MXPA97006854A - Factor 14 of fibroblas growth - Google Patents

Factor 14 of fibroblas growth

Info

Publication number
MXPA97006854A
MXPA97006854A MXPA/A/1997/006854A MX9706854A MXPA97006854A MX PA97006854 A MXPA97006854 A MX PA97006854A MX 9706854 A MX9706854 A MX 9706854A MX PA97006854 A MXPA97006854 A MX PA97006854A
Authority
MX
Mexico
Prior art keywords
polypeptide
cells
polynucleotide
dna
sequence
Prior art date
Application number
MXPA/A/1997/006854A
Other languages
Spanish (es)
Other versions
MX9706854A (en
Inventor
M Greene John
J Dillon Patrick
Original Assignee
J Dillon Patrick
M Greene John
Human Genome Sciences Inc
Filing date
Publication date
Priority claimed from PCT/US1995/006730 external-priority patent/WO1996039506A1/en
Application filed by J Dillon Patrick, M Greene John, Human Genome Sciences Inc filed Critical J Dillon Patrick
Publication of MX9706854A publication Critical patent/MX9706854A/en
Publication of MXPA97006854A publication Critical patent/MXPA97006854A/en

Links

Abstract

A polypeptide of fibroblast growth factor 14, human and DNA (RNA) encoding this polypeptide is disclosed. A method for producing this polypeptide by recombinant techniques is also provided. Methods for using this polypeptide to promote wound healing are also described, for example as a result of sunburn burns to prevent neuronal damage due to / associated with stroke and promote neuronal growth, and to prevent skin aging and loss of hair, to stimulate angiogenesis, mesodermal induction in early embryos and the regeneration of limbs. Antagonists are also described against these polypeptides and their use as a therapeutic to prevent abnormal cell proliferation, hypervascular diseases and the proliferation of epithelial crystalline cells. Diagnostic methods are also described for detecting mutations in the coding sequence and alterations in the concentration of the polypeptides in a sample derived from a host.

