CA2220988A1 - Fibroblast growth factor 13 - Google Patents

Fibroblast growth factor 13 Download PDF

Info

Publication number
CA2220988A1
CA2220988A1 CA002220988A CA2220988A CA2220988A1 CA 2220988 A1 CA2220988 A1 CA 2220988A1 CA 002220988 A CA002220988 A CA 002220988A CA 2220988 A CA2220988 A CA 2220988A CA 2220988 A1 CA2220988 A1 CA 2220988A1
Authority
CA
Canada
Prior art keywords
polypeptide
cells
polynucleotide
dna
polypeptides
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
CA002220988A
Other languages
French (fr)
Inventor
John M. Greene
Joachim R. Gruber
Craig A. Rosen
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Human Genome Sciences Inc
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to CA002220988A priority Critical patent/CA2220988A1/en
Publication of CA2220988A1 publication Critical patent/CA2220988A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • 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
    • C07K14/475Growth factors; Growth regulators
    • C07K14/50Fibroblast growth factor [FGF]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Chemistry (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Epidemiology (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biophysics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Microbiology (AREA)
  • Analytical Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Biomedical Technology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Engineering & Computer Science (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Toxicology (AREA)
  • Cell Biology (AREA)
  • Food Science & Technology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Disclosed is a human Fibroblast growth factor-13 polypeptide and DNA (RNA) encoding such polypeptide. Also provided is a procedure for producing such polypeptide by recombinant techniques. Also disclosed are methods for utilizing such polypeptide for promoting wound healing for example as a result of burns and ulcers, to prevent neuronal damage associated with stroke and due to neuronal disorders and promote neuronal growth, and to prevent skin aging and hair loss, to stimulate angiogenesis, mesodermal induction in early embryos and limb regeneration. Antagonists against such polypeptides and their use as a therapeutic to prevent abnormal cellular proliferation, hyper-vascular diseases and epithelial lens cell proliferation are also disclosed.
Diagnostic methods for detecting mutations in the coding sequence and alterations in the concentration of the polypeptides in a sample derived from a host are also disclosed.

Description

W O 96~9508 PCT/U'~5,a7l08 FTR~QRT-~CT GROW~ F~CTOR 13 This invention relates to newly identified polynucleotides, polypeptides encoded by such polynucleotides, the use of such polynucleotides and polypeptides, as well as the production of such polynucleotides and polypeptides. More particularly, the polypeptide of the present invention have been putatively identified as fibroblast growth factor/heparin binding growth factor, hereinafter referred to as "FGF-13". The invention also relates to inhibiting the action of such polypeptides.
Fibroblast growth factors are a family of proteins characteristic of bin~;ng to heparin and are, therefore, also called heparin binding growth factors (B GF). Expression of different m~mh~rs of these proteins are found in various tissues and are under particular temporal and spatial control. These proteins are potent mitogens for a variety of cells of mesodermal, ectodermal, and endodermal origin, including fibroblasts, corneal and vascular endothelial cells, granulocytes, adrenal cortical cells, ~hon~rocytes, myoblasts, vascular smooth muscle cells, lens epithelial cells, mel~nocytes~ keratinocytes, oligodendrocytes, astrocytes, osteoblasts, and hem.~topoietic cells;
Each memher has functions overlapping with others and also has its unique spectrum of functions. In addition to W O 96~9S08 PCT/U~9SJ~7108 the ability to stimulate proliferation of vascular endothelial cells, both FGF-1 and 2 are chemotactic for endothelial cells and FGF-2 has been shown to enable endothelial cells to penetrate the basement membrane.
Consistent with these properties, both FGF-1 and 2 have the capacity to stimulate angiogenesis. Another important feature of these growth factors is their ability to promote wound healing. Many other m~mhP~s of the FGF family share similar activities with FGF-1 and 2 such as promoting angiogenesis and wound healing. Several m~mh~rs of the FGF
family have been shown to induce mesoderm formation and to modulate differentiation of neuronal cells, adipocytes and skeletal muscle cells.
Other than these biological activities in normal tissues, FGF proteins have been implicated in promoting tumorigenesis in carc;no~ and sarcomas by promoting tumor vascularization and as transforming proteins when their expression is deregulated.
The FGF family presently consists of eight structurally-related polypeptides: basic FGF, acidic FGF, int 2, hst 1/k-FGF, FGF-5, FGF-6, keratinocyte growth factor, AIGF (FGF-8) and recently a glia-activating factor has been shown to be a novel heparin-h;n~;ng growth factor which was purified from the culture supernatant of a human glioma cell line (Miyamoto, M. et al., Mol. and Cell. Biol., 13(7):4251-4259 (1993). The genes for each have been cloned and sequenced.
Two of the members, FGF-1 and FGF-2, have been characterized under many names, but most often as acidic and basic fibroblast growth factor, respectively. The normal gene products influence the general proli~eration capacity of the majority of mesoderm and neuroectoderm-derived cells. They are capable of inducing angiogenesis in vivo and may play important roles in early development (Burgess, W.H. and Maciag, T., Annu. Rev. Biochem., 58:575-606, (1989)).

W O 96~9508 PCTlU~Slv7108 Many of the above-identified members of the FGF family also bind to the same receptors and elicit a second message through binding to these receptors.
A eukaryotic expression vector encoding a secreted form o~ FGF-1 has been introduced by gene transfer into porcine arteries. This model defines gene function in the arterial ~ wall in vivo. FGF-1 expression induced intimal thickening in porcine arteries 21 days after gene transfer (Nabel, E.G., et al., Nature, 362:844-6 (1993)). It has further been ~mon~trated that basic fibroblast growth factor may regulate glioma growth and progression independent of its role in tumor angiogenesis and that basic fibroblast growth factor release or secretion may be required ~or these actions (Morrison, R.S., et al., J. Neurosci. Res., 34:502-9 (1993)).
Fibroblast growth factors, such as basic FGF, have further been implicated in the growth of Kaposi~s sarcoma cells in vi tro (Huang, Y.Q., et al., J. Clin. Invest., 91:1191-7 (1993)). Also, the cDNA sequence encoding hllm~n basic fibroblast growth factor has been cloned downstream of a transcription promoter recognized by the bacteriophage T7 RNA polymerase. Basic fibroblast growth factors so obtained have been shown to have biological activity indistinguishable from human placental fibroblast growth factor in mitogenicity, synthesis of pl ~mi nogen activator and angiogenesis assays (Squires, C.H., et al., J. Biol. Chem., 263:16297-302 (1988)).
U.S. Patent No. 5,155,214 discloses substantially pure m~mm~l ian basic fibroblast growth factors and their production. The amino acid sequences of bovine and hllm~n basic fibroblast growth factor are disclosed, as well as the DNA sequence encoding the polypeptide of the bovine species.
Newly discovered FGF-9 has around 30% sequence similarity to other members of the FGF ~amily. Two cysteine residues and other consensus sequences in family members were also well conserved in the FGF-9 sequence. FGF-9 was found W 0 96~9508 CA 02220988 1997-11-12 PcT/u'35~llo8 to have no typical signal sequence in its N termlnus like those in acidic and basic FGF. However, FGF-g was found to be secreted from cells after synthesis despite its lack of a typical signal sequence FGF (Miyamoto, M. et al., Mol. and Cell. Biol., 13(7):4251-4259 (1g93). Further, FGF-9 was found to stimulate the cell growth o~ oligodendrocyte type 2 astrocyte progenitor cells, BALB/c3T3, and PC-12 cells but not that of human umbilical vein endothelial cells (Naruo, K., et al., J. Biol. Chem., 268:2857-2864 (1993).
Basic FGF and acidic FGF are potent modulators of cell proliferation, cell motility, differentiation, and survival and act on cell types from ectoderm, mesoderm and endoderm.
These two FGFs, along with KGF and AIGF, were identified by protein purification. However, the other four members were isolated as oncogenes., expression of which was restricted to embryogenesis and certian types of cancers. FGF-9 was ~monqtrated to be a mitogen against glial cells. Members of the FGF family are reported to have oncogenic potency. FGF-9 has shown transforming potency when transformed into BALB/c3T3 cells (Miyamoto, M., et al., Mol. Cell. Biol., 13(7):4251-4259 (1993).
Androgen induced growth factor (AIGF), also known as FGF-8, was purified from a conditioned medium of mouse m~mm~ry carcinoma cells (SC-3) simulated with testosterone.
AIGF is a distinctive FGF-like growth factor, having a putative signal peptide and sharing 30-40~ homology with known members of the FGF family. ~mm~lian cells transformed with AIGF shows a remarkable stimulatory effect on the growth of SC-3 cells in the absence of androgen. Therefore, AIGF
mediates androgen-induced growth of SC-3 cells, and perhaps other cells, since it is secreted by the tumor cells themselves.
The polypeptide of the present invention has ~een putatively identified as a member of the FGF family as a W O 96~9508 PCTIU',~1~7108 result of amino acid sequence homology with other m~mh~s of the FGF ~amily.
In accordance with one aspect of the present invention, there are provided novel mature polypeptides as well as biologically active and diagnostically or therapeutically useful fragments, analog~ and derivatives thereof. The polypeptides of the present invention are of hllm~n origin.
In accordance with another aspect of the present invention, there are provided isolated nucleic acid molecules encoding the polypeptides of the present invention, including mRNAs, DNAs, cDNAs, genomic DNA, as well as antisense analogs thereof and biologically active and diagnostically or therapeutically useful fragments thereof.
In accordance with still another aspect of the present invention, there are provided processes for proAllc~ng such polypeptides by reco~h;n~nt techniques through the use of = recombinant vectors, such as cloning and expression plasmids useful as reagents in the recombinant production of the polypeptides of the present invention, as well as recombinant prokaryotic and/or eukaryotic host cells comprising a nucleic acid sequence encoding a polypeptide of the present invention.
In accordance with a further aspect of the present invention, there is provided a process for utilizing such polypeptides, or polynucleotides encoding such polypeptides, for screening for agonists and antagonists thereto and for therapeutic purposes, for example, promoting wound healing for example as a result of burns and ulcers, to prevent neuronal damage associated with stroke and due to neuronal disorders and promote neuronal growth, and to prevent skin aging and hair loss, to stimulate angiogenesis, mesodermal induction in early embryos and limb regeneration.
In accordance with yet a further aspect of the present invention, there are provided antibodies against such polypeptides.

