CA2430223A1 - Keratinocyte growth factor-2 - Google Patents

Keratinocyte growth factor-2 Download PDF

Info

Publication number
CA2430223A1
CA2430223A1 CA002430223A CA2430223A CA2430223A1 CA 2430223 A1 CA2430223 A1 CA 2430223A1 CA 002430223 A CA002430223 A CA 002430223A CA 2430223 A CA2430223 A CA 2430223A CA 2430223 A1 CA2430223 A1 CA 2430223A1
Authority
CA
Canada
Prior art keywords
antibody
fragment
protein
specifically binds
amino acid
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
CA002430223A
Other languages
French (fr)
Inventor
Joachim R. Gruber
Patrick J. Dillon
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
Human Genome Sciences, Inc.
Joachim R. Gruber
Patrick J. Dillon
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 Human Genome Sciences, Inc., Joachim R. Gruber, Patrick J. Dillon filed Critical Human Genome Sciences, Inc.
Priority claimed from CA002210444A external-priority patent/CA2210444C/en
Publication of CA2430223A1 publication Critical patent/CA2430223A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/50Fibroblast growth factors [FGF]

Abstract

A human polypeptide and DNA (RNA) encoding such polypeptide and procedure for producing such polypeptide by recombinant techniques is disclosed. Also disclosed are methods for utilizing such polypeptide for stimulating epithelial cell growth which may be used to stimulate wound healing, reduce scarring and prevent hair loss. Antagonists against such polypeptides and their use as a therapeutic to treat proliferative diseases such as cancer, psoriasis, Kaposi's sarcoma, keloids, retinopathy and restenosis are also disclosed. Diagnostic methods for detecting mutations in the KGF-2 coding sequence and alterations in the concentration of KGF-2 protein in a sample derived from a host are also disclosed.

Description

RERATINOCYTE GRO~Q"f8 FACTOR-2 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 is a Keratinocyte growth factor, sometimes hereinafter referred to as "KGF-2". The invention also relates to inhibiting the action of such polypeptides. ._ The fibroblast growth factor family has emerged as a large family of growth factors involved in soft-tissue growth and regeneration. It presently includes several members that share a varying degree of homology at the protein level, and that, with one exception, appear to have a similar broad mitogenic spectrum, i.e., they promote the proliferation of a variety of cells of mesodemial and neuroectodermal origin and/or promote angiogenesis.
The pattern of- expression of the different members of the family is very different, ranging from extremely restricted expressions of some stages of development, to rather ubiquitous expression in a variety of tissues and organs. All the members appear to~ bind heparin and heparin sulfate proteoglycans and glycosaminoglycans and strongly -la-concentrate in the extracellular matrix. KGF was originally identified as a member of the FGF family by sequence homology or factor purification and cloning. .
K.eratinocyte growth factor (KGF) was isolated as' a mitogen for a cultured marine keratinocyte line (Robin, J. S . , et al., PNAS, USA, 86:802-806 (1989)). Unlike the other members of the FGF family, it has little activity on mesenchyme-derived cells but stimulates the growth of epithelial cells. The Reratinocyte growth factor gene encodes a 194-amino acid polypeptide (Finch, P.W., et al., Science, 245:?52-X55 (1989)). The N-terminal 64 amino acids are unique, but the remainder of the proteia has about 30%
homology to bFGF. RGF is, the most divergent member of the FGF family. The molecule has a hydrophobic signal sequence and is - efficiently secreted. Post-translational modifications include cleavage of the signal sequence and N-linked glycosylation at one site, resulting in a protein of 28 kDa. Reratinocyte growth factor is produced by fibroblast derived from skin and fetal lung, (Rubin,~ et al. , (1989) ) . The Keratinocyte growth factor mRl~i was found, to be expressed in adult kidney, colon and ilium, but not in brain or lung (Finch, P.W., et al., Sc3.ence, 245:752-755 (1989) ) .
KGF displays the conserved regians within the FGF protein family. RGF binds to the FGF-Z receptor with high affinity.
Summary of tl~e Irwer~~ior~
It is an object of the present invention to provide keratinocyte growth factor-2. In accordance with an aspect of the present invention there is provided an isolated polynucleotide selected from the group consisting of a. a polynucleotide encoding the polypeptide having the deduced amino acid sequence of SEQ ID No. 2 or a fragment, analog or derivative of said polypeptide;
b. a polynucleotide encoding the polypeptide having the amino acid sequence encoded by the cDNA contained in ATCC Deposit No. 75977 or a fragment, analog or derivative of said polypeptide.