Description

FACTOR 14 OF GROWTH OF FIBROBLASTS This invention relates to newly identified polynucleotides, polypeptides encoded by these polynucleotides, the use of these polynucleotides and polypeptides, as well as the production of these polynucleotides and polypeptides. More particularly, the polypeptide of the present invention has been putatively identified as the fibroblast growth factor / heparin binding growth factor, hereinafter referred to as "FGF-14". The invention also relates to the inhibition of the action of these polypeptides. Fibroblast growth factors are a family of proteins that are characteristic of heparin binding and therefore are also called heparin binding growth factors (HBGF). The expression of different members of these proteins are found in various tissues and they are under particular temporal and spatial control. These proteins are potent mitogens for a variety of cells of mesodermal, ectodermal and endodermal origin, including fibroblasts, corneal and vascular endothelial cells, granulocytes, adrenal cortical cells, chondrocytes, myoblasts, REF: 25488 vascular smooth muscle cells, lens epithelial cells, melanocytes, keratinocytes, oligodendrocytes, astrocytes, osteoblasts, and hematopoietic cells. Each member has functions that overlap with others and also has its unique spectrum of functions. In addition to the ability to stimulate the proliferation of endothelial, vascular cells, both FGF-1 and 2 are chemotactic for endothelial cells and FGF-2 has been shown to allow endothelial cells to penetrate to base membrane. Consistent with these properties, both FGF-1 and 2 have the ability to stimulate angiogenesis. Another important feature of these growth factors is their ability to promote wound healing. Many other members of the FGF family share similar activities with FGF-1 and 2 such as the promotion of angiogenesis and wound healing. Several members of the FGF family have been shown to induce mesodermal formation to modulate the differentiation of neuronal cells, adipocytes, and skeletal muscle cells. Unlike these biological activities in normal tissues, FGF proteins have been implicated in the promotion of tumorigenesis in carcinomas and sarcomas by promoting tumor vascularization and as transformation proteins when their expression is deregulated. The FGF family currently consists of eight structurally related polypeptides: basic FGF, acid FGF, int 2, hst 1 / KFGF, FGF-5, FGF-6, keratinocyte growth factor, AIGF (FGF-8) and recently a factor glia-activation has been shown to be a new heparin-binding growth factor that was purified from the culture supernatant of a human glioma cell line (Miyamoto, M. et al., Mol.And Cell. Biol., 13 (7): 4251-4259 (1993). The genes for each one have been cloned and sequenced. Two of the members, FGF-1 and FGF-2, have been characterized under many names, but most frequently as the fibroblast growth factor, has been and basic, respectively. Normal gene products influence the overall proliferation capacity of most cells derived from mesoderm and neuroectoderm. They are capable of inducing angiogenesis in vivo and can play important roles in early development (Burgess, W.H. and Maciag, T., Annu. Rev. Biochem., 58: 575-606, (1989)).
Many of the previously identified members of the FGF family also bind to the same receptors and elicit a second message through binding to these receptors. A eukaryotic expression vector encoding a secreted form of FGF-1 has been introduced by gene transfer into porcine arteries. This model defines the gene function in the arterial wall in vivo. The expression of FGF-1 induced by intimal thickening in the porcine arteries, 21 days after the gene transfer (Nabel, E. G., et al., Nature, 362: 844-6 (1993)). It has been further demonstrated that the fibroblast growth factor can regulate glioma growth and progression independent of its role in tumor angiogenesis and that the release or secretion of fibroblast growth factor, basic may be required by these actions ( Morrison, RS, et al., J. Neurosci, Res., 34: 502-9 (1993)). Fibroblast growth factors, such as basic FGF, have been further implicated in the growth of Kaposi's sarcoma cells in vitro (Huang, YQ, et al., J. Clin. Invest., 91: 1191-7 ( 1993)). Also, the sequence of the cDNA encoding the basic, human fibroblast growth factor has been cloned downstream of a transcription promoter recognized by the bacteriophage T7 RNA polymerase. The basic fibroblast growth factors thus obtained have been shown to have biological activity indistinguishable from fibroblast growth factor, placental, human in mitogenicity, plasminogen activator synthesis and angiogenesis assays (Squires, CH, et al., J. Biol. Chem., 263: 16297-302 (1988) U.S. Patent No. 5,155,214 discloses fibroblast growth factors, basic, mammalian, substantially pure and their production. growth of fibroblasts, human, and bovine, as well as the DNA sequence encoding the bovine polypeptide The newly discovered FGF-9 has approximately 30% sequence similarity to other members of the FGF family. of cysteine and other consensus sequences in family members were also observed in the FGF-9 sequence.FGF-9 was found to have no sequence of typical signals in its N-term similar to that in the acidic and basic FGF. However, FGF-9 was found to be segregated from cell after synthesis in spite of the lack of a typical signal sequence FGF (Miya oto, M. et al., Mol.And Cell. Biol., 13 (7): 4251-4259 (1993) Additionally, FGF-9 was found to stimulate the growth of astrocyte progenitor cells, type 2 oligodendrocytes, BALB / c3T3, and PC-12 cells but not those of cells human umbilical vein endothelial cells (Naruo, K., et al., J. Biol. Chem., 268: 2857-2864 (1993).) Basic FGF and acid FGF are potent modulators of cell proliferation, cell motility, differentiation, and survival and act on the cell types of ectoderm, mesoderm and endoderm.These two FGFs, together with KFG and AIFG, were identified by protein purification.However, the other four members were isolated as oncogenes, expression of which was restricted to embryogenesis and certain types of cancer FGF-9 It was shown to be a mitogen against the gual cells.
Members of the FGF family are reported to have oncogenic potency. FGF-9 has been shown to transform the power when it is transformed into BALB / c3T3 cells (Miyamoto, M., et al., Mol.Cell., Biol., 13 (7): 4251-4259 (1993)).
The androgen-induced growth factor (AIGF), also known as FGF-8, was purified from a conditioned medium of testosterone-stimulated mouse mammary carcinoma cells (SC-3). AIGF is a growth factor similar to the FGF, distinctive, which has a putative signal peptide and shares 30-40% homology with known members of the FGF family. Mammalian cells transformed with AIGF show a remarkable stimulatory effect on the growth of SC-3 cells in the absence of androgen. Therefore, AIGF mediates the androgen-induced growth of SC-3 cells, and perhaps other cells, since it is secreted by the same tumor cells. The polypeptide of the present invention has been putatively identified as a member of the FGF family as a result of the homology of the amino acid sequence with other members of the FGF family. In accordance with one aspect of the present invention, new mature polypeptides as well as biologically active and diagnostically or therapeutically useful fragments, analogs and derivatives thereof are provided. The polypeptides of the present invention are of human origin. According to another aspect of the present invention, isolated nucleic acid molecules encoding the polypeptides of the present invention, including mRNA, DNA, cDNA, genomic DNA, as well as antisense analogues thereof and biologically active documents, are provided. and diagnostically or therapeutically useful thereof. According to yet another aspect of the present invention, processes are provided for producing these polypeptides by recombinant techniques through the use of recombinant vectors, such as cloning and expression plasmids useful as reagents in the recombinant production of the polypeptides of the present invention. , as well as prokaryotic and / or eukaryotic, recombinant host cells, comprising a nucleic acid sequence encoding a polypeptide of the present invention. According to a further aspect of the present invention, there is provided a process for using these polypeptides, or polynucleotides encoding these polypeptides, for screening or screening agonists and antagonists therefor and for therapeutic purposes, for example, promotion of the wound healing, for example as a result of burns and ulcers, to prevent neuronal damage due to the associated blow and promote neuronal growth, and prevent skin aging and hair loss, to stimulate angiogenesis, mesodermal induction in early embryos and the regeneration of limbs. In accordance with a further aspect of the present invention, antibodies against these polypeptides are provided. According to yet another aspect of the present invention, antagonists against these polypeptides are provided and processes for their use to inhibit the action of these polypeptides, for example, in the treatment of cell transformation, e.g. tumors, to reduce healing and treat hypervascular diseases. In accordance with another aspect of the present invention, nucleic acid probes are provided which comprise nucleic acid molecules of sufficient length to specifically hybridize to a polynucleotide encoding a polypeptide of the present invention. According to yet another aspect of the present invention, diagnostic assays are provided to detect diseases or susceptibility to diseases related to mutations in a nucleic acid sequence of the present invention and to detect overexpression of the polypeptides encoded by these sequences. According to another aspect of the present invention, a process for using these polypeptides or polynucleotides encoding these polypeptides is provided, for in vitro purposes related to scientific research, DNA synthesis 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 therein. The following drawings are given only as illustrations of the specific embodiments of the present invention and are not intended as limitations in any way.
Figure 1 depicts the corresponding deduced, cDNA sequence and amino acid sequence of FGF-14. The initial 26 amino acid residues represent a putative leader sequence.
According to one aspect of the present invention, isolated nucleic acid molecules (polynucleotides) encoding the mature polypeptide having the deduced amino acid sequence of Figure 1 (SEQID Nos. 2) or for the mature polypeptide are provided. encoded by the cDNA of the clone deposited in deposit number 97147 in the American Type Culture Collection (ATCC), 12301 Parklawn Drive, Rockville, Maryland 20852, United States of America, on 12 , JQ May 1995.
The polynucleotide encoding FGF-14 of this invention was initially discovered in a cDNA library or library derived from the tissue of the human cerebellum. It is structurally related to the members of the family of fibroblast growth factors and contains an open reading frame that encodes a polypeptide of 225 amino acids of which the first 26 amino acids represent a putative signal sequence so that the mature polypeptide comprises 199 amino acids. Among the maximum matches are: 1) 40% identity and 61% sequence similarity to human FGF-9 over a stretch of 126 amino acids; 2) 40% identity and 61% similarity to rat FGF-9 over a region of 126 amino acids; 3) 36% identity and 57 ° or similarity with human KGF over a stretch of 148 amino acids.
The identification signal of the FGF / HBGF family, GXLX (S, T, A, G) X6 (D, E) CXFXE is converted into the polypeptide of the present invention, (X means any amino acid residue; E), means a residue either D or E, X6 means any of 6 amino acid residues The polynucleotide of the present invention may be in the form of RNA or in the form of DNA, DNA including cDNA, genomic DNA, and Synthetic DNA DNA can be double-stranded or single-stranded The coding sequence encoding the mature polypeptide can be identical to the coding sequence shown in Figure 1 (SEQ ID NOS: 1) or that of the deposited clone or it may be a different coding sequence as a result of the redundancy or degeneracy of the genetic code, it encodes for the same mature polypeptide as the DNA of Figure 1 (SEQ ID NOS: 1) or the deposited cDNA The polynucleotides encoding the mature polypeptide of the Fi Figure 1 (SEQ ID NOS: 2) or for the mature polypeptides encoded by the deposited cDNA (s) may include: only the coding sequence for the mature polypeptide; the coding sequence for the mature polypeptide and the additional coding sequence such as a guiding or secretory sequence or a proprotein sequence; the coding sequence for the mature polypeptide (and the optionally additional coding sequence) and the non-coding sequence, such as in introns or the 5 'and / or 3' non-coding sequence of the coding sequence for the mature polypeptide . In this manner, the term "polynucleotide encoding a polypeptide" encompasses a polynucleotide that includes only the coding sequence for the polypeptide as well as a polynucleotide that includes the additional coding and / or non-coding sequence. The present invention further relates to variants of the polynucleotides described above that encode fragments, analogs and derivatives of the polypeptides having the deduced amino acid sequence of Figure 1 (SEQ ID NOS: 2) or the polypeptides encoded by the cDNAs ( s) of the deposited clone (s). the variants of the polynucleotide may be a naturally occurring allelic variant of the polynucleotide or a non-naturally occurring variant of the polynucleotide. Thus, the present invention includes polynucleotides that encode the same mature polypeptide as shown in Figure 1 (SEQ ID NOS: 2) or the same mature polypeptides encoded by the cDNA (s) of the clone. (en) deposited (s) as well as variants of these polypeptides, variants encoding a fragment, derivative or analogue of the polypeptide of Figure 1 (SEQ ID NOS: 2) or the polypeptides encoded by the cDNA (s) of the deposited clone (s). These nucleotide variants include deletion variants, substitution variants and addition or insertion variants. As indicated hereinabove, the polynucleotide may have a coding sequence which is a naturally occurring allelic variant of the coding sequence shown in Figure 1 (SEQ ID NOS: 1) or the coding sequence of the (s) clone (s) deposited (s). As is known in the art, an allelic variant is an alternate form of a polynucleotide sequence that can have a substitution, deletion or addition of one or more nucleotides, which does not substantially alter the function of the encoded polynucleotides. The present invention also includes polynucleotides, wherein the coding sequence for the mature polypeptides can be fused in the same reading frame to a polynucleotide sequence that aids in the expression and secretion of a polypeptide from a host cell, e.g. , a guiding sequence that functions as a secretory sequence to control the transport of a polypeptide from the cell. The polypeptide having a leader sequence is a preprotein and may have the leader sequence cleaved by the host cells, to form the mature form of the polypeptide. The polynucleotides can also code for a proprotein which is the lowest mature protein of additional 5 'amino acids. A mature protein that has a prosequence is a proprotein and is an inactive form of the protein. Once the prosequence is cleaved, the mature, active protein remains. Thus, for example, the polynucleotides of the present invention may code for a mature protein, or for a protein having a prosequence or for a protein having both a prosequence and a presequence (leader sequence). The polynucleotides of the present invention may also have the coding sequence fused in the structure to a marker sequence that allows purification of the polypeptide of the present invention. The marker sequence can be a hexa-histidine fragment supplied by a pQE-9 vector to provide for purification of the mature polypeptide fused to the label in the case of a bacterial host, or, for example, the marker sequence can be a segment of hemagglutinin (HA) when a mammalian host is used, e.g., COS-7 cells. The HA fragment corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson, I., et al., Cell, 37: 767 (1984)). The term "gene" means the DNA fragment comprised in the production of a polypeptide chain; it includes regions that precede and follow the coding region (guidance and towing) as well as intervening sequences (introns) between the individual coding segments (exons). Fragments of the full-length FGF-14 gene can be used as a hybridization probe for a cDNA library to isolate the full-length gene and to isolate other genes having a high sequence similarity to the gene or similar biological activity. Probes of this type preferably have at least 30 bases and may contain, for example, 50 or more bases. The probe can also be used to identify a cDNA clone that corresponds to a full-length transcript and a genomic clone or clones that contain the complete FGF-14 gene that includes the