A (en) 1997-05-13 1998-05-14 New human transmembrane protein

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US85551997A 1997-05-13 1997-05-13
US08/855,519 1997-05-13

Publications (2)

Publication Number Publication Date
WO1999000408A2 true WO1999000408A2 (fr) 1999-01-07
WO1999000408A3 WO1999000408A3 (fr) 1999-04-15

Family

ID=25321461

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/009878 WO1999000408A2 (fr) 1997-05-13 1998-05-13 Nouvelle proteine transmembranaire humaine

Country Status (2)

Country Link
AU (1) AU7572398A (fr)
WO (1) WO1999000408A2 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000026243A2 (fr) * 1998-10-29 2000-05-11 Incyte Pharmaceuticals, Inc. Proteines transmembranaires 4
FR2809734A1 (fr) * 2000-05-31 2001-12-07 Pf Medicament Proteine tetraspane humaine et son utilisation
WO2001096399A2 (fr) * 2000-06-15 2001-12-20 Curagen Corporation Nouvelle proteine de tretraspon induite par l'interferon et acides nucleiques codant pour cette proteine
WO2006093337A1 (fr) * 2005-03-03 2006-09-08 Takeda Pharmaceutical Company Limited Agent préventif/thérapeutique pour le cancer
US7563880B2 (en) 1999-11-30 2009-07-21 Corixa Corporation Compositions and methods for the therapy and diagnosis of breast cancer

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997039123A2 (fr) * 1996-04-18 1997-10-23 Genetics Institute, Inc. Proteines secretees
WO1998039448A2 (fr) * 1997-03-07 1998-09-11 Human Genome Sciences, Inc. 186 nouvelles proteines secretees

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997039123A2 (fr) * 1996-04-18 1997-10-23 Genetics Institute, Inc. Proteines secretees
WO1998039448A2 (fr) * 1997-03-07 1998-09-11 Human Genome Sciences, Inc. 186 nouvelles proteines secretees

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
EMBL database entry HS1207167; accession number AA410464; 5. May. 1997; Hillier et al.: 'WashU-Merck EST Project' XP002093353 *
EMBL database entry HS1249226; accession number AA450348; 10. June 1997; Hillier et al.: ?WashU-Merck EST Project 1997' XP002093354 *
EMBL database entry HS91684; accession number R11916; 21. April 1995; Hillier et al.: 'The WashU-Merck EST project.' XP002093355 *
MAECKER H T ET AL: "THE TETRASPANIN SUPERFAMILY: MOLECULAR FACILITATORS" FASEB JOURNAL, vol. 11, no. 6, May 1997, pages 428-442, XP002037857 *
WRIGHT M. D. ET AL.: "THE INS AND OUTS OF THE TRAMSMEMBRANE 4 SUPERFAMILY" IMMUNOLOGY TODAY, vol. 15, no. 12, 1994, pages 588-594, XP002066879 cited in the application *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000026243A2 (fr) * 1998-10-29 2000-05-11 Incyte Pharmaceuticals, Inc. Proteines transmembranaires 4
WO2000026243A3 (fr) * 1998-10-29 2000-10-05 Incyte Pharma Inc Proteines transmembranaires 4
US7563880B2 (en) 1999-11-30 2009-07-21 Corixa Corporation Compositions and methods for the therapy and diagnosis of breast cancer
FR2809734A1 (fr) * 2000-05-31 2001-12-07 Pf Medicament Proteine tetraspane humaine et son utilisation
WO2001096399A2 (fr) * 2000-06-15 2001-12-20 Curagen Corporation Nouvelle proteine de tretraspon induite par l'interferon et acides nucleiques codant pour cette proteine
WO2001096399A3 (fr) * 2000-06-15 2002-10-24 Curagen Corp Nouvelle proteine de tretraspon induite par l'interferon et acides nucleiques codant pour cette proteine
WO2006093337A1 (fr) * 2005-03-03 2006-09-08 Takeda Pharmaceutical Company Limited Agent préventif/thérapeutique pour le cancer

Also Published As

Publication number Publication date
WO1999000408A3 (fr) 1999-04-15
AU7572398A (en) 1999-01-19

Similar Documents

Publication Publication Date Title
WO1998038310A1 (fr) Antigene tm4sf humain associe aux tumeurs
US5922566A (en) Tumor-associated antigen
US5843717A (en) Rab protein
WO1998033818A1 (fr) Canal anionique humain
EP0981615A1 (fr) Proteine de type inhibiteur de proteine kinase c (ipkc-2)
WO1998044114A1 (fr) Proteine de fixation du calcium similaire de la reticulocalbine
WO1998045440A1 (fr) Proteine humaine fixatrice d'acides gras
WO1998050550A1 (fr) Proteine humaine associee a la translocation
WO1999000408A2 (fr) Nouvelle proteine transmembranaire humaine
WO1998036063A1 (fr) Nouvelle proteine humaine du genre h-rev 107
US5739010A (en) Human transcription factor
WO1998033817A1 (fr) Proteine membranaire humaine
WO1998041635A1 (fr) Proteine associee a la tumorogenese humaine, htap
EP1012288A1 (fr) Nouvelle proteine humaine de type cystatine
WO1998040481A1 (fr) Nouvelle proteine acide riche en proline
WO1998030691A1 (fr) Proteine humaine de canal de chlorure
WO1998045439A1 (fr) Proteine membranaire associee a une tumeur chez l'homme
WO1998021318A1 (fr) Enzyme conjuguant une ubiquitine telle que ubch7
US5849899A (en) Human tumor suppressor
US5854022A (en) Polynucleotides encoding a CD53-like transmembrane protein
US6033870A (en) Human transmembrane protein
WO1998049304A1 (fr) Proteine a matrice extracellulaire neuronale
WO1998049194A2 (fr) Nouvelle proteine de type ras
EP0972040A1 (fr) Proteine humaine fixatrice de gtp
WO1998050548A1 (fr) Proteine de transfert vesiculaire

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AT AU BR CA CH CN DE DK ES FI GB IL JP KR MX NO NZ RU SE SG US

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW SD SZ UG 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 BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

AK Designated states

Kind code of ref document: A3

Designated state(s): AT AU BR CA CH CN DE DK ES FI GB IL JP KR MX NO NZ RU SE SG US

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW SD SZ UG 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 BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
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: CA

NENP Non-entry into the national phase in:

Ref country code: JP

Ref document number: 1998550783

Format of ref document f/p: F

122 Ep: pct application non-entry in european phase