W O 96~9508 PCTAUS95/07108 In accordance with yet another aspect o~ the present invention, there are provided antagonists against such polypeptides and processes for their use to ; nh; h; t the action of such polypeptides, for example, in the treatment of cellular transformation, ~or example, tumors, to reduce scarring and treat hyper-vascular diseases.
In accordance with another aspect of the present invention, there are provided nucleic acid probes comprising nucleic acid molecules of sufficient length to specifically hybridize to a polynucleotide encoding a polypeptide of the present invention In accordance with yet another aspect of the present invention, there are provided diagnostic assays for detecting diseases or susceptibility to diseases related to mutations in a nucleic acid sequence of the present invention and ~or detecting over-expression of the polypeptides encoded by such sequences.
In accordance with another aspect of the present invention, there is provided a process for utilizing such polypeptides, or polynucleotides encoding such polypeptides, for in vi tro purposes related to scientific research, synthesis of DNA and manufacture of DNA vectors.
These and other aspects of the present invention should be apparent to those skilled in the art ~rom the teachings herein.
The following drawings are meant only as illustrations of specific embo~;mPnts of the present invention and are not meant as limitations in any m~nn~r.
Figure 1 depicts the cDNA sequence and corresponding deduced amino acid sequence of FGF-13. The initial 21 amino acid residues represent a putative leader sequence.

W O 96~9508 PCTIU',S~1108 In accordance with one a~pect of the pre~ent invention, there are provided isolated nucleic acids molecules (polynucleotides) which encode for the mature polypeptide having the deduced amino acid sequence of Figure 1 (SEQ ID
NOS:2) or for the mature polypeptide encoded by the cDNA of the clone deposited as ATCC Deposit No. 97147 on May 12, 1995.
The polynucleotide encoding FGF-13 of this invention was discovered in a cDNA library derived from hllm~n ovarian cancer tissue. The FGF-13 polypeptide is structurally related to all members of the fibroblast growth factor family and contains an open reading frame encoding a polypeptide of 212 amino acids of which the first 21 amino acids represent a putative leader sequence such that the mature polypeptide comprises 191 amino acids. Among the top matches are: 1) 69 ~ identity and 81 % similarity to mouse AIGF over a stretch of 185 amino acids; 2) 30 ~ identity and 56 % similarity with FGF-4 from chicken in a region of 82 amino acids; 3) 41 identity and 64 % similarity with human KGF over a stretch of 78 amino acids.
The FGF/B GF family signature, GXLX(S,T,A,G)X6(D,E)CXFXE
is conserved in the polypeptide of the present invention, (X
means any amino acid residue; (D,E) means either D or E
= residue; X6 means any 6 amino acid residues).
The polynucleotide of the present invention may be in the form of RNA or in the form of DNA, which DNA includes cDNA, genomic DNA, and synthetic DNA. The DNA may be double-stranded or single-str~n~. The coding sequence which encodes the mature polypeptide may be identical to the coding sequence shown in Figure 1 (SEQ ID NOS:1) or that of the deposited clone or may be a different coding sequence, as a result of the re~lln~ncy or degeneracy of the genetic code, encodes the same, mature polypeptide as the DNA of Figure 1, (SEQ ID NOS:l) or the deposited CDNA.

CA 02220988 1997-ll-12 W O 96~9508 PCTAUS95/07108 The polynucleotides which encodes for the mature polypeptide of Figure 1 (SEQ ID NOS:2) or for the mature polypeptides encoded by the deposited cDNA(s) may include:
only the coding sequence for the mature polypeptide; the coding sequence for the mature polypeptide and additional coding sequence such as a leader or secretory sequence or a proprotein sequencei the coding sequence for the mature polypeptide (and optionally additional coding sequence) and non-coding sequence, such as introns or non-coding sequence 5~ and/or 3~ of the coding sequence for the mature polypeptide.
Thus, the term "polynucleotide encoding a polypeptide"
encompasses a polynucleotide which includes only coding sequence for the polypeptide as well as a polynucleotide which includes additional coding and/or non-coding sequence.
The present invention further relates to variants of the hereinabove described polynucleotides which encode for fragments, analogs and derivatives of the polypeptides having the deduced amino acid sequence of Figure 1 (SEQ ID NOS:2) or the polypeptides encoded by the cDNA(s) of the deposited clone(s). The variants of the polynucleotide may be a naturally occurring allelic variant of the polynucleotide or a non-naturally occurring variant of the polynucleotide.
Thus, the present invention includes polynucleotides encoding the same mature polypeptide as shown in Figure 1 (SEQ ID NOS:2) or the same mature polypeptides encoded by the cDNA(s) of the deposited clone(s) as well as variants of such polynucleotides which variants encode for a fragment, derivative or analog of the polypeptide of Figure 1 (SEQ ID
NOS:2) or the polypeptides encoded by the cDNA(s) of the deposited clone(s). Such nucleotide variants include deletion variants, substitution variants and addition or insertion variants.
As hereinabove indicated, the polynucleotide may have a coding sequence which is a naturally occurring allelic -W O 96~9508 PCT/U',Sr~710~

variant of the coding sequence shown in Figure 1 (SEQ ID
NOS:1) or of the coding sequence of the deposited clone(s).
As known in the art, an allelic variant is an alternate form of a polynucleotide sequence which may have a substitution, deletion or addition of one or more nucleotides, which does not substantially alter the function of the encoded polypeptides.
The present invention also includes polynu~leotides, wherein the coding sequence for the mature polypeptides may be fused in the same reading frame to a polynucleotide sequence which aids in expression and secretion of a polypeptide from a host cell, for example, a leader sequence which functions as a secretory sequence for controlling transport of a polypeptide from the cell. The polypeptide having a leader sequence is a preprotein and may have the leader sequence cleaved by the host cell to fonm the mature form of the polypeptide. The polynucleotides may also encode for a proprotein which is the mature protein plus additional 5~ amino acid residues. A mature protein having a prosequence is a proprotein and is an inactive form of the protein. Once the prosequence is cleaved an active mature protein rem~ n~ .
Thus, for example, the polynucleotides of the present invention may encode for a mature protein, or for a protein having a prosequence or for a protein having both a prosequence and a presequence (leader sequence).
The polynucleotides of the present invention may also have the coding sequence fused in frame to a marker sequence which allows for purification of the polypeptide of the present invention. The marker sequence may be a hexa-histidine tag supplied by a pQE-9 vector to provide for purification of the mature polypeptide fused to the marker in the case of a bacterial host, or, for example, the marker sequence may be a hemagglutinin (HA) tag when a m~mm~ 1 ian host, e.g. COS-7 cells, is used. The HA tag corresponds to W O 96~9508 PCT~',5~7108 an epitope derived from the influenza hemagglutinin protein (Wilson, I., et al., Cell, 37:767 (1984)).
The term "gene" means the segment of DNA involved in producing a polypeptide chain; it includes regions preceding and following the coding region (leader and trailer) as well as intervening sequences (introns) between indlvidual coding segments (exons).
Fragments of the full length FGF-13 gene may be used as a hybridization probe for a cDNA library to isolate the full length gene and to isolate other genes which have a high sequence similarity to the gene or similar biological activity. Probes of this type preferably have at least 30 bases and may contain, for example, 50 or more bases. The probe may also be used to identify a cDNA clone corresponding to a full length transcript and a genomic clone or clones that contain the complete FGF-13 gene including regulatory and promotor regions, exons, and introns. An example of a screen comprises isolating the coding region of the FGF-13 gene by using the known DNA sequence to synthesize an oligonucleotide probe. Labeled oligonucleotides having a sequence complementary to that of the gene of the present invention are used to screen a library of human cDNA, genomic DNA or mRNA to determine which members of the library the probe hybridizes to.
The present invention further relates to polynucleotides which hybridize to the hereinabove-described sequences if there is at least 70~, preferably at least 90~, and more preferably at least 95% identity between the sequences. The present invention particularly relates to polynucleotides which hybridize under stringent conditions to the her~in~hove-described polynucleotides. As herein used, the term ~stringent conditions" means hybridization will occur only i~ there is at 'east 95% and preferably at least 97% identity between the sequences. The polynucleotides which hybridize to the hereinabove described polynucleotides W O 96~9508 PCTJ~v7108 in a preferred embodiment encode polypeptides which either retain substantially the same biological function or activity as the mature polypeptide encoded by the cDNAs of Figure l (SEQ ID N0:1) or the deposited cDNA(s).
Alternatively, the polynucleotide may have at least 20 bases, preferably 30 bases, and more pre~erably at least 50 bases which hybridize to a polynucleotide of the present invention and which has an identity thereto, as hereinabove described, and which may or may not retain activity. For example, such polynucleotides may be employed as probes for the polynucleotide of SEQ ID NO:1, for example, for recovery of the polynucleotide or as a diagnostic probe or as a PCR
primer.
Thus, the present invention is directed to polynucleotides having at least a 70~ identity, preferably at least 90% and more preferably at least a 95~ identity to a polynuclectide which encodes the polypeptide of SEQ ID NO:2 as well as fragments thereof, which fragments have at least 30 bases and preferably at least 50 bases and to polypeptides encoded by such polynucleotides.
The deposit(s) referred to herein will be ~-;nt~n~d under the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for thP purposes of Patent Procedure. These deposits are provided merely as a convenience and are not an admission that a deposit is required under 35 U.S.C. 112. The sequence of the polynucleotides contained in the deposited materials, as well as the amino acid sequence of the polypeptides encoded thereby, are incorporated herein by reference and are controlling in the event of any conflict with the description of sequences herein. A license may be required to make, use or sell the deposited materials, and no such license is hereby granted.
The present invention further relates to an FGF
polypeptide which has the deduced amino acid sequence of W O 96~9508 PcT/u~J~ o8 Figure 1 (SEQ ID NOS:2) or which has the amino acid sequence encoded by the deposited cDNA(s), as well as fragments, analogs and derivatives of such polypeptides.
The terms "fragment," "derivative'l and ''analogll when referring to the polypeptide of Figure 1 (SEQ ID NOS:2) or those encoded by the depo~ited cDNA(s), means polypeptides which retains essentially the same biological function or activity as such polypeptides. Thus, an analog includes a proprotein which can be activated by cleavage of the proprotein portion to produce an active mature polypeptide.
The polypeptides of the present invention may be recombinant polypeptides, natural polypeptides or synthetic polypeptides, preferably recombinant polypeptides.
The fragment, derivative or analog o~ the polypeptide of Figure 1 (SEQ ID NOS:2) or that encoded by the deposited cDNA(s) may be (i) one in which one or more of the amino acid residues are substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code, or (ii) one in which one or more of the amino acid residues includes a substituent group, or (iii) one in which the mature polypeptide is fused with another compound, such as a compound to increase the half-life o~ the polypeptide (for example, polyethylene glycol), or (iv) one in which the additional amino acids are fused to the mature polypeptide, such as a leader or secretory sequence or a sequence which is employed for purification o~ the mature polypeptide or a proprotein sequence. Such fragments, derivatives and analogs are deemed to be within the scope of those skilled in the art from the teachings herein.
The polypeptides and polynucleotides of the present invention are preferably provided in an isolated form, and preferably are purified to homogeneity.