regulatory and promoter regions, exons and introns. An example of an experiment comprises the isolation of the coding region and an FGF-14 gene by using the known DNA sequence to synthesize an oligonucleotide probe. The labeled oligonucleotides having a sequence complementary to that of the gene of the present invention are used to examine a library of human cDNA, genomic DNA or mRNA to determine which members of the library or library hybridize to the probe. The present invention further relates to polynucleotides that hybridize to the sequence described above if there is at least 70%, preferably at least 90%, and most preferably at least 95% identity between the sequences. The present invention relates particularly to polynucleotides that hybridize under severe conditions to the polynucleotides described above. As used herein, the term "stringent conditions" means that the hybridization will occur only if there is at least 95% and preferably at least 97% identity between the sequences. The polynucleotides that hybridize to the polynucleotides described above in a preferred embodiment code for polypeptides that either substantially maintain the same biological function or activity as the mature polypeptide encoded by the cDNAs of Figure 1 (SEQ ID NO: 1) or the ) Deposited cDNA (s). Alternatively, the polynucleotide can have at least 20 bases, preferably 30 bases, and most preferably at least 50 bases that hybridize to a polynucleotide of the present invention and have an identity therefor, as described above in the present, and that can maintain the activity or not. For example, these polynucleotides can be used as probes for the polynucleotide of SEQ ID NO: 1, for example, for the recovery of the polynucleotide or as a diagnostic probe or as a PCR primer. Thus, the present invention is directed to polynucleotides having at least 70% identity, preferably at least 90% and most preferably at least 95% identity to a polynucleotide encoding the SEQ polypeptide. ID NO: 2 as well as fragments thereof, fragments having at least 30 bases and preferably at least 50 bases and polypeptides encoded by these polynucleotides. The deposit (s) referred to herein shall be maintained under the Budapest Treaty in the International Recognition of the Deposit of Microorganisms for purposes of patent procedure. These deposits are provided only as a convenience and are not an admission that a deposit is required under 35 U.S.C. § 112. The sequence of the polynucleotides contained in the deposited materials, as well as the amino acid sequence of the polypeptides encoded by them, are incorporated herein by reference and are controlled in the case of any conflict with the sequence description in the present. A license may be required to make, use or sell the deposited materials, and this license is not granted in this manner. The present invention further relates to an FGF polypeptide having the reduced amino acid sequence of Figure 1 (SEQ ID NOS: 2) or having the amino acid sequence encoded by the deposited cDNA (s) as well as fragments , analogs and derivatives of these polypeptides.
The terms "fragment", "derivative" and "analogue" when referring to the polypeptide of Figure 1 (SEQ ID NOS: 2) or those encoded by the deposited cDNA (s), means polypeptides that maintain essentially the same biological function or activity as these polypeptides. In this manner, an analog includes a proprotein that can be activated by cleavage of the proprotein portion to produce a mature, active polypeptide. The polypeptides of the present invention can be recombinant polypeptides, natural polypeptides or synthetic polypeptides, preferably recombinant polypeptides. The fragment, an analog derivative of the polypeptide of Figure 1 (SEQ ID NOS: 2) or that encoded by the deposited cDNA (s) can be (i) one in which one or more of the residues of amino acids are substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and this substituted amino acid residue may or may not be one encoded by the genetic code, or (ii) one in which one or more of the amino acid residues includes a substituent group, or (iii) one in which the mature polypeptide is fused to another group, such as a compound to increase the half-life of the polypeptide (by example, polyethylene glycol), or (iv) one in which the additional amino acids are fused to the mature polypeptide, such as a guiding or secretory sequence or a sequence that is employed for the purification of the mature polypeptide or a proprotein sequence. These fragments, derivatives and analogs are considered 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 are preferably purified to homogeneity. The term "isolated" means that the material is removed from its natural environment (for example, the natural environment if it occurs naturally). For example, a polynucleotide or polypeptide of natural origin present in a living animal is not isolated, but the same polynucleotide or DNA or polypeptide is isolated, separating some or all of the coexisting materials in the natural system. This polynucleotide could be part of a vector and / or this polynucleotide or polypeptide could be part of a composition, and still be isolated since this vector or composition is not part of its natural environment.
The polypeptides of the present invention include the polypeptide of SEQ ID NOS: 2 (in particular, the mature polypeptide) as well as polypeptides having at least 70% similarity (preferably at least 70% identity) to the polypeptide of SEQ ID NO. : 2 and preferably at least one similarity of 90% (more preferably at least 90% identity) to the polypeptide of SEQ ID NO: 2 and even more preferably at least one similarity of 95% (even more preferably at minus a 95% identity) to the polypeptide of SEQ ID NO: 2 and also includes portions of these polypeptides with these portions of the polypeptide that generally contain at least 30 amino acids and more preferably at least 50 amino acids. As is known in the art "similarity" between two polypeptides is determined by comparing the amino acid sequence and its conserved amino acid substitutes of a polypeptide to the sequence of a second polypeptide. The fragments or fractions of the polypeptides of the present invention can be used to produce the full-length polypeptide corresponding to the peptide synthesis, therefore, the fragments can be used as intermediates to produce the full-length polypeptides. The fragments or portions of the polynucleotides of the present invention can be used to synthesize full-length polynucleotides of the present invention. The present invention also relates to vectors that include polynucleotides of the present invention, host cells that are genetically engineered with vectors of the invention and the production of polypeptides of the invention by recombinant techniques. The host cells can be genetically engineered (transduced or transformed or transfected) with the vectors of this invention which can be, for example, a cloning vector or an expression vector. The vector can be, for example, in the form of a plasmid, a viral particle, a phage, etc. The managed host cells can be cultured in a modified conventional nutrient medium as appropriate for promoter activation, transformant selection or amplification of the FGF genes. The culture conditions, such as temperature, pH and the like are those previously used with the host cell selected for expression, and will be apparent to those skilled in the art. The polynucleotide of the present invention can be used to produce a polypeptide by recombinant techniques. Thus, for example, the polynucleotide sequence can be included in any of a variety of expression vehicles, in particular vectors or plasmids to express a polypeptide. These vectors include chromosomal, non-chromosomal and synthetic DNA sequences, eg, SV40 derivatives, bacterial plasmids, phage DNA, yeast plasmids; vectors derived from combinations of plasmids and phage DNA, viral DNA such as vaccinia, adenovirus, contagious epithelioma virus and pseudorabies. However, any other vector or plasmid can be used as long as they are replicable and viable in the host. The appropriate DNA sequence can be inserted into the vector by a variety of methods. In general, the DNA sequence is inserted into the restriction endonuclease sites, appropriate by procedures known in the art. These and other procedures are contemplated to be within the reach of those skilled in the art.
The DNA sequence in the expression vector is operably linked to an appropriate expression control sequence (s) (promoter) to direct mRNA synthesis. As representative examples of these promoters, there may be mentioned: the LTR or SV40 promoter, the lac or trp of E. Coli, the PL lambda phage promoter and other known promoters that control the expression of genes in prokaryotic or eukaryotic cells or their viruses. The special vector also contains a ribosome binding site for translation initiation and a transcription terminator. The vector may also include appropriate sequences to amplify the expression. In addition, expression vectors preferably contain a gene to provide a phenotypic trait for the selection of transformed host cells such as dihydrofolate reductase or neomycin resistance for the culture of eukaryotic cells, or such as resistance to tetracycline or ampicillin in E. Coli The vector containing the appropriate DNA sequence as described hereinbefore, as well as an appropriate promoter or control sequence, can be employed to transform an appropriate host to allow the host to express the protein. As representative examples of appropriate hosts, there may be mentioned: bacterial cells, such as E. coli, Salmonella, typhimurium, Streptomyces; fungal cells such as yeast; insect cells, such as Drosophila S2 and Spodoptera Sf9; animal cells such as CHO, COS or Bowes melanoma; adenovirus; plant cells, etc. The selection of an appropriate host is thought to be within the reach 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 described above in the broadest manner. The constructs comprise a vector, such as a plasmid or viral vector, in which a sequence of the invention has been inserted, in a direct or inverse orientation. In a preferred aspect of this embodiment, the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the sequence. Large numbers of suitable vectors and promoters are known to those skilled in the art and are commercially available. The following vectors are provided by way of example. Bacterial: pQE70, pQE60, pQE-9 (Qiagen), pBS, phagescript, psiX174, pBluescript SK, pBsKS, pNH8a, pNH16a, pNHlda, pNH46a, (Stratagene); pTRC99A,? KK223-3, pKK233-3, pDR540, pRIT5 (Pharmacia). Eukaryotic: pWLneo, pSV2cat, pOG44, pXTl, pSG (Stratagene) pSVK3, pBPV, pMSG, sSVL (Pharmacia). However, any other plasmid or vector can be used as long as they are replicable and viable in the host. The promoter regions can be selected from any desired gene using CAT (chloramphenicol transferase) vectors or other vectors with selectable markers. Two appropriate vectors are pKK232-8 and pCM7. The named, particular bacterial promoters include lacl, lacZ, T3, T7, gpt, lambda PR PL and trp. Eukaryotic promoters include immediate early CMV, HSV thymidine kinase, early and late SV40, retrovirus LTR, and mouse metallothionein-I. The selection of the appropriate vector and promoter is also within the level of one skilled in the art. In a further embodiment, the present invention relates to host cells containing the constructions described above. 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. The introduction of the construction into the host cell can be effected by transfection with calcium phosphate, transfection mediated by DEAE-Dextran, or electroporation (Davis, L., Dibner, M., Battey, I., Basic Methods in Molecular Biology, (1986)). Constructs in the 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 produced synthetically by conventional peptide synthesizers. Mature proteins can be expressed in mammalian cells, yeast, bacteria or other cells under the control of appropriate promoters. Cell-free translation systems can also be employed to produce those proteins using RNAs derived from the DNA constructs of the present invention. Suitable cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrook, et al., Molecular Cloning: A Laboratory Manual, Second Edition, (Col Spring Harbor, NY, 1989), the description of which is incorporated herein by reference.
The transcription of a DNA encoding the polypeptides of the present invention by higher eukaryotes is increased by inserting an enhancer sequence into the vector. Enhancers are cis-action elements of DNA, usually about 10 to 300 bp, that act on a promoter to increase its transcription. Examples include the SV40 enhancer on the anterior side of the Origin of replication (bp 100 to 270), an early promoter enhancer of cytomegalovirus, a polyoma enhancer on the anterior side of the replication origin, and adenovirus enhancers. In general, recombinant expression vectors will include origins of replication and selectable markers that allow the transformation of the host cell, for example, the E. coli ampicillin resistance gene of the S. cerevisiae TRP1 gene, and a derivative promoter. of a gene highly expressed to direct the transcription of a structural sequence in the 3 'direction. These promoters can be derived from operons that code for glycolytic enzymes such as 3-phosphoglycerate kinase (PGK), a-factor, acid phosphatase, or heat shock proteins, among others. The heterologous structural sequence is assembled in the appropriate phase with the translation, initiation and termination sequences, and preferably, a guiding sequence capable of directing the secretion of the translated protein into the periplasmic space or the extracellular medium. Optionally, the heterologous sequence can encode a fusion protein that includes an n-terminal identification peptide that imparts the desired characteristics, for example, stabilization or simplified purification of the expressed recombinant product. Expression vectors useful for bacterial use are constructed by inserting a DNA, structural sequence encoding a desired protein together with translation, initiation and termination signals, suitable in the operable reading phase with a functional promoter. The vector will contain one or more selectable, phenotypic markers and an origin of replication to ensure maintenance of the vector and to provide, if desired, amplification within the host. Prokaryotic hosts suitable for transformation include E. Coli, Bacillus subtilis, Salmonella typhimurium and several species within the genera Pseudomonas, Streptomyces and Stapohylococcus, although others may also be used as a lesson material.
As a representative but not limiting example, expression vectors useful for bacterial use may comprise a selectable marker and the bacterial origin of replication derived from commercially available plasmids comprising genetic elements of the well-known cloning vector pBR322 (ATCC 37017). These commercial vectors include, for example, pKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and GEMI (Promega Biotec, Madison, Wl, USA). These "skeleton" sections of pBR322 are combined with an appropriate promoter and the structural sequence to be expressed. After transformation of a suitable host strain and growth of the host strain to an appropriate cell density, the selected promoter is derepressed by an appropriate means (e.g., temperature change or chemical induction) and the cells are cultured for a time. additional. The cells are typically collected by centrifugation, broken by physical or chemical means, and the resulting crude extract is preserved for further purification. The microbial cells used in protein expression can be broken by any convenient method, which includes the freeze-thaw cycle, sound treatment, mechanical breakdown, or the use of cell lysis agents. Various mammalian cell culture systems may also be employed to express the recombinant protein. Examples of mammalian expression systems include COS-7 lines of monkey kidney fibroblasts, described by Gluzman, Cell, 23: 175 (1981), and other cell lines capable of expressing a compatible vector, for example, the cell lines C127, 3T3, CHO, HeLa and BHK. The mammalian expression vectors will contain a suitable origin of replication, a promoter and enhancer, and also any of the necessary ribosome binding sites, polyadenylation site, donor and binding acceptor sites, transcriptional termination sequences, and non-specific sequences. transcribed flanking 5 '. The DNA sequences derived from the SV40 viral genome, eg, SV40 origin, early promoter, enhancer, junction or splice and polyadenylation sites can be used to provide the required non-transcribed genetic elements. The polypeptide of the present invention can be recovered and purified from recombinant cell cultures by methods used to date, including precipitation with ammonium sulfate or ethanol, acid extraction, anionic or cation exchange chromatography, phosphocellulose chromatography , chromatography with hydrophobic interaction, affinity chromatography, hydroxyapatite chromatography and lectin chromatography. The steps of protein refolding can be used, as necessary, in the termination of the mature protein configuration. Finally, high performance liquid chromatography (HPLC) can be used for the final purification steps. The polypeptide of the present invention may be a naturally purified product, or product of synthetic chemical procedures, produced by recombinant techniques from a prokaryotic or eukaryotic host (eg, by bacterial cells of yeast, higher plants, insects and mammals in culture). Depending on the host employed in the recombinant production method, the polypeptides of the present invention can be glycosylated with mammalian or other eukaryotic carbohydrates or can be non-glycosylated. The polypeptides of the invention may also include an initial methionine amino acid residue.
The polypeptide of the present invention, as a result of the ability to stimulate the growth of endothelial, vascular cells, can be employed in the treatment to stimulate revascularization of ischemic tissues due to various disease conditions such as thrombosis, arteriesclerosis, and other conditions cardiovascular These polypeptides can also be used to stimulate engiogenesis and limb regeneration. - The polypeptide can also be used to treat wounds due to damage, burns, repair of post-operative tissue, and ulcers since they are mitogenic to several cells of different origins, such as fibroblast cells and skeletal muscle cells, therefore facilitate the repair or replacement of damaged or diseased tissue. The polypeptide of the present invention can also be used to stimulate neuronal growth and treat and prevent neuronal damage associated with stroke and which occurs in certain neuronal disorders or neurodegenerative conditions such as Alzheimer's disease, Parkinson's disease, and complexes related to AIDS. FGF-14 has the ability to stimulate the growth of chondrocytes, therefore, it can be used to improve bone and periodontal regeneration and aid in tissue transplants or bone grafts. The polypeptide of the present invention can also be used to prevent skin aging due to sunburn by stimulating the growth of keratinocytes. The FGF-14 polypeptide can also be used to prevent hair loss, since members of the FGF family activate the hair-forming cells and promote the growth of the melanocytes. Along with the same lines, the polypeptides of the present invention can be used to stimulate the growth and differentiation of hematopoietic cells and bone marrow cells when used in combination with other cytokines. The FGF-14 polypeptide can also be used to maintain the organs before transplantation or to support cell culture of primary tissues. The polypeptide of the present invention can also be used to induce tissue of mesodermal origin to differentiate into early embryos. According to yet a further aspect of the present invention, there is provided a process for using these polypeptides, or polynucleotides encoding these polypeptides, for in vitro purposes related to scientific research, DNA synthesis, manufacture of DNA vectors and for the purpose of providing diagnostics and therapeutic products for the treatment of human disease. This invention provides a method for the identification of the receptors for the polypeptides of the present invention. The genes encoded for the receptor can be identified by numerous methods known to those skilled in the art, for example, ligand washing and FACS classification (Coligan, et al., Current Protocols in Immun., 1 (2), Chapter 5 , (1991)). Preferably, expression cloning is employed wherein the polyadenylated RNA is prepared from a cell responsive to the polypeptides, for example, NIH3T3 cells that are known to contain multiple receptors for the FGF family proteins, and cells SC-3, and a cDNA library created from this RNA is divided into mixtures and used to transfect COS cells or other cells that are not sensitive to the polypeptides. Transfected cells that are grown on glass slides are exposed to the polypeptide of the present invention, after they have been labeled. The polypeptides can be labeled by a variety of means including iodination or inclusion in a recognition site for a site-specific protein kinase. After fixation and incubation, the slides are subjected to autoradiographic analysis. Positive mixtures are identified and sub-mixtures are prepared and re-transfected using an interactive sub-mixing and re-examination process, eventually producing individual clones that encode the putative receptor. As an alternative approach for the identification of the receptor, the tagged polypeptides can be linked by photoaffinity with the cell membrane or extract preparations expressing the receptor molecule. The crosslinked material is redissolved by PAGE analysis and exposed to an X-ray film. The labeled labeled complex containing the polypeptide receptors can be excised, redissolved in peptide fragments, subjected to protein microsequencing. The amino acid sequence obtained from the microsequencing will be used to design a set of degenerate oligonucleotide probes to examine a cDNA library or library to identify the genes encoding the putative receptors. This invention provides a method for screening compounds to identify those that modulate the action of the polypeptide of the present invention. An example of this assay comprises combining a fibroblast, mammalian cell, the polypeptide of the present invention, the compound to be examined and 3 [H] thymidine under cell culture conditions where the fibroblast cells would normally proliferate. A control assay can be performed in the absence of the compound to be examined and compared to the amount of proliferation of the fibroblast in the presence of the compound to determine whether the compound stimulates proliferation by determining the admission of 3 [H] thymidine in each case. The amount of fibroblast cell proliferation is measured by liquid scintillation chromatography which measures the incorporation of 3 [H] thymidine. The compounds, both agonists and antagonists, can be identified by this procedure. In another method, a membrane or mammalian cell preparation that expresses a receptor for a polypeptide of the present invention is incubated with a labeled polypeptide of the present invention in the presence of the compound. The ability of the compound to improve or block this interaction could then be measured. Alternatively, the response of a second known messenger system is measured after the interaction of a compound to be examined and the FGF-14 receptor, and the ability of the compound to bind to the receptor and produce a second messenger response is measured. to determine if the compound is a potential agonist or antagonist. These second messenger systems include but are not limited to cAMP guanylate cyclase, ion channels, phosphorylation with tyrosine or hydrolysis with phosphoinositide. Examples of antagonist compounds include antibodies, or in some cases, oligonucleotides, which bind to the receptor for the polypeptide of the present invention so as not to produce a second messenger response or to bind to the FGF-14 polypeptide itself. Alternatively, a potential antagonist may be a mutant form of the polypeptide that binds to the receptors, however, the second messenger response does not occur and therefore, the action of the polypeptide is effectively blocked.
Another antagonist compound for the FGF-14 gene and the gene product is an antisense construct prepared using the antisense technology. Antisense technology can be used to control gene expression through triple helix formation or DNA or antisense RNA, both methods that rely on the binding of a polynucleotide to DNA or RNA. For example, the 5 'coding portion of the polynucleotide sequence, which codes for the mature polypeptides of the present invention, is used to design an antisense RNA oligonucleotide of about 10 to 40 base pairs in length. A DNA oligonucleotide is designed to be complementary to a region of the gene comprised in transcription, (triple helix, see Lee et al., Nucí Acids Res., 6: 3073 (1979); Cooney et al., Science, 241: 456 (1988), and Dervan et al., Science, 251: 1360 (1991)), thereby having the transcription and production of the polypeptides of the present invention. The antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks the translation of the mRNA molecule into the polypeptide (Antisense - Okano, J. Neurochem., 56: 560 (1991); Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression, CRC Press, Boca Ratón, FL (1988)). The oligonucleotides described above can also be delivered to cells such that the antisense RNA or DNA can be expressed in vivo for the inhibition of polypeptide production. Potential antagonist compounds also include small molecules that bind and occupy the binding site of the receptors, thereby rendering the receptor inaccessible to their polypeptide such that normal biological activity is prevented. Examples of small molecules include, but are not limited to, small peptides or peptide-like molecules. Antagonist compounds can be employed to inhibit cell growth and proliferation effects of the peptides of the present invention from neoplastic cells and tissues, i.e., the stimulation of tumor angiogenesis, and therefore, the retardation or prevention of growth. and abnormal cell proliferation, for example, in the formation or growth of tumors. Antagonists can also be used to prevent hypervascular diseases, and to prevent the proliferation of epithelial crystalline cells after extracapsular cataract surgery. The prevention of mitogenic activity of the polypeptides of the present invention may also be desirable in cases such as restenosis after balloon angioplasty. Antagonists can also be used to prevent the growth of healing tissue during wound healing. Antagonists can be employed in a composition with a pharmaceutically acceptable carrier, for example, as described hereinafter. The polypeptides, agonists and antagonists of the present invention can be used in combination with a suitable pharmaceutical carrier to comprise a pharmaceutical composition for parenteral administration. These compositions comprise a therapeutically effective amount of the polypeptide, agonist or antagonist and a pharmaceutically acceptable carrier or excipient. This carrier includes but is not limited to saline, buffered saline, dextrose, water, glycerol, ethanol and combinations thereof. The formulation must be appropriate to the mode of administration. The invention also provides a pharmaceutical package or equipment comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention. Associated with this (these) recipient (s) may be a notice in the form prescribed by the government agency that regulates the manufacture, use or sale of pharmaceutical products or biological products, notice that reflects approval by the manufacturing agency, or use or sale for human administration. In addition, the polypeptides, agonists and antagonists of the present invention can be used in conjunction with other therapeutic compounds. The pharmaceutical compositions can be administered in a convenient manner such as by the oral, typical, intravenous, intraperitoneal, intramuscular, subcutaneous, intranasal or intradermal routes. The pharmaceutical compositions are administered in an amount that is effective to treat and / or for the prophylaxis of the specific indication. In general, they are administered in an amount of at least about 10 μg / kg of body weight and in most cases will be administered in an amount not to exceed about 8 mg / kg of body weight per day. In most cases, the dose is from about 10 μg / kg to about 1 mg / kg of body weight per day, taking into account the routes of administration, symptoms, etc. In the specific case of topical administration, the doses are preferably administered from about 0.1 μg to 9 mg per cm2. The polypeptide of the invention and the agonist and antagonist compounds which are polypeptides, can also be employed according to the present invention by the expression of this polypeptide in vivo, which is often referred to as a "gene therapy". In this way, for example, the cells can be engineered with a polynucleotide (DNA or RNA) encoding the polypeptide displayed, the managed cells are then provided to a patient to be treated with the polypeptide. These methods are well known in the art. For example, the cells can be engineered by procedures known in the art by the use of retroviral particles containing the RNA encoding the polypeptide of the present invention. Similarly, the cells can be engineered in vivo for polypeptide expression in vivo, for example, by procedures known in the art. As is known in the art, a producer cell for producing a retroviral particle containing the RNA encoding the polypeptide of the present invention can be administered to a patient by engineered cells in vivo and expression of the polypeptide in vivo. These and other methods for administering a polypeptide of the present invention by these methods should be apparent to those skilled in the art from the teachings of the present invention. For example, the expression vehicle for the engineered cells can be different from a retroviral particle, for example, an adenovirus, which can be used to engineered cells in vivo after combination with a suitable delivery vehicle. Retroviruses from which the retroviral plasmid vectors mentioned hereinabove can be derived, include, but are not limited to, Moloney murine leukemia virus, vessel necrosis virus, retrovirus such as Rous Sarcoma virus. , Harvert sarcoma virus, bird leukosis virus, Gibbon monkey leukemia virus, human immunodeficiency virus, adenovirus, Myoloproliferative sarcoma virus, and mammary tumor virus. In one embodiment, the retroviral plasmid vector is derived from Moloney murine leukemia virus. The vector includes one or more promoters. Suitable promoters that may be employed include, but are not limited to, retroviral LTR; the SV40 promoter; and the human cytomegalovirus (CMV) promoter described in Miller, et al., Biotechniques, Vol. 7. No. 9, 980-990 (1989), or any other promoter (eg, cellular promoters such as eukaryotic cell promoters that include , but are not limited to, the promoters of histone, pol III, and β-actin). Other viral promoters that may be employed include, but are not limited to, adenovirus promoters, thymidine kinase (TK) promoters, and parvovirus B19 promoters. The selection of a suitable promoter will be apparent to those skilled in the art from the teachings contained herein. The nucleic acid sequence encoding the polypeptide of the present invention is under the control of a suitable promoter. Suitable promoters that can be employed, include, but are not limited to, adenovirus promoters, such as the anterior, main, adenoviral promoter; or heterologous promoters, such as the cytomegalovirus (CMV) promoter; the promoter of the respiratory syncytial virus (RSV, for its acronym in English); inducible promoters, such as the MMT promoter, the metallothionein promoter; heat stroke promoters; the albumin promoter, the ApoAI promoter; human globin promoters; viral thymidine kinase promoters, such as the simple herpes thymidine kinase promoter; Retroviral LTRs (including the retroviral, modified LTRs described above); the β-actidine promoter; and the promoters of human growth hormone. The promoter may also be the native promoter that controls the gene encoding the polypeptide. The retroviral plasmid vector is used to transduce packaging cell lines to form producer cell lines. Examples of cell lines that can be transfected include, but are not limited to, PE501 cell lines., PA317,? -2,? -AM, PA12, T19-14X, VT-19-17-H2,? CRE,? CRIP, GP + E-86, GP + envAml2, and DAN, as described in Miller Human Gene Therapy, Vol. 1, pgs. 5-14 (1990), which is incorporated herein by reference in its entirety. The vector can transduce the packaging cells through any means known in the art. This medium includes, but is not limited to, electroporation, the use of liposomes, and CaP04 precipitation. In an alternative, the retroviral plasmid vector can be encapsulated in a liposome, or coupled to a liquid, and then administered to a host. The producer cell line generates retroviral vector, infectious vectors that include the nucleic acid sequence (s) encoding the polypeptides. These retroviral vector particles can then be used to transduce eukaryotic cells, either in vitro or in vivo. The eukaryotic cells transduced will then give the sequence (s) of nucleic acids that qualify for the polypeptide. Eukaryotic cells that can be transduced include, but are not limited to, embryonic stem cells, embryonic carcinoma cells, as well as hematopoietic hemocytoblasts, hepatocytes, fibroblasts, myoblasts, keratinocytes, endothelial cells and bronchoal epithelial cells. This invention also relates to the use of genes of the present invention as part of a diagnostic assay for detecting diseases or susceptibility to diseases related to the presence of mutations in the nucleic acid sequences encoding the polypeptide of the present invention.
Individuals having mutations in a gene of the present invention can be detected at the DNA level by a variety of techniques. Nucleic acids for diagnosis can be obtained from the cells of a patient, such as blood, urine, saliva, tissue biopsy and autopsy material. Genomic DNA can be used directly for detection or can be amplified enzymatically by using PCR (Saiki et al., Nature, 324: 163-166 (1986)) before analysis. RNA or cDNA can also be used for the same purpose. As an example, PCR primers complementary to the nucleic acid encoding a polypeptide of the present invention can be used to identify and analyze the mutations. For example, deletions and insertions may be attempted by a change in the size of the amplified product compared to the normal genotype. The point mutations can be identified by hybridizing the amplified DNA to the radio labeled RNA or alternatively, the radio-labeled antisense, DNA sequences. The perfectly matched sequences can be distinguished from the doubles uneven by pancreatic ribonuclease digestion or by differences in the melting temperatures.
Genetic testing based on differences in DNA sequences can be achieved by detecting the alteration in the electrophoretic mobility of DNA fragments and gels with or without denaturing agents. Small deletions or sequence insertions can be visualized by high resolution gel electrophoresis. DNA fragments of different sequence can be distinguished in denaturing formamide gradient gels in which the mobilities of the different DNA fragments are delayed in the gel at different positions according to their partial melting or specific melting temperatures ( see, for example, Myers et al., Science, 230: 1242 (1985)). Sequence changes at specific locations can also be revealed by nuclease protection assays, such as protection with pancreatic ribonuclease and SI or the chemical cleavage method (eg, Cotton et al., PNAS, USA, 85: 4397-4401 ( 1985)). In this way, detection of a specific DNA sequence can be achieved by methods such as hybridization, pancreatic ribonuclease protection, chemical cleavage, direct DNA sequencing or the use of restriction enzymes (eg, Restriction Fragment Length Polymorphisms (RFLP)). ) and transfer by Southern adsorption of the genomic DNA. In addition to more conventional gel electrophoresis and DNA sequencing, mutations can also be detected by in situ analysis. The present invention also relates to a diagnostic assay for detecting altered levels of FGF-14 proteins in various tissues due to overexpression of the proteins compared to normal control tissue samples can detect the presence of abnormal cell proliferation, for example, a tumor. The assays used to detect levels of protein in a sample derived from a host are well known to those skilled in the art and include radioimmunoassays, competitive binding assays, Western blot analysis, ELISA assays and "sandwich" assays. an ELISA assay (Coligan, et al., Current Protocols in Immunology, 1 (2), Chapter 6, (1991)) comprises initially preparing an antigen-specific antibody for the polypeptides of the present invention, preferably a monoclonal antibody. In addition, an indicator antibody against the monoclonal body is prepared. A detectable reagent such as radioactivity, fluorescence, or, in this example, a horseradish peroxidase enzyme is attached to the indicator antibody. A sample of a host is removed and incubated on a solid support, for example, a polystyrene disk, which binds the proteins in the sample. Any of the free protein binding sites on the disk are then covered by incubating with a bovine serum albumin similar to non-specific protein. Then, the monoclonal antibody is incubated in the disk, during which time the monoclonal antibodies bind to any of the polypeptides of the present invention attached to the polystyrene disk. All unbound monoclonal antibody is washed with buffer. The reporter antibody bound to horseradish peroxidase is now placed on the disc resulting in the binding of the reporter antibody to any monoclonal antibody bound to the protein of interest. The unbound reporter antibody is then washed. The peroxidase substrates are then added to the disk and the amount of color developed in a given period of time is a measure of the amount of a polypeptide of the present invention present in a given volume of a patient sample when compared against a curve standard.
A competition assay may be employed wherein antibodies specific for a polypeptide of the present invention bind to a solid support and labeled FGF-13 and a sample derived from the host are passed on a solid support and the amount of the label detected, by For example, by liquid scintillation chromatography, an amount of a polypeptide of the present invention can be correlated with the sample. - A "sandwich" assay is similar to an ELISA assay. In a "sandwich" assay a polypeptide of the present invention is passed over a solid support and bound to an antibody bound to the solid support. It then binds a second antibody to the polypeptide of interest. A third antibody that is labeled and specific to the second antibody is then passed over the solid support and binds to the second antibody and then an amount can be quantified. The sequences of the present invention are also valuable for the identification of chromosomes. The sequence is specifically designated and can hybridize to a particular location on an individual human chromosome. In addition, there is a current need to identify the particular sites on the chromosome. Currently few chromosome labeling reagents are available based on current sequence data (repeat polymorphism) to mark the chromosomal location. The correlation of the DNAs to the chromosomes according to the present invention is an important first step in the correlation of those sequences with genes associated with disease. Briefly, the sequences can correlate to the chromosomes when preparing PCR primers (preferably 15-25 bp) of the cDNA. Computer analysis of the 3 'untranslated region is used to rapidly select the primers so that it encompasses more than one exon in the genomic DNA, thereby complicating the amplification process. These primers are then used for the PCR examination of somatic cell hybrids containing individual human chromosomes. Only those hybrids that contain the human gene that corresponds to the primer will produce an amplified fragment. The PCR correlation of somatic cell hybrids is a rapid process for assigning a particular DNA to a particular chromosome. Using the present invention with the same oligonucleotide primers, sublocalization can be accomplished with panels of specific chromosome fragments or mixtures of large genomic clones in an analogous manner. Other correlation strategies that can be used similarly to correlate to their chromosome include in situ hybridization, pre-examination with flow-labeled chromosomes, labeling and prescreening by hybridization to construct chromosome-specific cDNA libraries. Fluorescence in situ hybridization (FISH) from a cDNA clone to a chromosomal metaphase spread can be used to provide a precise chromosomal location in one step. This technique can be used with cDNA as short as 50 or 60 bases. For a review of this technique see Verma et al., Human Chromosomes: a Manual of Basic Techniques, Pergamon Press, New York (1988). Once a sequence has been correlated to a precise chromosomal location, the physical position of the sequence on the chromosome can be correlated with the data on the genetic map. (These are found, for example, in V McKusick, Mendelian Inheritance in Man (available online through the Johns Hopkins University Welch Medical Library.) The relationship between genes and diseases that have been correlated to the same chromosomal region is then identify through linkage analysis (common inheritance of physically adjacent genes) .Then, it is necessary to determine the differences in the cDNA or genomic sequence between affected and unaffected individuals.If a mutation is observed in some or all individuals affected, but not in any of the normal individuals, then the mutation will probably be the agent that provokes the disease. With the current resolution of the physical correlation and genetic correlation techniques, a cDNA precisely localized to a chromosomal region associated with the disease could be one of 50 to 500 potential, provocative genes. (This assumes the correlation resolution of 1 megabase 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 antibodies therefor. 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 fragments, or the product of a Fab expression library. Various methods known in the art can be used for the production of these antibodies and fragments. Antibodies raised against 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 not a human. The antibody thus obtained will then bind to the polypeptides themselves. In this way, a sequence encoding only a fragment of the polypeptides can still be used to generate the antibodies that bind to the complete native polypeptides. These antibodies can then be used to isolate the polypeptide from the tissue expressing that polypeptide. For the preparation of monoclonal antibodies, any technique that provides antibodies produced by continuous cell line cultures can be used. Examples include the hybridoma technique (Kohler and Milstein, 1975, Nature, 256: 495-497), the trioma technique, the human B-cell hybridoma technique (Kozbor et al., 1983, Immunology Today 4:72), and the EBV-hybridoma technique for producing human monoclonal antibodies (Colé, et al., 1985, in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96). The techniques described for the production of single chain antibodies (US Patent No. 4,946,778) can be adapted to produce single chain antibodies for the immunogenic polypeptide products of this invention. Also, transgenic mice can be used to express the humanized antibodies to the immunogenic polypeptide products of this invention. The present invention will be further described with reference to the following examples; however, it should be understood that the present invention is not limited to these examples. All parts or quantities, unless otherwise specified, are by weight. In order to facilitate understanding of the following examples, certain methods and / or terms that occur frequently will be described. "Plasmids" are designated by a lowercase letter p preceded and / or followed by uppercase letters and / or numbers. The plasmids of initiation herein are either commercially available, publicly available on an unrestricted base, or can be constructed from available plasmids according to published procedures. In addition, plasmids equivalent to those described are known in the art and will be apparent to the person skilled in the art. "Digestion" of DNA refers to the catalytic cleavage of DNA with a restriction enzyme that acts only in certain sequences in DNA. The various restriction enzymes used herein are commercially available and their conditions of attraction, with factors and other requirements were used as would be known to the person skilled in the art. For analytical purposes, 1 μg of DNA plasmid with approximately 2 units of enzyme in approximately 20 μl of buffer was typically used. For the purpose of isolating the DNA fragments for the construction of plasmids, typically from 5 to 50 μg of DNA are digested with 20 to 250 units of enzyme in a larger volume. Suitable buffers and substrate amounts for the particular restriction enzymes are specified by the manufacturer. Incubation times of approximately 1 hour at 37 ° C are commonly used, but may vary according to the supplier's instructions. After digestion, the reaction is subjected to electrophoresis directly on a polyacrylamide gel to isolate the desired fragment. Size separation of the incision fragments is performed using the 8% polyethylated go gel described by Goeddel, D., et al. Nucleic Acids Res., 8: 4057 (1980). The "oligonucleotides" refer to either an individual-stranded polydeoxynucleotide or two complementary polydeoxynucleotide strands that can be chemically synthesized. These synthetic oligonucleotides do not have 5'-phosphate and thus will not be ligated to another oligonucleotide without adding a phosphate with an ATP in the presence of a kinase. A synthetic oligonucleotide will bind to a fragment that has not been dephosphorylated. "Ligation" refers to the process of forming phosphodiester linkages between two double-stranded acetic acid fragments (Maniatis, T., et al., Id., P.146). Unless provided otherwise, ligation can be achieved using known buffers with 10 units of T4 DNA ligase ("ligase") per 0.5 μg of approximately equimolar amounts of the DNA fragments to be ligated.
Unless stated otherwise, the transformation is performed as described by the method of Graham, F and Van der Eb, A., Virology, 52: 456-457 (1973).
Example 1 Bacterial Expression and Purification of the FGF-14 Protein The DNA sequence that encodes ATCC # 97147 of FGF-14, is initially amplified using PCR oligonucleotide primers that correspond to the 5 'sequence of the processed protein (minus the signal from the peptide sequence) and the 3' vector sequences to the gene. Additional nucleotides corresponding to the gene are added to the 5 'and 3' sequence. The 5 'oligonucleotide primer has the 5' sequence GCCAGAGCATGCAGCGGCGCGTGTGTCCCCGC 3 '(SEQ ID NO: 3) and contains a Sph1 restriction enzyme site. The 3 ', 5' sequence GCCAGAAGATCTGGGGGCAGGGGGACTGGAAGG 3 '(SEQ ID NO: 4) contains sequences complementary to a BglII site followed by 21 nucleotides of the coding sequence of FGF-14. The restriction enzyme sites correspond to the restriction enzyme sites in the bacterial expression vector pQE70 (Qiagen, Inc. Chatsworth, CA 91311). pQE-70 codes for antibiotic resistance (Ampr), a bacterial origin of replication (ori), a promoter operator (P / 0) regulable with IPTG, a ribosome binding site (RBS), a 6-His fragment and restriction enzyme sites. pQE-70 was then digested with Ncol and BglII. The amplified sequences were ligated into pQE-70 and inserted into the structure with the sequence encoding the histidine fragment and the ribosome binding site (RBS). The ligation mixture is then used to transform E. coli strain M15 / rep 4 (Qiagen, Inc.) by the procedure described in Sambrook, J. et al., Molecular Cloning: A Laboratory Manual, Cold Spring Laboratory Press, (1989). M15 / rep4 contains multiple copies of the pREP4 plasmid that expresses the lacl repressor and also confers resistance to kanamycin (Kanr). Transformants are identified by their ability to grow on LB plates and are selected in colonies resistant to ampicillin / kanamycin. The plasmid DNA is isolated and confirmed by restriction analysis. Clones containing the desired constructs are grown overnight (O / N) in liquid culture in LB medium supplemented with both Amp (100 μg / ml) and Kan (25 μg / ml). The O / N culture is used to inoculate a large crop at a ratio of 1: 100 to 1: 250. The cells are cultured at an optical density of 600 (O.D.600) of between 0.4 A 0.6. then IPTG ("Isopropyl-B-D-thiogalacto pyranoside") is added to a final concentration of 1 mM. IPTG is induced by inactivating the lacl repressor, cleansing the P / O guide to increased gene expression. The cells are grown for an additional 3 to 4 hours. The cells are then harvested by centrifugation. The cell pellet is solubilized in the chaotropic agent Guanidine HCl 6 Molar. After clarification, solubilized FGF-14 is purified from this solution by nickel-chelate column chromatography under conditions that allow for safe binding by proteins containing the 6-His fragment (Hochuli, E. et al., J. Chromatography 411: 177-184 (1984)). The proteins are eluted from the column in 6 molar guanidine-HCl, pH 5.0 and for the purpose of renaturation, it is adjusted to 3 molar guanidine-HCl, 100 mM sodium phosphate, 10 molar glutadione (reduced) and 2 mmolar glutathione (rusty) After incubation in this solution for 12 hours the proteins are dialyzed to 10 molar sodium phosphate.
EXAMPLE 2 Cloning and expression of FGF-14 using the baculovirus expression system The DNA sequence encoding the full-length FGF-14 protein, ATCC # 97147, is amplified using the PCR oligonucleotide primers corresponding to the 5 'and 3' sequence of the gene: The FGF-14 primer 'has the sequence 5' CTAGTGGATCCCATCATGGCGGCGCTGGCCAGT 3 '(SEQ ID NO: 5) for the vector pA2 and contains a BamHI restriction enzyme site (fresh) followed by 4 nucleotides which appear to be an efficient signal for the initiation of translation of the eukaryotic cells (Kozak, M., J. Mol. Biol., 196: 947-950 (1987) which fits behind the first 18 nucleotides of the gene (the initiation codon for the production of "ATG" is accentuated). For the vector pA2gp the 5 'primer has the sequence 5' CGACTGGATCCCCAGCGGCGCGTGTGTCCC 3 '(SEQ ID NO: 6). The 3 'primer has the sequence 5' CGACTTCTAGAATCAGGGGGCAGGGGGACTGGA 3 '(SEQ ID NO: 7) and contains the site of restriction endonuclease Xbal (fresh) and 22 nucleotides complementary to the 3' untranslated sequence of the gene.
The amplified sequences are isolated from a 1% agarose gene using commercially available equipment ("Geneclean," BIO 101 Inc., La Jolla, Ca.). The fragment is then digested with the respective endonucleases again purified on a 1% agarose gel. This fragment is designated F2. The vector pA2gp (and pA2) (modifications of vector pVL941, discussed below) is used for the expression of proteins using the baculovirus expression system (for review see: Summers, MD and Smith, GE 1987, A manual of methods for baculovirus vectors and insect cell culture procedures, Texas Agricultural Experimental Station Bulletin No. 1555). That expression vector contains the strong polyhedrin promoter of autologous caliform nuclear polyhedrosis virus (AcMNPV) followed by the recognition sites for the restriction endonucleases BamHI and Xbal. The polyadenylation site of simian virus (SV) 40 is used for efficient polyadenylation. For easy selection of the recombinant virus, the beta-galactosidase gene from E. coli is inserted in the same orientation as the polyhedrin promoter followed by the polyadenylation signal of the polyhedrin gene. The polyhedrin sequences are flanked on both sides by viral sequences by homologous recombination deviated by cells from the wild-type, co-transfected viral DNA. Many other bacoluvirus vectors could be used in place of pA2 such as pRGl, pAc373, pVL941 and pAcIMl (Luckow, V.A. and Summers, M.D., Virology, 170: 31-39). The plasmid is digested with restriction enzymes and dephosphorylated using calf intestinal phosphate by procedures known in the art. The DNA is then isolated from a 1% agarose gel using commercially available equipment ("Geneclean" BIO 101 Inc., La Jolla, Ca.). This DNA vector is designated V2. The fragment F2 and the dephosphorylated plasmid V2 are ligated with T4 DNA ligase. DH5a cells of E. coli are then transformed and the identified bacteria containing the plasmid (pBacFGF-14) using the respective restriction enzymes. The sequence of the cloned fragment is confirmed by DNA sequencing. 5 μg of plasmid pBacFGF-14 is co-transfected with 1.0 μg of commercially available linearized baculovirus ("BaculoGold ™ Baculovirus DNA", Pharmingen, San Diego, CA.) using a lipofection method (Felgner et al. Proc. Nati. Acad. Sci USA, 84: 7413-7417 (1987)). 1 μg of the BaculoGold ™ virus DNA and 5 μg of the plasmid are mixed in a sterile well of microtiter plates containing 50 μl of serum free Grace medium (Life Technologies Inc., Gaithersburg, MD). Then add 10 μl of Lipofectin plus 90 μl of Grace's medium, mix and incubate for 15 minutes at room temperature. Then, the transfection mixture is added dropwise to Sf9 insect cells (ATCC CRL 1711) seeded in 35 mm tissue culture plates with 1 ml of Grace's medium without serum. The plates are rocked back and forth to mix the recently added solution. The plates are then incubated for 5 hours at 27 ° C. After 5 hours, the transfection solution is removed from the plate and 1 ml of Grace's insect medium supplemented or supplemented with 10% fetal calf serum is added. Plates are placed back in an incubator and culture is continued at 27 ° C for four days. After four days, the supernatant is collected and plaque assays performed without looking as described by Summer and Smith (supra). As a modification, an agarose gel with "Blue Gal" is used (Life Technologies Inc., Gaithersburg) that allows easy isolation of the blue-bonded plates. (A detailed description of a "plaque assay" can also be found in the user guide for the cultivation of insect cells and baculovirology distributed by Life Technologies Inc., Gaithersburg, page 9-10). Four days after the serial dilution, the virus is added to the cells and the plates stained blue are ordered with the tip of an Eppendorf pipette. The agar containing the recombinant viruses is then redispersed or suspended in an Eppendorf tube containing 200 μl of Grace's medium. The agar is removed by brief centrifugation and the supernatant containing the recombinant bacoluvirus is used to infect Sf9 cells seeded on 35 mm discs. Four days later, the supernatants of these culture discs are harvested and then stored at 4 ° C. Sf9 cells are cultured in Grace's medium supplemented with 10% heat-inactivated FBS. The cells are infected with the recombinant bacoluvirus V-FGF-14 at a multiplicity of infection (MOI) of 2. Six hours later, the medium is removed and replaced with the SF900 II medium minus methionine and cysteine (Life Technologies Inc. , Gaithersburg). 42 hours later, 5 μCi of 35S-methionine and 5 μCi of 35S cysteine (Amersham) are added. The cells are further incubated for 16 hours before they are harvested by centrifugation and the labeled proteins are visualized by SDS-PAGE and autoradiography.
EXAMPLE 3 Expression of Recombinant FGF-14 in COS Cells The expression of the plasmids FGF-14-HA derived from a vector pcDNA3 / Amp (Invitrogen) that contains: 1) the origin of replication of SV40, 2) the ampicillin resistance gene, 3) the origin of replication of E. coli , 4) the CMV promoter followed by a polylinker region, an SV40 intron and a polyadenylation site. The DNA fragments encode the complete FGF-14 precursor and an HA fragment fused in the structure to the 3 'end is cloned into the polylinker region of the vector, therefore, the expression of the recombinant protein is directed under the CMV promoter. The HA fragment corresponds to an epitope derived from the glutinin protein of influence as previously described (I. Wilson, H. Niman, R. Heighten, A. Cherenson, M. Connolly, and R. Lerner, 1984, Cell 37: 767, (1984)). Infusion of the HA fragment to the target protein allows the detection phase of the recombinant protein with an antibody that recognizes the HA epitope. The construction strategy of the plasmid is described as follows: The DNA sequence encoding FGF-14, ATCC # 97147, are constructed by PCR with two primers: the 5 'capstan, 5' CTAGTGGATCCCATCATGGCGGCGCTGGCCAGT 3 '(SEQ ID NO: 8) contains a BamHI site followed by 18 nucleotides of the coding sequence starting from the codon of initiation. The sequence 3 ', 5' GATTTACTCGAGGGGGGCAGGGGGACTGGA 3 '(SEQ ID NO: 9) contains sequences complementary to the Xhol site, the stop codon of translation, the fragment HA and the last 18 nucleotides of the coding sequence of FGF-14 (which does not include the stop codon). Therefore, the PCR product contains a BamHI site, the coding sequence followed by the HA fragment fused to the structure, a translation stop stop codon next to the HA fragment, and an XHol site. The DNA fragments amplified by PCR and the vector, pcDNA3 / Amp, are digested with the respective restriction enzymes and ligated. The ligation mixture is transformed into the SURE strain of E. coli (available from Stratagene Cloning Systems, La Jolla, CA 92037), the transformed culture is seeded into plates of the ampicillin medium and the resistant colonies are selected. The plasmid DNA is isolated from the transformants and examined by restriction analysis for the presence of a correct fragment. For the expression of the recombinant FGF-14 COS cells, they are transfected with the expression vector by the DEAE-DEXTRAN method (J. Sambrook, E. Fritsch, T. Maniatis, Molecular Cloning: A Laboratory Manual, Cold Spring Laboratory Press , (1989)). The expression of the FGF-14HA protein is detected by radiolabelling and the immunoprecipitation method (E. Harlow, D. Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, (1988)). The cells are labeled for 8 hours with 35S-cysteine, two days after transfection. The culture medium is then collected and the cells are smoothed with detergent (RIPA buffer (150 mM NaCl, 1% NP-40, 0.1% SDS, 1% NP-40, 0.5% DOC, 50 mM Tris, pH 7.5) (Wilson, I. et al., Id. 37: 767 ( 1984)). Both the cell lysate and the culture medium are precipitated with an HA-specific monoclonal antibody. Precipitated proteins are analyzed on 15% SDS-PAGE gels.
Example 5 Expression Via Gene Therapy Fibroblasts are obtained from a subject by skin biopsy. The resulting tissue is placed in the tissue culture medium and separated into small pieces. Small pieces of tissue are placed on a wet surface in a tissue culture flask, approximately ten pieces are placed in each flask. The flask is placed on its head, sealed tightly and left at room temperature overnight. After 24 hours at room temperature, the flask is inverted and the pieces of tissue remain fixed to the bottom of the flask and the fresh medium (for example, Ham's F12 medium, with 10% FBS, penicillin and streptomycin, is added). This is then incubated at 37 ° C for about a week. At this time, fresh medium is added and changed subsequently every several days. After a further two weeks in culture, a monolayer of fibroblasts emerges. The monolayer is trypsin and scaled in larger flasks. PMV-7 (Kirschemeier, PT et al., DNA, 7: 219-25 (1988) flanked by long terminal repeats of Moloney murine sarcoma virus, is decided with EcoRI and HindIII and is subsequently treated with calf intestinal phosphatase. The linear vector is fractionated on agarose gel and purified using glass beads The cDNA encoding a polypeptide of the present invention is amplified using PCR primers corresponding to the 5 'and 3' end sequences, respectively. 5 'primer contains an EcoRI site and the 3' capstan containing a HindIII site Equal amounts of the linear skeleton of the Malonoey murine sarcoma virus and the EcoRI and HindIII fragment are added together, in the presence of T4 DNA ligase. it is maintained under appropriate conditions for the ligation of the two fragments.The ligation mixture is used to transform the HB101 bacteria, and then plated in kanamycin containing p-agar. for the purpose of confirming that the vector has the gene of interest properly inserted. Amphotropic amphotropic pA317 or GP + aml2 packing cells were grown in tissue culture at the confluent density in the Dulbecco Modified Eagles Medium (DMEM) with 10 j% calf serum (CS), penicillin and streptomycin. The MSV vector containing the gene was then added to the medium and the packaging cells were transduced with the vector. The packaging cells now produce particular infectious virals that contain the gene (packaging cells are now referred to as reproductive cells). Fresh medium was added to the transduced producer cells, and subsequently, the medium was collected from a 10 cm plate of the confluent producer cells. The spent medium, which contains the infectious viral particles, was filtered through the millipore filter to remove the isolated reproductive cells and this medium was then used to infect fibroblast cells. The medium was removed from a sub-confluent plate of fibroblasts and rapidly replaced with the medium from the producer cells. This medium was removed and replaced with fresh medium. If the virus titer is high, then virtually all the fibroblasts were infected and no selection is required. If the titer is very low, then it is necessary to use a retroviral vector that has a selectable marker, such as neo or his. The engineered fibroblasts were then injected into the host, either alone or after being cultured at confluence on microcarrier beads of cytodex 3. Fibroblasts now produce the protein product.
Numerous modifications and variations of the present invention are possible in view of the foregoing teachings and therefore, within the scope of the appended claims, the invention may be practiced otherwise than as described in particular manner.
SEQUENCE LIST (1) GENERAL INFORMATION: (i) APPLICANT: HU, ET AL (ii) TITLE OF THE INVENTION: Fibroblast Growth Factor 14 (iii) NUMBER OF SEQUENCES: 8 (iv): ADDRESS OF THE RECIPIENT: (A) ) RECIPIENT: CARELLA, BYRNE, BAIN, GILFILLAN, CECCHI, STEWART & OLSTEIN (B) STREET: 6 BECKER FARM ROAD (C) CITY: ROSELAND (D) STATE: NEW JERSEY (E) COUNTRY: UNITED STATES OF AMERICA (F) POSTAL CODE 07068 (v) LEGIBLE FORM ON COMPUTER: (A) TYPE MEDIUM: 3.5 INCH DISC (B) COMPUTER: IBM PS / 2 (C) OPERATING SYSTEM: MS-DOS (D) PROGRAM: WORD PERFECT 5.1 (vi) CURRENT APPLICATION DATA: (A) APPLICATION NUMBER: (B) ) DATE OF SUBMISSION: CONCURRENTLY (C) CLASSIFICATION: (vii) DATA OF THE PREVIOUS APPLICATION: (A) NUMBER OF APPLICATION: 08 / 207,412 (B) DATE OF SUBMISSION: MARCH 8, 1994 (viii) INFORMATION OF THE AGENT / LAWYER (A) NAME: FERRARO, GREGORY D. (B) REGISTRATION NUMBER: 36,134 (C) ORDER NUMBER / REFERENCE: 325800-402 (ix) TELECOMMUNICATION INFORMATION: (A) TELEPHONE: 201-994-1700 (B) TELEFAX: 201-994-1744 (2) INFORMATION FOR SEQ ID NO: l: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 680 PAIRS OF BASE (B) TYPE: NUCLEIC ACID (C) TYPE OF HEBRA: SIMPLE D) TOPOLOGY ÍA: LINEAR (ii) TYPE OF MOLECULE: cDNA (ix) DESCRIPTION OF THE SEQUENCE: SEQ ID NO: l: ATGGCGGCGC TGGCCAGTAG CCTGATCCGG CAGAAGCGGG AGCTCCGCGA GCCCGGGGGC 60 AGCCGGCCGG TCrTCGGCOCA GCOQCGCOTG TGTCCCCGCG GCACCAAGTC CCTTTGCCAG 120 AAGCAGCTCC TCATCCTGCT GTCCAACJGTO CGACTGTOCs GOGOGOGßCCCC CGOSCßßCCG 180 GACCGCGGCC CGGASCCTCA GCTCAAAGGC ATCGTCACCA AACTGTTCTG CCGCCACGGT 240 TTCTACCTCC AGGCGAATCC CGACOGAAGC ATCCAGGGCA CCCCAGAGG? TACCAGCTCC 300 TTCACCCACT TC? ACCTG? T CCCTOTOaGC CTCCGTGtOO TC? CCATCCA OAGCGCCAAG 360 CTGGGTCACt ACATßßCCAT GAATGCTQAG GGACTOCTCT ACAGTTCGCC GCATTTCACA 420 GC ß? G sTC GCTTrAAGQA GTGTßTCTTT GAGAATTACT ACOTCCTOTA CGCCTCTGCT 480 CTCTACCGCC AOCGTCGTTC TOOCCGOGCC TQ CACCtCG GCCTQGACAA OGAßGOCCAG 540 GTCATGAAGG GAAACCGAGT TAAGAAGACC AAGGCAGCTG CCCACTTTCT GCCCAAGCTC 600 crasAaoTOG CCATGTACCA ooAG < _cttct CTCX-ACAOGG tccccoAßsc CTCCCCTTCC ßßo AGTCCCCCTG CCCCCCTOAA «80 (2) INFORMATION FOR SEQ ID NO: 2: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 225 AMINO ACIDS (B) TYPE: AMINO CIDOS (C) TYPE OF HEBRA: D) TOPOLOGY: LINEAR (ii) TYPE OF MOLECULE: PROTEIN (ix) DESCRIPTION OF THE SEQUENCE: SEQ ID NO: 2: Met Wing Wing Leu Wing Being Ser Leu He Arg Gln Lys Arg Glu Val -25 -20 -15 Arg Glu Pro Gly Gly Ser Arg Pro Val Ser Wing Gln Arg Arg Val -10 -5 1 Cyß Pro Arg Gly Thr Lys Ser Leu cyß Gln Lys Oln Leu Leu He 10 15 Leu Leu Ser Lys Val Arg Leu Cyß Gly Gly Arg- Pro Ala Arg Pro 25 30 Aßp Arg Gly Pro Glu Pro Gln Leu Lys Gly lie val Thr Lya Leu 40 45 Phe Cys Arg Gln Gly Phe Tyr Leu Gln Wing Aßn Pro? ßp Gly Ser 50 55 60 He Gln Gly Thr Pro Glu Aßp Thr Ser Ser Phe Thr Hiß Phe Aßn 65 70 75 Leu He Pro Val Gly Leu Arg Val Val Thr He Gln Ser Ala Lys ao 85 90 Leu Gly Hiß Tyr Met Ala Met Aßn Ala Glu Gly Leu Leu Tyr Ser 95 100 105 Ser Pro His Phe Thr Ala Glu Cyß Arg Phe Lyß Glu Cyß Val Phe 110 115 120 Glu Aßn Tyr Tyr Val Leu Tyr Ala Ser Ala Leu Tyr Arg Gln Arg 125 130 135 Arg Ser Gly Arg Wing Trp Tyr Leu Gly Leu Aßp Lyß Glu Gly Gln 140 145 150 Val Met Lys ßly Asn Arg val Lys Lyß Thr Lyß Ala Ala Ala His 155 160 165 Phe Leu Pro Lys Leu Leu Glu Val Wing Met Tyr Gln Glu Pro Ser (2) INFORMATION FOR SEQ ID NO: 3: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 32 BASE PAIRS (B) TYPE: NUCLEIC ACID (C) TYPE OF HEBRA: SIMPLE D) TOPOLOGY: LINEAR ( ii) TYPE OF MOLECULE: Oligonucleotide (ix) DESCRIPTION OF SEQUENCE: SEQ ID NO: 3: GCCAGAGCAT GCAGCGGCGC GTGTGTCCCC GC 32 (2) INFORMATION FOR SEQ ID NO: 4: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 33 PAIRS OF BASE (B) TYPE: NUCLEIC ACID (C) TYPE OF HEBRA: SIMPLE D) TOPOLOGY: LINEAR ( ii) TYPE OF MOLECULE: Oligonucleotide (ix) DESCRIPTION OF SEQUENCE: SEQ ID NO: 4: GCCAGAAGAT CTGGGGGCAG GGGGACTGGA AGG 33 (2) INFORMATION FOR SEQ ID NO: 5: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH : 33 BASE PAIRS (B) TYPE: NUCLEIC ACID (C) TYPE OF FLEECE: SIMPLE D) TOPOLOGY: LINEAR (ii) TYPE OF MOLECULE: Oligonucleotide (ix) DESCRIPTION OF SEQUENCE: SEQ ID NO: 5: CTAGTGGATC CCATCATGGC GGCGCTGGCC AGT 33 (2) INFORMATION FOR SEQ ID NO: 6: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 30 PAIRS OF BASE (B) TYPE: NUCLEIC ACID (C) TYPE OF HEBRA: SIMPLE D) TOPOLOGY: LINEAR ( ii) TYPE OF MOLECULE: Oligonucleotide (ix) DESCRIPTION OF SEQUENCE: SEQ ID NO: 6: CGACTGGATC CCCAGCGGCG CGTGTGTCCC 30 (2) INFORMATION FOR SEQ ID NO: 7: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 33 BASE PAIRS (B) TYPE: NUCLEIC ACID (C) TYPE OF HEBRA: SIMPLE D) TOPOLOGY: LINEAR ( ii) TYPE OF MOLECULE: Oligonucleotide (ix) DESCRIPTION OF SEQUENCE: SEQ ID NO: 7: CGACTTCTAG AATCAGGGGG CAGGGGGACT GGA 33 (2) INFORMATION FOR SEQ ID NO: 8: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH : 33 BASE PAIRS (B) TYPE: NUCLEIC ACID (C) TYPE OF HEBRA: SIMPLE D) TOPOLOGY: LINEAR (ii) TYPE OF MOLECULE: Oligonucleotide (ix) DESCRIPTION OF SEQUENCE: SEQ ID NO: 8: CTAGTGGATC CCATCATGGC GGCGCTGGCC AGT 33 (2) INFORMATION FOR SEQ ID NO: 9: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 30 PAIRS OF BASE (B) TYPE: NUCLEIC ACID (C) TYPE OF HEBRA: SIMPLE D) TOPOLOGY: LINEAR ( ii) TYPE OF MOLECULE: Oligonucleotide (ix) DESCRIPTION OF THE SEQUENCE: SEQ ID NO: 9: GATTTACTCG AGGGGGGCAG GGGGACTGGA 30 It is noted that in relation to this date, the best method known by the applicant to carry out the present invention is that which is clear from the present description of the invention.
Having described the invention as above, the content of the following is claimed as property:

Claims (20)

1. An isolated polynucleotide, characterized in that it comprises a member selected from the group consisting of: (a) a polynucleotide encoding the polypeptide comprising amino acid 26 to amino acid 199 as set forth in SEQ ID NO: 2; (b) a polynucleotide encoding the polypeptide comprising amino acid 1 to amino acid 199 as set forth in SEQ ID NO: 2 (c) a polynucleotide capable of hybridizing and which is at least 70% identical to the polynucleotide of (a) or (b); and (d) a polynucleotide fragment of the polynucleotide of (a), (b) or (c).
2. The polynucleotide according to claim 1, characterized in that the polynucleotide is DNA.
3. The polynucleotide according to claim 1, characterized in that it comprises from the polynucleotide 1 to the polynucleotide 680 as set forth in SEQ ID NO: 1.
4. The polynucleotide according to claim 1, characterized in that it comprises from nucleotide 79 to nucleotide 680 as set forth in SEQ ID NO: 1.
5. An isolated polynucleotide comprising a member selected from the group consisting of: (a) a polynucleotide encoding a mature polypeptide encoded by the DNA contained in deposit number 97147 of ATCC; (b) a polynucleotide encoding the polypeptide expressed by the DNA contained in deposit number 97147 of ATCC; (c) a polynucleotide capable of hybridizing and which is at least 70% identical to the polynucleotide of (a) or (b); and (d) a polynucleotide fragment of the polynucleotide of (a), (b) or (c).
6. A vector, characterized in that it contains the DNA of claim 2.
7. A host cell, characterized in that it is genetically engineered with the vector of claim 6.
8. A process for producing a polypeptide characterized in that it comprises: expressing from the host cell of claim 7 the polypeptide encoded by the DNA.
9. A process for producing cells capable of expressing a polypeptide, characterized in that it comprises genetically managing the cells in the vector of claim 6.
10. A polypeptide characterized in that it comprises a member selected from the group consisting of (i) a polypeptide having the deduced amino acid sequence of SEQ ID NO: 2 and fragments, analogs and derivatives thereof; and (ii) a polypeptide encoded by the cDNA of Deposit No. 97147 of ATCC and fragments, analogs and derivatives of the polypeptide.
11. An antibody characterized in that it is to be against the polypeptide of the claim 10.
12. A compound characterized in that it inhibits the biological actions of the polypeptide of claim 10.
13. A compound characterized in that it activates a receptor for the polypeptide of the claim 10.
14. A method for the treatment of a patient in need of a FGF-14 polypeptide, characterized in that it comprises: administering to the patient a therapeutically effective amount of the polypeptide of claim 10.
15. A method for the treatment of a patient in need of inhibiting a FGF-14 polypeptide, characterized in that it comprises: administering to the patient a therapeutically effective amount of the compound of claim 12.
16. The method according to claim 14, characterized in that the therapeutically effective amount of the polypeptide is administered by providing the patient with the DNA encoding the polypeptide and expressing the polypeptide in vivo.
17. The method according to claim 15, characterized in that the compound is a polypeptide of a therapeutically effective amount of the compound is administered by providing the patient with the DNA encoding the antagonist and expressing the antagonist in vivo.
18. A process for identifying active compounds as agonists to the polypeptide of claim 10, characterized in that it comprises: (a) combining a compound to be examined and a reaction mixture containing cells under conditions wherein the cells are normally stimulated by the polypeptide , the reaction mixture containing a tag incorporated into the cells as they proliferate; and (b) determining the degree of proliferation of the cells to identify whether the compound is an effective agonist.
19. A process for identifying active compounds as antagonists to the polypeptide of claim 10, characterized in that it comprises: (a) combining a compound to be examined, the polypeptide and a reaction mixture containing cells under conditions where the cells are normally stimulated by the polypeptide, the reaction mixture containing a tag incorporated into the cells as they proliferate; and (b) determining the degree of proliferation of the cells to identify whether the compound is an effective antagonist.
20. A process for diagnosing a disease or a susceptibility to a disease related to a sub-expression of the polypeptide of claim 10, characterized in that it comprises: determining a mutation in the nucleic acid sequence encoding the polypeptide.
MXPA/A/1997/006854A 1997-09-09 Factor 14 of fibroblas growth MXPA97006854A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US1995/006730 WO1996039506A1 (en) 1995-06-05 1995-06-05 Fibroblast growth factor-14

Publications (2)

Publication Number Publication Date
MX9706854A MX9706854A (en) 1997-11-29
MXPA97006854A true MXPA97006854A (en) 1998-07-03

Family

ID=

Similar Documents

Publication Publication Date Title
US5773252A (en) Fibroblast growth factor 15
US20040086967A1 (en) Human criptin growth factor
AU702131B2 (en) Fibroblast growth factor-14
US20090311263A1 (en) Human vascular ibp-like growth factor
AU712297B2 (en) Fibroblast growth factor 11
US5728546A (en) Fibroblast growth factor 13
AU702682B2 (en) Fibroblast growth factor 13
WO1996039509A1 (en) Fibroblast growth factor 15
AU714165B2 (en) Human criptin growth factor
US20030022312A1 (en) Human hepatoma-derived growth factor-2
US7338934B2 (en) Fibroblast growth factor-14
AU711647C (en) Fibroblast growth factor 15
AU748429B2 (en) Fibroblast growth factor 11
AU752592B2 (en) Fibroblast growth factor 15
MXPA97006854A (en) Factor 14 of fibroblas growth
MXPA97009217A (en) Growth factor 13 of fibroblas
MXPA97007897A (en) Factor-11 fibroblas growth
MXPA97008493A (en) Factor 15 of fibroblas growth
CA2220988A1 (en) Fibroblast growth factor 13
MXPA97009570A (en) Growth factor 3 endothelial, vascular, hum
AU2003252757A1 (en) Human Criptin Growth Factor
KR19980703870A (en) Fibroblast Growth Factor-11