In accordance with another aspect of the present invention there is provided a polypeptide 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. 75977 and fragments, analogs and derivatives of said polypeptide.
In accordance with another aspect of the present invention there is provided a process for identifying compounds active as agonists to KGF-2 comprising:
a. combining a compound to be screened, and a reaction mixture containing cells under conditions where the cells are normally stimulated by KGF-2, 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.
The polypeptide of the present invention has been putatively identified as a member of the FGF family, more ' particularly the polypeptide has been putatively identified as KGF-2 as a result of amino acid sequence homology with other members of the FGF family.
In accordance with one aspect of the present invention, there are provided novel mature polypeptides which are KGF-2 ' as well as biologically act~.ve and diagnostically" or~
therapeutically useful fragments, analogs and derivatives thereof. The polypeptides of the present invention are of human origin.
-2a-In accordance with one aspect of the present invention, there are. provided isolated nucleic~acid molecules encoding human RGF-2, 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 another aspect of the present invention, there is provided a process for producing such w polypeptide by recombinant techniques through the use of recombinant vectors, such as cloning and expression plasmids useful as reagents in the rec~nbinant production of KGF-2 proteins, as well .as recombinant prokaryotic and/or eukaryotic host cells comprising a human KGF-2 nucleic acid sequence.
In accordance with yet a further aspect of the present invention, there is provided a process for utilizing such polypeptide, or polynucleotide encoding such polypeptide for therapeutic'purposes, for example, to stimulate epithelial cell proliferation for the purpose of wound healing, and to stimulate hair follicle production and healing of dermal wounds. .
In accordance with yet a further aspect of the present invention, there are provided antibodies against ~ such polypeptides. .' 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 human RGF-2 sequences.
In accordance with yet another aspect of the present invention, there are provided antagonists to such polypeptides, which may be used to inhibit the action of such polypeptides, for example, to reduce scarring during the wound healing process and to prevent and/or treat tumor proliferation, diabetic retinopathy, rheumatoid arthritis and tumor growth-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 RGF-2 nucleic acid sequences or over-expression of the polypeptides encoded by such sequences. -In accordance with another aspect of the present invention, there is provided a process fvr utilizing such polypeptides, or polynucleotides encod3.ng such polypeptides, for ire of tzn 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 from the teachings -herein.
The following drawings are illustrative of embodiments of the invention and are not meant to limit the scope of the invention as encompassed by the claims.
Figure 1 illustrates the cDNA and corresponding deduced amino acid sequence of the polypeptide of the present invention. The initial 36 amino acid .residues.represent the putative leader sequence (underlined). The standard one-letter abbreviations for amino acids are used. Sequencing inaccuracies are a common problem when attempting to -determine polynucleotide sequences. Sequencing was performed using a 373 Automated DNA sequencer (Applied Biosystems, Inc.). Sequencing accuracy is predicted to be greater than 97% accurate.
Figure 2 is an illustration of a comparison of the amino acid sequence of the pol~rpeptide of the present invention and other fibroblast growth factors.
In accordance with an aspect of the present invention, there is provided an isolated nucleic acid (polynucleotide) which encodes for the mature polypeptide having the deduced amino acid sequence of Figure 1 (S8~ ID No. 2? or for the mature polypeptide encoded by the cDNA of the clone deposited as ATCC Deposit No. ?5977 on December 16, 1994.
A polynucleotide encoding a polypeptide of the present invention may be obtained from a human prostate and fetal lung. A fragment of the eDNA encoding the polypeptide was initially isolated fram a library derived from a human normal prostate. The open reading frame encoding the full-length protein was subsequently isolated from a randomly primed human fetal lung-cDNA library. it is structurally related to the FGF family. It contains an open reading frame encoding a protein of 208 amino acid residues of which approximately the first 36 amino acid residues are the putative leader sequence such that the mature protein comprises 1T2 amino acids. The protein exhibits the. highest degree of homology to human keratinocyte growth factor with 45 % identity and 82 % similarity over a 206 amino acid stretch. It is also important that sequences that are conserved through the FGF
family are found to be consezved in the protein of the present invention.
The polynucleotide of the present invention may be in the form of RNA or in the foam of DNA, which DNA includes cDNA, genomic DNA, and synthetic DNA. The DNA may be double-stranded or single-stranded, and if single stranded may be the coding strand or non-coding (anti-sense) strand. The coding sequence which encodes the mature polypeptide may be identical to the coding sequence shown in Figure 1 (SFQ ID
No. 1) or that of the deposited clone or may be a different coding sequence which coding sequence, as a result of. the redundancy or degeneracy of the genetic code, encodes the same mature polypeptide as the DNA of Figure 1 (S8Q ID No. 1) or the deposited cDNA.
The polynucleotide which encodes for the mature polypeptide of Figure 1 (SBQ ID No. 2) or for the mature polypeptide encoded by the deposited cDNA 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 ar secretory sequence or a proprotein sequence; 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 polypeptide having the deduced amino acid sequence of Figure l (S8Q ID No. 2) or the polypeptide encoded by the cDNA of the deposited clone.
The variant 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 polynucieotides encoding the same mature polypeptide as shown in -Figure 1 (S8Q ID No. 2) or the same mature polypeptide encoded by the cDNA of the deposited clone as well as variants of such polynucleotides which variants encode for a fragment, derivative or analog of the polypept:ide of Figure 2 (SEQ ID
No. 2) or the polypeptide encoded by the cDNA of the deposited clone. Such nucleotide variants include deleta.on variants, substitution variants and addition or insertion variants .
As hereinabove indicated, the polynucleotide may have a coding sequence which is a naturally occurring allelic variant of the coding sequence shown in Figure 1 (SEQ TD No.
1) or of the coding sequence of the deposited clone. As known in the art, an allelic variant is an alternate forni of a polynucleotide sequence which may have a substitution, deletion or addition of one or more nucleotides, which does -s-not substantially alter the' funr_tion of the. encoded polypeptide.
The present invention also includes polynucleotides, wherein the coding sequence for the mature polypeptide 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, aleader sequence which functions as a secretory sequence for controlling transport of a polypeptide from the cell. The pol~rpeptide having a leader sequence is a preprotein and may have the leader sequence cleaved by the host cell to form the mature form of the polypeptide. The polynu"~cleotides may also encode far a proprotein which is the mature protein plus additional S' amino acid residues. A mature protein haviag a prosequence is a proprotein and is an inactive form of the protein. Once the prosequence is cleaved an active mature protein remains.
Thus, for example, the polynucleotide of the present invention may encode for a mature protein, or for a protein having a pros~equence or for a protein having both a prosequence and a presequenee (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 hexes.-histidine tag supplied by a pQB-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 I,HA) tag when a mammalian host, e.g. COS-7 cells, is used. The HA tag corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson, I., et al., Cell, 37:767 (1984)).
The present invention further relates 'to polynucleotides which hybridize to the hereinabove-described sequences if there is at least 50% and preferably 70%
identity between the sequences. The present invention particularly relates to polynucleotides which hybridize under '. stringent . conditions to the hereinabove-described polynucleotides. As herein used, the term "stringent conditions" means hybridization w:i.ll occur only if .there is at least 95 % and preferably at least 97 % identity'between the sequences: The polynucleotides which hybridize to the hereinabove described polynucleotides in a preferred embodiment encode polypeptides which retain substantially the same biological. function or activity as the mature polypeptide encoded .by the cDNA of Figure 1 (SEQ ID No. 1) or the deposited cDI~.
The deposits) referred to~herein will be maintained under the terns of t:he Budapest Treaty on the International Recognition of the Deposit of Micro-organisms for purposes of Patent Procedure. These deposits are provided merely as convenience to those of skill in the art and are not an admission that a deposit is required under S. 27{3) of thePaten~Act.
DNA Plasmid 366885A was deposited on December 16, 1994 with the American Type Culture Collection {ATCC), 1230T Parklawn Drive, Rockville, MD 20852, under accession number ATCC 75977.
The sequence of the polynucleotides contained in the deposited materials, as well as the amino acid sequence of .
the polypeptides~encoded thereby, are controlling, in the event of any conflict with any 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 a polypepti.de which has the deduced amino acid sequence of Figure 1 (S$Q ID
No. 2) or which has the amino acid sequence encoded by the deposited cDNA, as well as fragments, analogs and derivatives of such polypeptide The terms "fragment, " !'derivative" and '!analog" when referring to the polyp.eptide of Figure 1 (SEQ ID No. 2) or that encoded by the deposited cDNA, means a polypeptide which retains essentially the same biological function or activity as such polypeptide. Thus, an a~.~alog includes a proprotein -a -which can be activated by cleavage of the proproteiu portion to produce an active mature polypeptide.
The polypeptide of the present invention may be a recombinant polypeptide, a natural polypeptide or a synthetic polypeptide, preferably a recombinant polypeptide.
The fragrneat, derivative or analog of the polypeptide of Figure 1 (SBQ ID No. 2) or that encoded by the deposited cDNA may be ti) one in which one or more of the amino acid residues are substituted with a co~aserved 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 groug, or (iii) one in which the mature polypeptide is fused with another compound, such as a compound to increase the half-life of 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 ~of 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.
The term "isolated" means that the material is removed from its original environment (e. g., the natural environment if it is naturally occurring). For example, a naturally-occurring polynucleotide or polyppptide present in a living animal is not isolated, but the same polynucleotide or polypeptide, separated from some or all of the coexisting materials in the natural system, is isolated. Such polynucleotides could be part of a vector and/or such polynucleotides or polypeptides could be part of a _g_ composition, and still be isolated in that such vector or composition is not part of its natural environment.
The present invention also relates to vectors which include polynucleotides of the present invention, host cells which are genetica3ly engineered With vectors of the invention and the production of polypeptides of the invention by recombinant techniques.
Host cells are -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 KGF-2 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 polynucleotides of the present invention may be employed for producing polypep~ides by recombinant .techniques. Thus, for example, the polynucleotide may be included in any one of a variety of expression vectors for expressing a polypeptide. Such vectors include chromosomal, nonchromosomal and synthetic DNA sequences, e.g., derivatives of SV40; bacterial plasmids; phage DNA; baculovirus; 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 may be used as long as it is 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 )sown in the art. Such . 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 appropriate expression control sequences) (promoter) to direct mRNA synthesis. As representative examples of such promoters, there may be mentioned: LTR or SV40 promoter, the B. coli: ac or ~ pr , the phage lambda PL
promoter and other promoters known to control expression of genes in prokaryotic or eukaryotic cells or their viruses.
The expression vector also contaias 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 one or more selectable marker genes to provide a phenotypic trait for selection of transfornted host cells such as dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, or such as tetracycline or ampicillin resistance in .~. coli.
The vector containing the appropriate DNA sequence as hereinabove described, as well as an appropriate promoter or control sequence, may be employed to transform an appropriate host to pezmit the host to express the protein.
As representative examples of appropriate hosts, there may be mentioned: bacterial cells, such as F. coli, Strep_tonnvces, Salmonella typhimuri.um; fungal cells, such as yeast; insect cells such as Drosophila S2 and Snodogtera Sf9;
animal cells such as CHO, COS or Bowes melanoma;
adenvviruses; 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 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 sequence. barge numbers of suitable vectors and promoters are known to those of skill in the art, and are comzaercially available. The following vectors are provided by way of example; Bacterial: pQ$70, pQ$60, pQF-9 (Qiagen), pBS, pDlO, phagescript, psiX174, pbluescript SR, pbsks, pN88A, pNHl6a, pN~l8A, pN846A (Stratagene); ptrc99a, pKK223-3, gRR233-3, pDR540, pRIT5 (Phanaacia) ; B'uka~ryotic: pWLNSO, pSV2CAT, pOG44, pXTl, pSG (Stratagene) pSVK3, pBPV, pMSG, -pSVIs (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 (chloramphenicol transferase) vectors or other vectors with selectable markers. Two appropriate vectors are pKR232-8 and pCM7. Particular named bacterial promoters include lacI, lacZ, T3., T7, gpt, lambda PR, PL and trp.
Bukaryotic 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 3evel of ordinary skill in the art.
In a further embodiment, the present invention relates to host cells containing the above-described constructs. The host cell can be a higher eukaryotic cell, such as a mammalian 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, DEAR-Dextran mediated transfection, or electroporation (Davis, L., Dibner, M., Battey, I., Basic Methods in Molecular Biology, (1986) ) .

the constructs in host cells can be used in a conventional manner to produce the gene product encoded by the recombinant sequence. Alternatively, the polypeptides of the invention can be synthetically produced by conventional peptide synthesizers.
Mature proteins can be expressed in mammalian cells, yeast, bacteria, or other cells under the control of appropriate promoters. Celi-free translation systems can also_be employed to produce such proteins using RHAs derived from the DNA constructs of the present invention. ' Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrooh, et al., Molecular Cloning: A Laboratory Manual, Second 8dition, Cold Spring Harbor, N.Y., (1989)..
Transcription of the DNA encoding the polypeptides of the present invention by higher eukaryotes is increased by inserting an enhancer- sequence into the vector. Enhancers are cis-acting elements of DNA, usually about from 10 to 3 00 by that act on . a promoter to. i.n.crease its transcription.
Examples including the SV40 enhancer on the late side of the replication origin by 100 to 270, a cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovinis enhancers.
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 TRPl gene, and a promoter derived from a highly-expressed gene~to direct transcription of a downstream structural sequence. Such gromoters can be derived from operons encoding glycolytic enzymes such as 3-phosphoglycerate kinase (PGK), a-factor, acid phosphatase, or heat shack proteins, among others. Th.e heterologous structural sequence ~s assembled in appropriate phase ~rith translation initiation and termination sequences, and preferably, a leader sequence eapable of directing secretion of translated protein inta the periplasmic space or extracellular medium. Optionally, the heterologous sequence can encode a fusion protein including an N-texminal identi.ficativn peptide imparting desired characteristics, e.g., stabilization or simplified purification of expressed recombinant 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 R. coli, Bacillus subtilis, Salmonella typhiimirium and various species within the genera Pseudomonas, 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 GSM1 (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 induced by appropriate means (e. g., temperature shift or chemical induction) and cells are cultured for an additional period.

Cells are typically harvested by centrifugation, disrupted by physical or chemical iaeans, and the resulting crude extract retained 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 lysiag agents, such methods are well know to those skilled in the art.
Various mammalian cell culture systems can also be employed to express recombinant protein. ales of mammalian 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, far example, the 0127, 3T3, CHO, HeLa and BHK cell lines. Mammalian expression vectors will comprise an origin of replication, a suitable promoter and enhancer, and also any necessary ribos~ne - binding sites, polyadenylation site, splice donor and acceptor sites, transcriptional termination sequences, and 5' flanking nontranscribed sequences. DNA sequences derived from the, SV40 splice, and polyadenylation sites may be used to grovide the required nontranscribed genetic elements.
The KGF-2 polypeptide can be recovered and purified from recombinant cell cultures by methods including am~uonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite 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 polypeptides of the present invention may be a naturally purified product, or a product of chemical synthetic procedures, or produced by recombinant techniques from'~a' prokaryotic or eukaryotic host (for example, by bacterial, yeast, higher plant, insect and mammalian cells in culture . Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be ~glycosylated or may be non-glycosylated.
Polypeptides of the imrention may also include an initial methionine amino acid residue.
The polypegtide of the present invention may be employed to stimulate new blood vessel growth or angiogenesis.
Particularly, the polypeptide of the present invention may stimulate keratinocyte cell growth and proliferation.
Accordingly, the polypeptide of the present invention may be used to stimulate wound healing, and also to stimulate Reratinocytes which is related to the prevention of hair loss.
The polypeptide of the present invention may also be employed to heal dermal wounds by stimu:Lating epithelial cell proliferation.
The polypegtide of the present invention may also be employed to stimulate differentiation of cells, for example, muscle cells and nervous tissue, prostate cells and lung cells.
The signal sequence of RGF-2 encoding amino acids 1 through 36 may be employed to identify secreted proteins in general by hybridization and/or computational search algorithms.
The nucleotide sequence of RGF-2, could be employed to isolate S' sequences by hybridisation. Plasmids comprising the RGF-2 gene under the control of its native promoter/enhancer sequences could then be used in in vitro studies aimed at the identification of endogenous cellular and viral transactivators of RGF-2 gene expression.
The RGF-2 protein may also be employed as a positive control in experiments designed to identify peptido-mimetics acting upon the RGF-2 receptor.

In accordance with yet a further aspect of 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 )JNA vectors and for the purpose~of providing diagnostics and therapeutics for the treatment of human disease. ' Fragments of the full length R~GF-2 gene may be used as a hybridization probe for a cDNA library to isolate the full length KGF-2 genes and to isolate other genes which have a high sequence similarity to these genes or similar biological activity. Probes of this type generally have at least 20 bases. Preferably, however, the probes have at least 30 bases and generally do not exceed 50 bases, although they may have a greater number of 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 RGF-2 genes including regulatory and promotor regions, exons, and introns. An example of a screen comprises isolating the .coding region of the RGF--2 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.
This invention provides a method for identification of the receptors for the RGF-2 polypeptide. The gene encoding the receptor can be identified by numerous methods known to those of skill in the art, for e~cample, ligand panning and FAGS 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, 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 labeled polypeptides. The 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 autoradiographic 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 PAGF
analysis and exposed to x-ray film. The labeled complex containing the receptors of the polypeptides can be excised, resolved into peptide fragments, and subjected to protein microsequencing. The amino acid sequence obtained from microsequencing would be used to design a set of degenerate oligonucleotide probes to screen a cDI.~TA library to identify the genes encoding the putative receptors.
This invention provides a method of screening compounds to identify those which agonize the action of KGF-2 or block the function of KGF-2. An example of such an assay comprises combining'a mammalian Keratinocyte cell, the compound to be screened and '[H] thymidine under cell culture conditions where the keratinocyte 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 keratinocyte proliferation in the presence of the compound to determine if the compound stimulates proliferation of Keratinocytes.
To screen for antagonists, the same assay may be prepared in the presence of KGF-2 and the ability of the compound to prevent Keratinocyte proliferation is measured _18_ and a determination of antagonist: ability is made. The amount of Keratinocyte cell proliferation is measured by liquid scintillation chromatography which measures the incorporation of 3i81 thymidine.
In another method, a mammalian cell or membrane preparation expressing the KGF-2 receptor would be incubated with labeled KGF-2 in the presence of the compound. The ability of the compound to enhance or block this interaction could then be measured. Alternatively, the response of a known second messenger system following interaction of KGF-2 -and receptor would be measured and compared in the presence or absence of the compound. Such second messenger systems .
include but are not limited to, cPW guanyiate cycla.se, ion channels or phosphoinositide hydrolysis.
$xamples of potential RGF-2 antagonists include an antibody, or in some cases, an oligonucleotide, which binds to the polypeptide. Alternative3y, a potential KGF-2 antagonist may be a mutant fozm of KGF-Z which binds to KGF-2 receptors, however, no second messenger response is elicited and therefore the action of KGF-2 is effectively blocked.
Another potential KGF-2 antagonist is an antisense construct prepared using antisense technology. Antisense __ technology can be used to control gene expression throu3h 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 aligonucleotide is designed 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 a3., Science, 251: 1360 (1991)), thereby preventing transcription and the production of KGF-2. The _Zg_ antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into~KGF-2 polypeptide (Antisense - Okano, J. Neurochem., 56:560 11991);
Oligodeoxynucleotides as Antisense Inhibitors of Gene $xpression, 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 inhibit production of KGF-2.
Potential KGF-2 antagonists include small molecules which bind to and occupy the bindiug site of the RGF-2 receptor thereby making the receptor inaccessible to KGF-2 such that normal biological activity is prevented. 8xamples of small molecules include but are not limited to small peptides or peptide-like molecules.
The KGF-2 antagonists may be employed to prevent the induction of new blood vessel growth or angiogenesis in tumors. Angiogenesis stimulated by KGF-2 also contributes to several pathologies which may also be treated by the antagonists of the present invention, including diabetic retinopathy, and inhibition of the growth of pathological ' tissues, such as in rheumatoid arthritis.
KGF-2 antagonists may also be employed to treat glomerulonephritis, which is characterized by the marked proliferation of giomerular epithelial cells which form a cellular mass filling Bowman~s space.
The antagonists may also be employed to inhibit the over-production of scar tissue seen in keloid formation after surgery, fibrosis after myocardial infarction or fibrotic lesions associated with pulmonary fibrosis and restenosis.
KGF-2 antagonists may also be employed to treat other proliferative diseases which are stimulated by KGF-2', including cancer and Kaposi's sarcoma.
KGF-2 antagonists may also be employed to treat keratrtis which is a chronic infiltration of the deep layers of the cornea with uveal inflamoaation characterised by epithelial cell prolifezation.
The antagonists may be employed in a composition With a pharmaceutically acceptable carrier, e.g., as hereinafter described.
The polypeptides, agonists rind antagonists of the present invention may be employed in combination with a suitab~:e pharmaceutical carrier to comprise a pharmaceutical composition. Such c~npositions coaaprise 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 c~ administration. .
The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of. the invention. Associated with such containers) can be a notice 'in the form prescribed by a govezumental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of .
manufacture, use or sale for human administration, In addition, the polygeptides, agonists and antagonists of the present invention may be employed in conjunction with other therapeutic compounds.
The pharmaceutical compositions may be administered in a convenient manner such as by the oral; topical, intravenous, intraperitoneal, intramuscular, subcutaneous, intranasal or intradermal routes. The pharmaceutical compositions are administered in an amount which is effective for treating and/or prophylaxis of the specific indication.
In general, they are administered in an amount of at least about 10 ~g/kg body weight and in mast cases they will be administered in an. amount not in excess of about 8 mg/Kg body weight per day. In most cases, the dosage is from about -10.
.t ~g/kg to about 1 mg/kg body weight daily, taking into account the routes of administration, symptoms, etc. In the specifis case of topical administration dosages are preferably administered from about 0.1 ~Cg to 9 mg per cni .
The KGF-2 polypeptides, agonists and antagonists which are polypeptides may also be employed in accordance with the present iuveation by expression of such polypeptides in vivo, which is oftea referred to as gene therapy."
Thus, for- example, cells from a patient may be engineered with a polynucleotide tDWA or RNA) encoding a polypeptide ex vivo; with the engineered cells then being provided to a patient to be treated with the polypeptide.
Such methods are well-kaown in the art. For example, cells may be engineered by procedures known in the art by use of a retroviral particle containing RNA encoding a polypeptide of the present invention.
Similarly, cells may be engineered an vivo for expression of a polypeptide in vivo by, for example, procedures known in the art. As known in the art, a producer cell for producing a retroviral particle containing RNA
.encoding the polypeptide of the present invention may be __ administered to a patient for engineering cells .in vivo and expression of the polypeptide in ~ivo. These and other methods for administering a polypeptide of the present invention by such method 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 retrovirus, for example,, an adenovirus which may be used to engineer cells in vivo after combination with a suitable delivery vehicle. Examples of other delivery vehicles include an HSV-based vector system, adeno-associated virus vectors, and inert vehicles, for example, dextran coated ferrite particles.

This invention is.also related to the use of the KGF-2 gene as part of a diagnostic assay for detecting diseases or susceptibility to diseases related to the presence of mutations in the KGF-2 nucleic acid sequences:
Individuals carrying mutations in the KGF-2 gene~may be detected at the DNA level by a variety of techniques.
Nucleic acids for diagnosis may be obtained from'a patients cel7Ls, such as from.biood, urine, saliva, tissue biopsy and autopsy material. The genomic DNA may be used directly for detection or tray be amplified enzymatically by using PCR
(Saiki et al., Nature,.324:163-166 (X986)? prior to analysis.
RNA or cDNA may also be used for the same purpose. As an example,, PCR primers complementary to the nucleic acid encoding KGF-2 can be used to identify and analyze KGF-2 mutations. For example, deletions and insertions can be detected by a change in size of the amplified product in comparison to the normal genotype. Point mutations can be identified by hybridizing amplified DNA to radiolabeled RGF-2 RNA or alternatively, radiolabeled KGF-2 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 el:ectrophoretic 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 .__.._..__.~.~n.-~~"~~N.~~"~-_..__. _.,.~.,-protection 4r the chemical'cleavage method (e.g., Cotton et al., PNAS, iTSA, $5:439'7-4401 (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 wse of restriction enzymes, (e. g., Restriction Fragment Length Polymoxphisms (RFLPy) and Southern blotting of genomic DNA.
In~ addition to snore conventional gel-electrophoresis and DNA sequencing, mutations can also be detected by in situ analysis.
The ~ gresent invention also relates to a diagnostic assay for detecting altered levels of RGF-2 protein in various tissues since an over-expression of the proteins compared to no~aal control tissue samples may detect the presence of a disease or susceptibility to a disease, for example, a tumor.
Assays used to detect levels of RGF-~2 protein in a sample derived from a host are well-known to those of skill in the art and include ~adioimmunoassays, competitive-binding assays, Western Blot analysis, SLISA assays and "sandwich"
assay. An BLISA assay (Coligan, et al.., Current Protocols in Immunology, 1(2), Chapter 6, (1991)) initially comprises preparing an antibody specific to the RGF-2 antigen, 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 binding sites on the dish are then covered by incubating with a non-specific grotein like bovine serum albumen. Next, the monoclonal antibody is incubated in the dish during which time the monoclonal antibodies attach to any RGF-2 proteins 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 RGF-2.
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 RGF-2 protein present in a given volume of patient sample when compared against a standard curve.
A competition assay may be employed wherein antibodies specific to KGF-2 are attached to a solid support and labeled RGF-2 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, caa be correlated to a quantity of RGF-2 in the sample.
A "sandwich" assay is similar to an BhISA assay. In a "sandwich" assay RGF-2 is passed over a solid support and binds to antibody attached to a solid support. A second antibody is then bound to the RGF-2. A third antibody which is labeled and specific to the second antibody is then passed over the solid support and binds t:o 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 s~gecifical~.y 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 polymorphisms) are presently available for 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 containing individual human chromosomes. Only those hybrids containing the humaa gene corresponding to the primer will yield an amplified fragment.
PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular DNA toga particular chromosome.
Using t-he present invention with the same oligonucleotide primers, sublocalization can be achieved with panels of fragments from specific chromosomes or' pools of large genomic clones in an analogous manner. Other uaapping strategies that can similarly be used to map to its chromosome include in si to hybridization, prescreening with labeled flaw-sorted chromosomes and preselection by hybridization to construct chromosome specific-cDNA libraries.
Fluorescence in situ hybridization (FISH) of a cDHA
clone to a metaphase chromosomal spread can be used to provide a precise chromosomal location in one steg. This technique can be used with cDNA as short as 500 or 600 bases;
however, clones larger than 2, 000 by have a higher likelihood of binding to a unique chromosomal location with sufficient signal intensity for simple detection. FISH requires use of the clones from Which the EST was derived, and the longer the better. For example, 2,000 by is gaod, 4,000 is better, and more than 4, 000 is probably not necessary to get good results a reasonable percentage of the time. For a review of this technique, see Verina et al., Human Chromosomes: a Manual of Basic Techniquese Pergamon Press, New York (I988).
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 Library). The relationship between genes and diseases that have been mapped to the same chromosomal region are then identified through linkage analysis (coinheritance 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 m~utatian is obsexved in some or all of the offected individuals but not in any noxmal 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 eDNA precisely localized to a chromosomal region associated with the disease.could be one of between 50 and 500 potential causative genes. (This assumes 1 megabase mapping resolution and one gene per 20 kb) .
The polypeptides, their fragments or other derivatives, or analogs thereof, or cells expressing them can be used as an immunogen to produce antibodZ.es thereto. These antibodies can be, for example, polyclonal or monoclonal antibodies.
The present invention also includes chimeric, single chain, and humanized antibodies, as well as Fab fragcaents, or the product of an Fab expression library. Various procedures known in the art may be used for the production of such antibodies and fragments.
Antibodies generated against the polypeptides corresponding to a sequence of the present invention can be obtained by direct injection of the polypeptides into an animal or by administering the polypeptides to an animal, preferably a nonhuman. The antibody so obtained will then bind the polypeptides itself. In this manner, even a sequence encoding only a fragment of the polypeptides can be used to generate antibodies binding the whole native polypeptides. Such antiboda.es 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 8-cell hybridoma technique (Kozbor et al. , 1983, It~aaunology Today 4: 72) , aad the 8BV-hybridoma technique to produce human monoclonal antibodies (Cole, et al., 1985, in Monoclonal .Antibodies and Cancer Therapy;, A3an 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 immunogenic polypeptide products of this invention: .Also, transgenic mice may be used to express humanized antibodies to iu~nunogenic 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 understanding 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. Tn addition, equivalent glasmids 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 availab7.e and their reaction conditions, cofactors and other requirements were used as ' would be known to the ordinarily skilled artisan. For analytical purposes, typically 1 ~Cg of plasmid or DNA
fragment is used with about 2 units of enzyme in about 20 ~C1 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 eazyme 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 accordaace with the supplier's instructions. After digestion the reaction is electrophoresed directly on a polyacrylami.de gel to isolate the desired fragment.
Size separation of the cleav~:d 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 stranded 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 a1_, Id., p. I46). Unless otherwise provided, ligation may be accomplished using known buffers and conditions With 30 units of T4 DNA ligase ("ligase") per 0.5 ~.g of approximately equimolar amounts of the DNA fragments to be ligated.
A cel3 has been ~'transfornied" by exogenous DNA when scich exogenous DNA has been introduced inside the cell membrane.
Exogenous DISA may or may not be integrated (covalently linked) inter-chromosomal DNA making the genome of the cell.
Prokaryote and yeast, fvr example, the exogenous DNA may be maintained on an episomal element, such a plasmid. With respect to eukaryotic cells, a stably transfoztned or transfected cell is one in which the exogenous DNA has-become integrated into the chromosome so that it is inherited by daughter cells through chr~aosome replication. This ability is demonstrated by the ability of the eukaryotic cell to establish cell lines or clones comprised of a population of daughter cell containing the exogenous DNA. An ale of - transformation is exhibited in Graham, F. and Van der Eb, A., Virology, 52:456-457 (I973).
"Transduction" or "transduced" refers to a process by which cells take up foreign DNA and integrate that foreign DNA into their chromosome. Transduction can be accomplished, for example, by transfection, which refers to various techniques by which cells take up DNA, or infection, by which viruses are used to transfer DNA into cells.
- 8xample Z
Bacterial $xpression and Purification of RGF-2 The DNA sequence encoding RGF-2, ATCC # 75977, is initially amplified using PCR ol.igonucleotide primers corresponding to the 5' and 3' end sequences of the processed KGF-2 cDNA (including the signal peptide sequence). The 5' oligonucleotide primer has the sequence 5' CCCCACATGTGGAAATGGATACTGACACATTGTGCC 3' (SFQ ID. No. 3) contains an Afl III restriction enzyme site including and followed by 30 nucleotides of KGF-2 coding sequence starting from the presumed initiation codon..' The 3' sequence 5' CCCAAGCTTCCACA.AAC,GTTGCCTTCCTCTATGAG 3 ' ( SEQ ID No . 4 ) contains complementary sequences to Hind III site andwis followed by 26 nucleotides of KGF-2. The restriction enzyme sites are compatible with the restriction enzyme sites on the bacterial expression vector pQE-60 (Qiagen, Inc. Chatswor~h, CA) . gQ$-60 encodes antibiotic resistance (A~p'~ , a bacterial origin of replication ~(ori), an IPTG-regulatable promoter operator (P/o) , a ribosome binding site (RBS) , a 6-Fiis tag and restriction enzyme sites. pQS-60 is then digested with Ncol and HindIII. The amplified sequences are ligated into p~8-60 and are inserted in frame. The ligation mixture is then used to transform ~. coli strain M15/rep 4 (Qiagen, Inc.) by the procedure' described in Sambrook, J: et al., Molecular Cloning: A haboratory Manual, Cbld~ Sgring habOratory Press, (1989). MIS/rep4 contains multiple copies of the plastaid pREP4, which expresses the lacI repressor and also confers kanamycin resistance (Kanr) . Transfoximants are identified by their ability to grow on h8 plates and ampicillin/kanamycin resistant colonies are selected.
Plasmid D1~ is isolated and confirmed by restriction analysis. Clones containing the desired constructs are grown overnight (0/N) in liquid culture in LB media supplemented with both Amp (loo ug/ml) and Kan (25 ug/m1). The O/N
culture is used to inoculate a large culture at a ratio of 1:100 to 3:250. The cells are grown to an optical density 600 (0.D.6°°) of between 0.4 and 0.6. IPTG ("Isopropyl-B-D-thiogalacto. pyranosiden) is then added to a final concentration of Z mM. IPTG induces by inactivating the lacl repressor, clearing the P/0 leading to increased gene expression. Cells are grown an extra 3 to 4 hours. Cells ate then harvested by centrifugation. The cell pellet is solubilized in the chaotropi.c agent 6 Molar Guanidine HCl.
After clarification, solubilized KGF-2 is purified from this solution by chromatography on a Heparin affinity column under conditions that allow for tight binding of the proteins (Hochuli, 8. et al., J. Chromatography 411:177-184 (1984)).
KGF-2 ( 75 % pure) is eluted from the column by high salt buffer.

Example 2 Bacterial ression aiid Purification of a truncated version of RGF-2 The DNA sequence encoding KGF-2, ATCC # 75977, is initially amplified using PCR oligonucleotide primers corresponding to the 5' and 3' sequences of the truncated version of the RGF-2 polypeptide. The truncated version comprises the polypeptide minus the S6 amino acid signal sequence, with a methionine and alanine residue being added just before the cysteine residue which comprises amino acid 3? of the full-length protein. The 5' oligonucleotide primer has the sequence 5' CATGCCATGGCGTGCC~A.GCCCTTGGTCAGGACATG 3' (SEQ ID No. 5) contains an Ncol restriction enzyme site including and followed by 24 nucleotides of RGF-2 coding sequence. The 3' sequence S' CCCAAGCTTCCACAAACGTTGCCTTCCTC
TATGAG 3' (SEQ ID No. 6) contains complementary seguences to Hind III site and is followed by 26 nucleotides of the KGF-2 gene. The restriction enzyme sites axe compatible with the restriction enzyme sites on the bacterial expression vector pQE-60 (Qiagen, Inc. Chatsworth, CA). ,pQE-60 encodes antibiotic resistance (Amp'), a bacterial origin of replication (ori), an IPTG-regulatable promoter operator (P/0) , a ribosome binding site (RFtS) , a 6-Hi.s tag and restriction enzyme sites. pQE-60 is then digested with Ncol and HindIII. The amplified sequences are ligated into pQE-60 and are inserted in frame. The ligation mixture is then used to transform 8. coli strain Mi5/rep 4 (Qiagen, Inc.) by the procedure described in Sambrook, J. et al., Molecular Cloning: A Laboratory Manual, Cold Spring Laboratory Press, (1989). M15/rep4 contains multiple copies of the plasmid pREP4, which expresses the lacI repressor and also confers kanamycin resistance (Kan') . Transformants are identified,- by their ability to grow on LB plates and ampicillin/kanamycin resistant colonies are selected. Plasmid DNA is isolated and confirmed by restriction analysis. Clones containing the desired constructs are grown overnight (0/N) -in liquid culture in LB media supplemented~with both Amp (1o0 ug/ml) and Ran (25 ug/ml). The O/N culture is used to inoculate a large culture at a ratio of 1:140 to 1:250. The cells are grown to an optical density 600 (0.D.6°") 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 KGF-2 is purified from this solution by chromatography on a Heparin affinity column under conditions that allow for tight binding the proteins (Hochuli, E. et al.; J. Chromatography 411:177-184 (1984)). RGF-2 protein is eluted from. the column by high salt buffer.
Example 3 Clonin and cession of RGF-2 usin the baculovirus ex»ression system The DNA sequence encoding the full length KGF-2 protein, ATCC # 75977, is amplified using PCR oligonucleotide primers corresponding to the 5' and 3' sequences of the gene:
The 5' primer has the sequence 5' GCGGGATCCGCCATCAT'GTGGAAATGGATACTCAC 3' (S$Q ID No. 7) and contains a BamHI restriction enzyme site (in bold) followed by 6 nucleotides resembling an efficient signal for the initiation of translation in eukaryotic cells (Roaak, M., J.
Mol. Biol . , 196 : 947-950 (1.987? . and just behind the first 17 nucleotides of the KGF-2 gene (the initiation codon for translation ~~ATG" is underlined).
The 3' primer has the sequence 5' GCGCGGTACCACAAACGTTGCCTTCCT 3' (SBQ ID No. 8) and contains the cleavage site for the restriction endonuclease Asp718 and 19 nucleotides complementary to the 3' non-translated sequence of the KGF-2 gene. The amplified sequences are isolated from a 1% agarose gel using a commercially available kit from Qiagen, Inc., Chatsworth, CA. The fragment is then digested with the endonucleases BamHI and Asp~l8 and then purified again on a 1% agarose gel. 'This fragment is designated F2.
The vector pAx (modification of p~Th941 vector, discussed below) is used for the expression of the RGF-2 protein using the baculovirus expression system (for review see: Summers, M.D. and Smith, G.E. 1987, A manual of methods for baculovirus vectors and insect cell culture procedures, Texas Agricultural Experimental Station Bulletin No. 1555). This expression vector contains the strong polyhedrin promoter of the Autographs californica nuclear polyhidrosis virus (AcMNPV) followed by the recognition sites for the restriction endonucleases BamHI and Asp718. The polyadenylation site of the simian virus (SV)40 is used for efficient polyadenylation. For an easy selection of recombinant viruses 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-trarisfected wild-type viral DNA. Many other baculovirus vectors could be used in place of pRGl such as pAc373, pVL941 and pAcIMi (Luckow, V.A. and Suna~ers, M.D., Virology, 170:31-39).
The plasrnid is digested with the restriction enzymes BamHI and Asp718. The DNA is then isolated from a 1% agarose gel using the commercially available kit (Qiagen, Inc., Chatsworth, CA?. This vector DNA is designated V2.
Fragment F2 and the piasmid V2 are ligated with T4 DNA
ligase . E . coli HBi01 cells are then transfozmed and bacteria identified that contained the plasmid (pBacKGF-2) with the KGF-2 gene using PCR with both cloning oligonucleotides. The sequence of the cloned fragment is confixmed ~by DNA
sequencing.
beg of the plasmid pBacKGF-2 is co-transfected~with 1.0 ~g of a commercially available linearized' bacalovirus ("BaculoGold'" baculovirus DNA", Phar<ni.ngen, San Diego, CA. ) using the lipofection method (Felgner et al..Proc. Natl.
Acad. Sci. I1SA, 84 : ?413-?41? (198?) ) .
leg of BaculoGold°' virus DNA and 5 ~g of the plasmid pBacKGF-2 are mixed in a sterile well of a microtiter plate containing 50 gel of serum free Grace's medium (Life Technologies Inc., Gaithersburg, MD). Afterwards 10 ~C1 Lipof ectin~ plus 90 ~Cl Grace' s medium are added, mixed and incubated for 15 uzinutes at room. temperature . Then the transfection mixture is added drop-wise to the Sf9 insect cells tATCC CRL 1711) seeded in a 35 mttt tissue culture plate with Z ml Grace's medium without serum. The plate is rocked back and forth to mix~the newly added solution. The plate is then incubated for 5 hours at 27°C. After 5 hours the transfection salution is removed from the plate and 1 ml of Grace's insect medium supplemented with 10% fetal calf serum is added. The plate is put ba~..k into an incubator and cultivation continued at 27°C for four days.
After four days the supernatant is collected and a plaque assay performed sim3.lar 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 "plaque assay" can also be found a.n the user's guide for insect cell culture and baculovirology distributed by Life Technologies Inc., Gaithersburg, page 9-10) .
Four days after the serial dilution, the viruses are added to the cells and blue stained plaques are picked with the tip of an Bppendorf pipette. The agar containing the recombinant viruses is then resuspended in an Bppendorf tube containing 200 ~Cl of Grace s medium. The agar is~ removed ~by a brief centrifugation and the supernatant containing 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 recombinant baculovirus V-RGF-2 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 ~xCi of 35S-methionine and S ~xCi ~S cysteine (Amersham) are added. The cells are further incubated for 16 hours before they are harvested by centrifugation and the labelled proteins visualized by SD&-PAGE and autoradiography.
Example 4 Expression of Recombinant KGF-2 in COS cells The expression of plasmid, KGF-2 HA is derived from a vector pcDNAI/Amp (Invitrogen) containing: 1) SV40 origin of replication, 2) ampici.llin resistance gene, 3) E.coli replication origin, 4) CMV gromoter followed by a polylinker region, a SV40 intron and polyadenylation site. A DNA
fragment encoding the entire RGF-2 precursor and a HA tag fused in frame to its 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 correspond 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, L984, Cell 37:'76'7, (1.984) ) . The infusion of IiA tag to the target protein allows easy detection of the recombinant protein with an antibody that recognizes the HA epitope.

The plasmid construction strategy is described as follows:
The DNA sequence encoding KGF-2, ATCC # 75977, is constructed by PCR using two primers: the 5' primer S' CCCAAGCTTATGThGAAATGGATACTGACACATTGTGCC 3' (SEQ ID .No. 9) contains a Hind III site followed by 30 nucleotides of KGF-2 coding sequence starting from the initiation codon; the.3' sequence 5' TGCTCTAGACTAAGCGTAGT'C'PGC~GACGTCGTATC,GGTATGAGTG
TACCACCATTG~AAGAAAGTGAGG 3' (SBQ ID .No. 10) contains complementary sequences to an I~aaI site, translation stog codon, DA tag and the last 32 nucleotides of the RGF-2 coding sequence (not including the stop codon). Therefore, the PCR
product contains a Ni:nd III site, RGF-2 coding sequence followed by HA tag fused in frame, a translation termination stop codon next to the HA tag, and an Xhal site . The PCR
amplified DNA fragment and the vector, pcDNAI/Amp, are digested with Hind III and Xba I restriction enzyme and ligated. The ligation mixture is transformed into fi. coli strain XL1 Blue (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 examined by PCR and restriction analysis for the presence of the correct fragment. For expression of the recombinant KGF-2, C4S cells are transfected with the expression vector by DEAF-DBXTRAN method (J. Sambrook, B. Fritsch, T. Maniatis, Molecular Cloning: A
Laboratory Manual, Cold Spring Laboratory Press, (1989)).
The expression of the I~GF-2 IiA protein is detected by radiolabelling and inanunoprecipitation method (E. Harlow, D.
' Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, (1988)). Cells are labelled for 8 hours with ~S-cysteine two days post transfection. Culture media are 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 ?.5) (Wilson, I. et al., Id. 37:767 (1984)).

Both cell lysate and culture media are precipitated with a ~iA
_ specific monoclonal antibody. Proteins precipitated are analyzed on 15% SDS-PAGIs gels.
Example 5 Transcription and translation of recombinant RGF-2 in vitro:
A PCR product is derived from the cloned cDNA in the pA2 vector used for insect cell expression of RGF-2. The grimers used for this PCR were:
5' ATTAACCCTCACTAAAGGGAGGCCA'fGTGC~AATGGATACTGACACATTGTGCC 3~
tSEQ ID No . 11 ) and 5' CCCAA.GCTTCCACAAACGTTGCCTTCCTCTATGAG 3 (SEQ ID No. 12).
The first primer contains the sequence of a T3 promoter 5' to the ATG initiation codon. The second primer is complimentary to the 3' end of the RGF-Z open reading frame, and encodes the reverse complement of a stop codon.
The resulting PCR product is purified using a commercially available kit from Qiagen. 0.5 ~Cg of this DNA
is used as a template for an in vitro transcription-translation reaction. The reaction is performed with a kit commercially available from Promega under the name of TNT.
The assay is performed as described in the instructions for the kit, using radioactively labe7:ed methionine as a substrate, with the exception that only 1/2 of the indicated volumes of reagents are used and that the reaction is allowed to proceed at 33°C for 1.5 hours.
Five ~l of the reaction is electrophoretically separated on a denaturing 10 to 15% polyacrylamide gel. The gel is fixed for 30 minutes in a mixture of water: Methanol: Acetic acid at 6:3:1 volumes respectively. The gel is then dried under heat and vacuum and subsequently exposed to wn X-ray film for 16 hours. The film is developed showing the presence of a radioactive protein band corresponding in size to the conceptually translated RGF-2, strongly suggesting _3g_ -..

_ that the cloned ~cDl~tA for KGF-2 contains an open reading frame that codes for a protein of the expected size.
Numerous modifications and variations of the present invention are possible in light of the above teachings and, therefore, within the scope of the appended claims, the invention may be practiced otherwise than as particularly described.

SgQ~~ LISTING
- (1) GgNERAL INFORMATION:
(i) APPLICANT: GRUH$R, BT AL.
(~1) TI~$ OF IION~ Kerat:inocyte Gro~h Factor-2 (iii) N~$R OF SEQUENCES: 12 (iv) CORRLSPONDENCS ADDR$SS:
(A) ADDRESSEE: CARBLLA, BYRtU3. SIN, GILFILLAN, ~~I ~ STgyIART & OLSTEIN
($) STgggT: 6 BBCKER FARM ROAD
(C1 CITY: ROSSLAND .
(D) STATE: N$W JERSEY
(g) COUNTRY: USA
(F) ZIP: 07068 .
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: 3.5 INCH'DISI~TTB
~~R: IHM PS/2TM
( C ) OPERATING SYSTEM : MS -DOS T'"t (D) SOFTWARE: WORD PERFECT 5.1~
( ) APPLICATION DATA:
(A) APPLICATION Nt3MH8R:
(g) FILING DATE: Concurrently (C) CLASSIFICATION:
(vi.i) PRIOR APpLI~TxON DATA
(A) APPLICATION NUMEER:
(g) FILING DATE:

(viii). ATTORNEY/AGENT INFORMATION:
- (A) NAME: FERRARO, GREGORY D.
(B) REGISTRATION NUMB$R: 36,134 (C) REFBRENCE/DOCKET NUMBER: 325840-261 (ix) TBLECOMMUNICATiON INFORMATION:
(A) TELEPHONE: 201-994-1700 (B) TBLEFAX: 201-994-1744 (2) INFORMATION FOR SEQ ID N0:1:
(i) SEQDENCE CHARACTERISTICS
(A) LENGTH: 627 BASE PAIRS
(B) TYPE: NUCLEIC ACID
(C) STRANDEDNBSS: SINGLE
(D) TOPOLOGY: LINEAR
ii ) MOLBCDLE TYPfi : _ cDNA
(xi) SEQUENCE DESCRIPTION: SEQ~ID NO:1:
ATGTGGAAAT GGATACTGAC ACATTl3TGCC TCAGCCTTTC CCCACCTGCC60 CGGCTGCTGC

CCAAGCCCIT

GGTCAGGACA TGGTGTCACC AGAGGCCACC AACTCTTCT'T CCTCCTCCTT180 CTCCTCTCCT

CCGCTGGAGA

AAGCTATTCT CTT!'CACCRA GTACTTTCTC AAGATTGAG,A AGAACGGG1~A300 GGTCAGCGGG

fiATCGGAGTT

GGGGAAACTC

GGAAAATGGA

TGTGGCATTG

CTCTGCTCAC

(2) INFORMATION FOR SBQ ID N0:2:
(i) SEQUSNC$ CF~ARACTERISTICS
(A) LENGTH : 2 08 AMINO ACIDS
(B) TYPE: AMINO ACID
C) STRANDBDNESS
(D) TOPOLOGY: LINEAR
(ii) MOL$COLB TYPE: PROTEIN
(xi.) SBQIIFsNCE DBS~tIP~'ION: SEQ ID N0:2:
Met Trp Lys Trp Ile Leu Thr His Cys Ala Ser Ala Phe Pro His Leu Pro Gly Cys Cys Cys Cys Cys Phe Leu Leu Leu Phe Leu Val Ser Ser Val Pro Val Thr Cys Gln Ala Leu Gly Gln Asp Met Val Ser Pro Glu Ala Thr Asn Ser SerSer Ser Ser Phe Ser Ser Pro Ser Ser Ala Gly Arg His Val Arg;SerTyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu PheSer Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys ValSer Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile?'hrSer Val Glu .IleGly Val Val Ala Val Lys Ala Ile Asn SerAsn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly SerLys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu GluAsn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn GlyArg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly.Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala Hi.s Phe Leu Pro Met val Val His Ser (2) INFORMATION FOR SEQ ID N0:3:
(i) SEQUENCE CHARACTERISTICS
(A) LENGTH: 36 BASE PAIRS
(B) TYPE: NUCLEIC ACID
(C) STRANDEDN'FsSS : SINGLE
(D) TOPOLOGY: LINEAR
(ii) MOLECULE TYPE: 0ligonucleotide (xi) SEQUENCE DESCRIPTION: SgQ ID N0:3:

(2) INFORMATION FOR SEQ ID N0:4:
(i) SEQUENCE CHARACTERISTICS
(A) LENGTH: 35 BASE PAIRS
(B) TYPE: NtTCLEIC ACID
(C) STR.ANDEDNFsSS: SINGLE
(D) TOPOLOGY: LTNEAR
(ii) MOLECULE TYPE: Oligonucleotide (xi) SEQUENCE DESCRIPTION: SEQ ID N0:4:
CCCAAGCTTC'CACAAACGTT GCC'I'TCCTCT ATGAG 35 (2) INFORMATION FOR SEQ ID N0:5:
(i) SEQUENCE CHARACTERISTICS
(A) LENGTH: 36 BASE PAIRS

(B) TYPE:. NUCLEIC ACID
{C) STR.ANDEDNESS: SINGLE
{D) TOPOLOGY: LINEAR
(ii) MOLECULE TYPE: Oligonucleotide (xi) SEQUENCE DESCRIPTION: SEQ ID N0:5:

(2) INFORMATION FOR SEQ ID NO:6:
(i) SEQUENCE CHARACTERISTICS
{A) LENGTH: 35 BASE PAIRS
(B) TYPE: NtTCLEIC ACID
(C) STRANDEDNESS: SINGLE
fD) TOPOLOGY: LINEAR
{ii) MOLECULE TYPE:- Oligonucleoti:de (xi) SEQUENCE DESCRIPTION: SEQ ID N0:6:

(2) INFORMATION FOR S8Q ID N0:7:
(i) SEQUENCE CHARACTERISTICS
(A) LENGTH: 35 BASE PAIRS
(B) TYPE: NUCLEIC ACID
fC) STRANDEDNESS: SINGLE
(D) TOPOLOGY: LINEAR
(ii) MOLECULE TYPE: Oligonucleotide (xi) SEQUENCE DESCRIPTION: SEQ ID N0:7:

(2) TNFORMATION FOR SEQ ID N0:8:
(i) SEQUENCE QiARACTERISTICS
(A) LENGTH: 26 BASE PAIRS
(B) TYPE: NUCLEIC ACID
(C) STRANDBDNESS: SINGLE
(D) TOPOLOGY: LINEAR
(ii) MOLECULE TYPE: Oligonucleotide (xi) SEQUENCE DsscRIPTxON; sEQ xD No: s:

(2) INFORMATION FOR SEQ ID N0:9:
(i) SEQUENCE CHARACTERISTICS
(A) LENGTH: 39 BASE PAIRS
(B) TYPE: NUCLEIC ACID
(C) STRANDEDNESS: SINGLE
(D) TOPOLOGY: LINEAR
(ii) MOLECULE TYPE: Oligonucleotide (xi) SEQUENCE DESCRIPTTON: SEQ ID N0:9:

(2) INFORMATION FOR SEQ ID N0:10:
(i) SBQUENCE CHARACTERISTICS
(A) LENGTH: 69 BASE PAIRS
(B) TYPE: NUCLEIC ACID
(C) STRANDEDNESS: SINGLE
(D) TOPOLOGY: LINEAR
(ii) MOLECULE TYPE: Oligonucleotide (xi) ssQc~rcE DESCRIPTION: sEQ =D NO: is:

AAAGTGAGG ~ 69 (2) INFORMATION FOR S8Q ID N0:11:
(i) SEQUENCE C'~ARACTERISTICS
(A) LENGTH: 54 BASE PAIRS
(B) TYPE: NUCLEIC ACID
(C) SfiR.ANDEDNESS : SINGLE
(D) TOPOLOGY: LINEAR
(ii) MOLECOLE TYPE: Oligonucleotide (xi) SEQUENCE DESCRIPTION: SEQ ID N0:11:

(2) INFORMATION FOR SEQ ID N0:12:
(i) SEQUENCE CHARACTBRISTICS
(A) LENGTH: 35 BASE PAIRS
(B) TYPE: NUCLEIC ACID
(C) STRANDEDNESS: SINGLE
(D) TOPOLOGY: LINEAR
(ii) MOLECULE TYPE: Oligonucleoti.de , (xi) SEQU&NCE DESCRIPTION: SEQ ID N0:12:

Claims (88)

1. An isolated antibody or fragment thereof that specifically binds to a protein selected from the group consisting of:
(a) a protein consisting of amino acid residues -36 to +172 of SEQ
ID NO:2;
(b) a protein consisting of amino acid residues 1 to +172 of SEQ
ID NO:2;
(c) a protein consisting of a portion of SEQ ID NO:2, wherein said portion comprises at least 30 contiguous amino acid residues of SEQ ID NO:2;
and (d) a protein consisting of a portion of SEQ ID NO:2, wherein said portion comprises at least 50 contiguous amino acid residues of SEQ ID NO:2.
2. The antibody or fragment thereof of claim 1, that specifically binds protein (a).
3. The antibody or fragment thereof of claim 1, that specifically binds protein (b).
4. The antibody or fragment thereof of claim 1, that specifically binds protein (c).
5. The antibody or fragment thereof of claim 1, that specifically binds protein (d).
6. The antibody or fragment thereof of claim 2, that specifically binds protein (b).
7. The antibody or fragment thereof of claim 3, wherein said protein bound by said antibody or fragment thereof is glycosylated.
8. The antibody or fragment thereof of claim 3, which is a human antibody.
9. The antibody or fragment thereof of claim 3, which is a polyclonal antibody.
10. The antibody or fragment thereof of claim 3, which is selected from the group consisting of:
(a) a chimeric antibody;
(b) a humanized antibody;
(c) a single chain antibody; and (d) a Fab fragment.
11. The antibody or fragment thereof of claim 3, which is attached to a detectable reagent.
12. The antibody or fragment thereof of claim 11, wherein the detectable reagent is selected from the group consisting of:
(a) an enzyme;
(b) a fluorescent label; and (c) a radioactive label.
13. The antibody or fragment thereof of claim 3, wherein said antibody or fragment thereof specifically binds to said protein in a Western blot.
14. The antibody or fragment thereof of claim 3, wherein said antibody or fragment thereof specifically binds to said protein in an ELISA.
15. An isolated cell that produces the antibody or fragment thereof of claim 3.
16. A hybridoma that produces the antibody or fragment thereof of claim 3.
17. A method of detecting Keratinocyte Growth Factor-2 protein in a biological sample comprising:
(a) contacting the biological sample with. the antibody or fragment thereof of claim 3; and (b) detecting the Keratinocyte Growth Factor-2 protein in the biological sample.
18. The method of claim 17, wherein the antibody or fragment thereof is a polyclonal antibody.
19. An isolated antibody or fragment thereof obtained from an animal that has been immunized with a protein selected from the group consisting of:
(a) a protein consisting of amino acid residues -36 to +172 of SEQ
ID NO:2;
(b) a protein consisting of amino acid residues 1 to +172 of SEQ
ID NO:2;
(c) a protein consisting of a portion of SEQ ID NO:2, wherein said portion comprises at least 30 contiguous amino acid residues of SEQ ID NO:2;
and (d) a protein consisting of a portion of SEQ ID NO:2, wherein said portion comprises at least 50 contiguous amino acid residues of SEQ ID NO:2;
wherein said antibody or fragment thereof specifically binds to said amino acid sequence.
20. The antibody or fragment thereof of claim 19, obtained from an animal immunized with protein (a).
21. The antibody or fragment thereof of claim 19, obtained from an animal immunized with protein (b).
22. The antibody or fragment thereof of claim 19, obtained from an animal immunized with protein (c).
23. The antibody or fragment thereof of claim 19, obtained from an animal immunized with protein (d).
24. The antibody or fragment thereof of claim 19, which is a monoclonal antibody.
25. The antibody or fragment thereof of claim 19, which is selected from the group consisting of:

(a) a chimeric antibody;

(b) a polyclonal antibody;

(c) a humanized antibody;

(d) a single chain antibody; and (e) a Fab fragment.
26. An isolated monoclonal antibody or fragment thereof that specifically binds to a protein selected from the group consisting of:

(a) a protein consisting of amino acid residues -36 to +172 of SEQ
ID NO:2;

(b) a protein consisting of amino acid residues 1 to +172 of SEQ
ID NO:2;

(c) a protein consisting of a portion of SEQ ID NO:2, wherein said portion comprises at least 30 contiguous amino acid residues of SEQ ID NO:2;
and (d) a protein consisting of a portion of SEQ ID NO:2, wherein said portion comprises at least 50 contiguous amino acid residues of SEQ ID NO:2.
27. The antibody or fragment thereof of claim 26, that specifically binds protein (a).
28. The antibody or fragment thereof of claim 26, that specifically binds protein (b).
29. The antibody or fragment thereof of claim 26, that specifically binds protein (c).
30. The antibody or fragment thereof of claim 26, that specifically binds protein (d).
31. The antibody or fragment thereof of claim 27, that specifically binds protein (b).
32. The antibody or fragment thereof of claim 28, wherein said protein bound by said antibody or fragment thereof is glycosylated.
33. The antibody or fragment thereof of claim 28, which is a human antibody.
34. The antibody or fragment thereof of claim 28, which is selected from the group consisting of:
(a) a chimeric antibody;
(b) a humanized antibody;
(c) a single chain antibody; and (d) a Fab fragment.
35. The antibody or fragment thereof of claim 28, which is attached to a detectable reagent.
36. The antibody or fragment thereof of claim 35, wherein the detectable reagent is selected from the group consisting of:

(a) an enzyme;

(b) a fluorescent label; and (c) a radioactive label.
37. The antibody or fragment thereof of claim 28, wherein said antibody or fragment thereof specifically binds to said protein in a Western blot.
38. The antibody or fragment thereof of claim 28, wherein said antibody or fragment thereof specifically binds to said protein in an ELISA.
39. An isolated cell that produces the antibody or fragment thereof of claim 28.
40. A hybridoma that produces the antibody or fragment thereof of claim 28.
41. A method of detecting Keratinoctye Growth Factor-2 protein in a biological sample comprising:

(a) contacting the biological sample with the antibody or fragment thereof of claim 28; and (b) detecting the Keratinocyte Growth Factor-2 protein in the biological sample.
42. An isolated antibody or fragment thereof that specifically binds to a protein selected from the group consisting of:

(a) a protein consisting of the full-length polypeptide encoded by the cDNA contained in ATCC Deposit Number 75977;

(b) a protein consisting of the mature form of the polypeptide encoded by the cDNA contained in ATCC Deposit Number 75977;

(c) a protein consisting of a portion of the polypeptide encoded by the cDNA contained in ATCC Deposit Number 75977, wherein said portion comprises at least 30 contiguous amino acid residues of the polypeptide encoded by the cDNA contained in ATCC Deposit Number 75977; and (d) a protein consisting of a portion of the polypeptide encoded by the cDNA contained in ATCC Deposit Number 75977, wherein said portion comprises at least 50 contiguous amino acid residues of the polypeptide encoded by the cDNA contained in ATCC Deposit Number 75977.
43. The antibody or fragment thereof of claim 42, that specifically binds protein (a).
44. The antibody or fragment thereof of claim 42, that specifically binds protein (b).
45. The antibody or fragment thereof of claim 42, that specifically binds protein (c).
46. The antibody or fragment thereof of claim 42, that specifically binds protein (d).
47. The antibody or fragment thereof of claim 43, that specifically binds protein (b).
48. The antibody or fragment thereof of claim 44, wherein said protein bound by said antibody or fragment thereof is glycosylated.
49. The antibody or fragment thereof of claim 44, which is a human antibody.
50. The antibody or fragment thereof of claim 44, which is a polyclonal antibody.
51. The antibody or fragment thereof of claim 44, which is selected from the group consisting of:

(a) a chimeric antibody;
(b) a humanized antibody;
(c) a single chain antibody; and (d) a Fab fragment.
52. The antibody or fragment thereof of claim 44, which is attached to a detectable reagent.
53. The antibody or fragment thereof of claim 52, wherein the detectable reagent is selected from the group consisting of:
(a) an enzyme;
(b) a fluorescent label; and (c) a radioactive label.
54. The antibody or fragment thereof of claim 44, wherein said antibody or fragment thereof specifically binds to said protein in a Western blot.
55. The antibody or fragment thereof of claim 44, wherein said antibody or fragment thereof specifically binds to said protein in an ELISA.
56. An isolated cell that produces the antibody or fragment thereof of claim 44.
57. A hybridoma that produces the antibody or fragment thereof of claim 44.
58. A method of detecting Keratinocyte Growth Factor-2 protein in a biological sample comprising:

(a) contacting the biological sample with the antibody or fragment thereof of claim 44; and (b) detecting the Keratinocyte Growth Factor-2 protein in the biological sample.
59. The method of claim 58, wherein the antibody or fragment thereof is a polyclonal antibody.
60, An isolated antibody or fragment thereof obtained from an animal that has been immunized with a protein selected from the group consisting of:

(a) a protein comprising the amino acid sequence of the full-length polypeptide encoded by the cDNA contained in ATCC Deposit Number 75977;

(b) a protein comprising the amino acid sequence of the mature form of the polypeptide encoded by the cDNA contained in ATCC Deposit Number 75977;

(c) a protein comprising the amino acid sequence of at least 30 contiguous amino acid residues of the polypeptide encoded by the cDNA
contained in ATCC Deposit Number 75977; and (d) a protein comprising the amino acid sequence of at least 50 contiguous amino acid residues the polypeptide encoded by the cDNA contained in ATCC Deposit Number 75977;

wherein said antibody or fragment thereof specifically binds to said amino acid sequence.
61. The antibody or fragment thereof of claim 60, obtained from an animal immunized with protein (a).
62. The antibody or fragment thereof of claim 60, obtained from an animal immunized with protein (b).
63. The antibody or fragment thereof of claim 60, obtained from an animal immunized with protein (c).
64. The antibody or fragment thereof of claim 60, obtained from an animal immunized with protein (d).
65. The antibody or fragment thereof of claim 60, which is a monoclonal antibody.
66. The antibody or fragment thereof of claim 60, which is selected from the group consisting of:

(a) a chimeric antibody;

(b) a polyclonal antibody;

(c) a humanized antibody;

(d) a single chain antibody; and (e) a Fab fragment.
67. An isolated monoclonal antibody or fragment thereof that specifically binds to a protein selected from the group consisting of:

(a) a protein consisting of the full-length polypeptide encoded by the cDNA contained in ATCC Deposit Number 75977;

(b) a protein consisting of the mature form of the polypeptide encoded by the cDNA contained in ATCC Deposit Number 75977;

(c) a protein consisting of a portion of the polypeptide encoded by the cDNA contained in ATCC Deposit Number 75977, wherein said portion comprises at least 30 contiguous amino acid residues of the polypeptide encoded by the cDNA contained in ATCC Deposit Number 75977; and (d) a protein consisting of a portion of the polypeptide encoded by the cDNA contained in ATCC Deposit Number 75977, wherein said portion comprises at least 50 contiguous amino acid residues of the polypeptide encoded by the cDNA contained in ATCC Deposit Number 75977.
68. The antibody or fragment thereof of claim 67, that specifically binds protein (a).
69. The antibody or fragment thereof of claim 67, that specifically binds protein (b).
70. The antibody or fragment thereof of claim 67, that specifically binds protein (c).
71. The antibody or fragment thereof of claim 67, that specifically binds protein (d).
72. The antibody or fragment thereof of claim 68, that specifically binds protein (b).
73. The antibody or fragment thereof of claim 69, wherein said protein bound by said antibody or fragment thereof is glycosylated.
74. The antibody or fragment thereof of claim 69, which is a human antibody.
75. The antibody or fragment thereof of claim 69, which is selected from the group consisting of:

(a) a chimeric antibody;
(b) a humanized antibody;
(c) a single chain antibody; and (d) a Fab fragment.
76. The antibody or fragment thereof of claim 69, which is attached to a detectable reagent.
77. The antibody or fragment thereof of claim 76, wherein the detectable reagent is selected from the group consisting of:

(a) an enzyme;
(b) a fluorescent label; and (c) a radioactive label.
78. The antibody or fragment thereof of claim 69, wherein said antibody or fragment thereof specifically binds to said protein in a Western blot.
79. The antibody or fragment thereof of claim 69, wherein said antibody or fragment thereof specifically binds to said protein in an ELISA.
80. An isolated cell that produces the antibody or fragment thereof of claim 69.
81. A hybridoma that produces the antibody or fragment thereof of claim 69.
82. A method of detecting Keratinocyte Growth Factor-2 protein in a biological sample comprising:

(a) contacting the biological sample with the antibody or fragment thereof of claim 69; and (b) detecting the Keratinocyte Growth Factor-2 protein in the biological sample.
83. An isolated antibody or fragment thereof that specifically binds a Keratinocyte Growth Factor-2 protein purified from a cell culture wherein the cells in said cell culture comprise a polynucleotide encoding amino acids 1 to 170 of SEQ ID
NO:2 operably associated with a regulatory sequence that controls the expression of said polynucleotide.
84. The antibody or fragment thereof of claim 83, which is a monoclonal antibody.
85. The antibody or fragment thereof of claim 83, which is a human antibody.
86. The antibody or fragment thereof of claim 83, which is selected from the group consisting of:

(a) a chimeric antibody;

(b) a polyclonal antibody;

(c) a humanized antibody;

(d) a single chain antibody; and (e) a Fab fragment.
87. The antibody or fragment thereof of claim 83, wherein said antibody or fragment thereof specifically binds to said protein in a Western blot.
88. The antibody or fragment thereof of claim 83, wherein said antibody or fragment thereof specifically binds to said protein in an ELISA.
CA002430223A 1995-02-14 1995-02-14 Keratinocyte growth factor-2 Abandoned CA2430223A1 (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CA002210444A CA2210444C (en) 1995-02-14 1995-02-14 Keratinocyte growth factor-2

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CA002210444A Division CA2210444C (en) 1995-02-14 1995-02-14 Keratinocyte growth factor-2

Publications (1)

Publication Number Publication Date
CA2430223A1 true CA2430223A1 (en) 1996-08-22

Family

ID=4161064

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002430223A Abandoned CA2430223A1 (en) 1995-02-14 1995-02-14 Keratinocyte growth factor-2

Country Status (1)

Country Link
CA (1) CA2430223A1 (en)

Similar Documents

Publication Publication Date Title
EP0837934B1 (en) Human vascular endothelial growth factor 2
US7482326B2 (en) Endothelial-monocyte activating polypeptide III
US20070154908A1 (en) Connective Tissue Growth Factor-2
US7576189B2 (en) Antibodies to human vascular endothelial growth factor 2 and methods of using the same
AU708868B2 (en) Keratinocyte growth factor-2
WO1997035976A9 (en) Epidermal differentiation factor
WO1996039424A1 (en) Natural killer cell enhancing factor c
US6537539B2 (en) Immune cell cytokine
EP0814661A1 (en) UBIQUITIN CONJUGATING ENZYMES 7, 8 and 9
US6057434A (en) Mammary transforming protein
AU753987B2 (en) Keratinocyte growth factor-2
AU716100B2 (en) Human vascular endothelial growth factor 3
CA2430223A1 (en) Keratinocyte growth factor-2
WO1996015147A1 (en) Human stanniocalcin-alpha
US20020119487A1 (en) Human stem cell antigen 2
EP1284292A2 (en) Human B-cell Translocation Gene-3
US20030027989A1 (en) Mammary transforming protein
EP0873349A1 (en) Endothelial-monocyte activating polypeptide iii
MXPA97006204A (en) Factor 2 growth of queratinoci
JP2002325590A (en) Keratinocyte growth factor-2

Legal Events

Date Code Title Description
EEER Examination request
FZDE Dead