-W O 96~9508 PCTIU',~1~7108 The term ~isolated" means that the material is removed from its original enviLu~l-"-L-t (e.g., the natural environment if it is naturally occurring). For example, a naturally-occurring polynucleotide or polypeptide present in a living nimAl is not isolated, but the same po-ynucleotide or DNA or polypeptide, separated from some or all of the coexisting materials in the natural system, is isolated. Such polynucleotide could be part of a vector and/or such polynucleotide or polypeptide could be part of a composition, and still be isolated in that such vector or composition is not part of its natural enviLolll-lent.
The polypeptides of the present invention include t.=
= polypeptide of SEQ ID NO:2 (in particular the mature polypeptide) as well as polypeptides which have at least 70~
similarity (preferably at least a 70~ identity) to the polypeptide of SEQ ID NO:2 and more preferably at least a 90%
similarity (more preferably at least a 90~ identity) to the polypeptide OL SEQ ID NO:2 and still more preferably at least a 95% similarity (still more preferably at least a 95~
identity) to the polypeptide of S~Q ID NO:2 and also include portions of such polypeptides with such portion of the polypeptide generally cont~ining at least 30 amino acids and more preferably at least 50 amino acids.
As known in the art ~simi 1 Arity~ between two polypeptides is determined by comrAring the amino acid sequence and its conserved amino acid substitutes of one polypeptide to the sequence of a second polypeptide.
Fragments or portions of the polypeptides of the present invention may be employed for producing the _orresponding full-length polypeptide by peptide synthesis; therefore, the fragments may be employed as intermediates for producing the full-length polypeptides. Fragments or portions of the polynucleotides of the present invention may be used to synthesize full-length polynucleotides of the present invention.

W O 96~9508 PCTAUS95/07108 The present invention also relates to vectors which include polynucleotides of the present invention, host cells which are genetically engineered with vectors of the invention and the production of polypeptides of the invention by recomh;nAnt techniques.
Host cells may be genetically engineered (transduced or transformed or transfected) with the vectors of this invention which may be, for example, a cloning vector or an expression vector. The vector may be, for example, in the form of a plasmid, a viral particle, a phage, etc. The engineered host cells can be cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transformants or amplifying the FGF
genes. The culture conditions, such as temperature, pH and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.
The polynucleotide of the present invention may be employed for producing a polypeptide by recombinant techniques. Thus, for example, the polynucleotide sequence may be included in any one of a variety of expression vehicles, in particular vectors or plasmids for expressing a polypeptide. Such vectors include chromosomal, nonchromosomal and synthetic DNA sequences, e.g., derivatives of SV40; bacterial plasmids; phage DNA; yeast plasmids; vectors derived from combinations of plasmids and phage DNA, viral DNA such as vaccinia, adenovirus, fowl pox virus, and pseudorabies. However, any other vector or plasmid may be used as long as they are replicable and viable in the host.
The appropriate DNA sequence may be inserted into the vector by a variety of procedures. In general, the DNA
sequence is inserted into an appropriate restriction endonuclease sites by procedures known in the art. Such W096~9508 PCT~U',S~1108 procedures and others are deemed to be within the scope of those skilled in the art.
The DNA sequence in the expression vector is operatively linked to an a~u~riate expression control ~equence(s) (promoter) to direct mRNA synthesis. As representative examples of such promoters, there may be mentioned: LTR or SV40 promoter, the E. coli. lac or trp, the phage ~ Amh~lA PL
promoter and other promoters known to control expression of genes in prokaryotic or eukaryotic cells or their viruses.
The expression vector also contains a ribosome binding site for translation initiation and a transcription terminator.
The vector may also include appropriate sequences for amplifying expression.
In addition, the expression vectors preferably contain a gene to provide a phenotypic trait for selection o~
transformed host cells such as dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, or such as tetracycline or ampicillin resistance in E. coli.
The vector cont~ning the appropriate DNA sequence as herein above described, as well as an appropriate promoter or control sequence, may be employed to transform an appropriate host to permit the host to express the protein. As representative examples of appropriate hosts, there may be mentioned: bacterial cells, such as E. coli, SA-monella tY~himurium, StreptomYces; fungal cells, such as yeast;
insect cells, such as Drosophila S2 and Spodo~tera Sf9;
An,mAl cells such as CH0, COS or Bowes mel~nomA;
adenoviruses; plant cells, etc. The selection of an appropriate host is deemed to be within the scope of those skilled in the art from the teachings herein.
More particularly, the present invention also includes recombinant constructs comprising one or more of the sequences as broadly described above. The constructs comprise a vector, such as a plasmid or viral vector, into which a sequence of the invention has been inserted, in a W O 96~9SO8 PCT/U',S~7108 forward or reverse orientation. In a preferred aspect of this embodiment, the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the se~uence. Large numbers of suitable vectors and promoters are known to those of skill in the art, and are co~m~rcially available. The following vectors are provided by way of example. Bacterial: pQE70, pQE60, pQE-9 (Qiagen), pBS, phagescript, psiX174, pBluescript SK, pBSKS, pNH8a, pNH16a, pNH18a, pNH46a (Stratagene); pTRC9 9A, pKK2 2 3 - 3, pKK233-3, pDR540, pRIT5 (Pharmacia). Eukaryotic: pWLneo, pSV2cat, pOG44, pXTl, pSG (Stratagene) pSVK3, pBPV, pMSG, pSVL (Pharmacia). However, any other plasmid or vector may be used as long as they are replicable and viable in the host.
Promoter regions can be selected from any desired gene using CAT (chlor~m~hen;col transferase) vectors or other vectors with selectable markers. Two appropriate vectors are pKK232-8 and pCM7. Particular named bacterial promoters include lacI, lacZ, T3, T7, gpt, ~mh~ PR~ PL and trp.
Eukaryotic promoters include CMV immediate early, HSV
thymidine kinase, early and late SV40, LTRs from retrovirus, and mouse metallothionein-I. Selection of the appropriate vector and promoter is well within the level of ordinary skill in the art.
In a further emboA;ment, the present invention relates to host cells cont~;n~ng the above-described construct. The host cell can be a higher eukaryotic cell, such as a m~mm~l ian cell, or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell. Introduction of the construct into the host cell can be effected by calcium phosphate transfection, DEAE-Dextran mediated transfection, or electroporation (Davis, L., Dibner, M., Battey, I., Basic Methods in Molecular Biology, 1986) ) .

W O 96~9508 PCT/U'~J~7108 The constructs in host cells can ~e used in a conventional m~nner to produce the gene product ~nCo~d by the recombinant sequence. Alternatively, the polypeptides of the invention can be synthetically produced by conventional peptide synthesizers.
Mature proteins can be expressed in m~mm~ lian cells, yeast, bacteria, or other cells under the control of appropriate promoters. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention.
Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrook, et al., Molecular Cloning: A Laboratory M~nl7~l, Second Edition, (Cold Spring Harbor, N.Y., 1989), the disclosure of which is hereby incorporated by reference.
Transcription of a DNA encoding the polypeptides of the present invention by higher eukaryotes is increased by inserting an ~nh~ncer sequence into the vector. ~nhAncers are cis-acting elements of DNA, usually about from 10 to 300 bp, that act on a promoter to increase its transcription.
Examples include the SV40 ~nh~ncer on the late side of the replication origin (bp 100 to 270), a cytomegalovirus early promoter ~nh~nc~, a polyoma ~nh~ncer on the late side of the replication origin, and adenovirus ~nh~ncers.
Generally, recombinant expression vectors will include origins of replication and selectable markers permitting transformation of the host cell, e.g., the ampicillin resistance gene of E. coli and S. cerevisiae TRP1 gene, and a promoter derived from a highly-expressed gene to direct transcription of a downstream structural sequence. Such promoters can be derived from operons encoding glycolytic enzymes such as 3-phosphoglycerate kinase (PGK), a factor, acid phosphatase, or heat shock proteins, among others. The heterologous structural sequence is assembled in appropriate phase with translation, initiation and termination sequences, W 0 96~9508 CA 02220988 1997-11-12 PCT/U~33/'~7108 and preferably, a leader sequence capable of directing secretion of translated protein into the periplasmic space or extracellular medium. Optionally, the heterologous sequence can encode a fusion protein including an N-terminal identification peptide imparting desired characteristics, e.g., stabilization or simplified purification of expressed reco~h~n~nt product.
Useful expression vectors for bacterial use are constructed by inserting a structural DNA sequence encoding a desired protein together with suitable translation, initiation and termination signals in operable reading phase with a functional promoter. The vector will comprise one or more phenotypic selectable markers and an origin of replication to ensure maintenance of the vector and to, if desirable, provide amplification within the host. Suitable prokaryotic hosts for transformation include E. coli, Bacillus subtilis, Salmonella tyPhimurium and various species within the genera Psell~omnn~ Streptomyces, and Staphylococcus, although others may also be employed as a matter of choice.
As a representative but nonlimiting example, useful expression vectors for bacterial use can comprise a selectable marker and bacterial origin of replication derived from commercially available plasmids comprising genetic elements of the well known cloning vector pBR322 (ATCC
37017). Such commercial vectors include, for example, pKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and GEM1 (Promega Biotec, Madison, WI, USA). These pBR322 "backbone~
sections are combined with an appropriate promoter and the structural sequence to be expressed.
Following transformation of a suitable host strain and growth of the host strain to an appropriate cell density, the selected promoter is derepressed by appropriate ~eans (e.g., temperature shift or chemical induction) and cells are cultured for an additional period.

W O 96~9508 PCTIU~ 7108 Cells are typically harvested by centri~ugation, disrupted by physical or chemical means, and the resulting crude extract ret~ne~ for further purification.
Microbial cells employed in expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents.
Various m~mm~ n cell culture systems can also be employed to express recombinant protein. Examples of m~mm~l ian expression systems include the COS-7 lines of monkey kidney fibroblasts, described by Gluzman, Cell, 23:175 (1981), and other cell lines capable of expressing a compatible vector, for example, the C127, 3T3, CHO, HeLa and BHK cell lines. ~mm~ian expression vectors will comprise an origin of replication, a suitable promoter and enh~ncer, and also any necessary ribosome binding sites, polyadenylation site, splice donor and acceptor sites, transcriptional termination sequences, and 5~ ~lanking nontranscribed sequences. DNA sequences derived from the SV40 viral genome, for example, SV40 origin, early promoter, enh~ncer, splice, and polyadenylation sites may be used to p~ovide the required nontranscribed genetic elements.
The polypeptide of ~he present invention may be recovered and purified fr~m recombinant cell cultures by methods used heretofore, including ~mmon; um sulfate or ethanol precipitation, acid extraction, anion or cation Pxch~nge chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxyapatite chromatography and lectin chromatography. Protein refolding steps can be used, as necessary, in completing configuration of the mature protein.
Finally, high performance liquid chromatography (HPLC) can be employed for final purification steps.
The polypeptide of the present invention may be a naturally purified product, or a product of chemical W O 96~9508 PCT~US95/07108 synthetic procedures, or produced by recombinant techniques from a prokaryotic or eukaryotic host (for example, by bacterial, yeast, higher plant, insect and m~mm~ n cells in culture). Dep~n~;ng upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated with m~mm~ n or other eukaryotic carbohydrates or may be non-glycosylated.
Polypeptides of the invention may also include an initial methionine amino acid residue.
The polypeptide of the present invention, as a result of the ability to stimulate vascular endothelial cell growth, may be employed in treatment for stimulating re-vascularization of ischemic tissues due to various disease conditions such as thrombosis, arteriosclerosis, and other cardiovascular conditions. These polypeptide may also be employed to stimulate angiogenesis and lim.b regeneration.
The polypeptide may also be employed for treating wounds due to injuries, burns, post-operative tissue repair, and ulcers since they are mitogenic to various cells of different origins, such as fibroblast cells and skeletal muscle cells, and therefore, facilitate the repair or replacement of damaged or diseased tissue.
The polypeptide of the present invention may also be employed stimulate neuronal growth and to treat and prevent neuronal damage associated with stroke and which occurs in certain neuronal disorders or neuro-degenerative conditions such as Al~he;m~r's disease, Parkinson's disease, and AIDS-related complex. FGF-13 has the ability to stimulate chon~rocyte growth, therefore, they may be employed to Pnh~nce bone and periodontal regeneration and aid in tissue transplants or bone grafts.
The polypeptide of the present invention may be also be employed to prevent skin aging due to sunburn by stimulating keratinocyte growth.

W096~9508 PCT/U',S~7108 The FGF-13 polypeptide may also be employed for preventing hair loss, since FGF family members activate hair-forming cells and promotes melanocyte growth. Along the same lines, the polypeptides of the present invention may be ~ employed to st;m~ te growth and differentiation of hematopoietic cells and bone marrow cells when used in ~omh~n~tion with other cytokines.
The FGF-13 polypeptide may also be employed to maintain organs before transplantation or for supporting cell culture of primary tissues.
The polypeptide of the present invention may also be employed for inducing tissue of mesodermal origin to differentiate in early embryos.
In accordance with yet a ~urther aspect o~ the present invention, there is provided a process for utilizing such polypeptides, or polynucleotides encoding such polypeptides, for in vitro purposes related to scientific research, synthesis of DNA, manufacture of DNA vectors and for the purpose of providing diagnostics and therapeutics for the treatment of human disease.
This invention provides a method for identification of the receptors for the polypeptides of the present invention.
The genes encoding the receptor can be identified by numerous methods known to those of skill in the art, for example, ligand panning and FACS sorting (Coligan, et al., Current Protocols in Immun., 1(2), Chapter 5, (1991)). Preferably, = expression cloning is employed wherein polyadenylated RNA is prepared from a cell responsive to the polypeptides, for example, NIH3T3 cells which are known to contain multiple receptors for the FGF family proteins, and SC-3 cells, and a cDNA library created from this RNA is divided into pools and used to transfect COS cells or other cells that are not responsive to the polypeptides. Transfected cells which are grown on glass slides are exposed to the the polypeptide of the present invention, after they have been labelled. The W O 96~9508 PCT/u',~ 7108 polypeptides can be labeled by a variety of means including iodination or inclusion of a recognition site for a site-specific protein kinase.
Following fixation and incubation, the slides are subjected to auto-radiographic analysis. Positive pools are identified and sub-pools are prepared and re-transfected using an iterative sub-pooling and re-screening process, eventually yielding a single clones that encodes the putative receptor.
As an alternative approach for receptor identification, the labeled polypeptides can be photoaffinity linked with cell membrane or extract preparations that express the receptor molecule. Cross-linked material is resolved by PAGE
analysis and exposed to X-ray film. The labeled complex cont~ining the receptors of the polypeptides can be excised, resolved into peptide fragments, and subjected to protein microsequencing. The amino acid sequence obtained ~rom microsequencing would be used to design a set of degenerate oligonucleotide probes to screen a cDNA library to identify the genes encoding the putative receptors.
This invention provides a method of screening compounds to identify those which modulate the action of the polypeptide of the present invention. An example of such an assay comprises com~ining a m~mm~l ian fibroblast cell, a the polypeptide of the present invention, the compound to be screened and 3 ~H] thymidine under cell culture conditions where the fibroblast cell would normally proliferate. A
control assay may be performed in the absence of the compound to be screened and compared to the amount of fibroblast proliferation in the presence of the compound to determine if the compound stimulates proliferation by determining the uptake of 3 [H] thymidine in each case. The amount of fibroblast cell proliferation is measured by liquid scintillation chromatography which measures the incorporation W O 96~9508 PCT/U~J51v~108 of 3 [H] thymidine. Both agonist and antagonist compounds may be identified by this procedure.
In another method, a m~mm~ n cell or membrane preparation expressing a receptor for a polypeptide of the present invention is incubated with a labeled polypeptide of the present invention in the presence of the compound. The ability of the compound to ~nh~nce or block this interaction could then be measured. Alternatively, the response of a known second messenger system following interaction of a compound to be screened and the FGF-13 receptor is measured and the ability of the compound to bind to the receptor and elicit a second messenger response is measured to determine if the compound is a potential agonist or antagonist. Such second messenger systems include but are not limited to, cAMP
guanylate cyclase, ion ch~nnels or phosphoinositide hydrolysis.
Examples of antagonist compounds include antibodies, or in some cases, oligonucleotides, which bind to the receptor for the polypeptide of the present invention but elicit no second messenger response or bind to the FGF-13 polypeptide itself. Alternatively, a potential antagonist may be a mllt~nt form of the polypeptide which binds to the receptors, however, no second messenger response is elicited and, therefore, the action of the polypeptide is effectively blocked.
Another antagonist compound to the FGF-13 gene and gene product is an antisense construct prepared using antisense technology. Antisense technology can be used to control gene expression through triple-helix formation or antisense DNA or RNA, both of which methods are based on binding of a polynucleotide to DNA or RNA. For example, the 5' coding portion of the polynucleotide sequence, which encodes for the mature polypeptides of the present invention, is used to design an antisense RNA oligonucleotide of from about 10 to 40 base pairs in length. A DNA oligonucleotide is designed =

W O 96~9508 PCT/U~ /108 to be complementary to a region of the gene involved in transcription (triple helix -see Lee et al., Nucl. Acids Res., 6:3073 (1979); Cooney et al, Science, 241:456 (1988);
and Dervan et al., Science, 251: 1360 (l991)), thereby preventing transcription and the production of the polypeptides of the present invention. The antisense RNA
oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into the polypeptide (Antisense - Okano, J. Neurochem., 56:560 (1991);
Oligodeoxynucleotides as Antisense Tnh; h~ tors of Gene Expression, CRC Press, Boca Raton, FL (1988)). The oligonucleotides described above can also be delivered to cells such that the antisense RNA or DNA may be expressed in vivo to ; nhi hi t production o~ the polypeptide.
Potential antagonist compounds also include small molecules which bind to and occupy the binding site of the receptors thereby making the receptor inaccessible to its polypeptide such that normal biological activity is prevented. Examples of small molecules include, but are not limited to, small peptides or peptide-like molecules.
Antagonist compounds may be employed to inhibit the cell growth and proli~eration e~ects of the polypeptides of the present invention on neoplastic cells and tissues, i.e.
stimulation of angiogenesis of tumors, and, therefore, retard or prevent abnormal cellular growth and proliferation, ~or example, in tumor formation or growth.
The antagonists may also be employed to prevent hyper-vascular diseases, and prevent the proliferation of epithelial lens cells after extracapsular cataract surgery.
Prevention of the mitogenic ac~ivity of the polypeptides of the present invention may also be desirous in cases such as restenosis after balloon angioplasty.
The antagonists may also be employed to prevent the growth of scar tissue during wound healing.

W O 96~9508 P ~/U',SJ~7108 The antagonists may be employed in a composition with a pharmaceutically acceptable carrier, e.g., as hereinafter described.
The polypeptides, agonists and antagonists of the ~ present invention may be employed in combination with a suitable pharmaceutical carrier to comprise a pharmaceutical - composition for parenteral A~m~n~stration. Such compositions comprise a therapeutically effective amount of the polypeptide, agonist or antagonist and a pharmaceutically acceptable carrier or excipient. Such a carrier includes but is not limited to saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof. The formulation should suit the mode of A~m;n~ stration.
The invention also provides a pharmaceutical pack or kit comprising one or more contA;ners filled with one or more of the ingredients of the pharmaceutical compositions of the invention. Associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manu~acture, use or sale o~ phanmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human A~m;n;stration. In addition, the polypeptides, agonists and antagonists of the present invention may be employed in conjunction with other therapeutic compounds.
The pharmaceutical compositions may be A~m; n~ stered in a convenient mAnner such as by the oral, topical, intravenous, intraperitoneal, intramuscular, subcutaneous, intranasal or intradermal routes. The pharmaceutical compositions are A~m;n~stered in an amount which is effective for treating and/or prophylaxis of the specific indication.
In gen~ral, they are ~m; n; stered in an amount of at least about 10 ~gtkg body weight and in most cases they will be A~mi n; stered in an amount not in excess of about 8 mg/Kg body weight per day. In most cases, the dosage is from about 10 ~g/kg to about 1 mg/kg body weight daily, taking into account W O 96~9508 PCT/U~,3~7108 the routes of ~m;n; stration, s ~ toms, etc. In the specific case of topical ~m; n; stration, dosages are prèferably administered from about 0.1 ~g to 9 mg per cm2.
The polypeptide of the invention and agonist and antagonist compounds which are polypeptides, may also be employed in accordance with the present invention by expression of such polypeptide in vivo, which is often referred to as "gene therapy.ll Thus, for example, cells may be engineered with a polynucleotide (DNA or RNA) encoding for the polypeptide ex vivo, the engineered cells are then provided to a patient to be treated with the polypeptide. Such methods are well-known in the art. For example, cells may be engineered by procedures known in the art by use of a retroviral particle cont~-ning RNA encoding for the polypeptide of the present invention.
Similarly, cells may be engineered in vivo for expression of the polypeptide in vivo, for example, by procedures known in the art. As known in the art, a producer cell for producing a retroviral particle cont~;n;ng RNA
encoding the polypeptide of the present invention may be ~m; n; stered to a patient for engineering cells in vivo and expression of the polypeptide in vivo. These and other methods for ~m~ n;stering a polypeptide of the present invention by such methods should be apparent to those skilled in the art from the teachings of the present invention. For example, the expression vehicle for engineering cells may be other than a retroviral particle, for example, an adenovirus, which may be used to engineer cells in vivo after combination with a suitable delivery vehicle.
Retroviruses from which the retroviral plasmid vectors hereinabove mentioned may be derived include, but are not limited to, Moloney Murine Leukemia Virus, spleen necrosis virus, retroviruses such as Rous Sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, gibbon ape leukemia W O 96~9508 PCTAJS95/07108 virus, human ~mmllnQdeficiency virus, adenovirus, MyeloproliferatiVe Sarcoma Virus, and m~m~ry tumor virus.
In one embo~;mPnt, the retroviral plasmid vector is derived from Moloney Murine Leukemia Virus.
The vector includes one or more promoters. Suitable promoters which may be employed include, but are not limited - to, the retroviral LTR; the Sv40 promoter; and the hllm~n cytomegalovirus (CMV) promoter described in Miller, et al., Biotechniques, Vol. 7, No. 9, 980-990 (1989), or any other promoter (e.g., cellular promoters such as eukaryotic cellular promoters including, but not limited to, the histone, pol III, and ~-actin promoters). Other viral promoters which may be employed include, but are not limited to, adenovirus promoters, thymidine kinase (TK) promoters, and B19 parvovirus promoters. The selection of a suitable promoter will be apparent to those skilled in the art ~rom the teachings cont~nP~ herein.
The nucleic acid sequence encoding the polypeptide of the present invention is under the control of a suitable promoter. Suitable promoters which may be employed include, but are not limited to, adenoviral promoters, such as the adenoviral major late promoter; or hetorologous promoters, such as the cytomegalovirus (CMV) promoter; the respiratory syncytial virus (RSV) promoter; ~n~llr~hle promoters, such as the MMT promoter, the metallothionein promoter; heat shock promoters; the albumin promoter; the ApoAI promoter; hllm~n globin promoters; viral thymidine kinase promoters, such as the Herpes Simplex thymidine kinase promoter; retroviral LTRs (including the modified retroviral LTRs hereinabove described); the ~-actin promoter; and human growth hormone promoters. The promoter also may be the native promoter which controls the gene encoding the polypeptide.
The retroviral plasmid vector is employed to transduce packaging cell lines to form producer cell lines. Examples of packaging cells which may be transfected include, but are W O 96~9508 PCT/U',;~'~7108 not limited to, the PE501, PA317, ~-2, ~-AM, PA12, T19-14X, VT-19-17-H2, ~CRE, ~CRIP, GP+E-86, GP+envAml2, and DAN cell lines as described in Miller, Human Gene Therapy, Vol. 1, pgs. 5-14 (1990), which is incorporated herein by reference in its entirety. The vector may transduce the packaging cells through any means known in the art. Such means include, but are not limited to, electroporation, the use of liposomes, and CaPO4 precipitation. In one alternative, the retroviral plasmid vector may be encapsulated into a liposome, or coupled to a lipid, and then ~min; stered to a host.
The producer cell line generates infectious retroviral vector particles which include the nucleic acid sequence(s) encoding the polypeptides. Such retroviral vector particles then may be employed, to transduce eukaryotic cells, either in vi tro or in vivo. The transduced eukaryotic cells will express the nucleic acid sequence(s) encoding the polypeptide. Eukaryotic cells which may be transduced include, but are not limited to, embryonic stem cells, embryonic carcinoma cells, as well as hematopoietic stem cells, hepctocytes, fibroblasts, myoblasts, keratinocytes, endothelial cells, and bronchial epithelial cells.
This i~vention is also related to the use of the genes of the present invention as part of a diagnostic assay for detecting diseases or susceptibility to diseases related to the presence of mutations in the nucleic acid sequences encoding the polypeptide of the present invention.
Individuals carrying mutations in a gene of the present invention may be detected at the DNA level by a variety of techniques. Nucleic acids for diagnosis may be obtained from a patient's cells, such as from blood, urine, saliva, tissue biopsy and autopsy material. The genomic DNA may be used directly for detection or may be amplified enzymatically by using PCR (Saiki et al., Nature, 324:163-166 (1986)) prior to analysis. RNA or cDNA may also be used for the same purpose.

-W O 96~9508 P~TSII',,;Iv~lO~

As an example, PCR primers complementary to the nucleic acid encoding a polypeptide of the present invention can be used to identify and analyze mutations. For example, deletions and insertions can be detected by a change in size of the amplified product in r~mr~riSon to the normal genotype.
Point mutations can be identified by hybridizing amplified DNA to radiolabeled RNA or alternatively, radiolabeled antisense DNA sequences. Perfectly matched sequences can be distinguished from mismatched duplexes by RNase A digestion or by differences in melting temperatures.
Genetic testing based on DNA sequence differences may be achieved by detection of alteration in electrophoretic mobility of DNA fragments in gels with or without denaturing agents. Small sequence deletions and insertions can be visualized by high resolution gel electrophoresis. DNA
fragments of different sequences may be distinguished on denaturing formamide gradient gels in which the mobilities of different DNA fragments are retarded in the gel at different positions according to their specific melting or partial melting temperatures (see, e.g., Myers et al ., Science, 230:1242 (1985)).
Sequence changes at specific locations may also be revealed by nuclease protection assays, such as RNase and S1 protection or the chemical cleavage method (e.g., Cotton et al ., PNAS, USA, 85:4397-4401 (1985)).
Thus, the detection of a specific DNA sequence may be achieved by methods such as hybridization, RNase protection, chemical cleavage, direct DNA sequencing or the use of restriction enzymes, (e.g., Restriction Fragment Length Polymorphisms (RFLP)) and Southern blotting of genomic DNA.
In addition to more conventional gel-electrophoresis and DNA sequencing, mutations can also be detected by in situ analysis.
The present invention also relates to a diagnostic assay for detecting altered levels of FGF-13 proteins in various tissues since an over-expression of the proteins compared to normal control tissue samples may detect the presence of abnormal cellular proliferation, for example, a tumor.
Assays used to detect levels of protein in a sample derived from a host are well-known to those of skill in the art and include radio;mml~no~qsays, competitive-h; n~;ng assays, Western Blot analysis, ELISA assays and ''sandwichll assay. An ELISA assay (Coligan, et al., Current Protocols in Tmmllnology, 1(2), Chapter 6, (1991)) initially comprises preparing an antibody specific to an antigen to the polypeptides of the present invention, preferably a monoclonal antibody. In addition a reporter antibody is prepared against the monoclonal antibody. To the reporter antibody is attached a detectable reagent such as radioactivity, fluorescence or, in this example, a horseradish peroxidase enzyme. A sample is removed from a host and incubated on a solid support, e.g. a polystyrene dish, that binds the proteins in the sample. Any free protein h; n~; ng sites on the dish are then covered by incubating with a non-specific protein like bovine serum albumen. Next, the monoclonal antibody is incubated in the dish during which time the monoc~-nal antibodies attach to any polypeptides of the present lnvention attached to the polystyrene dish. All unbound monoclonal antibody is washed out with buffer. The reporter antibody linked to horseradish peroxidase is now placed in the dish resulting in binding of the reporter antibody to any monoclonal antibody bound to the protein of interest.
Unattached reporter antibody is then washed out.
Peroxidase substrates are then added to the dish and the amount of color developed in a given time period is a measurement of the amount of a polypeptide of the present invention present in a given volume of patient sample when compared against a standard curve.

W O 96~9508 PCTAJS95/07108 A competition assay may be employed wherein antibodies speci~ic to a polypeptide of the present invention are attached to a solid support and labeled FGF-13 and a sample derived from the host are passed over the solid support and the amount of label detected, for example by liquid scintillation chromatography, can be correlated to a quantity - of a polypeptide of the present invention in the sample.
A "sandwich" assay is similar to an ELISA assay. In a "sandwich" assay a polypeptide of the present invention is passed over a solid support and binds to antibody attached to a solid support. A second antibody is then bound to the polypeptide of interest. A third antibody which is labeled and specific to the second antibody is then passed over the solid support and binds to the second antibody and an amount can then be quantified.
The sequences of the present invention are also valuable for chromosome identification. The sequence is specifically targeted to and can hybridize with a particular location on an individual human chromosome. Moreover, there is a current need for identifying particular sites on the chromosome. Few chromosome marking reagents based on actual sequence data (repeat polymorphism's) are presently available ~or marking chromosomal location. The mapping of DNAs to chromosomes according to the present invention is an important first step in correlating those sequences with genes associated with disease.
Briefly, sequences can be mapped to chromosomes by preparing PCR primers (preferably 15-25 bp) from the cDNA.
Computer analysis of the 3' untranslated region is used to rapidly select primers that do not span more than one exon in the genomic DNA, thus complicating the amplification process.
These primers are then used for PCR screening of somatic cell hybrids cont~; n~ ng individual human chromosomes. Only those hybrids cont~ining the human gene corresponding to the primer will yield an amplified fragment.

WO 96/39!;08 CA 02220988 1997 - 1 1 - 12 PCT/US95/07108 PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular DNA to a particular chromosome.
Using the present invention with the same oligonucleotide primers, sublocalization can be achieved with panels of fragments from specific chromosomes or pools o~ large genomic clones in an analogous m~nner. Other mapping strategies that can similarly be used to map to its chromosome include in situ hybridization, prescreening with labeled flow-sorted chromosomes and preselection by hybridization to construct chromosome specific-cDNA libraries.
Fluorescence in situ hybridization (FISH) o~ a cDNA
clone to a met~rh~se chromosomal spread can be used to provide a precise chromosomal location in one step. This technique can be used with cDNA as short as 50 or 60 bases.
For a review of this technique, see Verma et al., ~llm~n Chromosomes: a M~nll~ 1 of Basic Techniques, Pergamon Press, New York (1988).
Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be correlated with genetic map data. (Such data are found, for example, in V. McKusick, Mendelian Inheritance in Man (available on line through Johns Hopkins University Welch Medical ~ibrary). The relationship between genes and diseases that have been mapped to the same chromosomal region are then identified through linkage analysis (c nheritance of physically adjacent genes).
Next, it is necessary to determine the differences in the cDNA or genomic sequence between affected and unaffected individuals. If a mutation is observed in some or all of the affected individuals but not in any normal individuals, then the mutation is likely to be the causative agent of the disease.
With current resolution of physical mapping and genetic mapping techniques, a cDNA precisely localized to a chromosomal region associated with the disease could be one WO 96/39508 PCT/U~J51~7108 of between 50 and 500 pote~tial causative genes. ~This assumes 1 megabase mapping resolution and one gene per 20 kb)-The polypeptides, their fra~nts or other derivatives,or analogs thereof, or cells expressing them can be used as an ~mmllnogen to produce antibodies thereto. These antibodies = ~ can be, for example, polyclonal or monoclonal antibodies.
The present invention also includes ch;me~iC~ single chain, and hllm~n; zed antibodies, as well as Fab fragments, or the product of an Fab expression library. Various procedures known in the art may be used for the production of such antibodies and ~ragments.
Antibodies generated against the polypeptides corresponding to a sequence of the present invention can be obtained by direct injection o~ the polypeptides into an ~nim~l or by ~m;n~ stering the polypeptides to an ~n;m~l, preferably a nonhllm~n The antibody so obt~i n~ will then bind the polypeptides itself. In this m~nne~, even a sequence encoding only a fragment of the polypeptides can be used to generate antibodies b; n~; ng the whole native polypeptides. Such antibodies can then be used to isolate the polypeptide from tissue expressing that polypeptide.
For preparation of monoclonal antibodies, any technique which provides antibodies produced by continuous cell line cultures can be used. Examples include the hybridoma technique (Kohler and Milstein, 1975, Nature, 256:495-497), the trioma technique, the human B-cell hybridoma technique (Kozbor et al., 1983, T~mllnology Today 4:72), and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole, et al., 1985, in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96).
Techniques described for the production of single chain antibodies (U.S. Patent 4,946,778) can be adapted to produce single chain antibodies to ~mmllnogenic polypeptide products of this invention. Also, transgenic mice may be used to W O 96~9508 PCT/U~3S/~7108 express hl]m~n;zed antibodies to imml~nogenic polypeptide products of this invention.
The present invention will be further described with reference to the following examples; however, it is to be understood that the present invention is not limited to such examples. All parts or amounts, unless otherwise specified, are by weight.
In order to facilitate underst~nd~ng of the following examples, certain frequently occurring methods and/or terms will be described.
"Plasmids" are designated by a lower case p preceded and/or followed by capital letters and/or numbers. The starting plasmids herein are either commercially available, publicly available on an unrestricted basis, or can be constructed from available plasmids in accord with published procedures. In addition, equivalent plasmids to those described are known in the art and will be apparent to the ordinarily skilled artisan.
"Digestion" of DNA refers to catalytic cleavage of the DNA with a restriction enzyme that acts only at certain sequences in the DNA. The various restriction enzymes used herein are commercially available and their reaction conditions, cofactors and other requirements were used as would be known to the ordinarily skilled artisan. For analytical purposes, typically 1 ~g of plasmid or DNA
fragment is used with about 2 units of enzyme in about 20 ~1 of buffer solution. For the purpose of isolating DNA
fragments for plasmid construction, typically 5 to 50 ~g of DNA are digested with 20 to 250 units of enzyme in a larger volume. Appropriate buffers and substrate amounts for particular restriction enzymes are specified by the manufacturer. Incubation times of about 1 hour at 37~C are ordinarily used, but may vary in accordance with the supplier's instructions. After digestion the reaction is -W O 96~9508 PCT/U'~S~7108 electrophoresed directly on a polyacrylamide gel to isolate the desired fragment.
Size separation of the cleaved fragments is performed using 8 percent polyacrylamide gel described by Goeddel, D.
et al ., Nucleic Acids Res., 8:4057 (1980).
~ Oligonucleotides" refers to either a single str~n~
polydeoxynucleotide or two complementary polydeoxynucleotide strands which may be chemically synthesized. Such synthetic oligonucleotides have no 5' phosphate and thus will not ligate to another oligonucleotide without adding a phosphate with an ATP in the presence of a kinase. A synthetic oligonucleotide will ligate to a fragment that has not been dephosphorylated.
"Ligation" refers to the process of forming phosphodiester bonds between two double stranded nucleic acid fragments (Maniatis, T., et al., Id., p. 146). Unless otherwise provided, ligation may be accomplished using known buffers and conditions with 10 units of T4 DNA ligase ("ligase") per 0.5 ~g of approximately equimolar amounts of the DNA fragments to be ligated.
Unless otherwise stated, transformation was per~onmed as described by the method of Graham, F. and Van der Eb, A., Virology, 52:456-457 (1973).

Exam~Ple 1 Bacterial Expression and Purification of FGF-13 Protein The DNA sequence encoding FGF-13 ATCC # 97147, is initially amplified using PCR oligonucleotide primers corresponding to the 5' sequences of the processed protein (minus the signal peptide sequence) and the vector sequences 3' to the gene. Additional nucleotides correspon~tng to the gene are added to the 5' and 3' sequences respectively. The 5' oligonucleotide primer 5' GCCAGACCATGGAGAATCA~l~-lCCTAAT
3' (SEQ ID NO:3) cnnt~tnc a Nco restriction enzyme site. The 3~ sequence 5' GATTTAAGATCTCGTGAGGGGCTGGGGCCG 3' (SEQ ID

W 0 96~9508 PcT/u~J~ o8 NO:4) cont~;n~ complementary sequences to a BglII site and is ~ollowed by 18 nucleotides of FGF-13 coding sequence.
The restriction enzyme sites correspond to the restriction enzyme sites on the bacterial expression vector pQE-60 (Qiagen, Inc. Chatsworth, CA 91311). pQE-60 encodes antibiotic resistance (Ampr), a bacterial origin o~
replication (ori), an IPTG-regulatable promoter operator (P/O), a ribosome binding site (RBS), a 6-His tag and restriction enzyme sites. pQE-60 was then digested with NcoI
and BglII. The ampli~ied sequences are ligated into pQE-60 and are inserted in frame with the sequence encoding for the histidine tag and the ribosome hi n~l;ng site (RBS). The ligation mixture is then used to transform E. coli strain M15/rep 4 (Qiagen, Inc.) by the procedure described in Sambrook, J. et al., Molecular Cloning: A Laboratory ~n~
Cold Spring Laboratory Press, (1989). M15/rep4 contains multiple copies of the plasmid pREP4, which expresses the lacI repressor and also confers kanamycin resistance (Kanr).
Transformants are identified by their ability to grow on LB
plates and ampicillin/kanamycin resistant colonies were selected. Plasmid DNA is isolated and confirmed by restriction analysis. Clones cont~;n;ng the desired constructs are grown overnight (O/N) in liquid culturein LB
media supplemented with both Amp (100 ug/ml) and Kan (25 ug/ml). The O/N culture is used to inoculate a large culture at a ratio of 1:100 to 1: 250. The cells are grown to an optical density 600 (O.D.~) of between 0.4 and 0.6. IPTG
("Isopropyl-B-D-thiogalacto pyranoside") is then added to a final concentration of 1 mM. IPTG induces by inactivating the lacI repressor, clearing the P/O leading to increased gene expression. Cells are grown an extra 3 to 4 hours.
Cells are then harvested by centrifugation. The cell pellet is solubilized in the chaotropic agent 6 Molar Guanidine HCl.
After clarification, solubilized FGF-13 is purified from this solution by chromatography on a Nickel-Chelate column under W O 96~9508 PCT/U~ 7108 conditions that allow for tight hi n~i n~ by proteins cont~in;ng the 6-His tag (Hochuli, E. et al., J.
Chrom.atography 411:177-184 (1984)). The proteins are eluted from the column in 6 molar guanidine HCl pH 5.0 and for the purpose of renaturation adjusted to 3 molar guanidine HCl, lOOmM sodium phosphate, 10 mmolar glutathione ~reduced) and 2 mmolar glutathione (oxidized). After incubation in this solution for 12 hours the proteins are dialyzed to 10 mmolar sodium phosphate.

~xample 2 Cloninq and exPression of FGF-13 usinq the baculovirus expression system The DNA sequence encoding the full length FGF-13 protein, ATCC # 97147,is am.plified using PCR oligonucleotide primers corresponding to the 5' and 3' sequences of the gene:
The FGF-13 5' primer has the sequence 5' CTAGTGGATCCCGA
GAATCACCCGTCTCCT 3' (SEQ ID NO:5) and contains a BamHI
restriction enzyme site (in bold) such that cloning at this site will put the baculovirus signal sequence in frame with 18 nucleotides of the FGF-13 gene downstream of the putative FGF-13 signal peptide cleavage site.
The 3' primer has the sequence 5' CGA~ AGAACCT
CGGGGATCTGGCTCC 3' (SEQ ID NO:6) and contains the cleavage site for the restriction endonuclease XbaI and 18 nucleotides complementary to the 3' non-translated sequence of the gene.
The amplified sequences are isolated from a 1% agarose gel using a commercially available kit ("Geneclean," BIO 101 Inc., La Jolla, Ca.). The fragment is then digested with the respective ~n~nllcleases and purified again on a 1~ agarose gel. This fragment is designated F2.
The vector pA2gp (modification of pVh941 vector, discussed below) is used for the expression of the proteins using the baculovirus expression system (for review see:
Summers, M.D. and Smith, G.E. 1987, A m~nll~l of methods for W O 96~9508 PCT/u~ 7108 baculovirus vectors and insect cell culture procedures, Texas Agricultural Experimental Station Bulletin No. 1555). This expression vector cont~nC the strong polyhedrin promoter of the Autographa californica nuclear polyhedrosis virus (AcMNPV) followed by the recognition sites for the restriction endonucleases BamHI and XbaI. The polyadenylation site of the simian virus (SV)40 is used for efficient polyadenylation. For an easy selection of recombinant virus the beta-galactosidase gene from E.coli is inserted in the same orientation as the polyhedrin promoter followed by the polyadenylation signal of the polyhedrin gene. The polyhedrin sequences are flanked at both sides by viral sequences for the cell-mediated homologous recombination of co-transfected wild-type viral DNA. Many other baculovirus vectors could be used in place of pA2 such as pRG1, pAc373, pVL941 and pAcIM1 (Luckow, V.A. and Summers, M.D., Virology, 170:31-39).
The plasmid is digested with the restriction enzymes and dephosphorylated using calf intestinal phosphatase by procedures known in the art. The DNA is then isolated from a 1~ agarose gel using the romm~cially available kit (llGenecle~n" BIO 101 Inc., La Jolla, Ca.). This vector DNA
is designated V2.
Fragment F2 and the dephosphorylated plasmid V2 are ligated with T4 DNA ligase. E.coli DH5~ cells are then transformed and bacteria identified that contained the plasmid (pBacFGF-13) using the respective restriction enzymes. The sequence of the cloned fragment are confinmed by DNA se~uencing.
5 ~g of the plasmid pBacFGF-13 are co-transfected with 1.0 ~g of a commercially available linearized baculovirus ("BaculoGold~ baculovirus DNA", Pharmingen, San Diego, CA.) using the lipofection method (Felgner et al. Proc. Natl.
Acad. Sci. USA, 84:7413-7417 (1987)).

= W 0 96~9508 PCTI~'9iJ~710~

l~g o~ BaculoGold~ virus DNA and 5 ~g of the plasmids, in each case, are mixed in a sterile well of microtiter plates cont~;n;ng 50 ~l of serum free Grace's medium (Life Technologies Inc., Gaithersburg, MD). Afterwards 10 ~1 Lipo~ectin plus 90 ~l Grace's medium are added, m; ~ and incubated for 15 minutes at room temperature. Then the transfection mixture is added drop-wise to the Sf9 insect cells ~ATCC CRL 1711) seeded in 35 mm tissue culture plates with 1 ml Grace's medium without serum. The plates are rocked back and forth to mix the newly added solution. The plates are then incubated for 5 hours at 27~C. After 5 hours the transfection solution is removed from the plate and 1 ml of Grace's insect medium supplemented with 10~ fetal calf serum is added. The plates are put back into an incubator and cultivation continued at 27~C for four days.
After four days the supernatant is collected and plaque assays performed similar as described by Summers and Smith (supra). As a modification an agarose gel with "Blue Gal"
(Life Technologies Inc., Gaithersburg) is used which allows an easy isolation of blue stained plaques. (A detailed description of a llplaque assay" can also be found in the user's guide for insect cell culture and baculovirology distributed by Life Technologies Inc., Gaithersburg, page 9-10) .
Four days a~ter the serial dilution the virus is added to the cells and blue st~ine~ plaques are picked with the tip of an Eppendorf pipette. The agar cont~;n;ng the recombinant viruses is then resuspended in an Eppendorf tube cont~;n;ng 200 ~1 of Grace's medium. The agar is removed by a brief centrifugation and the supernatant cont~;n;ng the recombinant baculovirus is used to infect Sf9 cells seeded in 35 mm dishes. Four days later the supernatants of these culture dishes are harvested and then stored at 4~C.
Sf9 cells are grown in Grace's medium supplemented with 10% heat-inactivated FBS. The cells are infected with the W O 96~9508 PCTAUS95/07108 recombinant baculovirus V-FGF-13 at a multiplicity of infection (MOI) of 2. Six hours later the medium is removed and replaced with SF900 II medium minus methionine and cysteine (Life Technologies Inc., Gaithersburg). 42 hours later 5 ~Ci of 35S-methionine and 5 ~Ci 35S cysteine (Amersham) are added. The cells are further incubated for 16 hours before they are harvested by centrifugation and the labelled proteins visualized by SDS-PAGE and autoradiography.

Example 3 Expression of Recombinant FGF-13 in COS cells The expression of plasmids, FGF-13-HA derived from a vector pcDNA3/Amp (Invitrogen) cont~;n;ng: 1) SV40 origin of r~ ication, 2) ampicillin resistance gene, 3) E.coli replication origin, 4) CMV promoter followed by a polylinker region, an SV40 intron and polyadenylation site. DNA
~ragments encoding the entire FGF-13 precursor and an HA tag fused in frame to the 3' end is cloned into the polylinker region of the vector, therefore, the recombinant protein expression is directed under the CMV promoter. The HA tag corresponds to an epitope derived from the influenza hemagglutinin protein as previously described (I. Wilson, H.
Niman, R. Heighten, A Cherenson, M. Connolly, and R. Lerner, 1984, Cell 37:767, (1984)). The infusion of HA tag to the target protein allows easy detection of the recombinant protein with an antibody that recognizes the HA epitope.
The PCR amplified DNA fragments and the vector, pcDNA3/Amp, are digested with the respective restriction enzymes and ligated. The ligation mixture is transformed into E. coli strain SURE (Stratagene Cloning Systems, La Jolla, CA) the transformed culture is plated on ampicillin media plates and resistant colonies are selected. Plasmid DNA is isolated from transformants and ~m~ned by restriction analysis for the presence of the correct fragment. For expression of the recombinant FGF-13 COS cells are W096~9508 PCTIU~SI~7108 trans~ected with the expression vector by DBAE-DE~{TRAN method (J. Sambrook, E. Fritsch, T. Maniatis, Molecular Cloning: A
Laboratory ~nll~l, Cold Spring Laboratory Press, (1989)).
The expression of the FGF-13-HA protein is detected by radiolabelling and ;mmllnoprecipitation method (E. Harlow, D.
Lane, Antibodies: A Laboratory ~nll~l, Cold Spring Harbor Laboratory Press, (1988)). Cells are la~elled for 8 hours with 35S-cysteine two days post transfection. Culture media is then collected and cells are lysed with detergent (RIPA
buffer (150 mM NaCl, 1% NP-40, 0.1% SDS, 1~ NP-40, 0.5% DOC, 50mM Tris, pH 7.5) (Wilson, I. et al., Id. 37:767 (1984)).
Both cell lysate and culture media are precipitated with an HA specific monoclonal antibody. Proteins precipitated are analyzed on 15~ SDS-PAGE gels.

~xam~le 5 Expression via Gene Therapy Fibroblasts are obt~ine~ ~rom a subject by skin biopsy.
The resulting tissue is placed in tissue-culture medium and separated into small pieces. Small chunks of the tissue are placed on a wet surface of a tissue culture flask, approximately ten pieces are placed in each flask. The flask is turned upside down, closed tight and le~t at room temperature over night. After 24 hours at room temperature, the flask is inverted and the chunks of tissue remain fixed to the bottom of the flask and fresh media (e.g., Ham~s F12 media, with 10% FBS, penicillin and streptomycin, is added.
This is then incubated at 37~C for approximately one week.
At this time, fresh media is added and subsequently changed every several days. After an additional two weeks in culture, a monolayer of fibroblasts emerge. The monolayer is trypsinized and scaled into larger flasks.
pMV-7 (Kirschmeier, P.T. et al, DNA, 7:219-25 (1988) flanked by the long terminal repeats of the Moloney murine sarcoma virus, is digested with EcoRI and HindIII and W O 96~9508 PCT/U'J~ 7108 subsequently treated with calf intestinal phosphatase. The linear vector is fractionated on agarose gel and puri~ied, using glass beads.
The cDNA enco~ng a polypeptide of the present invention is amplified using PCR primers which correspond to the 5' and 3~ end sequences respectively. The 5~ primer cont~n~ng an EcoRI site and the 3' primer having contains a HindIII site.
Equal quantities of the Moloney murine sarcoma virus linear backbone and the BcoRI and HimdIII fragment are added together, in the presence of T4 DNA ligase. The resulting mixture is maintAlne~ under conditions appropriate for ligation of the two fragments. The ligation mixture is used to transform bacteria B 101, which are then plated onto agar-cont~;n~ng kanamycin for the purpose of confirming that the vector had the gene of interest properly inserted.
The amphotropic pA317 or GP+aml2 packaging cells are grown in tissue culture to confluent density in Dulbecco's Modified Eagles Medium (DMEM) with 10~ calf serum (CS), penicillin and streptomycin. The MSV vector cont~;n;ng the gene is then added to the media and the packaging cells are transduced with the vector. The packaging cells now produce infectious viral particles containing the gene (the packaging cells are now referred to as producer cells).
Fresh media is added to the transduced producer cells, and subsequently, the media is harvested from a 10 cm plate of confluent producer cells. The spent media, containing the infectious viral particles, is filtered through a millipore filter to remove detached producer cells and this media is then used to infect fibroblast cells. Media is removed from a sub-confluent plate of fibroblasts and quickly replaced with the media from the producer cells. This media is removed and replaced with fresh media. If the titer of virus is high, then virtually all fibroblasts will be infected and no selection is required. If the titer is very low, then it W O 96~9508 PCTIU',~7108 is necessary to use a retroviral vector that has a selectable marker, such as neo or his.
The engineered fibroblasts are then injected into the host, either alone or after having been grown to con~luence on cytodex 3 microcarrier beads. The fibroblasts now produce the protein product.
Numerous modificatior, and variations o~ the present invention are possible in light of the above teachings and, therefore, within the scope of the appended cl~im~, the invention may be practiced otherwise than as particularly = described.

W O 96~9508 PCT/U',~J'~7108 SEQUENCE LISTING
(1) GENER~L INFORMATION:
(i) APPLICANT: HU, ET AL.
(ii) TITLE OF lNv~NllON: Fibroblast Growth Factor-13 (iii) NUMBER OF SEQUENCES: 8 (iv) CO~.SPONDENCE ADDRESS:
(A) ADDRESSEE: CARELLA, BYRNE, BAIN, GILFILLAN, CECCHI, STEWART ~ OLSTEIN
(B) STREET: 6 BECKER FARM ROAD
(C) CITY: ROSELAND
(D) STATE: NEW JERSEY
(E) ~O~N-1-~Y: USA
(F) ZIP: 07068 (v) COM~ul~ READABLE FORM:
(A) MEDIUM TYPE: 3.5 INCH DISKETTE
(B) COM~U-1~K: IBM PS/2 (C) OPERATING SYSTEM: MS-DOS
(D) SOFTWARE: WORD PERFECT 5.1 (vi) CURRENT APPLICATION DATA:
(A) APPLICATION N ~3ER:
(B) FILING DATE: Concurrently (C) CLASSIFICATION:
(vii) PRIOR APPLICATION DATA
(A) APPLICATION NUMBER: 08/207,412 (B) FILING DATE: 8 MAR 1994 (viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: FERRARO, GREGORY D.
(B) REGISTRATION NUMBER: 36,134 (C) REFERENCE/DOCKET NUMBER: 325800-364 (ix) TELECOMMUNICATION INFORMATION:
(A) TEL~N~: 201-994-1700 (B) TELEFAX: 201-994-1744 (2) INFORMATION FOR SEQ ID NO:1:
(i) SEQUENCE CHARACTERISTICS
(A) LENGTH: 641 BASE PAIRS
(B) TYPE: NUCLEIC ACID
(C) STRANDEDNESS: SINGLE
(D) TOPOLOGY: LINEAR

CA 02220988 l997-ll-l2 W O 96~9508 PCTIU',SI~08 (ii) MOLECULE TYPE: cDNA
(xi) SEQU~N~ DESCRIPTION: SEQ ID NO:l:
CCCGCCTGCT GCCCAACCTC A--l~-l~lGCT TACAGCTGCT GAl-l~-l~lGC TGTCAAACTC 60 AGGGGGAGAA TCACC~l~-l CCTAATTTTA ACCAGTACGT GAGGGACCAG GGCGCCATGA 120 CCGACCAGCT GAGCAGGCGG CAGATCCGCG AGTACr~CT CTACAGCAGG ACCAGTGGCA 180 AGCACGTGCA ~lCCCCGGG CGTCGCATCT CCGCCACCGC CGAGGACGGC AACAAGTTTG 240 CCAAGCTCAT AGTGGAGACG GACACGTTTG G~AGCCGGGT TCGCATCAAA GGGGCTGAGA 300 GCA~AGACTG ~L~l-l~ACG GAGATCGTGC TGGAGAA~ CTATACGGCC TTCCAGAACG 420 CCCGGCACGA GGG~-L w l-lC Al W l~-l-l~ CGCGGCAGGG GCGGCCCCGC CAGG--L-lCCC 480 GCAGCCGCCA GAACCAGCGC GAGGCCCACT TCATCAAGCG CCTCTACCAA Gw CCAGCTGC 540 C~-l-l'CCC~AA CCACGCCGAG AAGCAGAAGC AGTTCGAGTT TGT w GCTCC GCCCCCACCC 60O

(2) INFORMATION FOR SEQ ID NO:2:
(i) SEQu~N~ CHARACTERISTICS
(A) LENGTH: 212 AMINO ACIDS
(B) TYPE: AMINO ACID
(C) STRANDEDNESS:
(D) TOPOLOGY: LINEAR
(ii) MOLECULE TYPE: PROTEIN

W O 96/39508 PCT~US95/07108 (xi) SEQu~N~ DESCRIPTION: SEQ ID NO:2:
Arg Leu Leu Pro Asn Leu Thr Leu Cys Leu Gln Leu Leu Ile Leu Cys Cys Gln Thr Gln Gly Glu Asn His Pro Ser Pro Asn Phe Asn Gln Tyr Val Arg Asp Gln Gly Ala Met Thr Asp Gln Leu Ser Arg Arg Gln Ile Arg Glu Tyr Gln Leu Tyr Ser Arg Thr Ser Gly Lys His Val Gln Val Pro Gly Arg Arg Ile Ser Ala Thr Ala Glu Asp Gly Asn Lys Phe Ala Lys Leu Ile Val Glu Thr Asp Thr Phe Gly Ser Arg Val Arg Ile Lys Gly Ala Glu Ser Glu Lys Tyr Ile Cys Met Asn Lys Arg Gly Lys Leu Ile Gly Lys Pro Ser Gly Lys Ser Lys Asp Cys Val Phe Thr Glu Ile Val Leu Glu Asn Asn Tyr Thr Ala Phe Gln Asn Ala Arg His Glu Gly Trp Phe Met Val Phe Thr Arg Gln Gly Arg Phe Arg Gln Ala Ser Arg Ser Arg Gln Asn Gln Arg Glu Ala His Phe Ile Lys Arg Leu Tyr Gln Gly Gln Leu Pro Phe Pro Asn His Ala Glu Lys Gln Lys Gln Phe Glu Phe Val Gly Ser Ala Pro Thr Arg Arg Thr Lys Arg Thr Arg Arg Pro Gln Pro Leu Thr (2) INFORMATION FOR SEQ ID NO:3:
(i) SEQUENCE CHARACTERISTICS
(A) LENGTH: 32 BASE PAIRS
(B) TYPE: NUCLEIC ACID
(C) STRANDEDNESS: SINGLE
(D) TOPOLOGY: LINEAR
(ii) MOLECULE TYPE: Oligonucleotide (xi) SEQUENCE DESCRIPTION: SEQ ID NO:3:

(2) INFORMATION FOR SEQ ID NO:4:
(i) SEQUENCE CHARACTERISTICS

W O 96~9508 P ~ nUS95~07108 (A) LENGTH: 30 BASE PAIRS
(B) TYPE: NUCLEIC ACID
(C) STR~Nn~n~S: SINGLE
(D) TOPOLOGY: LINEAR
(ii) MOLECULE TYPE: Oligonucleotide (xi) SEQUENCE DESCRIPTION: SEQ ID NO:4:

(2) INFORMATION FOR SEQ ID NO:5 (i) SEQU~N~ CHARACTERISTICS
(A) LENGTH: 30 BASE PAIRS
(B) TYPE: NUCLEIC ACID
(C) STRANDEDNESS: SINGLE
(D) TOPOLOGY: LINEAR
(ii) MOLECULE TYPE: Oligonucleotide (xi) SEQUENCE DESCRIPTION: SEQ ID NO:5:

(2) INFORMATION FOR SEQ ID NO:6:
(i) SEQuhN~ CHARACTERISTICS
(A) LENGTH: 30 BASE PAIRS
(B) TYPE: NUCLEIC ACID
(C) STRANDEDNESS: SINGLE
(D) TOPOLOGY: LINEAR
(ii) MOLECULE TYPE: Oligonucleotide (xi) SEQUENCE DESCRIPTION: SEQ ID NO:6:

(2) INFORMATION FOR SEQ ID NO:7:
(i) SEQUENCE CHARACTERISTICS
(A) LENGTH: BASE PAIRS
(B) TYPE: NUCLEIC ACID
(C) STRAN~N~SS: SINGLE
(D) TOPOLOGY: LINEAR
(ii) MOLECULE TYPE: Oligonucleotide (xi) SEQUENCE DESCRIPTION: SEQ ID NO:7:

W O 96~9508 PCT/U'3S~7108 (2) INFORMATION FOR SEQ ID NO:8:
(i) SEQUENCE CHARACTERISTICS
(A) LENGTH: 30 BASE PAIRS
(B) TYPE: NUCLEIC ACID
(C) STRANDEDNESS: SINGLE
(D) TOPOLOGY: LINEAR
(ii) MOLECULE TYPE: Oligonucleotide (xi) SEQ~N~ DESCRIPTION: SEQ ID NO:8:
GATTTACTCG AGCGTGAGGG GCTG&GGCCG 30

Claims (20)

WHAT IS CLAIMED IS:
1. An isolated polynucleotide comprising a member selected from the group consisting of:
(a) a polynucleotide encoding the polypeptide comprising amino acid -21 to amino acid 191 as set forth in SEQ ID NO:2;
(b) a polynucleotide encoding the polypeptide comprising amino acid 1 to amino acid 191 as set forth in SEQ ID NO:2;
(c) a polynucleotide capable of hybridizing to and which is at least 70% identical to the polynucleotide of (a) or (b); and (d) a polynucleotide fragment of the polynucleotide of (a), (b) or (c).
2. The polynucleotide of Claim 1 wherein the polynucleotide is DNA.
3. The polynucleotide of Claim 1 comprising from nucleotide 65 to nucleotide 641 as set forth in SEQ ID
No:1.
4. An isolated polynucleotide comprising a member selected from the group consisting of:
(a) a polynucleotide encoding a mature polypeptide encoded by the DNA contained in ATCC Deposit No. 97147;
(b) a polynucleotide encoding the polypeptide expressed by the DNA contained in ATCC Deposit No. 97147;
(c) a polynucleotide capable of hybridizing to and which is at least 70% identical to the polynucleotide of (a) or (b); and (d) a polynucleotide fragment of the polynucleotide of (a), (b) or (c).
5. A vector containing the DNA of Claim 2.
6. A host cell genetically engineered with the vector of Claim 5.
7. A process for producing a polypeptide comprising:
expressing from the host cell of Claim 6 the polypeptide encoded by said DNA.
8. A process for producing cells capable of expressing a polypeptide comprising genetically engineering cells with the vector of Claim 5.
9. A polypeptide comprising a member selected from the group consisting of (i) a polypeptide having the deduced amino acid sequence of SEQ ID NO:2 and fragments, analogs and derivatives thereof; and (ii) a polypeptide encoded by the cDNA of ATCC Deposit No. 97147 and fragments, analogs and derivatives of said polypeptide.
10. An antibody against the polypeptide of claim 9.
11. A compound which inhibits the biological actions of the polypeptide of claim 9.
12. An agonist mimetic to the polypeptide of claim 9.
13. A method for the treatment of a patient having need of an FGF-13 polypeptide comprising: administering to the patient a therapeutically effective amount of the polypeptide of claim 9.
14. A method for the treatment of a patient having need to inhibit an FGF-13 polypeptide comprising: administering to the patient a therapeutically effective amount of the compound of claim 11.
15. The method of claim 13 wherein said therapeutically effective amount of said polypeptide is administered by providing to the patient DNA encoding said polypeptide and expressing said polypeptide in vivo.
16. The method of claim 14 wherein said compound is a polypeptide and a therapeutically effective amount of the compound is administered by providing to the patient DNA
encoding said antagonist and expressing said antagonist in vivo.
17. A process for identifying compounds active as agonists to the polypeptide of claim 9 comprising:
(a) combining a compound to be screened and a reaction mixture containing cells under conditions where the cells are normally stimulated by said polypeptide, said reaction mixture containing a label incorporated into the cells as they proliferate; and (b) determining the extent of proliferation of the cells to identify if the compound is an effective agonist.
18. A process for identifying compounds active as antagonists to the polypeptide of claim 9 comprising:
(a) combining a compound to be screened, the polypeptide and a reaction mixture containing cells under conditions where the cells are normally stimulated by said polypeptide, said reaction mixture containing a label incorporated into the cells as they proliferate; and (b) determining the extent of proliferation of the cells to identify if the compound is an effective antagonist.
19. A process for diagnosing a disease or a susceptibility to a disease related to an under-expression of the polypeptide of claim 9 comprising:

determining a mutation in the nucleic acid sequence encoding said polypeptide.
20. A diagnostic process comprising:
analyzing for the presence of the polypeptide of claim 9 in a sample derived from a host.
CA002220988A 1995-06-05 1995-06-05 Fibroblast growth factor 13 Abandoned CA2220988A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CA002220988A CA2220988A1 (en) 1995-06-05 1995-06-05 Fibroblast growth factor 13

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CA002220988A CA2220988A1 (en) 1995-06-05 1995-06-05 Fibroblast growth factor 13

Publications (1)

Publication Number Publication Date
CA2220988A1 true CA2220988A1 (en) 1996-12-16

Family

ID=4161763

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002220988A Abandoned CA2220988A1 (en) 1995-06-05 1995-06-05 Fibroblast growth factor 13

Country Status (1)

Country Link
CA (1) CA2220988A1 (en)

Similar Documents

Publication Publication Date Title
US5773252A (en) Fibroblast growth factor 15
US20070154908A1 (en) Connective Tissue Growth Factor-2
EP0873348A1 (en) Human vascular endothelial growth factor 3
AU702131B2 (en) Fibroblast growth factor-14
AU712297B2 (en) Fibroblast growth factor 11
US5728546A (en) Fibroblast growth factor 13
AU702682B2 (en) Fibroblast growth factor 13
WO1996039509A1 (en) Fibroblast growth factor 15
US20030022312A1 (en) Human hepatoma-derived growth factor-2
US7338934B2 (en) Fibroblast growth factor-14
AU748429B2 (en) Fibroblast growth factor 11
AU711647C (en) Fibroblast growth factor 15
CA2220988A1 (en) Fibroblast growth factor 13
AU752592B2 (en) Fibroblast growth factor 15
MXPA97006854A (en) Factor 14 of fibroblas growth
MXPA97007897A (en) Factor-11 fibroblas growth
KR19980703870A (en) Fibroblast Growth Factor-11
MXPA97009217A (en) Growth factor 13 of fibroblas
KR19980703677A (en) Fibroblast Growth Factor-14

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued