WO2002048173A2 - Expression of a cadherin-like protein in benign prostatic hyperplasia - Google Patents

Expression of a cadherin-like protein in benign prostatic hyperplasia Download PDF

Info

Publication number
WO2002048173A2
WO2002048173A2 PCT/US2001/047949 US0147949W WO0248173A2 WO 2002048173 A2 WO2002048173 A2 WO 2002048173A2 US 0147949 W US0147949 W US 0147949W WO 0248173 A2 WO0248173 A2 WO 0248173A2
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
protein
seq
acid molecule
expression
Prior art date
Application number
PCT/US2001/047949
Other languages
French (fr)
Other versions
WO2002048173A3 (en
Inventor
William E. Munger
Prakash Kulkarni
Robert H. Getzenberg
Original Assignee
Gene Logic, Inc.
Japan Tobacco, 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 Gene Logic, Inc., Japan Tobacco, Inc. filed Critical Gene Logic, Inc.
Priority to AU2002229004A priority Critical patent/AU2002229004A1/en
Publication of WO2002048173A2 publication Critical patent/WO2002048173A2/en
Publication of WO2002048173A3 publication Critical patent/WO2002048173A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the invention relates generally to the changes in gene expression in prostate tissue removed from male patients with benign prostatic hyperplasia (BPH).
  • BPH benign prostatic hyperplasia
  • the invention specifically relates to a novel human gene which is differentially expressed in BPH tissue compared to normal prostate tissue.
  • BPH is the most common benign tumor in men 60 years of age or older. It is estimated that one in four men living to the age of 80 will require treatment for this disease. BPH is usually noted clinically after the age of 50, the incidence increasing with age, but as many as two thirds of men between the ages of 40 and 49 demonstrate histological evidence of the disease.
  • the anatomic location of the prostate at the bladder neck enveloping the urethra plays an important role in the pathology of BPH, including bladder outlet obstraction.
  • Two prostate components are thought to play a role in bladder outlet obstruction. The first is the relative increased prostate tissue mass.
  • the second component is the prostatic smooth muscle tone.
  • the causative factors of BPH in man has been intensively studied (see Ziada et al, Urology 53:1-6, 1999). In general, the two most important factors appear to be aging and the presence of functional testes. Although these factors appear to be key to the development of BPH, both appear to be nonspecific.
  • cell-cell adhesion plays an important role in a wide range of events including morphogenesis and organ formation, leukocyte extravasion, tumor metastasis and invasion, and the fo ⁇ nation of cell junctions. Additionally, cell-cell adhesion is crucial for the maintenance of tissue integrity, e.g., the maintenance of the intestinal epithelial barrier and the integrity of cardiac muscle, hitercellular adhesion is mediated by specific cell surface adhesion molecules
  • CAMs CAMs
  • immunoglobulin integrin
  • selectin integrin
  • cadherin superfamilies
  • Cadhenns are a rapidly expanding family of calcium-dependent CAMs.
  • the classical cadherins are integral membrane glycoproteins that generally promote cell adhesion through homophilic interactions. They have an N-temiinal extracellular domain that determines binding specificity, a hydrophobic membrane-spanning domain and a C-terminal cytoplasmic domain (highly conserved among the members of the superfamily) that interacts with the cytoskeleton through catenins and other cytoskeleton-associated proteins.
  • Some cadherins lack a cytoplasmic domain, however, and appear to function in cell-cell adhesion by a different mechanism than cadherins that do have a cytoplasmic domain. Calcium binding motifs are interspersed throughout the extracellular domain.
  • Cadherins have been shown to regulate epithelial, endothelial, neural and cancer cell adhesion, with different cadherins expressed on different cell types. For example, N (neural)- cadherin is predominantly expressed by neural cells, endothelial cells and a variety of cancer cell types. E (epithelial)-cadherin is predominantly expressed by epithelial cells. Other cadherins are P (placental)-cadherin, which is found in human skin and R (retinal)-cadherin.
  • the unique expression pattern of the different cadherins is particularly significant with respect to the role each subclass of cadherins may play in vivo in normal events (e.g., the maintenance of the intestinal epithelial ba ier) and in abnormal events (e.g., autoimmune diseases, tumor metastatis or inflammation).
  • normal events e.g., the maintenance of the intestinal epithelial ba ier
  • abnormal events e.g., autoimmune diseases, tumor metastatis or inflammation.
  • Different subclasses or combinations of subclasses of cadherins are likely to be responsible for different cell-cell adhesion events in which therapeutic detection and/or intervention may be desirable.
  • cadherins may have some regulatory activity in addition to adhesive activity. Thus, therapeutic intervention in the regulatory activities of cadherins expressed in specific tissues may also be desirable.
  • the present invention is based on the discovery of a new gene family that is differentially expressed in BPH tissue compared to normal prostate tissue.
  • the invention includes isolated nucleic acid molecules selected from the group consisting of an isolated nucleic acid molecule that encodes the amino acid sequence of SEQ ID NO: 2, an isolated nucleic acid molecule that encodes a fragment of at least 6 contiguous amino acids of SEQ ID NO: 2, an isolated nucleic acid molecule which hybridizes to the complement of a nucleic acid molecule comprising SEQ LD NO: 1 and an isolated nucleic acid molecule which hybridizes to the complement of a nucleic acid molecule that encodes the amino acid sequence of SEQ ID NO: 2.
  • Nucleic acid molecules of the invention may encode a protein having at least about
  • the present invention further includes the nucleic acid molecules operably linked to one or more expression control elements, including vectors comprising the isolated nucleic acid molecules.
  • the invention further includes host cells transformed to contain the nucleic acid molecules of the invention and methods for producing a protein comprising culturing a host cell transfonned with a nucleic acid molecule of the invention under conditions in which the protein is expressed.
  • the invention further provides an isolated polypeptide selected from the group consisting of an isolated polypeptide comprising the amino acid sequence of SEQ ID NO: 2, an isolated polypeptide comprising a functional or antigenic fragment of at least 10 contiguous amino acids of SEQ ID NO: 2, an isolated polypeptide comprising conservative amino acid substitutions of SEQ ID NO: 2 and an isolated polypeptide comprising naturally occumng amino acid sequence variants of SEQ ID NO: 2.
  • Polypeptides of the invention also include polypeptides with an amino acid sequence having at least about 50%, 60%, 65%, 70% or 75% amino acid sequence identity with the sequence set forth in SEQ ID NO: 2 more preferably at least about 80-85%, even more preferably at least about 90%, and most preferably at least about 95%o sequence identity with the sequence set forth in SEQ ID NO: 2.
  • the invention further provides an isolated antibody or antigen-binding fragment that specifically binds to a polypeptide of the invention, including monoclonal and polyclonal antibodies.
  • the invention further provides methods of identifying an agent which modulates the expression of a nucleic acid encoding a protein of the invention, comprising: exposing cells which express the nucleic acid to the agent; and determining whether the agent modulates expression of said nucleic acid, thereby identifying an agent which modulates the expression of a nucleic acid encoding the protein.
  • the invention further provides methods of identifying an agent which modulates the level of or at least one activity of a protein of the invention, comprising: exposing cells which express the protein to the agent; and determining whether the agent modulates the level of or at least one activity of said protein, thereby identifying an agent which modulates the level of or at least one activity of the protein.
  • the invention further provides methods of identifying binding partners for a protein of the invention, comprising: exposing said protein to a potential binding partner; and deteiTnining if the potential binding partner binds to said protein, thereby identifying binding partners for the protein.
  • the present invention further provides methods of modulating the expression of a nucleic acid encoding a protein of the invention, comprising administering an effective amount of an agent which modulates the expression of a nucleic acid encoding the protein of the invention.
  • the invention also provides methods of modulating at least one activity of a protein of the invention, comprising administering an effective amount of an agent which modulates at least one activity of the protein of the invention.
  • the present invention further includes non-human transgenic animals modified to contain the nucleic acid molecules of the invention or mutated nucleic acid molecules such that expression of the encoded polypeptides of the invention is prevented.
  • the present invention also includes non-human transgenic animals in which all or a portion of the gene comprising all or a portion of SEQ ID NO: 1 has been knocked out or deleted from the genome of the animal.
  • the invention further provides methods of diagnosing BPH or other disease states, comprising the steps of: acquiring a tissue, blood, urine or other sample from a subject; and determining the level of expression of a nucleic acid molecule of the invention or polypeptide of the invention.
  • compositions comprising a diluent and a polypeptide or protein selected from the group consisting of an isolated polypeptide comprising all or a portion of SEQ ID NO: 2, an isolated polypeptide comprising a fragment of at least 6 contiguous amino acids of SEQ ID NO: 2, an isolated polypeptide comprising one or more conservative ammo acid substitutions of SEQ ID NO: 2, naturally occurring amino acid sequence variants of SEQ ID NO: 2, and an isolated polypeptide with an amino acid sequence having at least about 50%, 60%, 70% or 75% amino acid sequence identity with the sequence set forth in SEQ ID NO: 2, preferably at least about 80%, more preferably at least about 90%, and most preferably at least about 95% sequence identity with the sequence set forth in SEQ ID NO: 2.
  • a polypeptide or protein selected from the group consisting of an isolated polypeptide comprising all or a portion of SEQ ID NO: 2, an isolated polypeptide comprising a fragment of at least 6 contiguous amino acids of SEQ ID NO: 2, an isolated polypeptide compris
  • Figure 1 shows Affymetrix GeneChip® data on the expression of a F13763 nucleic acid molecule (SEQ ID NO: 1) in prostate biopsy samples from normal human patients and BPH patients, including BPH samples from individuals without symptoms (BPH- w/o), samples from BPH patients without symptoms who were diagnosed with prostate cancer (BPH-Ca), and samples from BPH patients with symptoms (BPH-with).
  • BPH-w/o BPH samples from individuals without symptoms
  • BPH-Ca samples from BPH patients without symptoms who were diagnosed with prostate cancer
  • BPH-with samples from BPH patients with symptoms
  • the subregion of the prostate analyzed was the transitional zone.
  • JPN and BRF-55T are Japanese BPH samples and cell culture samples respectively. Both unadjusted values (upper panel; raw Average Difference values) and "floored" values are shown (lower panel; all Average Difference values less than 20 are assigned a value of 20).
  • Figure 2 shows the tissue distribution of RNA encoding the protein of F13763 (SEQ ID NO: 2) as analyzed by Northern blot. mRNA expression levels were measured across a panel of nom al tissues (ClonTech human mRNA blot). Lane M) RNA Marker; 1) brain; 2) heart; 3) placenta; 4) lung; 5) liver; 6) smooth muscle; 7) kidney; and 8) pancreas.
  • Figure 3 shows the results of semi-quantitative PCR for the expression of F 13763 (SEQ ID NO: 1) in various nomial tissues.
  • Left gel panel 25 cycles (25 ); right gel panel: 30 cycles (30 O).
  • Figure 4 shows the results of an electronic Northern assay, in which the expression of the nuclei acid of SEQ LD NO: 1 was measured across a panel of human normal and BPH tissues using the Affymetrix human 42k GeneChip® set. For each tissue type, horizontal bar represents the mean +/- SDM for samples obtained from 3 or more nomial individuals.
  • Figure 5 shows a hydrophobicity analysis (PEPPLOT) of the polypeptide of SEQ LD NO:2. Analysis was done using the methods of Goldman et al. and of
  • the present invention is based in part on the identification of a new gene family that is differentially expressed in human BPH tissue compared to nomial human prostate tissue. This gene family corcesponds to the human cDNA of SEQ ID NO: 1. Genes that encode the human protein of SEQ ID NO: 2 may also be found in other animal species, particularly mammalian species.
  • the proteins and nucleic acids of the invention may be used as diagnostic agents to detect BPH.
  • the proteins of the present invention can also serve as a target for agents that can be used to modulate the expression or activity of the proteins.
  • agents may be identified that modulate biological processes associated with prostate growth, including the hyperplastic process of BPH.
  • the present invention is further based on the development of methods for isolating binding partners that bind to the protein.
  • Probes based on the protein are used as capture probes to isolate potential binding partners, such as other proteins.
  • Dominant negative proteins, DNAs encoding these proteins, antibodies to these proteins, peptide fragments of these proteins or mimics of these proteins may be mtroduced into cells to affect function.
  • the protein provides novel targets for the screening of synthetic small molecules and combinatorial or naturally occumng compound libraries to discover novel therapeutics to regulate prostate function.
  • the present invention provides isolated proteins, allelic variants of the proteins, and conservative amino acid substitutions of the proteins.
  • the "protein” or “polypeptide” refers, in part, to a protein that has the human amino acid sequence depicted in SEQ ID NO: 2.
  • the terms also refer to naturally occumng allelic variants and proteins that have a slightly different amino acid sequence than that specifically recited above. Allelic variants, though possessing a slightly different amino acid sequence than those recited above, will still have the same or similar biological functions associated with these proteins.
  • the present invention also encompasses proteins translated from alternative splice variants of the genes encoding the identified proteins.
  • the family of proteins related to the human amino acid sequence of SEQ ID NO: 2 refers to proteins that have been isolated from organisms in addition to humans. The methods used to identify and isolate other members of the family of proteins related to these proteins are described below.
  • the proteins of the present invention are preferably in isolated fomi.
  • a protein is said to be isolated when physical, mechanical or chemical methods are employed to remove the protein from cellular constituents that are normally associated with the protein. A skilled artisan can readily employ standard purification methods to obtain an isolated protein.
  • the proteins of the present invention further include insertion, deletion or conservative amino acid substitution variants of SEQ ID NO: 2.
  • a conservative variant refers to alterations in the amino acid sequence that do not adversely affect the biological functions of the protein.
  • a substitution, insertion or deletion is said to adversely affect the protein when the altered sequence prevents or disrupts a biological function associated with the protein.
  • the overall charge, structure or hydrophobic/hydrophilic properties of the protein can be altered without adversely affecting a biological activity.
  • the amino acid sequence can be altered, for example to render the peptide more hydrophobic or hydrophilic, without adversely affecting the biological activities of the protein.
  • allelic variants, the conservative substitution variants, and the members of the protein family will have an amino acid sequence having at least about 50%, 60%, 65%, 70% or 75% amino acid sequence identity with the sequence set forth in SEQ ID NO: 2, more preferably at least about 80%, even more preferably at least about 90%, and most preferably at least about 95% sequence identity.
  • Identity or homology with respect to such sequences is defined herein as the percentage of amino acid residues in the candidate sequence that are identical with the known peptides, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent homology, and not considering any conservative substitutions as part of the sequence identity (see section B for the relevant parameters). Fusion proteins, or N-terminal, C-tei rrinal or internal extensions, deletions, or insertions into the peptide sequence shall not be construed as affecting homology.
  • proteins of the present invention include molecules having the amino acid sequence disclosed in SEQ ID NO: 2; fragments thereof having a consecutive sequence of at least about 3, 4, 5, 6, 10, 15, 20, 25, 30, 35 or more amino acid residues of these proteins; amino acid sequence variants wherein one or more amino acid residues has been inserted N- or
  • Such fragments may contain antigenic regions, functional regions of the protein identified as regions of the amino acid sequence which correspond to known protein domains, as well as regions of pronounced hydrophilicity. The regions are all easily identifiable by using commonly available protein sequence analysis software such as Mac Vector (Oxford Molecular).
  • Contemplated variants further include those containing predetemiined mutations by, e.g., homologous recombination, site-directed or PCR mutagenesis, and the corresponding proteins of other animal species, including but not limited to rabbit, mouse, rat, porcine, bovine, ovine, equine and non-human primate species, and the alleles or other naturally occumng variants of the family of proteins; and derivatives wherein the protein has been covalently modified by substitution, chemical, enzymatic, or other appropriate means with a moiety other than a naturally occumng amino acid (for example a detectable moiety such as an enzyme or radioisotope).
  • a detectable moiety such as an enzyme or radioisotope
  • members of the family of proteins can be used: (1) to identify agents which modulate at least one activity of the protein; (2) to identify binding partners for the protein, (3) as an antigen to raise polyclonal or monoclonal antibodies; (4) as a therapeutic agent or target; and (5) as a diagnostic agent or marker.
  • nucleic acid molecules that encode the protein having SEQ ID NO: 2 and the related proteins herein described, preferably in isolated fomi.
  • nucleic acid is defined as RNA or DNA that encodes a protein or peptide as defined above, is complementary to a nucleic acid sequence encoding such peptides, hybridizes to such a nucleic acid and remains stably bound to it under appropriate stringency conditions, or encodes a polypeptide sharing at least about 50%, 60%, 70% or 75% sequence identity, preferably at least about 80%, more preferably at least about 85%, and even more preferably at least about 90% or 95% or more identity with the peptide sequences.
  • nucleic acid molecules will have at least about 50%, 60%, 70% or 75% nucleotide sequence identity to
  • SEQ LO NO: 1 preferably about S0%, more preferably about 85%, and even more preferably
  • nucleotide sequence identity to SEQ ID NO: 1, particularly over the open reading frame of SEQ ID NO: 1.
  • genomic DNA cDNA, mRNA and antisense molecules, as well as nucleic acids based on alternative backbones or including alternative bases whether derived from natural sources or synthesized.
  • hybridizing or complementary nucleic acids are defined further as being novel and unobvious over any prior art nucleic acid including that wliich encodes, hybridizes under appropriate stringency conditions, or is complementary to nucleic acid encoding a protein according to the present invention.
  • BLAST Basic Local Alignment Search Tool
  • blastp, blastn, blastx, tblastn and tblastx the algorithm employed by the programs blastp, blastn, blastx, tblastn and tblastx
  • blastp, blastn, blastx, tblastn and tblastx the algorithm employed by the programs blastp, blastn, blastx, tblastn and tblastx
  • blastp, blastn, blastx, tblastn and tblastx Karlin et al, Proc Natl Acad Sci USA 87: 2264-2268, 1990, and Altschul, JMol Evol 36: 290-300, 1993, fully incorporated by reference
  • the approach used by the BLAST program is to first consider similar segments between a query sequence and a database sequence, then to evaluate the statistical significance of all matches that are identified, and finally to summarize only those matches wliich satisfy a preselected threshold of significance.
  • the scoring matrix is set by the ratios of M (i.e., the reward score for a pair of matching residues) to N (i.e., the penalty score for mismatching residues), wherein the default values for M and N are 5 and -4, respectively.
  • M i.e., the reward score for a pair of matching residues
  • N i.e., the penalty score for mismatching residues
  • “Stringent conditions” are those that (1) employ low ionic strength and high temperature for washing, for example, 0.015 M NaCl/0.0015 M sodium citrate/0.1% SDS at 50°C, or (2) employ during hybridization a denaturing agent such as formamide, for example,
  • Preferred molecules are those that hybridize under the above conditions to the complement of SEQ ID NO: 1 and which encode a functional protein. Even more preferred hybridizing molecules are those that hybridize under the above conditions to the complement strand of the open reading frame of SEQ ID NO: 1.
  • a nucleic acid molecule is said to be "isolated” when the nucleic acid molecule is substantially separated from contaminant nucleic acid molecules encoding other polypeptides.
  • the present invention further provides fragments of the encoding nucleic acid molecule.
  • a fragment of an encoding nucleic acid molecule refers to a small portion of the entire protein coding sequence. The size of the fragment will be determined by the intended use. For example, if the fragment is chosen so as to encode an active portion of the protein, the fragment will need to be large enough to encode the functional region(s) of the protein. For instance, fragments which encode peptides coiresponding to predicted antigenic regions may be prepared (see Figure 5). If the fragment is to be used as a nucleic acid probe or PCR primer, then the fragment length is chosen so as to obtain a relatively small number of false positives during probing/priming (see the discussion in Section H).
  • Fragments of the encoding nucleic acid molecules of the present invention i.e., synthetic oligonucleotides
  • PCR polymerase chain reaction
  • Fragments of the encoding nucleic acid molecules of the present invention can easily be synthesized by chemical techniques, for example, the phosphoramidite method of Matteucci et al., J Am Chem Soc 103: 3185-3191, 1981, or using automated synthesis methods.
  • larger DNA segments can readily be prepared by well known methods, such as synthesis of a group of oligonucleotides that define various modular segments of the gene, followed by ligation of oligonucleotides to build the complete modified gene.
  • the encoding nucleic acid molecules of the present invention may further be modified so as to contain a detectable label for diagnostic and probe purposes.
  • a detectable label for diagnostic and probe purposes.
  • labels include, but are not limited to, biotin, radiolabeled nucleotides and the like. A skilled artisan can readily employ any such label to obtain labeled variants of the nucleic acid molecules of the invention.
  • the identification and characterization of the human nucleic acid molecule having SEQ ID NO: 1 allows a skilled artisan to isolate nucleic acid molecules that encode other members of the protein family in addition to the sequences herein described.
  • nucleic acid molecules allow a skilled artisan to isolate nucleic acid molecules that encode other members of the family of proteins in addition to the protein having SEQ ID NO: 2.
  • amino acid sequence of SEQ ID NO: 2 can be readily used to generate antibody probes to screen expression libraries prepared from appropriate cells.
  • polyclonal antiserum from mammals such as rabbits immunized with the purified protein (as described below) or monoclonal antibodies can be used to probe a mammalian cDNA or genomic expression library, such as lambda gtll library, to obtain the appropriate coding sequence for other members of the protein family.
  • the cloned cDNA sequence can be expressed as a fusion protein, expressed directly using its own control sequences, or expressed by constructions using control sequences appropriate to the particular host used for expression of the enzyme.
  • coding sequence herein described can be synthesized and used as a probe to retrieve DNA encoding a member of the protein family from any mammalian organism. Oligomers containing approximately 18-20 nucleotides (encoding about a 6-7 amino acid stretch) are prepared and used to screen genomic DNA or cDNA libraries to obtain hybridization under stringent conditions or conditions of sufficient stringency to eliminate an undue level of false positives.
  • pairs of oligonucleotide primers can be prepared for use in a polymerase chain reaction (PCR) to selectively clone an encoding nucleic acid molecule.
  • PCR polymerase chain reaction
  • a PCR denature- anneal-extend cycle for using such PCR primers is well known in the art and can readily be adapted for use in isolating other encoding nucleic acid molecules.
  • Nucleic acid molecules encoding other members of the protein family may also be identified in existing genomic or other sequence information using any available computational method, including but not limited to: PSI-BLAST (Altschul, et al, Nucleic Acids Res 25:3389- 3402, 1997); PHI-BLAST (Zhang, et al. Nucleic Acids Res 26:3986-3990, 1998), 3D-PSSM (Kelly et al, J Mol Biol 299(2): 499-520, 2000); and other computational analysis methods (Shi et al, Biochem Biophys Res Commun 262(1):132-138, 1999, and Matsunami et. al, Nature 404(6778):601-604, 2000.
  • PSI-BLAST Altschul, et al, Nucleic Acids Res 25:3389- 3402, 1997
  • PHI-BLAST Zahang, et al. Nucleic Acids Res 26:3986-3990, 1998)
  • 3D-PSSM
  • rDNA molecules Containing a Nucleic Acid Molecule The present invention further provides recombinant DNA molecules (rDNAs) that contain a coding sequence.
  • a rDNA molecule is a DNA molecule that has been subjected to molecular manipulation in situ. Methods for generating rDNA molecules are well known in the art, for example, see Sambrook et al., Molecular Cloning - A Laboratory Manual. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989. h the prefeired rDNA molecules, a coding DNA sequence is operably linked to expression control sequences and/or vector sequences.
  • vector and/or expression control sequences to which one of the protein family encoding sequences of the present invention is operably linked depends directly, as is well known in the art, on the functional properties desired, e.g., protein expression, and the host cell to be transformed.
  • a vector contemplated by the present invention is at least capable of directing the replication or insertion into the host chromosome, and preferably also expression, of the structural gene included in the rDNA molecule.
  • Expression control elements that are used for regulating the expression of an operably linked protein encoding sequence are known in the art and include, but are not limited to, inducible promoters, constitutive promoters, secretion signals, and other regulatory elements.
  • the inducible promoter is readily controlled, such as being responsive to a nutrient in the host cell's medium.
  • the vector containing a coding nucleic acid molecule will include a prokaryotic replicon, i.e., a DNA sequence having the ability to direct autonomous replication and maintenance of the recombinant DNA molecule extrachromosomally in a prokaryotic host cell, such as a bacterial host cell, transformed therewith.
  • a prokaryotic replicon i.e., a DNA sequence having the ability to direct autonomous replication and maintenance of the recombinant DNA molecule extrachromosomally in a prokaryotic host cell, such as a bacterial host cell, transformed therewith.
  • a prokaryotic replicon i.e., a DNA sequence having the ability to direct autonomous replication and maintenance of the recombinant DNA molecule extrachromosomally in a prokaryotic host cell, such as a bacterial host cell, transformed therewith.
  • vectors that include a prokaryotic replicon may also include a gene whose expression confers a
  • Vectors that include a prokaryotic replicon can further include a prokaryotic or bacteriophage promoter capable of directing the expression (transcription and translation) of the coding gene sequences in a bacterial host cell, such as E. coli.
  • a promoter is an expression control element formed by a DNA sequence that permits binding of RNA polymerase and transcription to occur. Promoter sequences compatible with bacterial hosts are typically provided in plasmid vectors containing convenient restriction sites for insertion of a DNA segment of the present invention.
  • Typical of such vector plasmids are pUC8, pUC9, pBR322 and pBR329 available from BioRad Laboratories, (Richmond, CA), pPL and pKK223 available from Phannacia (Piscataway, NJ).
  • Expression vectors compatible with eukaryotic cells can also be used to fomi rDNA molecules that contain a coding sequence.
  • Eukaryotic cell expression vectors including viral vectors, are well known in the art and are available from several commercial sources. Typically, such vectors are provided containing convenient restriction sites for insertion of the desired DNA segment. Typical of such vectors are pSVL and pKSV-10 (Pharmacia), pBPV-l/pML2d (International Biotechnologies, Inc.), pTDTl (ATCC, #31255), the vector pCDM8 described herein, and the like eukaryotic expression vectors. Vectors may be modified to include prostate cell specific promoters if needed.
  • Eukaryotic cell expression vectors used to construct the rDNA molecules of the present invention may further include a selectable marker that is effective in an eukaryotic cell, preferably a drug resistance selection marker.
  • a prefened drug resistance marker is the gene whose expression results in neomycin resistance, i.e., the neomycin phosphotransferase (neo) gene (Southern et al, J Mol Anal Genet 1:327-341, 1982).
  • the selectable marker can be present on a separate plasmid, and the two vectors are introduced by co-transfection of the host cell, and selected by culturing in the appropriate drug for the selectable marker.
  • the present invention further provides host cells transformed with a nucleic acid molecule that encodes a protein of the present invention.
  • the host cell can be either prokaryotic or eukaryotic.
  • Eukaryotic cells useful for expression of a protein of the invention are not limited, so long as the cell line is compatible with cell culture methods and compatible with the propagation of the expression vector and expression of the gene product.
  • Preferred eukaryotic host cells include, but are not limited to, yeast, insect and mammalian cells, preferably vertebrate cells such as those from a mouse, rat, monkey or human cell line.
  • Prefeired eukaryotic host cells include Chinese hamster ovary (CHO) cells available from the ATCC as CCL61, NIH Swiss mouse embryo cells (NIH/3T3) available from the ATCC as CRL 1658, baby hamster kidney cells (BHK), and the like eukaryotic tissue culture cell lines.
  • Any prokaryotic host can be used to express a rDNA molecule encoding a protein of the invention.
  • the preferred prokaryotic host is E. coli.
  • Transfonnation of appropriate cell hosts with a rDNA molecule of the present invention is accomplished by well known methods that typically depend on the type of vector used and host system employed.
  • electroporation and salt treatment methods are typically employed, see, for example, Cohen et al. , Proc Natl Acad Sci USA 69:2110, 1972; and Sambrook et al, (supra).
  • electroporation, cationic lipid or salt treatment methods are typically employed (see, for example, Graham et al, Virol 52:456, 1973; Wigler et al, Proc Natl Acad Sci USA 16:1313-1316, 1979).
  • Successfully transformed cells i.e., cells that contain a rDNA molecule of the present invention
  • cells resulting from the introduction of an rDNA of the present invention can be cloned to produce single colonies. Cells from those colonies can be harvested, lysed and their DNA content examined for the presence of the rDNA using a method such as that described by Southern, JMol Biol 98:503, 1975, or Berent et al, Biotech 3:208, 1985, or the proteins produced from the cell assayed via an immunological method.
  • the present invention further provides methods for producing a protein of the invention using nucleic acid molecules herein described, hi general temis, the production of a recombinant form of a protein typically involves the following steps:
  • a nucleic acid molecule is first obtained that encodes a protein of the invention, such as a nucleic acid molecule comprising, consisting essentially of or consisting of SEQ ID NO: 1 or nucleotides 395-3790 (-3793 with the stop codon) of SEQ ID NO: 1. If the encoding sequence is uninterrupted by introns, as is this open-reading-frame, it is directly suitable for expression in any host.
  • the nucleic acid molecule is then preferably placed in operable linkage with suitable control sequences, as described above, to fom an expression unit containing the protein open reading frame.
  • the expression unit is used to transfomi a suitable host, and the transformed host is cultured under conditions that allow the production of the recombinant protein.
  • the recombinant protein is isolated from the medium or from the cells. Recovery and purification of the protein may not be necessary in some instances where some impurities may be tolerated.
  • the desired coding sequences may be obtained from genomic fragments and used directly in appropriate hosts.
  • expression vectors that are operable in a variety of hosts is accomplished using appropriate replicons and control sequences, as set forth above.
  • the control sequences, expression vectors, and transformation methods are dependent on the type of host cell used to express the gene and ⁇ vere discussed in detail earlier.
  • Suitable restriction sites can, if not nomially available, be added to the ends of the coding sequence so as to provide an excisable gene to insert into these vectors.
  • a skilled artisan can readily adapt any host/expression system known in the art for use with the nucleic acid molecules of the invention to produce recombinant protein.
  • Another embodiment of the present invention provides methods for isolating and identifying binding partners of proteins of the invention, hi general, a protein of the invention is mixed with a potential binding partner or an extract or fraction of a cell under conditions that allow the association of potential binding partners with the protein of the invention. After mixing, peptides, polypeptides, proteins or other molecules that have become associated with a protein of the invention are separated from the mixture. The binding partner that bound to the protein of the invention can then be removed and further analyzed. To identify and isolate a binding partner, the entire protein, for instance a protein comprising the entire amino acid sequence of SEQ ID NO: 2 can be used. Alternatively, a fragment of the protein can be used.
  • a cellular extract refers to a preparation or fraction which is made from a lysed or disrupted cell.
  • the prefeired source of cellular extracts will be cells derived from human prostate tissue or cells, for instance, biopsy tissue or tissue culture cells from subjects with BPH.
  • cellular extracts may be prepared from nomial human prostate tissue or available cell lines, particularly prostate derived cell lines.
  • a variety of methods can be used to obtain an extract of a cell.
  • Cells can be dismpted using either physical or chemical disruption methods. Examples of physical disruption methods include, but are not limited to, sonication and mechanical shearing. Examples of chemical lysis methods include, but are not limited to, detergent lysis and enzyme lysis.
  • a skilled artisan can readily adapt methods for preparing cellular extracts in order to obtain extracts for use in the present methods.
  • the extract is mixed with the protein of the invention under conditions in which association of the protein with the binding partner can occur.
  • a variety of conditions can be used, the most preferred being conditions that closely resemble conditions found in the cytoplasm of a human cell.
  • Features such as osmolarity, pH, temperature, and the concentration of cellular extract used, can be varied to optimize the association of the protein with the binding partner.
  • the bound complex is separated from the mixture.
  • a variety of techniques can be utilized to separate the mixture. For example, antibodies specific to a protein of the invention can be used to immunoprecipitate the binding partner complex. Alternatively, standard chemical separation techniques such as chromatography and density/sediment centrifugation can be used.
  • the binding partner can be dissociated from the complex using conventional methods. For example, dissociation can be accomplished by altering the salt concentration or pH of the mixture.
  • the protein of the invention can be immobilized on a solid support.
  • the protein can be attached to a nitrocellulose matrix or acrylic beads. Attachment of the protein to a solid support aids in separating peptide/binding partner pairs from other constituents found in the extract.
  • the identified binding partners can be either a single protein or a complex made up of two or more proteins. Alternatively, binding partners may be identified using a Far- Western assay according to the procedures of Takayama et al. , Methods Mol Biol 69:171-184, 1997, or Sauder et al, J Gen Virol 77:991-996, 1996, or identified through the use of epitope tagged proteins or GST fusion proteins.
  • the nucleic acid molecules of the invention can be used in a yeast two- hybrid system.
  • the yeast two-hybrid system has been used to identify other protein partner pairs and can readily be adapted to employ the nucleic acid molecules herein described.
  • Another embodiment of the present invention provides methods for identifying agents that modulate the expression of a nucleic acid encoding a protein of the invention such as a protein having the amino acid sequence of SEQ ID NO: 2. Such assays may utilize any available means of monitoring for changes in the expression level of the nucleic acids of the invention.
  • an agent is said to modulate the expression of a nucleic acid of the invention if it is capable of up- or down-regulating expression of the nucleic acid in a cell.
  • cell lines that contain reporter gene fusions between the open reading frame defined by nucleotides 395-3790 of SEQ ID NO: 1, and/or the 5 'and/or 3' regulatoiy elements and any assayable fusion partner may be prepared.
  • Numerous assayable fusion partners are known and readily available including the firefly luciferase gene and the gene encoding chloramphenicol acetyltransferase (Alam et al. , Anal Biochem 188:245-254, 1990).
  • Cell lines containing the reporter gene fusions are then exposed to the agent to be tested under appropriate conditions and time. Differential expression of the reporter gene between samples exposed to the agent and control samples identifies agents wliich modulate the expression of a nucleic acid of the invention.
  • Additional assay fonnats may be used to monitor the ability of the agent to modulate the expression of a nucleic acid encoding a protein of the invention, such as the protein having
  • mRNA expression may be niomtored directly by hybridization to the nucleic acids of the invention.
  • Cell lines are exposed to the agent to be tested under appropriate conditions and time and total RNA or mRNA is isolated by standard procedures such those disclosed in Sambrook et al. (supra).
  • the preferred cells will be those derived from human prostate tissue, for instance, prostate biopsy tissue or cultured prostate cells from nomial or BPH patients, for example
  • BRF-55T cells immortalized human prostate cells obtained from an individual with BPH
  • Probes to detect differences in RNA expression levels between cells exposed to the agent and control cells may be prepared from the nucleic acids of the invention. It is preferable, but not necessary, to design probes which hybridize only with target nucleic acids under conditions of high stringency. Only highly complementaiy nucleic acid hybrids form under conditions of high stringency. Accordingly, the stringency of the assay conditions determines the amount of complementarity which should exist between two nucleic acid strands in order to fomi a hybrid. Stringency should be chosen to maximize the difference in stability between the probe:target hybrid and probe:non-target hybrids. Probes may be designed from the nucleic acids of the invention through methods known in the art.
  • the G+C content of the probe and the probe length can affect probe binding to its target sequence.
  • Methods to optimize probe specificity are commonly available in Sambrook et al, (supra) or Ausubel et al, Current Protocols in Molecular Biology. Greene Publishing Co., 1995.
  • Hybridization conditions are modified using known methods, such as those described by Sambrook et al. and Ausubel et al. as required for each probe.
  • Hybridization of total cellular RNA or RNA enriched for polyA RNA can be accomplished in any available format.
  • total cellular RNA or RNA enriched for polyA RNA can be affixed to a solid support and the solid support exposed to at least one probe comprising at least one, or part of one of the sequences of the invention under conditions in which the probe will specifically hybridize.
  • nucleic acid fragments comprising at least one, or part of one of the sequences of the invention can be affixed to a solid support, such as a silicon chip or a porous glass wafer.
  • the solid support can then be exposed to total cellular RNA or polyA RNA from a sample under conditions in wliich the affixed sequences will specifically hybridize.
  • Such solid supports and hybridization methods are widely available, for example, those disclosed by
  • agents wliich up or down regulate the expression of a nucleic acid encoding the protein having the sequence of SEQ ID NO: 2 are identified.
  • Hybridization for qualitative and quantitative analysis of niRNAs may also be canied out by using a RNase Protection Assay (i.e., RPA, see Ma et al, Methods 10:273-238, 1996).
  • RPA RNase Protection Assay
  • an expression vehicle comprising cDNA encoding the gene product and a phage-specific DNA-dependent RNA polymerase promoter (e.g., TI, T3 or SP6 RNA polymerase) is linearized at the 3' end of the cDNA molecule, downstream from the phage promoter, wherein such a linearized molecule is subsequently used as a template for synthesis of a labeled antisense transcript of the cDNA by in vitro transcription.
  • a phage-specific DNA-dependent RNA polymerase promoter e.g., TI, T3 or SP6 RNA polymerase
  • the labeled transcript is then hybridized to a mixture of isolated RNA (i.e., total or fractionated mRNA) by incubation at 45 °C overnight in a buffer comprising 80% fomiamide, 40 mM Pipes, pH 6.4, 0.4 M NaCl and 1 mM EDTA.
  • the resulting hybrids are then digested in a buffer comprising 40 ⁇ g/ml ribonuclease A and 2 ⁇ g/ml ribonuclease. After deactivation and extraction of extraneous proteins, the samples are loaded onto urea polyaciylamide gels for analysis.
  • cells or cell lines are first identified which express the gene products of the invention physiologically, for example BRF-55T cells (e.g., by using assays of tissue distribution via Northern blot, although RPAs may serve the identical purpose of expression selection).
  • Cell and/or cell lines so identified would be expected to comprise the necessary cellular machinery such that the fidelity of modulation of the transcriptional apparatus is maintained with regard to exogenous contact of agent with appropriate surface transduction mechanisms and/or the cytosolic cascades.
  • such cells or cell lines would be transduced or transfected with an expression vehicle (e.g., a plasmid or viral vector) construct comprising an operable non- translated 5 '-promoter containing end of the structural gene encoding the instant gene products fused to one or more antigenic fragments, which are peculiar to the instant gene products, wherein said fragments are under the transcriptional control of said promoter and are expressed as polypeptides whose molecular weight can be distinguished from the naturally occumng polypeptides or may further comprise an immunologically distinct tag or other detectable marker.
  • an expression vehicle e.g., a plasmid or viral vector
  • Cells or cell lines transduced or transfected as outlined above are then contacted with agents under appropriate conditions; for example, the agent in a pharmaceutically acceptable excipient is contacted with cells in an aqueous physiological buffer such as phosphate buffered saline (PBS) at physiological pH, Eagles balanced salt solution (BSS) at physiological pH,
  • PBS phosphate buffered saline
  • BSS Eagles balanced salt solution
  • PBS or BSS comprising serum or conditioned media comprising PBS or BSS and/or serum incubated at 37 °C. Said conditions may be modulated as deemed necessary by one of skill in the art. Subsequent to contacting the cells with the agent, said cells will be dismpted and the polypeptides of the lysate are fractionated such that a polypeptide fraction is pooled and contacted with an antibody to be further processed by immuno logical assay (e.g., ELISA, immunoprecipitation or Western blot).
  • immuno logical assay e.g., ELISA, immunoprecipitation or Western blot.
  • the pool of proteins isolated from the "agent- contacted” sample will be compared with a control sample where only the excipient is contacted with the cells and an increase or decrease in the immunologically generated signal from the "agent-contacted” sample compared to the control will be used to distinguish the effectiveness of the agent.
  • Another embodiment of the present invention provides methods for identifying agents that modulate at least one activity of a protein of the invention such as the protein having the amino acid sequence of SEQ ID NO: 2. Such methods or assays may utilize any means of monitoring or detecting the desired activity.
  • the relative amounts of a protein of the invention between a cell population that has been exposed to the agent to be tested compared to an un-exposed control cell population may be assayed.
  • probes such as specific antibodies are used to monitor the differential expression of the protein in the different cell populations.
  • Cell lines or populations are exposed to the agent to be tested under appropriate conditions and time.
  • Cellular lysates may be prepared from the exposed cell line or population and a control, unexposed cell line or population. The cellular lysates are then analyzed with the probe.
  • Antibody probes are prepared by immunizing suitable mammalian hosts in appropriate imnumization protocols using the peptides, polypeptides or proteins of the invention if they are of sufficient length, or, if desired, or if required to enhance immunogenicity, conjugated to suitable earners. Methods for preparing immunogenic conjugates with earners such as BSA,
  • KLH, or other carrier proteins are well known in the art.
  • direct conjugation using, for example, carbodiimide reagents may be effective; in other instances linking reagents such as those supplied by Pierce Chemical Co. (Rockford, IL), may be desirable to provide accessibility to the hapten.
  • the hapten peptides can be extended at either the amino or carboxy terminus with a cysteine residue or interspersed with cysteine residues, for example, to facilitate linking to a carrier.
  • Administration of the immunogens is conducted generally by injection over a suitable time period and with use of suitable adjuvants, as is generally understood in the art. During the immunization schedule, titers of antibodies are taken to detennine adequacy of antibody fomiation.
  • Immortalized cell lines wliich secrete the desired monoclonal antibodies may be prepared using the standard method of Kohler and Milstein (Nature 256:495-497, 1975) or modifications which effect immortalization of lymphocytes or spleen cells, as is generally known.
  • the immortalized cell lines secreting the desired antibodies are screened by immunoassay in wliich the antigen is the peptide hapten, polypeptide or protein.
  • the cells can be cultured either in vitro or by production in ascites fluid.
  • the desired monoclonal antibodies are then recovered from the culture supernatant or from the ascites supernatant. Fragments of the monoclonal antibodies or the polyclonal antisera wliich contain the immunologically significant portion can be used as antagonists, as well as the intact antibodies. Use of immunologically reactive antibody fragments, such as the Fab, Fab', or F(ab') 2 fragments is often preferable, especially in a therapeutic context, as these fragments are generally less immunogenic than the whole immunoglobulin.
  • the antibodies or fragments may also be produced, using current technology, by recombinant means.
  • Antibody regions that bind specifically to the desired regions of the protein can also be produced in the context of chimeras with multiple species origin, such as humanized antibodies.
  • one or more activities of a protein of the invention may be assayed in the presence and absence of an agent to be tested to determine whether they are capable of modulating (i.e. enhancing or inhibiting) this activity, for example cell adhesion between cells expressing a protein of the invention.
  • the ability of an agent to modulate cell adhesion may generally be evaluated in vitro by assaying the effect of the agent on one or more of the following: (1) neurite outgrowth, (2) adhesion between endothelial cells, (3) adhesion between epithelial cells (e.g., nomial rat kidney cells and/or human skin), (4) adhesion between cancer cells, and/or (5) adhesion between other cell types, wherein the cells under study express a protein of the invention either endogenously or as a result of transfection with a nucleic acid of the invention.
  • an agent is considered to be a modulator of cell adhesion if, within one or more of these representative assays, contact of the test cells with the agent results in a discernible disruption of cell adhesion.
  • contact of the test cells with the agent results in a discernible disruption of cell adhesion.
  • Agents that are assayed in the above method can be randomly selected or rationally selected or designed.
  • an agent is said to be randomly selected when the agent is chosen randomly without considering the specific sequences involved in the association of a protein of the invention alone or with its associated substrates, binding partners, etc.
  • An example of randomly selected agents is the use a chemical library or a peptide combinatorial library, or a growth broth of an organism.
  • an agent is said to be rationally selected or designed when the agent is chosen on a nonrandom basis wliich takes into account the sequence of the target site and/or its conformation in connection with the agent's action.
  • Agents can be rationally selected or rationally designed by utilizing the peptide sequences that make up these sites.
  • a rationally selected peptide agent can be a peptide whose amino acid sequence is identical to or a derivative of any functional consensus site.
  • the agents of the present invention can be, as examples, peptides, small molecules, vitamin derivatives, as well as carbohydrates. Dominant negative proteins, DNAs encoding these proteins, antibodies to these proteins, peptide fragments of these proteins or mimics of these proteins may be introduced into cells to affect function. "Mimic” used herein refers to the modification of a region or several regions of a peptide molecule to provide a structure chemically different from the parent peptide but topographically and functionally similar to the parent peptide (see Grant GA in: Molecular Biology and Biotechnology. Meyers (ed.), pp. 659-664, VCH Publishers, New York, 1995). A skilled artisan can readily recognize that there is no limit as to the structural nature of the agents of the present invention.
  • the peptide agents of the invention can be prepared using standard solid phase (or solution phase) peptide synthesis methods, as is known in the art.
  • the DNA encoding these peptides may be synthesized using commercially available oligonucleotide synthesis instrumentation and produced recombinantly using standard recombinant production systems. The production using solid phase peptide synthesis is necessitated if non-gene- encoded amino acids are to be included.
  • Another class of agents of the present invention are antibodies immunoreactive with critical positions of proteins of the invention.
  • Antibody agents are obtained by inimunization of suitable mammalian subjects with peptides, containing as antigenic regions, those portions of the protein intended to be targeted by the antibodies.
  • the proteins and nucleic acids of the invention are differentially expressed in BPH tissue.
  • Agents that up-or-down-regulate or modulate the expression of the protein, or at least one activity of the protein, such as agonists or antagonists may be used to modulate biological and pathologic processes associated with the protein's function and activity.
  • a subject can be any mammal, so long as the mammal is in need of modulation of a pathological or biological process mediated by a protein of the invention.
  • the tenn "mammal" is defined as an individual belonging to the class Mammalia. The invention is particularly useful in the treatment of human subjects.
  • Pathological processes refer to a category of biological processes which produce a deleterious effect.
  • expression of a protein of the invention may be associated with prostate cell growth or hyperplasia.
  • an agent is said to modulate a pathological process when the agent reduces the degree or severity of the process.
  • BPH may be prevented or disease progression modulated by the administration of agents which up- or down-regulate or modulate in some way the expression or at least one activity of a protein of the invention.
  • the agents of the present invention can be provided alone, or in combination with other agents that modulate a particular pathological process.
  • an agent of the present invention can be administered in combination with other known drugs.
  • two agents are said to be administered in combination when the two agents are administered simultaneously or are administered independently in a fashion such that the agents will act at the same time.
  • the agents of the present invention can be administered via parenteral, subcutaneous, intravenous, intramuscular, intraperitoneal, transdermal, or buccal routes. Alternatively, or concurrently, administration may be by the oral route.
  • the dosage administered will be dependent upon the age, health, and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the nature of the effect desired.
  • the present invention further provides compositions containing one or more agents which modulate expression or at least one activity of a protein of the invention. While individual needs vary, detemiination of optimal ranges of effective amounts of each component is within the skill of the art.
  • Typical dosages comprise 0J to 100 ⁇ g/kg body wt.
  • the prefened dosages comprise 0J to 10 ⁇ g/kg body wt.
  • the most prefeired dosages comprise 0J to 1 ⁇ g/kg body wt.
  • compositions of the present invention may contain suitable pharmaceutically acceptable earners comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used phamiaceutically for delivery to the site of action.
  • suitable formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble fomi, for example, water-soluble salts.
  • suspensions of the active compounds as appropriate oily injection suspensions may be administered.
  • Suitable lipophilic solvents or vehicles include fatty oils, e.g., sesame oil, or synthetic fatty acid esters, e.g., ethyl oleate or triglycerides.
  • Aqueous injection suspensions may contain substances wliich increase the viscosity of the suspension include, for example, sodium carboxymethyl cellulose, sorbitol, and/or dextran.
  • the suspension may also contain stabilizers. Liposomes can also be used to encapsulate the agent for delivery into the cell.
  • the phamiaceutical formulation for systemic administration according to the invention may be formulated for enteral, parenteral or topical administration. Indeed, all three types of formulations may be used simultaneously to achieve systemic administration of the active ingredient.
  • Suitable formulations for oral administration include hard or soft gelatin capsules, pills, tablets, including coated tablets, elixirs, suspensions, syrups or inhalations and controlled release fomis thereof.
  • the compounds of this invention may be used alone or in combination, or in combination with other therapeutic or diagnostic agents.
  • the compounds of this invention may be coadministered along with other compounds typically prescribed for these conditions according to generally accepted medical practice.
  • the compounds of this invention can be utilized in vivo, ordinarily in mammals, such as humans, sheep, horses, cattle, pigs, dogs, cats, rats and mice, or in vitro.
  • Transgenic animals containing mutant, knock-out or modified genes corresponding to the cDNA sequence of SEQ ID NO: 1, or the open reading frame encoding the polypeptide sequence of SEQ ID NO: 2, or fragments thereof having a consecutive sequence of at least about 3, 4, 5, 6, 10, 15, 20, 25, 30, 35 or more amino acid residues, are also included in the invention.
  • Transgenic animals are genetically modified animals into which recombinant, exogenous or cloned genetic material has been experimentally transferred.
  • transgene Such genetic material is often referred to as a "transgene.”
  • the nucleic acid sequence of the transgene in some embodiments, all or a portion of SEQ ID NO: 1, may be integrated either at a locus of a genome where that particular nucleic acid sequence is not otherwise nonrially found or at the nomial locus for the transgene.
  • the transgene may consist of nucleic acid sequences derived from the genome of the same species or of a different species than the species of the target animal.
  • transgenic animals in which all or a portion of one or more genes comprising SEQ ID NO: 1 is deleted may be constructed.
  • the gene corresponding to SEQ ID NO: 1 contains one or more introns
  • the entire gene- all exons, introns and the regulatoiy sequences- may be deleted.
  • less than the entire gene may be deleted.
  • a single exon and/or intron may be deleted, so as to create an animal expressing a modified version of a protein of the invention.
  • the term "germ cell line transgenic animal” refers to a transgenic animal in which the genetic alteration or genetic infomiation was introduced into a germ line cell, thereby conferring the ability of the transgenic animal to transfer the genetic information to offspring. If such offspring in fact possess some or all of that alteration or genetic infomiation, then they too are transgenic animals.
  • the alteration or genetic information may be foreign to the species of animal to wliich the recipient belongs, foreign only to the particular individual recipient, or may be genetic information already possessed by the recipient. In the last case, the altered or introduced gene may be expressed differently than the native gene.
  • Transgenic animals can be produced by a variety of different methods including transfection, electroporation, microinjection, gene targeting in embryonic stem cells and recombinant viral and retroviral infection (see, e.g., U.S. Patent No. 4,736,866; U.S. Patent No.
  • mice A number of recombinant or transgenic mice have been produced, including those which express an activated oncogene sequence (U.S. Patent No. 4,736,866); express simian SV40 T-antigen (U.S. Patent No. 5,728,915); lack the expression of interferon regulatoiy factor 1 (IRF-1) (U.S. Patent No. 5,731,490); exhibit dopaminergic dysfunction (U.S. Patent No. 5J23J19); express at least one human gene which participates in blood pressure control (U.S. Patent No. 5,731,489); display greater similarity to the conditions existing in naturally occumng Alzheimer's disease (U.S. Patent No.
  • mice and rats remain the animals of choice for most transgenic experimentation, in some instances it is preferable or even necessary to use alternative animal species.
  • Transgenic procedures have been successfully utilized in a variety of non-murine animals, including sheep, goats, pigs, dogs, cats, monkeys, chimpanzees, hamsters, rabbits, cows and guinea pigs (see, e.g.
  • the method of introduction of nucleic acid fragments into recombination competent mammalian cells can be by any method which favors co-transfonuation of multiple nucleic acid molecules.
  • Detailed procedures for producing transgenic animals are readily available to one skilled in the art, including the disclosures in U.S. Patent No. 5,489,743 and U.S. Patent No. 5,602,307. L. Diagnostic Methods
  • the genes and proteins of the invention are differentially expressed in BPH tissue compared to nomial prostate tissue
  • the genes and proteins of the invention may be used to diagnose or monitor BPH, prostate function, or to track disease progression.
  • One means of diagnosing BPH using the nucleic acid molecules or proteins of the invention involves obtaining prostate tissue from living subjects. Obtaining tissue samples from living sources is problematic for tissues such as prostate. However, due to the nature of the treatment paradigms for BPH, biopsy may be necessary. When possible, urine, blood or peripheral lymphocyte samples may be used as the tissue sample in the assay.
  • nucleic acid probes comprising all or at least part of the sequence of SEQ ID NO: 1 may be used to detennine the expression of a nucleic acid molecule in forensic/pathology specimens.
  • nucleic acid assays may be canied out by any means of conducting a transcriptional profiling analysis.
  • forensic methods of the invention may target the proteins of the invention, particularly a protein comprising SEQ ID NO: 2, to detemiine up or down regulation of the genes (Shiverick et al, Biochim Biophys Ada 393, 124-133, 1975).
  • Methods of the invention may involve treatment of tissues with collagenases or other proteases to make the tissue amenable to cell lysis (Semenov et al, Biull Eksp Biol Med
  • Assays to detect nucleic acid or protein molecules of the invention may be in any available fomiat.
  • Typical assays for nucleic acid molecules include hybridization or PCR- based fomiats.
  • Typical assays for the detection of proteins, polypeptides or peptides of the invention include the use of antibody probes in any available fomiat such as in situ binding assays, etc. (see Harlow & Lane, Antibodies - A Laboratory Manual. Cold Spring Harbor Laboratoiy Press, Cold Spring Harbor, NY, 1988). hi prefened embodiments, assays are carried-out with appropriate controls.
  • the above methods may also be used in other diagnostic protocols, including protocols and methods to detect disease states in other tissues or organs, for example the tissues in which gene expression is detected.
  • Human tissue was obtained from the transitional zone of the prostate in biopsy samples from normal individuals and from patients with BPH or prostate cancer. BPH was defined histologically in all samples. Nonnal tissue and asymptomatic BPH samples came from individuals who died of trauma, and did not report symptoms. Patients having BPH with symptoms were defined as those with a need for frequent urination; in these patients a radical prostatectomy had been performed. Prostate cancer patients provided age-matched tissue samples for symptomatic BPH patients, but were without symptoms and without cancer in the transitional zone under histological examination. BRF-55T cells, an immortalized human prostate cell line from an individual with BPH, were also screened.
  • First strand cDNA synthesis was primed with a T7- (dT24) ohgonucleotide.
  • the cDNA was phenol-chlorofonn extracted and ethanol precipitated to a final concentration of 1 ⁇ g/ ⁇ l.
  • nucleotides Bio-11-CTP and Bio-16-UTP were added into the reaction. Following a 37°C incubation for six hours, impurities were removed from the labeled cRNA following the RNAeasy Mini kit protocol
  • cRNA was fragmented (5' fragmentation buffer consisting of 200 mM Tris-acetate
  • Affymetrix protocol 55 ⁇ g of fragmented cRNA was hybridized on the Human 42k 5-chip set and the HuGeneFL array for twenty-four hours at 60 rpm in a 45 °C hybridization oven. The chips were washed and stained with Streptavidin Phycoerythrin (S APE) (Molecular Probes) in
  • SAPE solution was added twice with an anti-streptavidin biotinylated antibody (Vector Laboratories) staining step in between.
  • Hybridization to the probe anays was detected by fluorometric scanning (Hewlett Packard Gene Array Scanner). Data was analyzed using Affymetrix GeneChip version 3.0 and Expression Data Mining Tool (EDMT) software (version 1.0).
  • EDMT Expression Data Mining Tool
  • SEQ ID NO: 1 (clone no. F 13763) is upregulated (4-fold) in prostate tissue samples from patients with BPH with symptoms (p-0.0002) and in samples from the non-cancerous hyperplastic region of prostate cancer patients, compared to nomial prostate tissue samples. It is not upregulated in BPH without symptoms.
  • SEQ ED NO: 1 is also expressed in BRF-55T cells, a prostate epithelial cell line immortalized from a patient with BPH.
  • the full length cDNA was obtained by the GeneTrapper ® method (Life Technologies).
  • a gene-specific oligo was designed based on the sequence of the clone no. F 13763 oligonucleotides in Example 1.
  • the oligo was labeled with biotin and used to hybridize with single strand plasmid DNA (cDNA-recombinants) from a human MRC-5 cDNA library following the GeneTrapper ® procedures.
  • the hybridized cDNAs were separated by streptavidin-conjugated paramagnetic beads and eluted by washing.
  • the eluted cDNA was converted to double strand plasmid DNA and used to transfomi E. coli cells (DH10B) and the longest cDNA was screened. After positive selection was confirmed by PCR using gene-specific primers, the cDNA clone was subjected to DNA sequencing.
  • the nucleotide sequences of the full-length human cDNA of clone no. F13763, conesponding to the differentially regulated niRNAs detected above, is set forth in SEQ ID NO: 1.
  • the cDNA comprises 5132 base pairs, with an open reading frame at nucleotides 395- 3790 encoding a protein of 1132 amino acids.
  • the amino acid sequence conesponding to the encoded protein is set forth in SEQ LD NO: 2.
  • the polypeptide of the invention also shares a low level of amino acid sequence similarity to human vascular cadherin-2 (GenBank accession number AB026893; 1184 amino acids) and mouse vascular cadherin-2 (GenBank accession number Y08715; 1180 amino acids).
  • RGD Arg-Gly-Asp tripeptide sequence starting at amino acid 872.
  • the cadherin-like motifs all fall between amino acids 120 and 580, i.e. within the putative extracellular region of the polypeptide of the invention.
  • the RGD peptide is considered a crucial sequence for the interaction of fibronectin with its cell surface receptor (an integrin protein) and has also been found in a number of other proteins with cell adhesion roles, such as fibrinogen, vitronectin and some collagens. Eight potenital N-glycosylation sites are present starting at the asparagine at amino acid position 100, 266, 417, 556, 580, 638, 763 and
  • a possible glycosaminoglycan attachment site is present at amino acid 870; a region resembling a carboxypeptidase zinc-binding signature motif is found starting at amino acid 636 of the polypeptide of SEQ ID NO: 2, and numerous potential N-myristolation sites are also present, at amino acids 38, 189, 478, 508, 522, 590, 620, 704, 785, 788, 848 and 1071.
  • Figure 2 shows relative clone no. F13763 mRNA levels in various normal human tissues determined via Northern Blot.
  • a probe based on SEQ ID NO: 1 randomly labeled was exposed to human mRNA blots (available from ClonTech) for 1 hour at 68°C in Clontech ExpressHyb hybridization buffer, following standard methodology as described by Sambrook et al. (supra).
  • the highest expression levels in nomial tissue were observed in the brain and in the placenta, with a lower but significant level in the liver. Still lower levels of expression were observed in the lung, kidney, pancreas, and smooth muscle.
  • the detection of more than one mRNA species per lane indicates the presence of splice variants. Such splice variants are within the scope of the present invention.
  • Figure 3 shows the results of the semi-quantitative PCR analysis of expression levels of mRNA conesponding to SEQ ID NO: 1 in various nomial human tissue samples. Real time
  • the 7700 measures the fluorescence intensity of the sample each cycle and is able to detect the presence of specific amplicons within the PCR reaction.
  • Each sample was assayed for the level of GAPDH and mRNA con-esponding to SEQ ID NO: 1.
  • GAPDH detection was performed using Perkin Elmer part #402869 according to the manufacturer's directions.
  • Primers were designed from SEQ ID NO: 1 using Primer Express, a program developed by PE to efficiently find primers and probes for specific sequences. These primers (BPH-F13763 F39/R40S) were used in conjunction with SYBR green (Molecular Probes), a nonspecific double stranded DNA dye, to measure the expression level of mRNA conesponding to 391 bp of SEQ ID NO: 1, which was noimalized to the GAPDH level in each sample.
  • SYBR green Molecular Probes
  • Figure 4 shows the results of an electronic Northern assay, in which the expression level was measured across a panel of human nomial and BPH tissues using the Affymetrix human 42k 5-chip GeneChip set.
  • SEQ ID NO: 1 is upregulated in non-cancerous BPH tissue adjacent to cancerous tissue, it is likely to be of diagnostic and prognostic significance in primary tumors and in metastatic tissues in which it is found (see Figures 2, 3, and 4). SEQ ID NO: 1 is, therefore, a potential target for therapeutic intervention in both inflammatory and hyperplastic diseases.
  • SEQ ID NO: 1 is detected in the epithelial prostate cell line BRF55-T, by both semi-quantitative PCR ( Figure 3) and by Affymetrix GeneChip analysis ( Figure 1), it could be used as a cell line for drug screening.

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

The invention relates generally to the changes in gene expression in Benign Prostatic Hyperplasia (BPH). The invention relates specifically to a human gene family which is differentially expressed in BPH compared to normal prostate tissue.

Description

EXPRESSION OF A CADHERIN-LIKE PROTEIN IN BENIGN PROSTATIC HYPERPLASIA
INVENTORS: William E. Munger, Prakash Kullcarni and Robert H. Getzenberg
RELATED APPLICATION
This application claims priority to U.S. Provisional Application 60/254,'928, filed December 13, 2000, which is herein incorporated by reference in its entirety.
FIELD OF THE INVENTION
The invention relates generally to the changes in gene expression in prostate tissue removed from male patients with benign prostatic hyperplasia (BPH). The invention specifically relates to a novel human gene which is differentially expressed in BPH tissue compared to normal prostate tissue.
BACKGROUND OF THE INVENTION
Benign Prostatic Hyperplasia (BPH)
BPH is the most common benign tumor in men 60 years of age or older. It is estimated that one in four men living to the age of 80 will require treatment for this disease. BPH is usually noted clinically after the age of 50, the incidence increasing with age, but as many as two thirds of men between the ages of 40 and 49 demonstrate histological evidence of the disease.
The anatomic location of the prostate at the bladder neck enveloping the urethra plays an important role in the pathology of BPH, including bladder outlet obstraction. Two prostate components are thought to play a role in bladder outlet obstruction. The first is the relative increased prostate tissue mass. The second component is the prostatic smooth muscle tone. The causative factors of BPH in man has been intensively studied (see Ziada et al, Urology 53:1-6, 1999). In general, the two most important factors appear to be aging and the presence of functional testes. Although these factors appear to be key to the development of BPH, both appear to be nonspecific.
Cadherins 7). vivo, cell-cell adhesion plays an important role in a wide range of events including morphogenesis and organ formation, leukocyte extravasion, tumor metastasis and invasion, and the foπnation of cell junctions. Additionally, cell-cell adhesion is crucial for the maintenance of tissue integrity, e.g., the maintenance of the intestinal epithelial barrier and the integrity of cardiac muscle, hitercellular adhesion is mediated by specific cell surface adhesion molecules
(CAMs). There are many different families of CAMs, including the immunoglobulin, integrin, selectin and cadherin superfamilies, and each cell type expresses a unique combination of these molecules.
Cadhenns are a rapidly expanding family of calcium-dependent CAMs. The classical cadherins are integral membrane glycoproteins that generally promote cell adhesion through homophilic interactions. They have an N-temiinal extracellular domain that determines binding specificity, a hydrophobic membrane-spanning domain and a C-terminal cytoplasmic domain (highly conserved among the members of the superfamily) that interacts with the cytoskeleton through catenins and other cytoskeleton-associated proteins. Some cadherins lack a cytoplasmic domain, however, and appear to function in cell-cell adhesion by a different mechanism than cadherins that do have a cytoplasmic domain. Calcium binding motifs are interspersed throughout the extracellular domain.
Cadherins have been shown to regulate epithelial, endothelial, neural and cancer cell adhesion, with different cadherins expressed on different cell types. For example, N (neural)- cadherin is predominantly expressed by neural cells, endothelial cells and a variety of cancer cell types. E (epithelial)-cadherin is predominantly expressed by epithelial cells. Other cadherins are P (placental)-cadherin, which is found in human skin and R (retinal)-cadherin.
The unique expression pattern of the different cadherins is particularly significant with respect to the role each subclass of cadherins may play in vivo in normal events (e.g., the maintenance of the intestinal epithelial ba ier) and in abnormal events (e.g., autoimmune diseases, tumor metastatis or inflammation). Different subclasses or combinations of subclasses of cadherins are likely to be responsible for different cell-cell adhesion events in which therapeutic detection and/or intervention may be desirable. Studies have also suggested that cadherins may have some regulatory activity in addition to adhesive activity. Thus, therapeutic intervention in the regulatory activities of cadherins expressed in specific tissues may also be desirable.
For recent reviews on cadherins, see Yagi et al, "Cadherin superfamily genes: functions, genomic organization, and neurologic activity "Genes Dev 14: 1169-1180, 2000;
Gumbiner, BM "Regulation of cadherin adhesive activity," J Cell Biol 148:399-404, 2000;
Dejana et al, "Vascular endothelial (VE)-cadherin: only an intercellular glue?" Exp Cell Res
252:13-19, 1999 and U.S. patent 5,639,634 (S. Suzuki), and references therein.
Molecular Changes in BPH
Little is known about the molecular changes in prostate cells associated with the development and progression of BPH. It has been demonstrated that the expression levels of a number of individual genes are changed compared to normal prostate cells. These changes in gene expression include a decreased level of Wilm's tumor gene (WT-1) and increased expression of insulin growth factor II (IGF-II) (Dong et al, J Clin Endocrin Metab 82:2198- 2203, 1997).
While the changes in the expression levels of a number of individual genes have been identified, the investigation of the global changes in gene expression has not been reported. Accordingly, there exists a need for the investigation of the changes in global gene expression levels as well as the need for the identification of new molecular markers associated with the development and progression of BPH. Furthemiore, if intervention is expected to be successful in halting or slowing down BPH, means of accurately assessing the early manifestations of BPH need to be established. One way to accurately assess the early manifestations of BPH is to identify markers which are uniquely associated with disease progression. Likewise, the development of therapeutics to prevent or stop the progression of BPH relies on the identification of genes responsible for BPH growth and function.
SUMMARY OF THE INVENTION The present invention is based on the discovery of a new gene family that is differentially expressed in BPH tissue compared to normal prostate tissue. The invention includes isolated nucleic acid molecules selected from the group consisting of an isolated nucleic acid molecule that encodes the amino acid sequence of SEQ ID NO: 2, an isolated nucleic acid molecule that encodes a fragment of at least 6 contiguous amino acids of SEQ ID NO: 2, an isolated nucleic acid molecule which hybridizes to the complement of a nucleic acid molecule comprising SEQ LD NO: 1 and an isolated nucleic acid molecule which hybridizes to the complement of a nucleic acid molecule that encodes the amino acid sequence of SEQ ID NO: 2. Nucleic acid molecules of the invention may encode a protein having at least about
50%, 60%, or 65% amino acid sequence identity to SEQ ID NO: 2 preferably at least about
70% or 75% sequence identity, more preferably at least about 80-85% sequence identity, and even more preferably at least about 90-95% sequence identity to SEQ ID NO: 2. The present invention further includes the nucleic acid molecules operably linked to one or more expression control elements, including vectors comprising the isolated nucleic acid molecules. The invention further includes host cells transformed to contain the nucleic acid molecules of the invention and methods for producing a protein comprising culturing a host cell transfonned with a nucleic acid molecule of the invention under conditions in which the protein is expressed.
The invention further provides an isolated polypeptide selected from the group consisting of an isolated polypeptide comprising the amino acid sequence of SEQ ID NO: 2, an isolated polypeptide comprising a functional or antigenic fragment of at least 10 contiguous amino acids of SEQ ID NO: 2, an isolated polypeptide comprising conservative amino acid substitutions of SEQ ID NO: 2 and an isolated polypeptide comprising naturally occumng amino acid sequence variants of SEQ ID NO: 2. Polypeptides of the invention also include polypeptides with an amino acid sequence having at least about 50%, 60%, 65%, 70% or 75% amino acid sequence identity with the sequence set forth in SEQ ID NO: 2 more preferably at least about 80-85%, even more preferably at least about 90%, and most preferably at least about 95%o sequence identity with the sequence set forth in SEQ ID NO: 2.
The invention further provides an isolated antibody or antigen-binding fragment that specifically binds to a polypeptide of the invention, including monoclonal and polyclonal antibodies.
The invention further provides methods of identifying an agent which modulates the expression of a nucleic acid encoding a protein of the invention, comprising: exposing cells which express the nucleic acid to the agent; and determining whether the agent modulates expression of said nucleic acid, thereby identifying an agent which modulates the expression of a nucleic acid encoding the protein.
The invention further provides methods of identifying an agent which modulates the level of or at least one activity of a protein of the invention, comprising: exposing cells which express the protein to the agent; and determining whether the agent modulates the level of or at least one activity of said protein, thereby identifying an agent which modulates the level of or at least one activity of the protein.
The invention further provides methods of identifying binding partners for a protein of the invention, comprising: exposing said protein to a potential binding partner; and deteiTnining if the potential binding partner binds to said protein, thereby identifying binding partners for the protein.
The present invention further provides methods of modulating the expression of a nucleic acid encoding a protein of the invention, comprising administering an effective amount of an agent which modulates the expression of a nucleic acid encoding the protein of the invention. The invention also provides methods of modulating at least one activity of a protein of the invention, comprising administering an effective amount of an agent which modulates at least one activity of the protein of the invention.
The present invention further includes non-human transgenic animals modified to contain the nucleic acid molecules of the invention or mutated nucleic acid molecules such that expression of the encoded polypeptides of the invention is prevented. The present invention also includes non-human transgenic animals in which all or a portion of the gene comprising all or a portion of SEQ ID NO: 1 has been knocked out or deleted from the genome of the animal.
The invention further provides methods of diagnosing BPH or other disease states, comprising the steps of: acquiring a tissue, blood, urine or other sample from a subject; and determining the level of expression of a nucleic acid molecule of the invention or polypeptide of the invention.
The invention further includes compositions comprising a diluent and a polypeptide or protein selected from the group consisting of an isolated polypeptide comprising all or a portion of SEQ ID NO: 2, an isolated polypeptide comprising a fragment of at least 6 contiguous amino acids of SEQ ID NO: 2, an isolated polypeptide comprising one or more conservative ammo acid substitutions of SEQ ID NO: 2, naturally occurring amino acid sequence variants of SEQ ID NO: 2, and an isolated polypeptide with an amino acid sequence having at least about 50%, 60%, 70% or 75% amino acid sequence identity with the sequence set forth in SEQ ID NO: 2, preferably at least about 80%, more preferably at least about 90%, and most preferably at least about 95% sequence identity with the sequence set forth in SEQ ID NO: 2. BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 Figure 1 shows Affymetrix GeneChip® data on the expression of a F13763 nucleic acid molecule (SEQ ID NO: 1) in prostate biopsy samples from normal human patients and BPH patients, including BPH samples from individuals without symptoms (BPH- w/o), samples from BPH patients without symptoms who were diagnosed with prostate cancer (BPH-Ca), and samples from BPH patients with symptoms (BPH-with). In all cases, the subregion of the prostate analyzed was the transitional zone. JPN and BRF-55T are Japanese BPH samples and cell culture samples respectively. Both unadjusted values (upper panel; raw Average Difference values) and "floored" values are shown (lower panel; all Average Difference values less than 20 are assigned a value of 20).
Figure 2 Figure 2 shows the tissue distribution of RNA encoding the protein of F13763 (SEQ ID NO: 2) as analyzed by Northern blot. mRNA expression levels were measured across a panel of nom al tissues (ClonTech human mRNA blot). Lane M) RNA Marker; 1) brain; 2) heart; 3) placenta; 4) lung; 5) liver; 6) smooth muscle; 7) kidney; and 8) pancreas.
Figure 3 Figure 3 shows the results of semi-quantitative PCR for the expression of F 13763 (SEQ ID NO: 1) in various nomial tissues. Left gel panel: 25 cycles (25 ); right gel panel: 30 cycles (30 O). Lanes: M) DNA ladder; 1) heart; 2) brain; 3) leukocytes; 4) lung; 5) liver; 6) fetal brain; 7) kidney; S) spleen; 9) placenta; 10) BRF-55T; 11) glomeruli; and 12) osteoblast.
Figure 4 Figure 4 shows the results of an electronic Northern assay, in which the expression of the nuclei acid of SEQ LD NO: 1 was measured across a panel of human normal and BPH tissues using the Affymetrix human 42k GeneChip® set. For each tissue type, horizontal bar represents the mean +/- SDM for samples obtained from 3 or more nomial individuals.
Figure 5 Figure 5 shows a hydrophobicity analysis (PEPPLOT) of the polypeptide of SEQ LD NO:2. Analysis was done using the methods of Goldman et al. and of
Kyle-Doolittle.
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT I. General Description
The present invention is based in part on the identification of a new gene family that is differentially expressed in human BPH tissue compared to nomial human prostate tissue. This gene family corcesponds to the human cDNA of SEQ ID NO: 1. Genes that encode the human protein of SEQ ID NO: 2 may also be found in other animal species, particularly mammalian species.
The proteins and nucleic acids of the invention may be used as diagnostic agents to detect BPH. The proteins of the present invention can also serve as a target for agents that can be used to modulate the expression or activity of the proteins. For example, agents may be identified that modulate biological processes associated with prostate growth, including the hyperplastic process of BPH.
The present invention is further based on the development of methods for isolating binding partners that bind to the protein. Probes based on the protein are used as capture probes to isolate potential binding partners, such as other proteins. Dominant negative proteins, DNAs encoding these proteins, antibodies to these proteins, peptide fragments of these proteins or mimics of these proteins may be mtroduced into cells to affect function. Additionally, the protein provides novel targets for the screening of synthetic small molecules and combinatorial or naturally occumng compound libraries to discover novel therapeutics to regulate prostate function.
II. Specific Embodiments
A. The Proteins Associated with BPH
The present invention provides isolated proteins, allelic variants of the proteins, and conservative amino acid substitutions of the proteins. As used herein, the "protein" or "polypeptide" refers, in part, to a protein that has the human amino acid sequence depicted in SEQ ID NO: 2. The terms also refer to naturally occumng allelic variants and proteins that have a slightly different amino acid sequence than that specifically recited above. Allelic variants, though possessing a slightly different amino acid sequence than those recited above, will still have the same or similar biological functions associated with these proteins.
The present invention also encompasses proteins translated from alternative splice variants of the genes encoding the identified proteins.
As used herein, the family of proteins related to the human amino acid sequence of SEQ ID NO: 2 refers to proteins that have been isolated from organisms in addition to humans. The methods used to identify and isolate other members of the family of proteins related to these proteins are described below.
The proteins of the present invention are preferably in isolated fomi. As used herein, a protein is said to be isolated when physical, mechanical or chemical methods are employed to remove the protein from cellular constituents that are normally associated with the protein. A skilled artisan can readily employ standard purification methods to obtain an isolated protein.
The proteins of the present invention further include insertion, deletion or conservative amino acid substitution variants of SEQ ID NO: 2. As used herein, a conservative variant refers to alterations in the amino acid sequence that do not adversely affect the biological functions of the protein. A substitution, insertion or deletion is said to adversely affect the protein when the altered sequence prevents or disrupts a biological function associated with the protein. For example, the overall charge, structure or hydrophobic/hydrophilic properties of the protein can be altered without adversely affecting a biological activity. Accordingly, the amino acid sequence can be altered, for example to render the peptide more hydrophobic or hydrophilic, without adversely affecting the biological activities of the protein.
Ordinarily, the allelic variants, the conservative substitution variants, and the members of the protein family, will have an amino acid sequence having at least about 50%, 60%, 65%, 70% or 75% amino acid sequence identity with the sequence set forth in SEQ ID NO: 2, more preferably at least about 80%, even more preferably at least about 90%, and most preferably at least about 95% sequence identity. Identity or homology with respect to such sequences is defined herein as the percentage of amino acid residues in the candidate sequence that are identical with the known peptides, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent homology, and not considering any conservative substitutions as part of the sequence identity (see section B for the relevant parameters). Fusion proteins, or N-terminal, C-tei rrinal or internal extensions, deletions, or insertions into the peptide sequence shall not be construed as affecting homology.
Thus, the proteins of the present invention include molecules having the amino acid sequence disclosed in SEQ ID NO: 2; fragments thereof having a consecutive sequence of at least about 3, 4, 5, 6, 10, 15, 20, 25, 30, 35 or more amino acid residues of these proteins; amino acid sequence variants wherein one or more amino acid residues has been inserted N- or
C-terminal to, or within, the disclosed coding sequence; and amino acid sequence variants of the disclosed sequence, or their fragments as defined above, that have been substituted by at least one residue. Such fragments, also referred to as peptides or polypeptides, may contain antigenic regions, functional regions of the protein identified as regions of the amino acid sequence which correspond to known protein domains, as well as regions of pronounced hydrophilicity. The regions are all easily identifiable by using commonly available protein sequence analysis software such as Mac Vector (Oxford Molecular).
Contemplated variants further include those containing predetemiined mutations by, e.g., homologous recombination, site-directed or PCR mutagenesis, and the corresponding proteins of other animal species, including but not limited to rabbit, mouse, rat, porcine, bovine, ovine, equine and non-human primate species, and the alleles or other naturally occumng variants of the family of proteins; and derivatives wherein the protein has been covalently modified by substitution, chemical, enzymatic, or other appropriate means with a moiety other than a naturally occumng amino acid (for example a detectable moiety such as an enzyme or radioisotope).
As described below, members of the family of proteins can be used: (1) to identify agents which modulate at least one activity of the protein; (2) to identify binding partners for the protein, (3) as an antigen to raise polyclonal or monoclonal antibodies; (4) as a therapeutic agent or target; and (5) as a diagnostic agent or marker.
B. Nucleic Acid Molecules The present invention further provides nucleic acid molecules that encode the protein having SEQ ID NO: 2 and the related proteins herein described, preferably in isolated fomi. As used herein, "nucleic acid" is defined as RNA or DNA that encodes a protein or peptide as defined above, is complementary to a nucleic acid sequence encoding such peptides, hybridizes to such a nucleic acid and remains stably bound to it under appropriate stringency conditions, or encodes a polypeptide sharing at least about 50%, 60%, 70% or 75% sequence identity, preferably at least about 80%, more preferably at least about 85%, and even more preferably at least about 90% or 95% or more identity with the peptide sequences. Alternatively, nucleic acid molecules will have at least about 50%, 60%, 70% or 75% nucleotide sequence identity to
SEQ LO NO: 1, preferably about S0%, more preferably about 85%, and even more preferably
90% or 95% nucleotide sequence identity to SEQ ID NO: 1, particularly over the open reading frame of SEQ ID NO: 1. Specifically contemplated are genomic DNA, cDNA, mRNA and antisense molecules, as well as nucleic acids based on alternative backbones or including alternative bases whether derived from natural sources or synthesized. Such hybridizing or complementary nucleic acids, however, are defined further as being novel and unobvious over any prior art nucleic acid including that wliich encodes, hybridizes under appropriate stringency conditions, or is complementary to nucleic acid encoding a protein according to the present invention.
Homology or identity at the nucleotide or amino acid sequence level is deteπnined by BLAST (Basic Local Alignment Search Tool) analysis using the algorithm employed by the programs blastp, blastn, blastx, tblastn and tblastx (Karlin et al, Proc Natl Acad Sci USA 87: 2264-2268, 1990, and Altschul, JMol Evol 36: 290-300, 1993, fully incorporated by reference) which are tailored for sequence similarity searching. The approach used by the BLAST program is to first consider similar segments between a query sequence and a database sequence, then to evaluate the statistical significance of all matches that are identified, and finally to summarize only those matches wliich satisfy a preselected threshold of significance. For a discussion of basic issues in similarity searching of sequence databases, see Altschul et al., Nature Genetics 6: 119-129, 1994, which is fully incorporated by reference. The search parameters for histogram, descriptions, alignments, expect (i.e., the statistical significance threshold for reporting matches against database sequences), cutoff, matrix and filter are at the default settings. The default scoring matrix used by blastp, blastx, tblastn, and tblastx is the BLOSUM62 matrix (Henikoff et al, Proc Natl Acad Sci USA 89: 10915-10919, 1992, fully incorporated by reference). For blastn, the scoring matrix is set by the ratios of M (i.e., the reward score for a pair of matching residues) to N (i.e., the penalty score for mismatching residues), wherein the default values for M and N are 5 and -4, respectively. Four blastn parameters were adjusted as follows: Q=10 (gap creation penalty); R=10 (gap extension penalty); wink=l (generates word hits at every wink* position along the query); and gapw=16 (sets the window width witliin wliich gapped alignments are generated). The equivalent Blastp parameter settings were Q=9; R=2; wink=l; and gapw=32. A Bestfit comparison between sequences, available in the GCG package version 10.0, uses DNA parameters GAP=50 (gap creation penalty) and LEN=3 (gap extension penalty) and the equivalent settings in protein comparisons are GAP=8 and LEN=2.
"Stringent conditions" are those that (1) employ low ionic strength and high temperature for washing, for example, 0.015 M NaCl/0.0015 M sodium citrate/0.1% SDS at 50°C, or (2) employ during hybridization a denaturing agent such as formamide, for example,
50% (vol/vol) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM NaCl, 75 mM sodium citrate at 42°C. Another example is hybridization in 50% fonnamide, 5x SSC (0.75 M
NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5x Denhardt's solution, sonicated salmon sperm DNA (50 μg/ml), 0.1% SDS, and 10% dextran sulfate at 42°C, with washes at 42°C in 0.2x SSC and 0.1% SDS. A skilled artisan can readily detemiine and vary the stringency conditions appropriately to obtain a clear and detectable hybridization signal. Preferred molecules are those that hybridize under the above conditions to the complement of SEQ ID NO: 1 and which encode a functional protein. Even more preferred hybridizing molecules are those that hybridize under the above conditions to the complement strand of the open reading frame of SEQ ID NO: 1.
As used herein, a nucleic acid molecule is said to be "isolated" when the nucleic acid molecule is substantially separated from contaminant nucleic acid molecules encoding other polypeptides. The present invention further provides fragments of the encoding nucleic acid molecule. As used herein, a fragment of an encoding nucleic acid molecule refers to a small portion of the entire protein coding sequence. The size of the fragment will be determined by the intended use. For example, if the fragment is chosen so as to encode an active portion of the protein, the fragment will need to be large enough to encode the functional region(s) of the protein. For instance, fragments which encode peptides coiresponding to predicted antigenic regions may be prepared (see Figure 5). If the fragment is to be used as a nucleic acid probe or PCR primer, then the fragment length is chosen so as to obtain a relatively small number of false positives during probing/priming (see the discussion in Section H).
Fragments of the encoding nucleic acid molecules of the present invention (i.e., synthetic oligonucleotides) that are used as probes or specific primers for the polymerase chain reaction (PCR), or to synthesize gene sequences encoding proteins of the invention, can easily be synthesized by chemical techniques, for example, the phosphoramidite method of Matteucci et al., J Am Chem Soc 103: 3185-3191, 1981, or using automated synthesis methods. In addition, larger DNA segments can readily be prepared by well known methods, such as synthesis of a group of oligonucleotides that define various modular segments of the gene, followed by ligation of oligonucleotides to build the complete modified gene. The encoding nucleic acid molecules of the present invention may further be modified so as to contain a detectable label for diagnostic and probe purposes. A variety of such labels are known in the art and can readily be employed with the encoding molecules herein described. Suitable labels include, but are not limited to, biotin, radiolabeled nucleotides and the like. A skilled artisan can readily employ any such label to obtain labeled variants of the nucleic acid molecules of the invention.
Modifications to the primary structure itself by deletion, addition, or alteration of the amino acids incorporated into the protein sequence during translation can be made without destroying the activity of the protein. Such substitutions or other alterations result in proteins having an amino acid sequence encoded by a nucleic acid falling within the contemplated scope of the present invention.
C. Isolation of Other Related Nucleic Acid Molecules
As described above, the identification and characterization of the human nucleic acid molecule having SEQ ID NO: 1 allows a skilled artisan to isolate nucleic acid molecules that encode other members of the protein family in addition to the sequences herein described.
Further, the presently disclosed nucleic acid molecules allow a skilled artisan to isolate nucleic acid molecules that encode other members of the family of proteins in addition to the protein having SEQ ID NO: 2.
Essentially, a skilled artisan can readily use the amino acid sequence of SEQ ID NO: 2 to generate antibody probes to screen expression libraries prepared from appropriate cells. Typically, polyclonal antiserum from mammals such as rabbits immunized with the purified protein (as described below) or monoclonal antibodies can be used to probe a mammalian cDNA or genomic expression library, such as lambda gtll library, to obtain the appropriate coding sequence for other members of the protein family. The cloned cDNA sequence can be expressed as a fusion protein, expressed directly using its own control sequences, or expressed by constructions using control sequences appropriate to the particular host used for expression of the enzyme. Alternatively, a portion of the coding sequence herein described can be synthesized and used as a probe to retrieve DNA encoding a member of the protein family from any mammalian organism. Oligomers containing approximately 18-20 nucleotides (encoding about a 6-7 amino acid stretch) are prepared and used to screen genomic DNA or cDNA libraries to obtain hybridization under stringent conditions or conditions of sufficient stringency to eliminate an undue level of false positives.
Additionally, pairs of oligonucleotide primers can be prepared for use in a polymerase chain reaction (PCR) to selectively clone an encoding nucleic acid molecule. A PCR denature- anneal-extend cycle for using such PCR primers is well known in the art and can readily be adapted for use in isolating other encoding nucleic acid molecules.
Nucleic acid molecules encoding other members of the protein family may also be identified in existing genomic or other sequence information using any available computational method, including but not limited to: PSI-BLAST (Altschul, et al, Nucleic Acids Res 25:3389- 3402, 1997); PHI-BLAST (Zhang, et al. Nucleic Acids Res 26:3986-3990, 1998), 3D-PSSM (Kelly et al, J Mol Biol 299(2): 499-520, 2000); and other computational analysis methods (Shi et al, Biochem Biophys Res Commun 262(1):132-138, 1999, and Matsunami et. al, Nature 404(6778):601-604, 2000.
D. rDNA molecules Containing a Nucleic Acid Molecule The present invention further provides recombinant DNA molecules (rDNAs) that contain a coding sequence. As used herein, a rDNA molecule is a DNA molecule that has been subjected to molecular manipulation in situ. Methods for generating rDNA molecules are well known in the art, for example, see Sambrook et al., Molecular Cloning - A Laboratory Manual. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989. h the prefeired rDNA molecules, a coding DNA sequence is operably linked to expression control sequences and/or vector sequences.
The choice of vector and/or expression control sequences to which one of the protein family encoding sequences of the present invention is operably linked depends directly, as is well known in the art, on the functional properties desired, e.g., protein expression, and the host cell to be transformed. A vector contemplated by the present invention is at least capable of directing the replication or insertion into the host chromosome, and preferably also expression, of the structural gene included in the rDNA molecule. Expression control elements that are used for regulating the expression of an operably linked protein encoding sequence are known in the art and include, but are not limited to, inducible promoters, constitutive promoters, secretion signals, and other regulatory elements.
Preferably, the inducible promoter is readily controlled, such as being responsive to a nutrient in the host cell's medium.
In one embodiment, the vector containing a coding nucleic acid molecule will include a prokaryotic replicon, i.e., a DNA sequence having the ability to direct autonomous replication and maintenance of the recombinant DNA molecule extrachromosomally in a prokaryotic host cell, such as a bacterial host cell, transformed therewith. Such replicons are well known in the art. In addition, vectors that include a prokaryotic replicon may also include a gene whose expression confers a detectable marker such as a drug resistance. Typical bacterial dmg resistance genes are those that confer resistance to ampicillin or tetracycline.
Vectors that include a prokaryotic replicon can further include a prokaryotic or bacteriophage promoter capable of directing the expression (transcription and translation) of the coding gene sequences in a bacterial host cell, such as E. coli. A promoter is an expression control element formed by a DNA sequence that permits binding of RNA polymerase and transcription to occur. Promoter sequences compatible with bacterial hosts are typically provided in plasmid vectors containing convenient restriction sites for insertion of a DNA segment of the present invention. Typical of such vector plasmids are pUC8, pUC9, pBR322 and pBR329 available from BioRad Laboratories, (Richmond, CA), pPL and pKK223 available from Phannacia (Piscataway, NJ).
Expression vectors compatible with eukaryotic cells, preferably those compatible with vertebrate cells, such as prostate cells, can also be used to fomi rDNA molecules that contain a coding sequence. Eukaryotic cell expression vectors, including viral vectors, are well known in the art and are available from several commercial sources. Typically, such vectors are provided containing convenient restriction sites for insertion of the desired DNA segment. Typical of such vectors are pSVL and pKSV-10 (Pharmacia), pBPV-l/pML2d (International Biotechnologies, Inc.), pTDTl (ATCC, #31255), the vector pCDM8 described herein, and the like eukaryotic expression vectors. Vectors may be modified to include prostate cell specific promoters if needed.
Eukaryotic cell expression vectors used to construct the rDNA molecules of the present invention may further include a selectable marker that is effective in an eukaryotic cell, preferably a drug resistance selection marker. A prefened drug resistance marker is the gene whose expression results in neomycin resistance, i.e., the neomycin phosphotransferase (neo) gene (Southern et al, J Mol Anal Genet 1:327-341, 1982). Alternatively, the selectable marker can be present on a separate plasmid, and the two vectors are introduced by co-transfection of the host cell, and selected by culturing in the appropriate drug for the selectable marker.
E. Host Cells Containing an Exogenously Supplied Coding Nucleic Acid Molecule
The present invention further provides host cells transformed with a nucleic acid molecule that encodes a protein of the present invention. The host cell can be either prokaryotic or eukaryotic. Eukaryotic cells useful for expression of a protein of the invention are not limited, so long as the cell line is compatible with cell culture methods and compatible with the propagation of the expression vector and expression of the gene product. Preferred eukaryotic host cells include, but are not limited to, yeast, insect and mammalian cells, preferably vertebrate cells such as those from a mouse, rat, monkey or human cell line. Prefeired eukaryotic host cells include Chinese hamster ovary (CHO) cells available from the ATCC as CCL61, NIH Swiss mouse embryo cells (NIH/3T3) available from the ATCC as CRL 1658, baby hamster kidney cells (BHK), and the like eukaryotic tissue culture cell lines. Any prokaryotic host can be used to express a rDNA molecule encoding a protein of the invention. The preferred prokaryotic host is E. coli. Transfonnation of appropriate cell hosts with a rDNA molecule of the present invention is accomplished by well known methods that typically depend on the type of vector used and host system employed. With regard to transfonnation of prokaryotic host cells, electroporation and salt treatment methods are typically employed, see, for example, Cohen et al. , Proc Natl Acad Sci USA 69:2110, 1972; and Sambrook et al, (supra). With regard to transformation of vertebrate cells with vectors containing rDNAs, electroporation, cationic lipid or salt treatment methods are typically employed (see, for example, Graham et al, Virol 52:456, 1973; Wigler et al, Proc Natl Acad Sci USA 16:1313-1316, 1979).
Successfully transformed cells, i.e., cells that contain a rDNA molecule of the present invention, can be identified by well known techniques including the selection for a selectable marker. For example, cells resulting from the introduction of an rDNA of the present invention can be cloned to produce single colonies. Cells from those colonies can be harvested, lysed and their DNA content examined for the presence of the rDNA using a method such as that described by Southern, JMol Biol 98:503, 1975, or Berent et al, Biotech 3:208, 1985, or the proteins produced from the cell assayed via an immunological method.
F. Production of Recombinant Proteins using a rDNA Molecule The present invention further provides methods for producing a protein of the invention using nucleic acid molecules herein described, hi general temis, the production of a recombinant form of a protein typically involves the following steps:
A nucleic acid molecule is first obtained that encodes a protein of the invention, such as a nucleic acid molecule comprising, consisting essentially of or consisting of SEQ ID NO: 1 or nucleotides 395-3790 (-3793 with the stop codon) of SEQ ID NO: 1. If the encoding sequence is uninterrupted by introns, as is this open-reading-frame, it is directly suitable for expression in any host.
The nucleic acid molecule is then preferably placed in operable linkage with suitable control sequences, as described above, to fom an expression unit containing the protein open reading frame. The expression unit is used to transfomi a suitable host, and the transformed host is cultured under conditions that allow the production of the recombinant protein. Optionally the recombinant protein is isolated from the medium or from the cells. Recovery and purification of the protein may not be necessary in some instances where some impurities may be tolerated. Each of the foregoing steps can be done in a variety of ways. For example, the desired coding sequences may be obtained from genomic fragments and used directly in appropriate hosts. The construction of expression vectors that are operable in a variety of hosts is accomplished using appropriate replicons and control sequences, as set forth above. The control sequences, expression vectors, and transformation methods are dependent on the type of host cell used to express the gene and λvere discussed in detail earlier. Suitable restriction sites can, if not nomially available, be added to the ends of the coding sequence so as to provide an excisable gene to insert into these vectors. A skilled artisan can readily adapt any host/expression system known in the art for use with the nucleic acid molecules of the invention to produce recombinant protein.
G. Methods to Identify Binding Partners
Another embodiment of the present invention provides methods for isolating and identifying binding partners of proteins of the invention, hi general, a protein of the invention is mixed with a potential binding partner or an extract or fraction of a cell under conditions that allow the association of potential binding partners with the protein of the invention. After mixing, peptides, polypeptides, proteins or other molecules that have become associated with a protein of the invention are separated from the mixture. The binding partner that bound to the protein of the invention can then be removed and further analyzed. To identify and isolate a binding partner, the entire protein, for instance a protein comprising the entire amino acid sequence of SEQ ID NO: 2 can be used. Alternatively, a fragment of the protein can be used.
As used herein, a cellular extract refers to a preparation or fraction which is made from a lysed or disrupted cell. The prefeired source of cellular extracts will be cells derived from human prostate tissue or cells, for instance, biopsy tissue or tissue culture cells from subjects with BPH. Alternatively, cellular extracts may be prepared from nomial human prostate tissue or available cell lines, particularly prostate derived cell lines.
A variety of methods can be used to obtain an extract of a cell. Cells can be dismpted using either physical or chemical disruption methods. Examples of physical disruption methods include, but are not limited to, sonication and mechanical shearing. Examples of chemical lysis methods include, but are not limited to, detergent lysis and enzyme lysis. A skilled artisan can readily adapt methods for preparing cellular extracts in order to obtain extracts for use in the present methods. Once an extract of a cell is prepared, the extract is mixed with the protein of the invention under conditions in which association of the protein with the binding partner can occur. A variety of conditions can be used, the most preferred being conditions that closely resemble conditions found in the cytoplasm of a human cell. Features such as osmolarity, pH, temperature, and the concentration of cellular extract used, can be varied to optimize the association of the protein with the binding partner.
After mixing under appropriate conditions, the bound complex is separated from the mixture. A variety of techniques can be utilized to separate the mixture. For example, antibodies specific to a protein of the invention can be used to immunoprecipitate the binding partner complex. Alternatively, standard chemical separation techniques such as chromatography and density/sediment centrifugation can be used.
After removal of non-associated cellular constituents found in the extract, the binding partner can be dissociated from the complex using conventional methods. For example, dissociation can be accomplished by altering the salt concentration or pH of the mixture.
To aid in separating associated binding partner pairs from the mixed extract, the protein of the invention can be immobilized on a solid support. For example, the protein can be attached to a nitrocellulose matrix or acrylic beads. Attachment of the protein to a solid support aids in separating peptide/binding partner pairs from other constituents found in the extract. The identified binding partners can be either a single protein or a complex made up of two or more proteins. Alternatively, binding partners may be identified using a Far- Western assay according to the procedures of Takayama et al. , Methods Mol Biol 69:171-184, 1997, or Sauder et al, J Gen Virol 77:991-996, 1996, or identified through the use of epitope tagged proteins or GST fusion proteins.
Alternatively, the nucleic acid molecules of the invention can be used in a yeast two- hybrid system. The yeast two-hybrid system has been used to identify other protein partner pairs and can readily be adapted to employ the nucleic acid molecules herein described.
H. Methods to Identify Agents that Modulate the Expression of a Nucleic Acid Encoding the Genes Associated BPH
Another embodiment of the present invention provides methods for identifying agents that modulate the expression of a nucleic acid encoding a protein of the invention such as a protein having the amino acid sequence of SEQ ID NO: 2. Such assays may utilize any available means of monitoring for changes in the expression level of the nucleic acids of the invention. As used herein, an agent is said to modulate the expression of a nucleic acid of the invention if it is capable of up- or down-regulating expression of the nucleic acid in a cell. In one assay format, cell lines that contain reporter gene fusions between the open reading frame defined by nucleotides 395-3790 of SEQ ID NO: 1, and/or the 5 'and/or 3' regulatoiy elements and any assayable fusion partner may be prepared. Numerous assayable fusion partners are known and readily available including the firefly luciferase gene and the gene encoding chloramphenicol acetyltransferase (Alam et al. , Anal Biochem 188:245-254, 1990). Cell lines containing the reporter gene fusions are then exposed to the agent to be tested under appropriate conditions and time. Differential expression of the reporter gene between samples exposed to the agent and control samples identifies agents wliich modulate the expression of a nucleic acid of the invention.
Additional assay fonnats may be used to monitor the ability of the agent to modulate the expression of a nucleic acid encoding a protein of the invention, such as the protein having
SEQ ID NO: 2. For instance, mRNA expression may be niomtored directly by hybridization to the nucleic acids of the invention. Cell lines are exposed to the agent to be tested under appropriate conditions and time and total RNA or mRNA is isolated by standard procedures such those disclosed in Sambrook et al. (supra).
The preferred cells will be those derived from human prostate tissue, for instance, prostate biopsy tissue or cultured prostate cells from nomial or BPH patients, for example
BRF-55T cells (immortalized human prostate cells obtained from an individual with BPH;
Type, PT et al, "Establishment and characterization of immortalized human cell lines from prostatic carcinoma and benign prostatic hyperplasia," Int J Oncol 12:257-263, 1998). Alternatively, other available cells or cell lines may be used.
Probes to detect differences in RNA expression levels between cells exposed to the agent and control cells may be prepared from the nucleic acids of the invention. It is preferable, but not necessary, to design probes which hybridize only with target nucleic acids under conditions of high stringency. Only highly complementaiy nucleic acid hybrids form under conditions of high stringency. Accordingly, the stringency of the assay conditions determines the amount of complementarity which should exist between two nucleic acid strands in order to fomi a hybrid. Stringency should be chosen to maximize the difference in stability between the probe:target hybrid and probe:non-target hybrids. Probes may be designed from the nucleic acids of the invention through methods known in the art. For instance, the G+C content of the probe and the probe length can affect probe binding to its target sequence. Methods to optimize probe specificity are commonly available in Sambrook et al, (supra) or Ausubel et al, Current Protocols in Molecular Biology. Greene Publishing Co., 1995. Hybridization conditions are modified using known methods, such as those described by Sambrook et al. and Ausubel et al. as required for each probe. Hybridization of total cellular RNA or RNA enriched for polyA RNA can be accomplished in any available format. For instance, total cellular RNA or RNA enriched for polyA RNA can be affixed to a solid support and the solid support exposed to at least one probe comprising at least one, or part of one of the sequences of the invention under conditions in which the probe will specifically hybridize. Alternatively, nucleic acid fragments comprising at least one, or part of one of the sequences of the invention can be affixed to a solid support, such as a silicon chip or a porous glass wafer. The solid support can then be exposed to total cellular RNA or polyA RNA from a sample under conditions in wliich the affixed sequences will specifically hybridize. Such solid supports and hybridization methods are widely available, for example, those disclosed by
Beattie, WO 95/11755 (1995). By examining for the ability of a given probe to specifically hybridize to an RNA sample from an untreated cell population and from a cell population exposed to the agent, agents wliich up or down regulate the expression of a nucleic acid encoding the protein having the sequence of SEQ ID NO: 2 are identified.
Hybridization for qualitative and quantitative analysis of niRNAs may also be canied out by using a RNase Protection Assay (i.e., RPA, see Ma et al, Methods 10:273-238, 1996). Briefly, an expression vehicle comprising cDNA encoding the gene product and a phage- specific DNA-dependent RNA polymerase promoter (e.g., TI, T3 or SP6 RNA polymerase) is linearized at the 3' end of the cDNA molecule, downstream from the phage promoter, wherein such a linearized molecule is subsequently used as a template for synthesis of a labeled antisense transcript of the cDNA by in vitro transcription. The labeled transcript is then hybridized to a mixture of isolated RNA (i.e., total or fractionated mRNA) by incubation at 45 °C overnight in a buffer comprising 80% fomiamide, 40 mM Pipes, pH 6.4, 0.4 M NaCl and 1 mM EDTA. The resulting hybrids are then digested in a buffer comprising 40 μg/ml ribonuclease A and 2 μg/ml ribonuclease. After deactivation and extraction of extraneous proteins, the samples are loaded onto urea polyaciylamide gels for analysis. In another assay format, cells or cell lines are first identified which express the gene products of the invention physiologically, for example BRF-55T cells (e.g., by using assays of tissue distribution via Northern blot, although RPAs may serve the identical purpose of expression selection). Cell and/or cell lines so identified would be expected to comprise the necessary cellular machinery such that the fidelity of modulation of the transcriptional apparatus is maintained with regard to exogenous contact of agent with appropriate surface transduction mechanisms and/or the cytosolic cascades. Further, such cells or cell lines would be transduced or transfected with an expression vehicle (e.g., a plasmid or viral vector) construct comprising an operable non- translated 5 '-promoter containing end of the structural gene encoding the instant gene products fused to one or more antigenic fragments, which are peculiar to the instant gene products, wherein said fragments are under the transcriptional control of said promoter and are expressed as polypeptides whose molecular weight can be distinguished from the naturally occumng polypeptides or may further comprise an immunologically distinct tag or other detectable marker. Such a process is well known in the art (see Sambrook et al, supra).
Cells or cell lines transduced or transfected as outlined above are then contacted with agents under appropriate conditions; for example, the agent in a pharmaceutically acceptable excipient is contacted with cells in an aqueous physiological buffer such as phosphate buffered saline (PBS) at physiological pH, Eagles balanced salt solution (BSS) at physiological pH,
PBS or BSS comprising serum or conditioned media comprising PBS or BSS and/or serum incubated at 37 °C. Said conditions may be modulated as deemed necessary by one of skill in the art. Subsequent to contacting the cells with the agent, said cells will be dismpted and the polypeptides of the lysate are fractionated such that a polypeptide fraction is pooled and contacted with an antibody to be further processed by immuno logical assay (e.g., ELISA, immunoprecipitation or Western blot). The pool of proteins isolated from the "agent- contacted" sample will be compared with a control sample where only the excipient is contacted with the cells and an increase or decrease in the immunologically generated signal from the "agent-contacted" sample compared to the control will be used to distinguish the effectiveness of the agent.
I. Methods to Identify Agents that Modulate at Least One Activity of the BPH Associated Proteins Another embodiment of the present invention provides methods for identifying agents that modulate at least one activity of a protein of the invention such as the protein having the amino acid sequence of SEQ ID NO: 2. Such methods or assays may utilize any means of monitoring or detecting the desired activity.
In one format, the relative amounts of a protein of the invention between a cell population that has been exposed to the agent to be tested compared to an un-exposed control cell population may be assayed. In this fonnat, probes such as specific antibodies are used to monitor the differential expression of the protein in the different cell populations. Cell lines or populations are exposed to the agent to be tested under appropriate conditions and time. Cellular lysates may be prepared from the exposed cell line or population and a control, unexposed cell line or population. The cellular lysates are then analyzed with the probe.
Antibody probes are prepared by immunizing suitable mammalian hosts in appropriate imnumization protocols using the peptides, polypeptides or proteins of the invention if they are of sufficient length, or, if desired, or if required to enhance immunogenicity, conjugated to suitable earners. Methods for preparing immunogenic conjugates with earners such as BSA,
KLH, or other carrier proteins are well known in the art. In some circumstances, direct conjugation using, for example, carbodiimide reagents may be effective; in other instances linking reagents such as those supplied by Pierce Chemical Co. (Rockford, IL), may be desirable to provide accessibility to the hapten. The hapten peptides can be extended at either the amino or carboxy terminus with a cysteine residue or interspersed with cysteine residues, for example, to facilitate linking to a carrier. Administration of the immunogens is conducted generally by injection over a suitable time period and with use of suitable adjuvants, as is generally understood in the art. During the immunization schedule, titers of antibodies are taken to detennine adequacy of antibody fomiation.
While the polyclonal antisera produced in this way may be satisfactory for some applications, for phanuaceutical compositions, use of monoclonal preparations is prefened. Immortalized cell lines wliich secrete the desired monoclonal antibodies may be prepared using the standard method of Kohler and Milstein (Nature 256:495-497, 1975) or modifications which effect immortalization of lymphocytes or spleen cells, as is generally known. The immortalized cell lines secreting the desired antibodies are screened by immunoassay in wliich the antigen is the peptide hapten, polypeptide or protein. When the appropriate immortalized cell culture secreting the desired antibody is identified, the cells can be cultured either in vitro or by production in ascites fluid.
The desired monoclonal antibodies are then recovered from the culture supernatant or from the ascites supernatant. Fragments of the monoclonal antibodies or the polyclonal antisera wliich contain the immunologically significant portion can be used as antagonists, as well as the intact antibodies. Use of immunologically reactive antibody fragments, such as the Fab, Fab', or F(ab')2 fragments is often preferable, especially in a therapeutic context, as these fragments are generally less immunogenic than the whole immunoglobulin.
The antibodies or fragments may also be produced, using current technology, by recombinant means. Antibody regions that bind specifically to the desired regions of the protein can also be produced in the context of chimeras with multiple species origin, such as humanized antibodies.
In another assay format, one or more activities of a protein of the invention may be assayed in the presence and absence of an agent to be tested to determine whether they are capable of modulating (i.e. enhancing or inhibiting) this activity, for example cell adhesion between cells expressing a protein of the invention. The ability of an agent to modulate cell adhesion may generally be evaluated in vitro by assaying the effect of the agent on one or more of the following: (1) neurite outgrowth, (2) adhesion between endothelial cells, (3) adhesion between epithelial cells (e.g., nomial rat kidney cells and/or human skin), (4) adhesion between cancer cells, and/or (5) adhesion between other cell types, wherein the cells under study express a protein of the invention either endogenously or as a result of transfection with a nucleic acid of the invention. In general, an agent is considered to be a modulator of cell adhesion if, within one or more of these representative assays, contact of the test cells with the agent results in a discernible disruption of cell adhesion. For example, see U.S. Pat. No. 6,031,072; U.S. Pat. No. 5,891,706; Urushihara et al, "Transformation of Cell Adhesion Properties by Exogenously Introduced E-cadherin cDNA," Dev Biol 70: 206-216, 1979; Nagafuchi et al., Nature, 329:341-343, 1987.
Agents that are assayed in the above method can be randomly selected or rationally selected or designed. As used herein, an agent is said to be randomly selected when the agent is chosen randomly without considering the specific sequences involved in the association of a protein of the invention alone or with its associated substrates, binding partners, etc. An example of randomly selected agents is the use a chemical library or a peptide combinatorial library, or a growth broth of an organism. As used herein, an agent is said to be rationally selected or designed when the agent is chosen on a nonrandom basis wliich takes into account the sequence of the target site and/or its conformation in connection with the agent's action. Agents can be rationally selected or rationally designed by utilizing the peptide sequences that make up these sites. For example, a rationally selected peptide agent can be a peptide whose amino acid sequence is identical to or a derivative of any functional consensus site.
The agents of the present invention can be, as examples, peptides, small molecules, vitamin derivatives, as well as carbohydrates. Dominant negative proteins, DNAs encoding these proteins, antibodies to these proteins, peptide fragments of these proteins or mimics of these proteins may be introduced into cells to affect function. "Mimic" used herein refers to the modification of a region or several regions of a peptide molecule to provide a structure chemically different from the parent peptide but topographically and functionally similar to the parent peptide (see Grant GA in: Molecular Biology and Biotechnology. Meyers (ed.), pp. 659-664, VCH Publishers, New York, 1995). A skilled artisan can readily recognize that there is no limit as to the structural nature of the agents of the present invention.
The peptide agents of the invention can be prepared using standard solid phase (or solution phase) peptide synthesis methods, as is known in the art. In addition, the DNA encoding these peptides may be synthesized using commercially available oligonucleotide synthesis instrumentation and produced recombinantly using standard recombinant production systems. The production using solid phase peptide synthesis is necessitated if non-gene- encoded amino acids are to be included.
Another class of agents of the present invention are antibodies immunoreactive with critical positions of proteins of the invention. Antibody agents are obtained by inimunization of suitable mammalian subjects with peptides, containing as antigenic regions, those portions of the protein intended to be targeted by the antibodies.
J. Uses for Agents that Modulate the Expression or at Least One Activity of the Proteins As provided in the Examples, the proteins and nucleic acids of the invention, such as the protein having the amino acid sequence of SEQ LD NO: 2, are differentially expressed in BPH tissue. Agents that up-or-down-regulate or modulate the expression of the protein, or at least one activity of the protein, such as agonists or antagonists, may be used to modulate biological and pathologic processes associated with the protein's function and activity. As used herein, a subject can be any mammal, so long as the mammal is in need of modulation of a pathological or biological process mediated by a protein of the invention. The tenn "mammal" is defined as an individual belonging to the class Mammalia. The invention is particularly useful in the treatment of human subjects.
Pathological processes refer to a category of biological processes which produce a deleterious effect. For example, expression of a protein of the invention may be associated with prostate cell growth or hyperplasia. As used herein, an agent is said to modulate a pathological process when the agent reduces the degree or severity of the process. For instance, BPH may be prevented or disease progression modulated by the administration of agents which up- or down-regulate or modulate in some way the expression or at least one activity of a protein of the invention.
The agents of the present invention can be provided alone, or in combination with other agents that modulate a particular pathological process. For example, an agent of the present invention can be administered in combination with other known drugs. As used herein, two agents are said to be administered in combination when the two agents are administered simultaneously or are administered independently in a fashion such that the agents will act at the same time. The agents of the present invention can be administered via parenteral, subcutaneous, intravenous, intramuscular, intraperitoneal, transdermal, or buccal routes. Alternatively, or concurrently, administration may be by the oral route. The dosage administered will be dependent upon the age, health, and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the nature of the effect desired. The present invention further provides compositions containing one or more agents which modulate expression or at least one activity of a protein of the invention. While individual needs vary, detemiination of optimal ranges of effective amounts of each component is within the skill of the art. Typical dosages comprise 0J to 100 μg/kg body wt. The prefened dosages comprise 0J to 10 μg/kg body wt. The most prefeired dosages comprise 0J to 1 μg/kg body wt.
In addition to the phamiacologically active agent, the compositions of the present invention may contain suitable pharmaceutically acceptable earners comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used phamiaceutically for delivery to the site of action. Suitable formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble fomi, for example, water-soluble salts. In addition, suspensions of the active compounds as appropriate oily injection suspensions may be administered. Suitable lipophilic solvents or vehicles include fatty oils, e.g., sesame oil, or synthetic fatty acid esters, e.g., ethyl oleate or triglycerides. Aqueous injection suspensions may contain substances wliich increase the viscosity of the suspension include, for example, sodium carboxymethyl cellulose, sorbitol, and/or dextran. Optionally, the suspension may also contain stabilizers. Liposomes can also be used to encapsulate the agent for delivery into the cell.
The phamiaceutical formulation for systemic administration according to the invention may be formulated for enteral, parenteral or topical administration. Indeed, all three types of formulations may be used simultaneously to achieve systemic administration of the active ingredient. Suitable formulations for oral administration include hard or soft gelatin capsules, pills, tablets, including coated tablets, elixirs, suspensions, syrups or inhalations and controlled release fomis thereof. h practicing the methods of this invention, the compounds of this invention may be used alone or in combination, or in combination with other therapeutic or diagnostic agents. In certain prefeired embodiments, the compounds of this invention may be coadministered along with other compounds typically prescribed for these conditions according to generally accepted medical practice. The compounds of this invention can be utilized in vivo, ordinarily in mammals, such as humans, sheep, horses, cattle, pigs, dogs, cats, rats and mice, or in vitro.
K. Transgenic Animals Transgenic animals containing mutant, knock-out or modified genes corresponding to the cDNA sequence of SEQ ID NO: 1, or the open reading frame encoding the polypeptide sequence of SEQ ID NO: 2, or fragments thereof having a consecutive sequence of at least about 3, 4, 5, 6, 10, 15, 20, 25, 30, 35 or more amino acid residues, are also included in the invention. Transgenic animals are genetically modified animals into which recombinant, exogenous or cloned genetic material has been experimentally transferred. Such genetic material is often referred to as a "transgene." The nucleic acid sequence of the transgene, in some embodiments, all or a portion of SEQ ID NO: 1, may be integrated either at a locus of a genome where that particular nucleic acid sequence is not otherwise nonrially found or at the nomial locus for the transgene. The transgene may consist of nucleic acid sequences derived from the genome of the same species or of a different species than the species of the target animal.
In some embodiments, transgenic animals in which all or a portion of one or more genes comprising SEQ ID NO: 1 is deleted may be constructed. In those cases where the gene corresponding to SEQ ID NO: 1 contains one or more introns, the entire gene- all exons, introns and the regulatoiy sequences- may be deleted. Alternatively, less than the entire gene may be deleted. For example, a single exon and/or intron may be deleted, so as to create an animal expressing a modified version of a protein of the invention.
The term "germ cell line transgenic animal" refers to a transgenic animal in which the genetic alteration or genetic infomiation was introduced into a germ line cell, thereby conferring the ability of the transgenic animal to transfer the genetic information to offspring. If such offspring in fact possess some or all of that alteration or genetic infomiation, then they too are transgenic animals. The alteration or genetic information may be foreign to the species of animal to wliich the recipient belongs, foreign only to the particular individual recipient, or may be genetic information already possessed by the recipient. In the last case, the altered or introduced gene may be expressed differently than the native gene. Transgenic animals can be produced by a variety of different methods including transfection, electroporation, microinjection, gene targeting in embryonic stem cells and recombinant viral and retroviral infection (see, e.g., U.S. Patent No. 4,736,866; U.S. Patent No.
5,602,307; Mullins et al, Hypertension 22:630-633, 1993; Brenin et al, Surg Oncol 6:99-110,
1997; Tuan, Recombinant Gene Expression Protocols, Methods in Molecular Biology, Humana Press, 1997).
A number of recombinant or transgenic mice have been produced, including those which express an activated oncogene sequence (U.S. Patent No. 4,736,866); express simian SV40 T-antigen (U.S. Patent No. 5,728,915); lack the expression of interferon regulatoiy factor 1 (IRF-1) (U.S. Patent No. 5,731,490); exhibit dopaminergic dysfunction (U.S. Patent No. 5J23J19); express at least one human gene which participates in blood pressure control (U.S. Patent No. 5,731,489); display greater similarity to the conditions existing in naturally occumng Alzheimer's disease (U.S. Patent No. 5,720,936); have a reduced capacity to mediate cellular adhesion (U.S. Patent No. 5,602,307); possess a bovine growth hom one gene (Clutter et al, Genetics 143:1753-1760, 1996); or, are capable of generating a fully human antibody response (McCarthy, Lancet 349:405, 1997).
While mice and rats remain the animals of choice for most transgenic experimentation, in some instances it is preferable or even necessary to use alternative animal species. Transgenic procedures have been successfully utilized in a variety of non-murine animals, including sheep, goats, pigs, dogs, cats, monkeys, chimpanzees, hamsters, rabbits, cows and guinea pigs (see, e.g. , Kim et al, Mol Reprod Dev 46:515-526, 1997; Houdebine, Reprod Nutr Dev 35:609-617, 1995; Petters, Reprod Fertil Dev 6:643-645, 1994; Schnieke et al, Science 278:2130-2133, 1997; and Amoa J Animal Science 15:518-585, 1997).
The method of introduction of nucleic acid fragments into recombination competent mammalian cells can be by any method which favors co-transfonuation of multiple nucleic acid molecules. Detailed procedures for producing transgenic animals are readily available to one skilled in the art, including the disclosures in U.S. Patent No. 5,489,743 and U.S. Patent No. 5,602,307. L. Diagnostic Methods
As the genes and proteins of the invention are differentially expressed in BPH tissue compared to nomial prostate tissue, the genes and proteins of the invention may be used to diagnose or monitor BPH, prostate function, or to track disease progression. One means of diagnosing BPH using the nucleic acid molecules or proteins of the invention involves obtaining prostate tissue from living subjects. Obtaining tissue samples from living sources is problematic for tissues such as prostate. However, due to the nature of the treatment paradigms for BPH, biopsy may be necessary. When possible, urine, blood or peripheral lymphocyte samples may be used as the tissue sample in the assay.
The use of molecular biological tools has become routine in forensic technology. For example, nucleic acid probes comprising all or at least part of the sequence of SEQ ID NO: 1 may be used to detennine the expression of a nucleic acid molecule in forensic/pathology specimens. Further, nucleic acid assays may be canied out by any means of conducting a transcriptional profiling analysis. In addition to nucleic acid analysis, forensic methods of the invention may target the proteins of the invention, particularly a protein comprising SEQ ID NO: 2, to detemiine up or down regulation of the genes (Shiverick et al, Biochim Biophys Ada 393, 124-133, 1975).
Methods of the invention may involve treatment of tissues with collagenases or other proteases to make the tissue amenable to cell lysis (Semenov et al, Biull Eksp Biol Med
104:113-116, 1987). Further, it is possible to obtain biopsy samples from different regions of the prostate for analysis.
Assays to detect nucleic acid or protein molecules of the invention may be in any available fomiat. Typical assays for nucleic acid molecules include hybridization or PCR- based fomiats. Typical assays for the detection of proteins, polypeptides or peptides of the invention include the use of antibody probes in any available fomiat such as in situ binding assays, etc. (see Harlow & Lane, Antibodies - A Laboratory Manual. Cold Spring Harbor Laboratoiy Press, Cold Spring Harbor, NY, 1988). hi prefened embodiments, assays are carried-out with appropriate controls. The above methods may also be used in other diagnostic protocols, including protocols and methods to detect disease states in other tissues or organs, for example the tissues in which gene expression is detected. Without further description, it is believed that one of ordinary skill in the art can, using the preceding description and the following illustrative examples, make and utilize the compounds of the present invention and practice the claimed methods. The following working examples therefore, specifically point out prefened embodiments of the present invention, and are not to be construed as limiting in any way the remainder of the disclosure.
EXAMPLES
Example 1 Identification of Differentially Expressed BPH mRNA
Human tissue was obtained from the transitional zone of the prostate in biopsy samples from normal individuals and from patients with BPH or prostate cancer. BPH was defined histologically in all samples. Nonnal tissue and asymptomatic BPH samples came from individuals who died of trauma, and did not report symptoms. Patients having BPH with symptoms were defined as those with a need for frequent urination; in these patients a radical prostatectomy had been performed. Prostate cancer patients provided age-matched tissue samples for symptomatic BPH patients, but were without symptoms and without cancer in the transitional zone under histological examination. BRF-55T cells, an immortalized human prostate cell line from an individual with BPH, were also screened. Microarray sample preparation was conducted with minor modifications, following the protocols set forth in the Affymetrix GeneChip® Expression Analysis Manual. Frozen tissue was ground to a powder using a Spex Certiprep 6800 Freezer Mill. Total RNA was extracted from ground tissue or cultured cells with Trizol® (GibcoBRL) utilizing the manufacturer's protocol. The total RNA yield for each tissue sample was 200-500 μg per 300 mg tissue weight. mRNA was isolated using the Oligotex mRNA Midi kit® (Qiagen) followed by ethanol precipitation. Double stranded cDNA was generated from mRNA using the Superscript Choice® system (GibcoBRL). First strand cDNA synthesis was primed with a T7- (dT24) ohgonucleotide. The cDNA was phenol-chlorofonn extracted and ethanol precipitated to a final concentration of 1 μg/μl. From 2 μg of cDNA, cRNA was synthesized using Ambion's T7 MegaScript in vitro Transcription Kit®.
To biotin label the cRNA, nucleotides Bio-11-CTP and Bio-16-UTP (Enzo Diagnostics) were added into the reaction. Following a 37°C incubation for six hours, impurities were removed from the labeled cRNA following the RNAeasy Mini kit protocol
(Qiagen). cRNA was fragmented (5' fragmentation buffer consisting of 200 mM Tris-acetate
(pH 8.1), 500 mM KOAc, 150 mM MgOAc) for thirty-five minutes at 94°C. Following the
Affymetrix protocol, 55 μg of fragmented cRNA was hybridized on the Human 42k 5-chip set and the HuGeneFL array for twenty-four hours at 60 rpm in a 45 °C hybridization oven. The chips were washed and stained with Streptavidin Phycoerythrin (S APE) (Molecular Probes) in
Affymetrix fluidics stations. To amplify staining, SAPE solution was added twice with an anti-streptavidin biotinylated antibody (Vector Laboratories) staining step in between.
Hybridization to the probe anays was detected by fluorometric scanning (Hewlett Packard Gene Array Scanner). Data was analyzed using Affymetrix GeneChip version 3.0 and Expression Data Mining Tool (EDMT) software (version 1.0).
Differential expression of genes between the BPH and normal prostate samples was determined using the Affymetrix GeneChip 42K human gene 5-chip set by the following criteria: (1) For each gene, Affymetrix GeneChip average difference values were determined by standard Affymetrix EDMT software algorithms, which also made "Absent" (=not detected), "Present" (=detected) or "Marginal" (=not clearly Absent or Present) calls for each GeneChip element; (2) all negative values (= Absent) were raised to a floor of +20 (positive 20) so that fold change calculations could be made where values were not already greater than or equal to +20; (3) median levels of expression were compared between the normal control group and the BPH with symptoms disease group to obtain greater than or equal 1.5 -fold up/down values; (4) The median value for the higher expressing group needed to be greater or equal to 200 average difference units in order to be considered for statistical significance; (5) Genes passing the criteria of #1-4 were analyzed for statistical significance using a two-tailed T test and deemed statistically significant if p < 0.05. The average difference values demonstrate that SEQ ID NO: 1 (clone no. F 13763) is upregulated (4-fold) in prostate tissue samples from patients with BPH with symptoms (p-0.0002) and in samples from the non-cancerous hyperplastic region of prostate cancer patients, compared to nomial prostate tissue samples. It is not upregulated in BPH without symptoms. SEQ ED NO: 1 is also expressed in BRF-55T cells, a prostate epithelial cell line immortalized from a patient with BPH.
Example 2 Cloning of a Full Length Human cDNA Conesponding to the Differentially Expressed mRNA species
The full length cDNA was obtained by the GeneTrapper® method (Life Technologies).
Briefly, a gene-specific oligo was designed based on the sequence of the clone no. F 13763 oligonucleotides in Example 1. The oligo was labeled with biotin and used to hybridize with single strand plasmid DNA (cDNA-recombinants) from a human MRC-5 cDNA library following the GeneTrapper® procedures. The hybridized cDNAs were separated by streptavidin-conjugated paramagnetic beads and eluted by washing. The eluted cDNA was converted to double strand plasmid DNA and used to transfomi E. coli cells (DH10B) and the longest cDNA was screened. After positive selection was confirmed by PCR using gene-specific primers, the cDNA clone was subjected to DNA sequencing.
The nucleotide sequences of the full-length human cDNA of clone no. F13763, conesponding to the differentially regulated niRNAs detected above, is set forth in SEQ ID NO: 1. The cDNA comprises 5132 base pairs, with an open reading frame at nucleotides 395- 3790 encoding a protein of 1132 amino acids. The amino acid sequence conesponding to the encoded protein is set forth in SEQ LD NO: 2.
A Blast of this sequence against GenBank databases indicated that the polypeptide of SEQ ID NO: 2 has unique sequence in the N-terminal 423 amino acids followed by a C- terminal region identical to a "hypothetical protein" that is encoded by a nucleotide sequence that matches SEQ ID NO: 1 only from nucleotide 1664-3954 (GenBank accession number AL137371). This hypothetical protein has no known function and does not match or disclose the open reading frame defined in SEQ ID NO: 1. The polypeptide of the invention also shares a low level of amino acid sequence similarity to human vascular cadherin-2 (GenBank accession number AB026893; 1184 amino acids) and mouse vascular cadherin-2 (GenBank accession number Y08715; 1180 amino acids).
Signal peptide and hydrophobicity analyses (see the hydrophobicity plot in Figure 5) of the amino acid sequence suggest that the novel polypeptide of SEQ ID NO: 2 has a signal peptide and a transmembrane region around amino acid 700. This is similar to a cadherin protein structure, having a signal peptide followed by a propeptide of about 130 residues, then an extracellular domain of about 600 residues (with motifs supporting a calcium-binding role), a transmembrane region and a cytoplasmic domain.
Analysis of the amino acid sequence of SEQ LD NO: 2 predicts the presence of 5 domains similar to the cadherin extracellular repeated domain signature starting at amino acids
122, 231, 339, 450 and 561, as well as an RGD (Arg-Gly-Asp) tripeptide sequence starting at amino acid 872. The cadherin-like motifs all fall between amino acids 120 and 580, i.e. within the putative extracellular region of the polypeptide of the invention. The RGD peptide is considered a crucial sequence for the interaction of fibronectin with its cell surface receptor (an integrin protein) and has also been found in a number of other proteins with cell adhesion roles, such as fibrinogen, vitronectin and some collagens. Eight potenital N-glycosylation sites are present starting at the asparagine at amino acid position 100, 266, 417, 556, 580, 638, 763 and
813. The sequence thus suggests a cadherin-like structure for the protein of the invention. Other motifs identified by primary peptide sequence analysis suggest potential cAMP- and cGMP-dependent protein kinase phosphoiylation sites at amino acids 728, 748 and 979; numerous casein kinase II (ck2) phosphoiylation sites (amino acids 157, 174, 185, 207, 214, 232, 26S, 465, 485, 487, 500, 562, 647, 866, 880, 888, 963, 987, 1019 and 1024); Protein Kinase C phosphoiylation sites at amino acids 60, 287, 293, 298, 444, 549, 845, 854, 866, 870 and 1079; and tyrosine kinase phosphoiylation motifs starting at amino acids 184, 406, 723 and 734. A possible glycosaminoglycan attachment site is present at amino acid 870; a region resembling a carboxypeptidase zinc-binding signature motif is found starting at amino acid 636 of the polypeptide of SEQ ID NO: 2, and numerous potential N-myristolation sites are also present, at amino acids 38, 189, 478, 508, 522, 590, 620, 704, 785, 788, 848 and 1071.
Example 3
Quantitative Analysis of Expression Levels
Figure 2 shows relative clone no. F13763 mRNA levels in various normal human tissues determined via Northern Blot. A probe based on SEQ ID NO: 1 (randomly labeled) was exposed to human mRNA blots (available from ClonTech) for 1 hour at 68°C in Clontech ExpressHyb hybridization buffer, following standard methodology as described by Sambrook et al. (supra). The highest expression levels in nomial tissue were observed in the brain and in the placenta, with a lower but significant level in the liver. Still lower levels of expression were observed in the lung, kidney, pancreas, and smooth muscle. The detection of more than one mRNA species per lane indicates the presence of splice variants. Such splice variants are within the scope of the present invention. Figure 3 shows the results of the semi-quantitative PCR analysis of expression levels of mRNA conesponding to SEQ ID NO: 1 in various nomial human tissue samples. Real time
PCR detection was accomplished by the use of the ABI PRISM 7700 Sequence Detection
System. The 7700 measures the fluorescence intensity of the sample each cycle and is able to detect the presence of specific amplicons within the PCR reaction. Each sample was assayed for the level of GAPDH and mRNA con-esponding to SEQ ID NO: 1. GAPDH detection was performed using Perkin Elmer part #402869 according to the manufacturer's directions.
Primers were designed from SEQ ID NO: 1 using Primer Express, a program developed by PE to efficiently find primers and probes for specific sequences. These primers (BPH-F13763 F39/R40S) were used in conjunction with SYBR green (Molecular Probes), a nonspecific double stranded DNA dye, to measure the expression level of mRNA conesponding to 391 bp of SEQ ID NO: 1, which was noimalized to the GAPDH level in each sample.
Expression of a 391 bp mRNA species was observed at high levels in the osteoblast (lane 12) and placental cells (lane 9) and at lower levels in heart (lane 1), brain (lane 2), lung (lane 4), liver (lane 5), fetal brain (lane 6), kidney (lane 7), and glomemli (lane 11) but not in leukocytes (lane 3) or spleen tissue (lane 8). The gene of the invention was also expressed in the prostate epithelial cell line BRF-55T (lane 10). Thus, based upon the tissue panels tested, expression is restricted in some nomial tissues.
Figure 4 shows the results of an electronic Northern assay, in which the expression level was measured across a panel of human nomial and BPH tissues using the Affymetrix human 42k 5-chip GeneChip set. The order of expression (high to low) across the nomial tissues in Figure 4 is: endometrium >myometriunι >breast =cervix =ovary >kidney =colon =small intestine =normal prostate >stomach =esophagus =lung =liver =spleen =muscles >thymus =brain. Because BPH with symptoms involves a chronic inflammatory response, the tissue distribution results indicate that the upregulation of SEQ ID NO: 1 is likely to be of diagnostic significance in inflammatory responses in tissues in which SEQ ID NO: 1 is found. Also, because SEQ ID NO: 1 is upregulated in non-cancerous BPH tissue adjacent to cancerous tissue, it is likely to be of diagnostic and prognostic significance in primary tumors and in metastatic tissues in which it is found (see Figures 2, 3, and 4). SEQ ID NO: 1 is, therefore, a potential target for therapeutic intervention in both inflammatory and hyperplastic diseases.
Because SEQ ID NO: 1 is detected in the epithelial prostate cell line BRF55-T, by both semi-quantitative PCR (Figure 3) and by Affymetrix GeneChip analysis (Figure 1), it could be used as a cell line for drug screening.
Although the present invention has been described in detail with reference to examples above, it is understood that various modifications can be made without departing from the spirit of the invention. Accordingly, the invention is limited only by the following claims. All cited patents, patent applications and publications refened to in this application are herein incorporated by reference in their entirety.

Claims

What Is Claimed:
1. An isolated nucleic acid molecule selected from the group consisting of: (a) an isolated nucleic acid molecule that encodes the amino acid sequence of SEQ ID NO: 2; (b) an isolated nucleic acid molecule that encodes a fragment of at least 6 contiguous amino acids of SEQ ID NO: 2; (c) an isolated nucleic acid molecule which hybridizes to the complement of a nucleic acid molecule comprising SEQ ID NO: 1; (d) an isolated nucleic acid molecule which hybridizes to the complement of a nucleic acid molecule that encodes the amino acid sequence of SEQ ID NO: 2; and (e) an isolated nucleic acid molecule that encodes a protein that exhibits at least about 50% amino acid sequence identity to SEQ ID NO: 2.
2. The isolated nucleic acid molecule of claim 1, wherein the nucleic acid molecule comprises nucleotides 395-3790 of SEQ ID NO: 1.
3. The isolated nucleic acid molecule of claim 1, wherein the nucleic acid molecule consists of nucleotides 395-3790 of SEQ ID NO: 1.
4. The isolated nucleic acid molecule of claim 1, wherein the nucleic acid molecule comprises nucleotides 395-3793 of SEQ ID NO: 1.
5. The isolated nucleic acid molecule of any one of claims 1-4, wherein said nucleic acid molecule is operably linked to one or more expression control elements.
6. A vector comprising an isolated nucleic acid molecule of any one of claims 1-4.
7. A host cell transfoπned to contain the nucleic acid molecule of any one of claims 1- 4.
8. A host cell comprising the vector of claim 6.
9. The host cell of claim 8, wherein said host is selected from the group consisting of prokaryotic host cells and eukaryotic host cells.
10. A method for producing a polypeptide comprising culturing a host cell transformed with the nucleic acid molecule of any one of claims 1-4 under conditions in which the protein encoded by said nucleic acid molecule is expressed.
11. The method of claim 10, wherein said host cell is selected from the group consisting of prokaryotic host cells and eukaryotic host cells.
12. An isolated polypeptide produced by the method of claim 10.
13. An isolated polypeptide or protein selected from the group consisting of an isolated polypeptide comprising the amino acid sequence of SEQ ID NO: 2, an isolated polypeptide comprising a fragment of at least 6 contiguous amino acids of SEQ ID NO: 2, an isolated polypeptide comprising conseivative amino acid substitutions of SEQ ID NO: 2, an isolated polypeptide comprising naturally occurring amino acid sequence variants of SEQ ID NO: 2, and an isolated polypeptide exhibiting at least about 50% amino acid sequence identity with SEQ ID NO: 2.
14. An isolated antibody that binds to a polypeptide of either claim 12 or 13.
15. An antibody of claim 14 wherein said antibody is a monoclonal or a polyclonal antibody.
16. A method of identifying an agent which modulates the expression of a nucleic acid encoding a protein of claim 13, comprising: exposing cells which express the nucleic acid to the agent; and detem ining whether the agent modulates expression of said nucleic acid, thereby identifying an agent which modulates the expression of a nucleic acid encoding the protein.
17. A method of identifying an agent wliich modulates the level of or at least one activity of a protein of claim 13, comprising: exposing cells which express the protein to the agent; detemiining whether the agent modulates the level of or at least one activity of said protein, thereby identifying an agent which modulates the level of or at least one activity of the protein.
18. The method of claim 17, wherein the agent modulates one activity of the protein.
19. A method of identifying binding partners for a protein of claim 13, comprising: exposing said protein to a potential binding partner; and determining if the potential binding partner binds to said protein, thereby identifying binding partners for the protein.
20. A method of modulating the expression of a nucleic acid encoding a protein of claim 13, comprising: administering an effective amount of an agent which modulates the expression of a nucleic acid encoding the protein.
21. A method of modulating at least one activity of a protein of claim 13, comprising: administering an effective amount of an agent which modulates at least one activity of the protein.
22. A non-human transgenic animal modified to contain the nucleic acid molecule of any of claims 1-4.
23. A non-human transgenic animal modified to contain the nucleic acid molecule of any of claims 1 -4, wherein all or a portion of SEQ ID NO: 1 has been knocked out.
24. A method of diagnosing a disease state in a subject, comprising determining the level of expression of a nucleic acid molecule or protein of any one of claims 1-4 or 13.
25. The method of claim 24, wherein the disease state is benign prostatic hyperplasia.
26. The method of claim 24, wherein the disease state is benign prostatic hyperplasia with symptoms.
27. The method of claim 24, wherein the disease state is benign prostatic hyperplasia with prostate cancer.
PCT/US2001/047949 2000-12-13 2001-12-13 Expression of a cadherin-like protein in benign prostatic hyperplasia WO2002048173A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2002229004A AU2002229004A1 (en) 2000-12-13 2001-12-13 Expression of a cadherin-like protein in benign prostatic hyperplasia

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US25492800P 2000-12-13 2000-12-13
US60/254,928 2000-12-13

Publications (2)

Publication Number Publication Date
WO2002048173A2 true WO2002048173A2 (en) 2002-06-20
WO2002048173A3 WO2002048173A3 (en) 2003-10-23

Family

ID=22966135

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/047949 WO2002048173A2 (en) 2000-12-13 2001-12-13 Expression of a cadherin-like protein in benign prostatic hyperplasia

Country Status (2)

Country Link
AU (1) AU2002229004A1 (en)
WO (1) WO2002048173A2 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001051514A1 (en) * 2000-01-07 2001-07-19 Millennium Pharmaceuticals, Inc. Novel genes encoding proteins having prognostic, diagnostic, preventive, therapeutic, and other uses

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001051514A1 (en) * 2000-01-07 2001-07-19 Millennium Pharmaceuticals, Inc. Novel genes encoding proteins having prognostic, diagnostic, preventive, therapeutic, and other uses

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
1995, WILEY & SONS INC. article AUSUBEL F.M. ET AL.: 'Short protocols in molecular biology', pages 15.3-5 - 16.58-62, XP002967111 3rd edition *
DATABASE GENBANK [Online] 12 September 2001 XP002967106 Database accession no. (AAD09283) *
DATABASE GENBANK [Online] 12 September 2001 XP002967107 Database accession no. (AAE05293) *
DATABASE GENBANK [Online] 22 February 2001 NAGASE T. ET AL.: 'Homo sapians mRNA for KIAA1562 protein', XP002967108 Database accession no. (AB046782) *
DATABASE GENBANK [Online] 24 February 2000 XP002967109 Database accession no. (AAA16643) *
DATABASE GENBANK [Online] 24 February 2000 XP002967110 Database accession no. (AAY94923) *

Also Published As

Publication number Publication date
AU2002229004A1 (en) 2002-06-24
WO2002048173A3 (en) 2003-10-23

Similar Documents

Publication Publication Date Title
US20130059751A1 (en) Gene family (lbfl313) associated with pancreatic cancer
JP2003525634A (en) Protein named FCTRX and nucleic acid encoding the same
US20050037454A1 (en) Gene associated with bone disorders
US7718787B2 (en) Gene families associated with cancers
WO2001098456A2 (en) IDENTIFICATION OF cDNAs ASSOCIATED WITH BENIGN PROSTATIC HYPERPLASIA
WO2002048173A2 (en) Expression of a cadherin-like protein in benign prostatic hyperplasia
WO2002010338A2 (en) Expression of gage/page-like protein in benign prostatic hyperplasia
WO2003008561A2 (en) Genes associated with benign prostatic hyperplasia
US20060121471A1 (en) Gene families associated with stomach cancer
WO2002046362A2 (en) Gene associated with benign prostatic hyperplasia in humans
WO2002077162A2 (en) Gene (t23490) associated with benign prostatic hyperplasia
WO2003025135A2 (en) Genes associated with malignant neoplasms
US20060242721A1 (en) Gene families associated with liver cancer
WO2002036826A2 (en) Del-1 and benign prostatic hyperplasia
WO2001005803A9 (en) NOVEL cDNAs ASSOCIATED WITH RENAL DISEASE
US20030079239A1 (en) Gene Associated with bone disorders
WO2002012262A1 (en) IDENTIFICATION OF cDNAs ASSOCIATED WITH BENIGN PROSTATIC HYPERPLASIA
US20050147969A1 (en) Genes associated with mast cell activation
US20030166881A1 (en) Novel genes associated with allergic hypersensitivity and mast cell activation
WO2001041805A1 (en) Novel cdna 22360 associated with renal disease and other disease states
WO2000078788A1 (en) NOVEL cDNA ASSOCIATED WITH RENAL DISEASE
WO2001005804A1 (en) Novel genes associated with renal disease
WO2000052026A1 (en) IDENTIFICATION OF A cDNA ASSOCIATED WITH COMPENSATORY HYPERTROPHY IN RENAL TISSUE
WO2002012439A2 (en) Genes associated with renal disease
WO2003025152A2 (en) IDENTIFICATION OF A cDNA ASSOCIATED WITH ISCHEMIA IN HUMAN HEART TISSUE

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

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

AL Designated countries for regional patents

Kind code of ref document: A2

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
NENP Non-entry into the national phase in:

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP