MXPA97009217A - Growth factor 13 of fibroblas - Google Patents

Growth factor 13 of fibroblas

Info

Publication number
MXPA97009217A
MXPA97009217A MXPA/A/1997/009217A MX9709217A MXPA97009217A MX PA97009217 A MXPA97009217 A MX PA97009217A MX 9709217 A MX9709217 A MX 9709217A MX PA97009217 A MXPA97009217 A MX PA97009217A
Authority
MX
Mexico
Prior art keywords
polypeptide
cells
polynucleotide
dna
sequence
Prior art date
Application number
MXPA/A/1997/009217A
Other languages
Spanish (es)
Other versions
MX9709217A (en
Inventor
John M Greene
Joachim R Gruber
Craig A Rosen
Original Assignee
John M Greene
Joachim R Gruber
Human Genome Sciences Inc
Craig A Rosen
Filing date
Publication date
Application filed by John M Greene, Joachim R Gruber, Human Genome Sciences Inc, Craig A Rosen filed Critical John M Greene
Priority to MXPA/A/1997/009217A priority Critical patent/MXPA97009217A/en
Publication of MX9709217A publication Critical patent/MX9709217A/en
Publication of MXPA97009217A publication Critical patent/MXPA97009217A/en

Links

Abstract

A polypeptide of human fibroblast growth factor 13, and DNA (RNA) encoding this polypeptide is disclosed. A method for producing this polypeptide by recombinant techniques is also provided. Methods for using such a polypeptide to promote wound healing, for example as a result of sunburn burns, to prevent the nueronal damage associated with strokes and due to neuronal disorders and to promote neuronal growth, and to prevent neuronal growth are also disclosed. aging of the skin and hair loss, to stimulate angiogenesis, mesodermal induction in primary embryos and the regeneration of mienbros. Antagonists against such polypeptides and their use as a therapeutic agent to prevent abnormal cell proliferation, hypervascular diseases and proliferation of lens-like epithelial cells are also disclosed. Diagnostic methods are also revealed to detect mutations in the coding sequence and alterations in the concentration of the polypeptides in a sample derived from a host.

Description

GROWTH FACTOR 13 OF FIBROBLASTS This invention relates to newly identified polynucleotides, polypeptides encoded by such polynucleotides, the use of such polynucleotides and polypeptides, as well as the production of these polynucleotides and polypeptides. More particularly, the polypeptides of the present invention have been putatively identified as fibroblast growth factors / heparin binding growth factors, hereinafter referred to as "FGF-13". The invention also relates to the inhibition of the action of these polypeptides. The growth factors of fibroblasts are a family of proteins characteristic for the binding to heparin and, therefore, are also named heparin binding growth factors (HBGF). The expression of different members of these proteins was found in various tissues and under particular control, temporal and spatial. 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, vascular smooth muscle cells, lens-like epithelial cells , melanocytes, keratinocytes, oligodendrocytes, astrocytes, osteoblasts and hematopoietic cells. Each member has functions that overlap with others and also have their unique spectrum of functions. The addition to the ability to stimulate the proliferation of endothelial cells, both FGF-1 and 2, is chemotactic for endothelial cells and FGF-2 has been shown to make it possible for endothelial cells to penetrate the basement 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 and modulate the differentiation of neuronal cells, adipocytes and skeletal muscle cells. In addition to these biological activities in normal tissues, FGF proteins have been implicated in promoting tumorigenesis in carcinomas and sarcomas, promoting the vascularization of tumors and as transformation proteins, when their expression is deregulated.
The FGF family consists presently of eight structurally related polypeptides: basic FGF, acid FGF, int 2, hst 1 / k-FGF, FGF-5, FGF-6, keratinocyte growth factor, AIGF (FGF-8) and recently, a spinning factor has been shown to be a novel heparin-binding growth factor, which was purified from the culture supernatant of a human glioma cell line (Miyamoto, M et al., Mol. and Cell. Biol. ., 13 (7) .4251-4259 (1993) .The genes for each have been cloned and sequenced.Two of the members, FGF-1 and FGF-2, have been characterized under many names, but, more often, as growth factors of fibroblasts, acid and basic, respectively.The products of the normal gene have influence on the general proliferation capacity of most of the cells derived from mesoderm and neuroectoderm.They are able to induce angiogenesis in I live and can play important roles in the 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 link to the same recipients and produce a second message through the league to these recipients. A eukaryotic expression vector, which encodes the secreted form of FGF-1, has been introduced by gene transfer in porcine arteries. This model defines the function of the gene in the arterial wall in vivo. The expression of FGF-1 induced intimal thickening in the arteries of swine, 21 days after the gene transfer (Nabel, E. G., et al., Na ture, 362: 844-6 (1993)). It has further been shown that the basic growth factor of fibroblasts can regulate glioma growth and independent progression of its role in tumor angiogenesis and that the release or secretion of the basic growth factor of fibroblasts can be required for these actions ( Morrison, RS, et al., J. Neurosci. Res., 34: 502-9 (1992)). Fibroblast growth factors, such as basic FGF, have also been implicated in the growth of Kaposi's sarcoma cells in vitro (Huang, YQ, et al., J. Clin. Invest., 91_1191-7 (1993 )). Also, the cDNA sequence encoding the basic growth factor of fibroblasts has been cloned downstream of a transcription promoter recognized by the bacteriophage T7 RNA polymerase. The basic growth factors of fibroblasts, thus obtained, have been shown to have a biological activity indistinguishable from human placental fibroblast growth factor in the itogenicity, plasminogen activator synthesis and angiogenesis assays (Squires, CH, et al., J. Biol. Chem., 263: 16297-302 (1988)). The U.A. Patent No. 5,155,214 discloses basic growth factors of substantially pure mammalian fibroblasts and their production. The amino acid sequences of the basic growth factor of bovine and human fibroblasts are revealed, as is the DNA sequence encoding the bovine species polypeptide. The newly discovered FGF-9 has about 30% sequence similarity to other members of the FGF family. Two cysteine residues and other consensus sequences in the family members were also well conserved in the sequence of FGF-9. This FGF-9 was found to have no typical signal sequence at its N-terminus as that in the acidic and basic FGF. However, FGF-9 was found to be secreted from cells after synthesis despite its lack of a typical signal sequence FGF (Miyamoto, M, et al., Mol. And Cell. Biol., 13 (7). ): 4251-4259 (1993) In addition, FGF-9 was found to stimulate the cell growth of progenitor cells of type 2 astrocyte of the oligondendrocyte, BALB / c3T3, and PC-12 cells, but not those of the endothelial cells of the human umbilical vein (Naruo, K., et al., J. Biol. Chem., 268: 2857-2864 (1993).
The basic FGF and the acid FGF are powerful modulators of cell proliferation, cell motility, differentiation and survival and act on cell types of the ectoderm, mesoderm and endoderm. These two FGFs, together with the KGF and AIGF, were identified by the purification of proteins. However, the other four members were isolated as oncogenes, whose expression was restricted to embryo-genesis and certain types of cancers. FGF-9 was shown to be a mitogen against glial cells. Members of the FGF family were reported to have oncogenic potency. FGF-9 has shown transformation potency when transformed into BALB / c3T3 cells (Miyamoto, M. et al., Mol.Cell. Biol., 13 (7): 4251-4259 (1993). Androgen induced (AIGF), also known as FGF-8, was purified from the conditioned medium of mouse mammary carcinoma cells (SC-3) simulated with testosterone.AIGF is a distinctive FGF type growth factor, which has a putative signal peptide that shares 30 to 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, DIGF mediates the androgen-induced growth of SC-3 cells and perhaps other cells, since it is secreted by the tumor cells themselves The polypeptide of the present invention has been allegedly identified as a member of the family ia of the FGF 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, novel mature polypeptides are provided as well as biologically active and diagnostically and therapeutically useful fragments, their analogs and derivatives. The polypeptides of the present invention are of human origin. According to another aspect of the present invention, isolated molecules of the nucleic acid encoding the polypeptides of the present invention, including the mRNAs, DNAs, cDNAs, genomic DNA, as well as their antisensing analogues and their biologically active fragments are supplied and Diagnostically or therapeutically useful. According to yet another aspect of the present invention, processes are provided to produce such polypeptides, by recombinant techniques, through the use of recombinant vectors, such as the cloning and expression plasmids, useful as reagents in the recombinant production of polypeptides of the present invention, as well as the prokaryotic and / or eukaryotic host cells, comprising a nucleic acid sequence encoding a polypeptide of the present invention. According to a further aspect of the present invention, a process is provided for the use of such polypeptides or polynucleotides encoding these polypeptides, for the classification of agonists and antagonists therein and for therapeutic purposes, for example, promoting wound healing, for example, as a result of burns and ulcers, to prevent neuronal damage associated with apoplectic attacks and due to neuronal disorders and to promote neuronal growth, and to prevent skin aging and hair loss, to stimulate angiogenesis, Mesodermal induction in early embryos and limb regeneration. According to yet a further aspect of the present invention, antibodies against such polypeptides are supplied. According to yet another aspect of the present invention, antagonists are provided against such polypeptides and processes for their use in inhibiting the action of these polypeptides, for example in the treatment of cell transformation, eg tumors, to reduce scars and treat diseases hyper-vascular 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 over-expression of the polypeptides encoded by such sequences. According to another aspect of the present invention, a process is provided for the use of such polypeptides, or polynucleotides encoding these polypeptides, for in vitro purposes related to scientific research, DNA synthesis and manufacture of DNA vectors. These and other aspects of the present invention will be apparent to those skilled in the art from the teachings of the present invention. The following drawings are only illustrations of specific embodiments of the present invention and do not attempt to limit it in any way. Figure 1 illustrates the cDNA sequence and the corresponding deduced amino acid sequence of FGF-13.
The 21 initial amino acid residues represent a putative guiding 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 (SEQ ID NOS: 2) or for the mature polypeptide encoded by the cDNA of the clone deposited as ATCC Deposit No. 97147, on May 12, 1995. The polynucleotide encoding the FGF-13 of this invention was discovered in a cDNA library derived from human ovarian cancer tissue. The FGF-13 polypeptide is structurally related to all members of the fibroblast growth factor family and contains an open reading frame that encodes a 212 amino acid polypeptide, of which the first 21 amino acids represent a putative guidance sequence , so that the mature polypeptide comprises 191 amino acids. Among the main correspondences are: 1) 69% identity and 81% similarity to mouse AIGF over an extension of 185 amino acids; 2) 30% identity and 56% similarity with chicken FGF-4 in a region of 82 amino acids; 3) 41% identity and 64% similarity with human KGF over an extension of 78 amino acids.
The identity of the FGF / HBGF family, GXLX (S, T, A, G) X6 (D, E), CXFXE is conserved in the polypeptide of the present invention,. (X means any amino acid residue; (D, E) means any residue D or E; X6 means any residue of 6 amino acids). The polynucleotide of the present invention may be in the form of the RNA or in the DNA form, this DNA includes the cDNA, genomic DNA, and synthetic DNA. The DNA can be double-stranded or single-strand. The coding sequence encoding the mature polypeptide may be identical to the coding sequence shown in Figure 1 (SEA ID NOS: 1) or that of the deposited clone or may be a different coding sequence, as a result of redundancy or degeneracy of the genetic key, which encodes the same, the mature polypeptide as the DNA of Figure 1 (SEQ ID NOS: l) or the deposited cDNA. Polynucleotides encoding the mature polypeptide of Figure 1 (SEQ ID NOS: 2) or for the mature polypeptides encoded by the deposited cDNA (s) may include: only the coding sequence for the mature polypeptide; the coding sequence for the mature polypeptide and the additional coding sequence, such as a guiding or secretory sequence or a proprotein sequence; the coding sequence for the mature polypeptide (and, optionally, the additional coding sequence) and the non-coding sequence, such as the instrons or 5 'and / or 3' non-coding sequences of the coding sequence for the mature polypeptide. Thus, the term "polynucleotide encoding a polypeptide" encompasses a polynucleotide which includes only the coding sequence for the polypeptide as well as a polynucleotide which includes the additional coding sequence and / or non-coding sequence. The present invention further relates to variants of the polynucleotides described above, which encode the fragments, analogs and derivatives of the polypeptides having the deduced amino acid sequence of Figure 1 (SEQ ID NOS: 2) or the polypeptides encoding pro the cDNA (s) of the deposited clone (s). The variants of the polynucleotides may be naturally occurring allelic variants of the polynucleotide or a variant that occurs not naturally of the polynucleotide. Thus, the present invention includes polynucleotides encoding the same mature polypeptide as shown in Figure 1 (SEQ ID NOS: 2) or the same mature polypeptides encoded by the cDNA (s) of the deposited clones, as well as variants of such polynucleotides, these variants encode a fragment, derivative or analog of the polypeptide of Figure 1 (SEQ ID NOS: 2) or the polypeptides encoded by the cDNA (s) of the deposited clones. 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 an allelic variant occurring naturally of the coding sequence shown in Figure 1 (SEQ ID NOS: 1) or of the coding sequence of the (s) clone (s) deposited (s). As is known in the art, an allelic variant is an alternative form of a polynucleotide sequence which may have a substitution, deletion or addition of one or more nucleotides, which do not substantially alter the function of the encoded polypeptides. The present invention also includes polynucleotides, in which 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, for example, a guide 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 split by the host cell to obtain the mature form of the polypeptide. The polynucleotides can also encode a proprotein which is the mature protein plus the additional 5 'amino acid residues. A mature protein that has a prosequence is a proprotein and is an inactive form of the protein. Once the prosequence is unfolded, an active mature 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 frame to a marker sequence, which allows purification of the polypeptide of the present invention. The marker sequence may be a hexahistidine tag supplied by a pQE-9 vector, to deliver the purification of the mature fused polypeptide to the tag in the case of a bacterial host or, for example, the tag sequence may be a hemagglutinin tag (HA ) when a mammalian host is used, for example COS-7 cells. The HA tag corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson, I., et al., Cell, 37: 767 (1984)). The term "gene" means the segment of the DNA involved in producing a polypeptide chain; it includes the regions that precede and follow the coding region (guide and drag) as well as the intervening sequences (introns) between the individual coding segments (exons). Fragments of the full-length FGF-13 gene can be used as a hybridization probe for a cDNA library to isolate the full-length gene and to isolate other genes that have a high sequence similarity to the gene or a 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, which contain the complete FGF-13 gene, which includes regulatory and promoter regions, exons and introns. An example of a protection comprises isolating the coding region of the FGF-13 gene, using the known DNA sequence to synthesize an oligonucleotide probe. The labeled oligonucleotides, which have a sequence complementary to that of the gene of the present invention, are used to protect a collection of the human cDNA, the genomic DNA or mRNA, to determine which members of the collection hybridize the probe. The present invention further relates to polynucleotides that hybridize to the sequences described above, if there is at least 70%, preferably at least 90%, and more preferably at least 95% identity between the sequences. The present invention particularly relates to polynucleotides that hybridize under severe conditions to the polynucleotides described herein above. As used herein, the term "severe conditions" means that 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, hereinabove described, in a preferred embodiment, encode polypeptides that retain substantially the same function or biological activity as the mature polypeptide encoded by the cDNAs of Figure 1 (SEQ ID NO: 1) or the cDNAs deposited. Alternatively, the polynucleotide may have at least 20 bases, preferably 30 bases and more preferably at least 50 bases, which hybridize to a polynucleotide of the present invention and which have an identity there, as described above, and which may or may not retain the activity. For example, each polynucleotide can be used as a probe for the polynucleotide of SEQ ID NO: 1, for example, to recover the polynucleotide or as a diagnostic probe or as a polymerase chain reaction (PCR) primer. Thus, the present invention is directed to polynucleotides having at least 70% identity, preferably at least 90% and more preferably at least 95% identity, to a polynucleotide encoding the polypeptide of SEQ ID NO. : 2, like their fragments, these fragments have at least 30 bases and preferably at least 50 bases and the polypeptides encoded by these polynucleotides. The deposits mentioned here will be kept under the Budapest Treaty in the International Recognition of the Deposit of Microorganisms for the purposes of the Patent Procedure. These deposits are provided merely as a convenience and not to admit that a deposit is required, according to Code 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 herein, are incorporated by reference and are the control in the case of any conflict with the description of the sequences herein. . A license may be required to obtain, use or sell the deposited materials and no license is granted hereby. The present invention furthermore relates to a FGF polypeptide having the deduced 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 the fragments , analogs and derivatives of these polypeptides.
The terms "fragments", "derivatives" and "analogs", when referring to the polypeptide of Figure 1 (SEQ ID NOS: 2) or those encoded by the deposited cDNA (s), mean the polypeptides that retain essentially the same function or biological activity of such polypeptides. Thus, an analog includes a proprotein that can be activated by cleavage of the proprotein portion to produce an active mature polypeptide. The polypeptides of the present invention can be recombinant polypeptides, natural polypeptides or synthetic polypeptides, preferably recombinant polypeptides. The fragments, derivatives or analogs of the polypeptide of Figure 1 (SEQ ID NOS: 2) or those encoded by the deposited cDNA (s), can be (i) that in which one or more of the amino acid residues are substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such a substituted amino acid residue may or may not be encoded by the genetic key or (ii) that one or more of the amino acid residues include a substituent group or (iii) that in which the mature polypeptide is fused with another compound, such as a compound to increase the half-life of the polypeptide (e.g., polyethylene glycol) or (iv) that in which 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. Such fragments, derivatives and the like are considered to be within the scope of those skilled in the art taking into account the present teachings. The polypeptides and polynucleotides of the present invention are preferably provided in an isolated form and preferably are purified to homogeneity. The term "isolated" means that the material is removed from its original environment (for example the natural environment if it occurs naturally). For example, a naturally occurring polynucleotide or polypeptide present in a living animal is not isolated, but this same polynucleotide or DNA or polypeptide, separated from it or from all materials coexisting in the natura system, is isolated. These polynucleotides can be part of a vector and / or such polynucleotides or polypeptides can be part of a composition and still be isolated in that that vector or composition is not part of their natural environment. The polypeptides of the present invention include the polypeptide of SEQ ID NO: 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 more preferably at least 90% similarity (more preferably at least 90% identity) to the polypeptide of SEQ ID NO: 2 and even more preferably at least 95% similarity (further preferably, at least 95% identity) to the polypeptide of SEQ ID NO: 2 and also include portions of such polypeptides with such a portion of the polypeptide generally containing at least 30 amino acids and more preferably at least 50 amino acids. As known in the art, the "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. Fragments or portions of the polypeptides of the present invention can be used to produce the corresponding full-length polypeptide by the synthesis of peptides; therefore, the fragments can be used as intermediates to produce the full-length polypeptides. 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 the polynucleotides of the present invention, the host cells that are genetically treated with the vectors of the invention and the production of the polypeptides of the invention by recombinant techniques. The host cells can be genetically treated (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 treated host cells can be cultured in conventional nutrient media, modified as appropriate, to activate the promoters, select the transformants or amplify 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 this will be apparent to one of ordinary skill 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. Such 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 vaccines, adenoviruses, virus of postulose eruptions of birds and pseudo-rabies. However, any other vector or plasmid can be used as long as it is duplicable and viable in the host. The appropriate sequence of the DNA can be inserted into the vector by a variety of procedures. In general, the DNA sequence is inserted into an appropriate restriction endonuclease site, by procedures known in the art. Such procedures and others are considered within the scope of those skilled in the art. The DNA sequence in the expression vector is operably linked to one or more appropriate expression control sequences (promoters) to direct the synthesis of the mRNA. As representative examples of such promoters, there may be mentioned, the promoter LTR or SV40, E. coli, lac or trp, the promoter Pj. of phage lambda and other known promoters in the control of gene expression in prokaryotic or eukaryotic cells or their viruses. The expression vector also contains a ribosome binding site for translation initiation and a transcription terminator. The vector may also include appropriate sequences to amplify expression. In addition, expression vectors preferably contain a gene for providing a phenotypic treatment for the selection of transformed host cells, such as dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, or resistance to tetracycline or ampicillin. in E. coli. The vector containing the appropriate DNA sequence, as described above, 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 yeasts; insect cells, such as Drosophila S2 t Spodoptera Sf9; animal cells, such as CHO, COS or Bowes melanoma; adenovirus; plant cells, etc. The selection of an appropriate host is considered to be within the scope of those skilled in the art, of the present teachings. More particularly, the present invention also includes recombinant constructs comprising one or more of the sequences as described broadly above. The builders comprise a vector, such as a plasmid or a viral vector, in which a sequence of the invention has been inserted, in a forward or reverse orientation. In a preferred aspect of this embodiment, the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the sequence.
Large numbers of suitable vectors and promoters are known to those skilled in the art, and are commercially available. The following vectors are provided in the form of an example. Bacteria: pQR70, pQE60, pQE-9 (qiagen), pBS, phagescript, psiX174, pBluescript SK, pBsKS, pNHSa, pNHlda, pNH18a, pNH46a (Stratagene); pTRC99A, pKK223-3, pKK233-3, pDR540, pRIT5 (Pharmacia). Eukaryotic: pWLneo, pSV2cat, pOG44, pXTl, pSG (Stratagene) pSVK3, pBPV, pMSG, pSVL (Pharmacia). However, any other plasmid or vector can be used as long as it is duplicable and viable in the host. The promoter regions can be selected from any desired gene using CAT vectors (chloramphenicol transferase) or other vectors with selectable markers. Two appropriate vectors are pKK232-8 and pCM7. Particularly named bacterial promoters include lacl, lacZ, T3, T7, gpt, lambda PR, PL and trp. Eukaryotic promoters include immediate primary DMV, HSV thymidine kinase, primary and late SV40, retrovirus LTRs and mouse metallothionein-I. The selection of the appropriate vector and promoter is well within the level of ordinarily skilled in the art. In a further embodiment, the present invention relates to host cells containing the above-described manufacturer. The host cell can be a higher eukaryotic cell, such as a mammalian cell, or a minor 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 builder into the host cell can be effected by transfection of calcium phosphate, transfection mediated by DEAE-Dextran, or electroporation (Davis, L., Dibner, M. Battey, I., Basic Methodos in Molecular Biology, (1986)). The builders in host cells can be pulsed 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 these proteins, using the RNAs derived from the DNA constructs of the present invention. Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrook, et al., Molecular Cloning: A Laboratory Manual, Second Edition (Cold Spring Harbor, NY, 1989), the disclosure of which is incorporated herein by reference. reference.
The transcription of a DNA encoding the polypeptides of the present invention by higher eukaryotes is enhanced by inserting an enhancer sequence into the vector. Enhancers are the cis-active elements of DNA, usually around 10 to 300 bp, that act on a promoter to increase its transcription. Examples include the SV40 enhancer on the latter side of the replication origin (bp 100 to 270), an early cytomegalovirus promoter enhancer, a polyoma enhancer on the latter side of the replication origin and the adenovirus enhancers. Generally, recombinant expression vectors will include replication origins and selectable markers that allow the transformation of the host cell, for example, the E. coli ampicillin resistance gene and the S. cerevisiae TRPI gene and a promoter derived from a gene highly engineered to direct the transcription of a downstream structural sequence. Such promoters can be derivatives of glycolytic enzymes that code operons, such as 3-phosphoglycerate kinase (PGK), factor a, acid phosphates or heat shock proteins, among others. The heterologous structural sequence is assembled in appropriate phase with the translation sequences, initiation and termination, and preferably, a guiding sequence, capable of directing the secretion of the protein translated in the periplasmic space or the extracellular medium. Optionally, the heterologous sequence can encode a fusion protein, which includes an N-terminal identification peptide, which imparts the desired characteristics, for example stabilization or simplified purification of the expressed recombinant product. Useful expression vectors for bacterial use are constructed by inserting a structural DNA sequence encoding a desired protein, together with suitable translational, initiation and termination signals in the operable reading tag with a functional promoter. The vector will comprise one or more selectable phenotypic markers and a replication origin to ensure maintenance of the vector and, if desired, to provide amplification within the host. Prokaryotic hosts suitable for transformation include E. coli, Bacillus subtilis, Salmonella typhimurium and several species within the genera of Pseudomonas, Streptomyces and Staphilococcus, although others may also be used as selection material. As a representative, but or limiting example, expression vectors useful for bacterial use may comprise a selectable marker and the bacterial origin of replicate derived from commercially available plasmids, comprising genetic elements of the well-known cloning vector pBR322 (ATCC 37017), Such commercial vectors include, for example, pKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and GEMI (promete Biotec, Madison, Wl, USA). These "skeleton" sections pBR322 are combined with an appropriate promoter and the structure sequence will be expressed. Following transformation of the appropriate host strain and growth of the host strain to an appropriate cell density, the selected promoter is stopped repressing by appropriate means (e.g., temperature shift or chemical induction) and the cells are cultured by an additional period. The cells are typically collected by centrifugation, disruption by physical or chemical means, and the resulting crude extract retained for further purification. The microbial cells used in the expression of proteins can be broken by any convenient method, which includes the freeze-melt cycle, sonication, mechanical disruption, or use of cell lysis agents. Several mammalian cell culture systems can also be employed to express the recombinant protein. Examples of mammalian expression systems include the COS-7 lines of monkey kidney fibroblasts, described by Gluzman, Cell 23: 175 (1981), and other cell lines capable of expressing a compatible vector, for example, the C127 cell lines. , 3T3, CHO, HeLA and BHK. Mammalian expression vectors will comprise a replication origin, a suitable promoter and enhancer, and also any necessary binding site of the ribosome, polyadenylation site, splice donor and acceptor sites, transcriptional termination sequences and non-transcribed flanking sequences , DNA sequences derived from the SV40 viral genome, eg, SV40 origin sites, early promoter, enhancer, overlap and poly-adenylation, can be used to deliver the required non-transcribed genetic elements. The polypeptide of the present invention can be recovered and purified from the recombinant cell cultures by the methods used hitherto, which include ammonium sulfate or ethanol precipitation, acid extraction, anion exchange or cation chromatography, Phosphocellulose, hydrophobic interaction chromatography, affinity chromatography, hydroxyapatite chromatography and lectin chromatography. The protein redoubling steps can be used, as necessary, to complete the mature protein configuration. Finally, high performance liquid chromatography (HPLC) can be used for the final purification step.
The polypeptide of the present invention may be a naturally purified product or a product of synthetic chemical processes or produced by the recombinant techniques of a prokaryotic or eukaryotic host (for example, by bacterial, yeast, higher plant, insect and mammals, in the crop). Depending on the host employed in the recombinant production method, the polypeptides of the present invention may be glycosylated with mammalian carbohydrates or other eukaryotic carbohydrates, or may 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 vascular endothelial cells, can be employed in treatment to stimulate the revascularization of ischemic tissues due to various disease conditions, such as thrombosis, arteriosclerosis and other cardiovascular conditions. These polypeptides can also be used to stimulate angiogenesis and regeneration of limbs. Polypeptides can also be used to treat wounds due to injuries, burns, post-operative tissue repairs, and ulcers, since they are mitogenic to several cells of different origins, such as fibroblast cells and skeletal muscle cells. , and, therefore, facilitate the repair or replacement of damaged or diseased tissues. The polypeptides of the present invention can also be employed in stimulating neuronal growth and treating and preventing neuronal damage associated with strokes and occurring in certain neuronal disorders or neurodegenerative conditions, such as Alzheimer's disease, Parkinson's disease and complexes related to AIDS. FGF-13 has the ability to stimulate the growth of chondrocytes, therefore, it can be used to increase bone and periodontal regeneration and aid in tissue transplants or bone grafts. The polypeptides of the present invention may also be employed to prevent skin aging due to sunburn, by stimulating the growth of keratinocytes. The FGF-13 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 melanocytes. Along 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-13 polypeptide can also be used to maintain the organs before transplantation or to supplement cell cultures of primary tissues. The polypeptide of the present invention can also be used to induce tissue of mesodermal origin to differentiate into embryo principles. According to yet a further aspect of the present invention, there is provided a process for using these polypeptides, or the polynucleotides encoding such polypeptides, for purposes related to scientific research, DNA synthesis, manufacture of DNA vectors, and for diagnostic purposes and therapeutic means for the treatment of human diseases. This invention provides a method for the identification of the receptors for the polypeptides of the present invention. The genes encoding the receptor can be identified by numerous methods known to those skilled in the art, for example, by washing the ligand and classifying the FACS (Coligan, et al., Current Protocols in Immun., 1 (2), Chapter 5 , (1991)). Preferably, expression cloning is employed in which the polyadenylated RNA is prepared from a cell responsive to the polypeptides, for example NIH3T3 cells, which are known to contain multiple receptors for proteins of the FGF family, and SC-3 cells, and a collection of the cDNA created by this RNA is divided into groups and used to transfect COS cells or other cells that are not sensitive to the polypeptides. Transfected cells growing on glass cursors 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 the iodination or inclusion of a site recognition site for a protein kinase. Following the fixation and incubation, the cursors are subjected to a self-radiographic analysis. The positive groups are identified and the sub-groups are prepared and re-transfected using an iterative sub-group and a re-classification process, which finally supplies simple clones that encode the supposed receptor. As an alternative approach for receptor identification, the tagged polypeptides can be linked by photoaffinity with a cell membrane or extract preparations that express the receptor molecule. The interlaced material is resolved by PAGE analysis and exposed to an X-ray film. The full labeling containing the polypeptide receptors can be excised, resolved into peptide fragments and subjected to protein microequence. The amino acid sequence obtained from the microsequence will be used to design a set of degenerate oligonucleotide probes to classify a collection of the cDNA to identify the genes encoding the presumed receptors. This invention provides a method for classifying compounds to identify those that modulate the action of the polypeptide of the present invention. An example of such an assay comprises combining the mammalian fibroblast cell, the polypeptide of the present invention, the compound to be classified and the [H] thymidine under cell culture conditions, where the fibroblast cell will normally proliferate. A control assay can be performed in the absence of the compound to be classified and compared to the amount of fibroblast proliferation in the presence of the compound, to determine whether the compound stimulates proliferation, determining the capture of the [H] ] thymidine in each case. The amount of fibroblast cell proliferation was measured by scintillation chromatography, which measures the incorporation of the 3 [H] thymidine. Both compounds, agonist and antagonist, can be identified by this procedure. In another method, a mammalian cell or membrane preparation, which 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 intensify or block this interaction can then be measured. Alternatively, the response of a second messenger system known at once to the interaction of a compound to be classified and the FGF-13 receptor was measured and the ability of the compound to bind to the receptor and produce the response of the second messenger was measured. to determine if the compound is a potential agonist or antagonist. These second messenger systems include, but are not limited to, the cAMP guanylate cyclase, the ion channels or the hydrolysis of the phosphoinositidine. Examples of antagonist compounds include antibodies or, in some cases, oligonucleotides, which bind to the receptor for the polypeptide of the present invention, but do not elicit response from the second messenger or bind to the polypeptide itself of FGF-13. Alternatively, a potential antagonist may be a mutant form of the polypeptide that binds to the receptors, however, no response of the second messenger occurs and, therefore, the action of the polypeptide is effectively blocked. Another compound antagonistic to the gene of FGF-13 and the product of the gene is an antisensitive constructor, prepared using antisensitive technology. This technology can be used to control the expression of the gene through triple helix formation or antisensitive DNA or RNA, both methods are based on the binding of a polynucleotide to DNA or RNA. For example, the 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. An oligonucleotide of DNA is designed to be complementary to the region of the gene involved in transcription (triple helix, see Lee et al., Nucí Acids Res., 6: 3073 (1979); Cooney et al., Science, 241: 456 (1938) and Dervan et al., Science 251: 1360 (1991)). thus preventing 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 the cells, so that the antisensible RNA or DNA can be expressed in vivo to inhibit the production of the polypeptide. Potential antagonist compounds also include small molecules that bind to and occupy the ligand site of the receptors, thereby rendering the receptor inaccessible to this polypeptide, so that normal biological activity is impeded. Examples of small molecules include, but are limited to, small peptides or peptide-like molecules. Antagonist compounds can be employed to inhibit the effects of cell growth and proliferation of the polypeptides of the present invention on neoplastic cells and tissues, i.e. the stimulation of tumor angiogenesis and, therefore, the retardation or prevention of growth and proliferation. abnormal cell, for example, in the formation or growth of tumors. Antagonists can also be employed to prevent hypervascular diseases and prevent the proliferation of epithelial lens cells, after extracapsular cataract surgery. The prevention of the mitogenic activity of the polypeptides of the present invention may also be desired in cases such as restenosis, after balloon-type angioplasty. Antagonists can also be employed to prevent tissue growth of scars during wound healing. Antagonists can be employed in a composition with a pharmaceutically acceptable carrier, for example those described above. 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. Such compositions comprise a therapeutically effective amount of the polypeptide, agonist or antagonist and a pharmaceutically acceptable carrier or excipient. Such a carrier includes, but is not limited to, a saline solution, regulated saline solution, dextrose, water, glycerol, ethanol, and combinations thereof. The formulation must be suitable for the mode of administration. The invention also provides a pharmaceutical package or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention. Associated with these containers may be a notification in the form prescribed by the government agency that regulates the manufacture, use or sale of pharmaceutical or biological products, this notification reflects the approval by the manufacturing, use or sale agency 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 oral, topical, intravenous, intraperitoneal, intramuscular, subcutaneous, intranasal or intradermal routes. The pharmaceutical compositions are administered in an amount that is effective for the treatment and / or 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 many cases, they are administered in an amount of no more than about 8 mg / kg of body weight per day. In many cases, the dose is approximately 10 μg / kg to 1 mg / kg of body weight per day, taking into account the route of administration, symptoms, etc. In the specific case of topical administration, preferred administered doses are from about 0.1 μg to 9 mg per cm. The polypeptide of the invention and the agonist and antagonist compounds, which are polypeptides, can also be used according to the present invention for the expression of such polypeptides in vivo, which is often referred to as the "gene therapy". Thus, for example, cells can be treated with a polynucleotide (DNA or RNA) encoding the polypeptide ex vivo, the treated cells are then provided to a patient to be treated with the polypeptide. These methods are well known in the art. For example, cells can be treated by methods known in the art, by the use of RNA containing a retroviral particle encoding the polypeptide of the present invention. Similarly, cells can be treated in vivo for expression of the polypeptide in vivo, for example by methods known in the art. As is known, a producer cell, to produce the RNA containing a retroviral particle, which encodes the polypeptide of the present invention, can be administered to a patient to treat the cells in vivo and the expression of the polypeptide in vivo. These and other methods of administration of the polypeptide of the present invention by such methods will be apparent to those skilled in the art from the teachings of the present invention. For example, the expression vehicle for the treated cells may be other than a retroviral particle, for example, an adenovirus, which can be used to treat the cells in vivo prior to combination with a suitable delivery vehicle. Retroviruses from which the aforementioned retroviral plasmid vectors can be derived, include, but are not limited to, Moloney's Lucemia virus from Moloney, spleen necrosis virus, retroviruses such as Rous sarcoma virus. , Harvey's sarcoma virus, bird leukosis virus, gibbon simian leukemia virus, human immunodeficiency virus, adenovirus, myeloproliferative sarcoma virus and mammary tumor virus. In one embodiment, the vector of retorviral plasmid is derived from Moloney's 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 by Miller et al., Biotechniques Vol. 7, No. 9, 980-990 (1989), or any other promoter (e.g., cellular promoters, such as eukaryotic cell promoters). , which include, but are not limited to, histone, pol III, and β-actin promoters). 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, adenoviral promoters, such as the adenoviral major late promoter; or heterologous promoters, such as the cytomegalovirus (CMV) promoter; the respiratory syncytial virus (RSV) promoter; inducible promoters, such as the MMT promoter, the metaltionein promoter; heat shock promoters; the promoter of albumin; the ApoAI promoter; the promoters of human globin; promoters of the viral thymidine kinase, such as the Herpes Simplex thymidine kinase promoter; Retroviral LTRs (including modified retroviral LTRs, described above); the β-actin 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 the packaging cell lines to form the cell lines of the producer. Examples of transferable packaging cells include, but are not limited to, cell lines PE501,? -2,? -AM, PA12, T19-14X, VT-19-17-H2,? CRE,? -CRIP , GP + E + 86, GP + envAml2 and DAN, as described in Miller Human Gene Theory, Vol. 1, pages 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. These means include, but are not limited to, electroporation, the use of liposomes, and CaPO precipitation. In an alternative, the retroviral plasmid vector may be encapsulated in a liposome, or coupled to a lipid, and then administered to a guest. The producer cell line generates infectious retroviral vector particles, which include one or more nucleic acid sequences, which encode the polypeptides. Such retroviral vector particles can then be used, to transduce the eukaryotic cells, or in vi tro or in vivo. The transduced eukaryotic cells will express one or more nucleic acid sequences encoding the polypeptide. Eukaryotic cells that can be transduced include, but are not limited to, embryonic stem cells, embryonic carcinoma cells, as well as hematopoietic stem cells, hepatocytes, fibroblasts, myoblasts, keratinocytes, endothelial cells and bronchial epithelial cells. This invention also relates to the use of the genes of the present invention as part of a diagnostic assay for detecting diseases or susceptibility to diseases related to the presence of mutations in the nucleic acid sequences encoding the polypeptide of the present invention. Individuals carrying mutations in a gene of the present invention can be detected at the DNA level by a variety of techniques. The nucleic acids for diagnosis can be obtained from the patient's cells, such as blood, urine, saliva, tissue biopsy and autopsy material. Genomic DNA can be used directly for detection or can be amplified enzymatically by the use of PCR (Saiki et al., Na ture, 324: 163-166 (1986)) before analysis. The RNA or the 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, deletion and insertions can be detected by a change in the size of the amplified product, compared to the normal genotype. Peak mutations can be identified by hybridization of amplified DNA to radiolabeled RNA or alternatively, radiolabelled anti-sensitive DNA sequences. The sequences that correspond perfectly can be distinguished from duplexes not in correspondence by the digestion of the RNase A or by differences in the fusion temperatures. Genetic testing based on DNA sequence differences can be achieved by detecting the alteration in the electrophoretic mobility of DNA fragments in gels with or without denaturing agents. Suppressions and insertions of small sequences can be visualized by high resolution gel electrophoresis. DNA fragments from different sequences can be distinguished from the gradient gels of denatured formamide in which the mobilities of the different DNA fragments are delayed in the gel at different positions, according to their specific melting temperatures or partial melting. (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 the protection of RNase and SI or the chemical cleavage method (eg, Cotton et al., PNAS, USA, 85: 4397-4401 (1985)). Thus, the detection of a specific sequence of DNA can be achieved by methods such as hybridization, RNase protection, chemical splitting, direct DNA sequencing or the use of restriction enzymes (for example, Fragment Length Polymorphisms).
Restriction (RFLP)) and the Southern blot Genomic DNA In addition to the more conventional gel electrophoresis and the DNA sequence, mutations can also be detected by in situ analysis. The present invention also relates to a diagnostic assay for detecting altered levels of FGF-13 proteins in various tissues, since an over-expression of the proteins, compared to normal control tissue samples, can detect the presence of abnormal cell proliferation, for example, a tumor. The assays used to detect the levels of the 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 assay" An ELISA assay (Coligan, et al., Current Protocols in Immunology, 1 (2), Chapter 6, (1991)), comprises initially preparing an antibody specific to an antigen to the polypeptides of the present invention, preferably a monoclonal antibody.In addition to a reporter antibody is prepared against the monoclonal antibody.To the reporter antibody is attached a detectable reagent, such as radioactivity, fluorescence or, in this example, a strong horseradish peroxidase enzyme. of a host and incubated on a solid support, for example a polystyrene disk, which binds the proteins in the sample.Any free protein binding site in the The disc is then covered by incubation with a non-specific protein, such as bovine serum albumin. Next, the monoclonal antibody is incubated on the disc during this time the monoclonal antibodies bind to any polypeptide of the present invention attached to the polystyrene disk. All unbound monoclonal antibody is washed and separated with a buffer solution. The reporter antibody bound to horseradish peroxidase is now placed on the disk, which results in the binding of the reporter antibody to any monoclonal antibody bound to the protein of interest. The unbound reporter antibody is then washed and separated. The peroxidase substrates are then added to the disk and the amount of color developed in a given period of time is a measurement of the amount of the polypeptide of the present invention, which is in a given volume in the patient sample, when compared against a standard curve. A competition assay may be employed, wherein the antibody specific to a polypeptide of the present invention binds to a solid support and the labeled FGF-13 and a sample derived from the host is passed over this solid support and the amount of label detected. , for example, by liquid scintillation chromatography, can be correlated to an amount of a polypeptide of the present invention in the sample. A "sandwich" assay is similar to an ELISA assay. In a "sandwich" assay a polypeptide of the present invention is passed over a solid support and attached to the attached antibody to a solid support. A second antibody then binds to the polypeptide of interest. A third antibody, which is labeled and is specific to the second antibody, is then passed over the solid support and bound to the second antibody and an amount can then be quantified. The sequences of the present invention are also valuable for the identification of chromosomes. The sequence is specifically the target and can be hybridized to a particular location on an individual human chromosome. Likewise, there is a current need to identify particular sites on the chromosome. Few chromosome labeling reagents, based on the actual sequence data (polymorphism repeat) are currently available to mark the location of chromosomes. The mapping of the DNA to the chromosomes, according to the present invention, is a first important step in correlating these sequences with the genes associated with diseases. In brief, the sequences can be mapped to chromosomes by preparing the PCR primers (preferably 15 to 25 bp) of the cDNA. Computer analysis of the 3 'untranslated region is used to quickly select the primers, so that they do not extend for more than one exon in the genomic DNA, thus complicating the amplification process. These primers are then used for the classification of hybrid somatic cell PCRs containing individual human chromosomes. Only those hybrids that contain the human gene that correspond to the initiator will supply an amplified fragment.
The mapping of the somatic cell hybrid PCR is a rapid procedure for assigning a particular DNA to a particular chromosome. Using the present invention with the same oligonucleotide primers, the sub-location can be accomplished with panels of specific chromosome fragments or groups of large genomic clones in an analogous manner. Other mapping strategies that can be used similarly for mapping your chromosome include hybridization in itself, previously classified with labeled labeled fluxes and pre-selection by hybridization to build collections of the chromosome-specific cDNA. Fluorescence in-situ hybridization (FISH) from a cDNA clone to a metaphase chromosomal extension can be used to deliver a precise chromosomal location in one step. This technique can be used with a 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 mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be correlated with genetic map data. (Such data are found, for example, in V. McKusick, Mendelian Inheritance in Man (available online through the Johns Hopkins University Welch Medical Library.) The relationship between genes and diseases that have been mapped to some chromosomal region are Then, it is necessary to determine the differences in the cDNA or the genomic sequence between affected and unaffected individuals, if a mutation is observed in some or all of the individuals. affected, but not in a normal individual, then the mutation is probably the causative agent of the disease.With the current resolution of the techniques of physical mapping and genetic mapping, a cDNA located precisely in a chromosomal region associated with the disease, can be one of between 50 and 500 potential cause genes. (This supposes a mapping resolution of 1 megabase and one gene per 20 kb). Oligoptides, their fragments or other derivatives, or their analogues, or cells expressing them, can be used as an immunogen to produce antibodies. These antibodies can be, for example, polyclonal or monoclonal. The present invention also includes chimeric single chains and humanized antibodies, as well as Fab fragments, or the product of an Fab expression library. Various methods known in the art can be used for the production of such 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 administration of the polypeptides to an animal, preferably non-human. The antibody, thus obtained, will then bind to the polypeptides themselves. In this way, even a sequence encoding only a fragment of the polypeptides can be used to generate antibody that bind to the total native polypeptides. Such antibodies can then be used to isolate the polypeptide from the tissue expressing that polypeptide. For the preparation of monoclonal antibodies, any technique that delivers antibodies produced by continuous cultures of cell lines can be used. Examples include the hybridoma technique (Kohier and Milstein, 1975, Na ture, 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, to produce human monoclonal antibodies (Colé et al., 1985, in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pages 77-96). The technique described for the production of single chain antibodies (patent of E. U. A., No. 4,946,778), can be adapted to produce single chain antibodies to the immunogenic polypeptide products of this invention. Likewise, transgenic mice can be used to express 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 will be understood that the present invention is not limited to such examples. All parts or quantities, unless otherwise specified, are by weight. In order to facilitate understanding of the following examples, certain methods that occur frequently and / or terms will be described. "Plasmids" are designated by a lowercase letter p preceded and / or followed by capital letters and / or numbers. The starting plasmids here are commercially available, with advertising available on an unrestricted basis, or plasmids can be constructed available, according to published procedures. In addition, plasmids equivalent to those described are known in the art and will be apparent to those of ordinary skill in the art. "Digestion" of DNA refers to the catalytic cleavage of DNA with a restriction enzyme, which acts only in certain sequences in DNA. The various restriction enzymes used herein are commercially available and their reaction conditions, cofactors and other requirements are used as they will be known to those of ordinary skill in the art. For analytical purposes, typically 1 μg of plasmid or DNA fragment is used with about 2 units of enzyme in about 20 μl of the buffer. For the purpose of isolating DNA fragments for the construction of plasmids, typically 5 to 50 μg of the DNA are digested with 20 to 250 units of enzyme in a larger volume. Appropriate amounts of regulators and substrates for the particular restriction enzymes are specified by the manufacturer. Incubation times of approximately 1 hour at 37 ° C are those used ordinarily, but may vary according to the instructions of the supplier. After digestion, the reaction is subjected directly to electrophoresis in a polyacrylamide gel to isolate the desired fragment. The size separation of the split fragments is carried out using 8 percent of the polyacrylamide gel described by Goeddel, D. et al., Nucleic Acids Res. , 8: 4057 (1980). "Oligonucleotides" refer to any single-stranded polydeoxynucleotide or two strands of complementary polydeoxynucleotides, which can be chemically synthesized. Such synthetic oligonucleotides do not have 5'-phosphate and thus do not bind to another oligonucleotide without adding a phosphate with ATP in the presence of a kinase. A synthetic oligonucleotide will be ligated to a fragment that has not been dephosphorylated. "Ligation" refers to the process of forming phosphodiester bonds between two double-stranded nucleic acid fragments (Maniatis, T., et al., Id., P.146). Unless otherwise mentioned, ligation can be accomplished using known regulators and conditions 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).
Ej-ampio 1 Bacterial Expression and Purification of the FGF-13 protein The DNA sequence that encodes the FGF-13, ATCC # 97147, was initially amplified using oligonucleotide primers from the PCR, which correspond to the 5"sequences of the processed protein (minus the signal peptide sequence) and the 3 'sequences of the vector to the gene. gene were added to the 5 'and 3' sequences respectively. The primer 5 'oligonucleotide, 5 'GCCAGACCATGGAGAATCACCCGTCTCCTAAT 3' (SEQ ID NO: 3) contains a Neo restriction enzyme site. The 3 '5' sequence GATTTAAGATCTCGTGAGAGGGGCTGGGGCCCC 3 '(SEQ ID NO: 4) contains sequences complementary to the BglII site and is followed by 18 nucleotides of the coding sequence of FGF-13. The restriction enzyme sites correspond to the restriction enzyme sites in the bacterial expression vector pQE-60 (Qiagen, Inc. Chatsworth, CA 91311). PQR-60 codes for resistance to antibiotics (Amp1), a bacterial origin of replication (ori), an operator (P / O) of the regulator promoter of IPTG, a ribosome binding site (RBS), a 6-His tag and sites of the restriction enzyme. The pQE-60 was then digested with Ncol and BglII. The amplified sequences were ligated into pQR-60 and inserted into the frame with the sequence coding for the histidine tag and the ribosome binding site (RBS). The ligation mixture was then used to transfer E. coli strain M15 / rep 4 (Qiagen, Inc.) by the procedure described by Sambrook, J et al., Molecular Cloning: A Labora tory Manual, - Cold Spring Laboratory Press, (1989). Ml5 / rep4 contains multiple copies of plasmid pREP4, which express the lacl repressor and also confer resistance to kanamycin (Kan1). Transformants were identified by their ability to grow on LB plates and the ampicillin / kanamycin resistant colonies were selected. Plasmid DNA was isolated and confirmed by restriction analysis. The clones containing the desired builders were grown overnight (0 / N = in an LB medium of liquid culture supplemented with both Amp (100 ug / ml) and Kan (25 ug / ml). The O / N culture was used for inoculate a large culture in a ratio of 1: 100 to 1: 250. The cells grew at an optical density of 600 (OD600) between 0.4 and 0.6, then the IPTG ("Isopropyl-BD-thiogalacto-pyranoside") was added. at a final concentration of 1 mm IPTG induces by inactivation the lacl repressor, clarifies the P / O that leads to an increased gene expression.The cells grew for 3 to 4 extra hours.These cells were then collected by centrifugation. The cell pellet was solubid in a 6 molar chaotropic guanidine-HCl agent After clarification the solubid FGF-13 was purified from this solution by chromatography on a Nickel-Chelate column under conditions that allow binding by tight bonding. proteins that contain the 6-H label is (Hochuli), E et al., J. Chroma tography 411: 177-184 (1984). The proteins were eluted from the column in 6 molar guanidine-HCl, pH 5.0 and for the purpose of renaturation, adjusted to 3 molar guanidine-HCl, with 100 mM sodium phosphate, 10 mmolar glutathione (reduced) and 2 mmolar of glutathione (oxidized).
After incubation in this solution for 12 hours, the proteins were dialyzed to 10 mmolar sodium phosphate.
Example 2 Cloning and Expression of FGF-13, with the use of the baculovirus expression system. The DNA sequence encoding the full length of the FGF-13 protein, ATCC # 97147, was amplified using the oligonucleotide primers of the PCR corresponding to the 5 'and 3' sequences of the gene: The 5 'primer of the FGF- 13 has the sequence 5 'CTAGTGGATCCCGAGAATCACCCGTCTCCT 3' (SEQ ID NO: 5) and contains a BamH1 restriction enzyme site (in bold) so that cloning at this site places the baculovirus signal sequence in frame with 18 nucleotides of the FGF-13 gene downstream of the peptide cleavage site of the putative FGF-13 signal. The 3 'initiator of the sequence 5' CGACTTCTAGAACCTCGGGGATCTGGCTCC 3 '(SEQ ID NO: 6) and contains the cleavage site for the restriction endonucleases XBal and 18 nucleotides complementary to the sequence not translated 3 'of the gene. The amplified sequences were isolated from a 1% agarose gel, using commercially available equipment ("Geneclean," BIO 101 Inc., La Jolla, Ca.). The fragment was then digested with the respective endonucleases and purified again on a 1% agarose gel. This fragment was designated F2. The vector pA2gp (modification of vector pVL941, discussed below) was used for the expression of proteins using the baculovirus expression system (for review, see: Summmers, MD and Smith, G E. 1987, A Manual Of Methods For Baculovirus Vectors and Insect Cell Cul ture Procedures, Texas Agricultural Experimental Station Bulletin No. 1555). This expression vector contains the strong polyhedrin promoter of Autographa californica nuclear polyhedrosis virus (AcMNPV), followed by the recognition sites for the restriction endonucleases BamHl and Xbal. The polyadenylation site of simian virus (SV) 40 was used for efficient polyadenylation. For an easy relation of the recombinant virus the E. coli beta-galactosidase gene was inserted in the same orientation as the polyhedrin promoter followed by the polyadenylation signal of the polyhedrin gene. The polyhedrin sequences were flanked on both sides by viral sequences for the cell-mediated homologous recombination of the co-transfected wild-type viral DNA. Many other baculovirus vectors can be used in place of pA2, such as pRG1, pAc373, pVL941 and pACIMl (Luckow, V.A. and Summers, M.D., Virology, 170: 31-39).
The plasmid was digested with the restriction enzymes and dephosphorylated using the calf intestinal phosphatase by procedures known in the art. The DNA was then isolated from the 1% agarose gel using commercially available equipment ("Geneclean" BIO 101 Inc., La Jolla ¡, Ca.). This vector DNA is designated V2. The F2 fragment and the dephosphorylated V2 plasmid were ligated with the T4 DNA ligase. The E. coli DH5a cells are then transformed and the identified bacteria containing the plasmid (pBacFGF-13) using the respective restriction enzymes. The sequence of the cloned fragment was confirmed by the DNA sequence. 5 μg of plasmid pBacFGF-13 was co-transfected with 1.0 μg of a commercially available linearized baculovirus ("BaculoGold® baculovirus DNA", Pharmingen, San Diego, CA.) using the 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 plasmids, in each case, were mixed in well sterilized microtiter plates, containing 50 μl of serum-free Grace's medium (Life Technologies Inc., Gaithersburg, MD). Next, 10 μl of Lipofectin plus 90 μl of Grace's medium were added, mixed and incubated for 15 minutes at room temperature. Then the transfection mixture was added in drops to the Sf9 insect cells (ATC CRL 1711) seeded in 35 mm tissue culture plates with 1 ml of Grace's medium without serum. The plates were oscillated back and forth to mix the newly added solution. The plates were then incubated for 5 hours at 27 ° C. After 5 hours, the transfection solution was removed from the plate and 1 ml of Grace's insect medium supplemented with 10% fetal calf serum were added. Plates were placed back in an incubator and culture was continued at 27 ° C for four days. After four days, the supernatant was collected and plaque assays performed similarly as described by Sumares and Smith (supra). As a modification, an agar gel with "Blue Gal" (Life Technologies Inc., Gaithersburg) was used, which allowed easy isolation of the blue-stained 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, pages 9-10). Four days later, serial dilution of the virus was added to the cells and the blue stained plates were collected with the tip of an Eppendorf pipette. The agar containing the recombinant viruses was then resuspended in an Eppendorf tube containing 200 μl of Grace's medium. The agar was removed by brief centrifugation and the supernatant containing the recombinant baculovirus was used to infect the Sf9 cells seeded on 35 mm discs. Four days later, the supernatants of these culture discs were collected and then stored at 4 ° C. The Sf9 cells grew in Grace's medium, supplemented with 10% heat-inactivated FBS. The cells were infected with the recombinant baculovirus V-FGF-13 at a multiplicity of infections (MOI) of 2. Six hours later, the medium was removed and replaced with the SF900 II medium minus methionine and cysteine (Life Technologies Inc. , Gaithersburg). 42 hours later, 5 μCi of the 35g_met-ionin and 5μCi of the 35s-cysteine (Amersham) were added. The cells were further incubated for 16 hours before being collected by centrifugation and the labeled proteins were visualized by SDS-PAGE and autoradiography.
Example 3 Expression of recombinant FGF-13 of FGF-13 in COS cells Expression of plasmids, FGF-13-HA, derived from a pcDNA3 / Amp vector (Invitrogen), containing: 1) origin SV40 of replica, 2 ) ampicillin resistant gene, 3) replication origin of E. coli, 4) CMV promoter followed by a polylinker region, an SV40 intron and polyadenylation site. The DNA fragments encoding the entire precursor of FGF-13 and the HA tag fused in the frame to the 3 'end were cloned into the polylinker region of the vector, therefore, the expression of the recombinant protein was directed under the promoter. CMV. The HA tag corresponds to an epitope derived from the influenza hemagglutinin protein, as previously described (I. Wilson, H. Niman, R. Heighten, A. Cherenson, M. Connolly and R. Lerner, 1984, Cell 31 : 161, (1984)). Infusion of the HA tag to the target protein allows easy detection of the recombinant protein with an antibody that recognizes the HA epitope. The DNA fragments amplified by the PCR and the pcDNA3 / Amp vector were digested with the respective restriction enzymes and ligated. The ligand mixture was transformed into the SURE E. coli strain (Stratagene Cloning Systems, La Jolla, CA) the transformed culture was plated on ampicillin medium and the resistant colonies were selected. The plasmid DNA was isolated from the transformants and examined by restriction analysis in the presence of the correct fragment. For expression of recombinant FGF-13, COS cells were transfected with the expression vector by the DEAE-DEXTRAN method (J. Sambrook, E. Fritsch, T. Maniatis, Molecular Cloning: A Laboratory Manual, Col Spring Laboratory Press (1989)). The expression of the FGF-13-HA protein was detected by radio-labeling and the immunoprecipitation method (E. Harlow, D. Lane, Antibodies: A Labora tory Manual, Cold Spring Harbor Laboratory Press, (1988)). Cells were labeled for 8 hours with 35g-cysteine two days after transfection. The culture media were then harvested and the cells were lysed with detergent (RIPA regulator (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 were precipitated with an HA-specific monoclonal antibody. Precipitated proteins were analyzed on SDS gels. 15% PAGE.
Ex amp 4 Expression by Way of Gene Therapy The fibroblasts of a subject were obtained by the biopsa of the skin. The resulting tissue was placed in the tissue culture medium and separated into small pieces. Small fragments of tissue were placed on a wet surface of a tissue culture flask, approximately ten pieces were placed in each flask. The flask was turned upside down, closed tightly and left at room temperature overnight. After 24 hours at room temperature, the flask was inverted and the tissue fragments remained fixed at the bottom of the flask and the fresh medium (e.g., Ham's F12 medium), with 10% FBS, penicillin and streptomycin, it was added. This was then incubated at 37 ° C for about a week. At this time, the fresh medium was added and changed subsequently every several days. After two more weeks in culture, a monolayer of fibroblasts emerged. The monolayer was trypsinized and related in larger flasks. PMV-7 (Kirschmeier, PT et al., DNA, 1: 219-25 (1988) flanked by the long terminal repeats of murine sarcoma virus, from Moloney, was digested with EcoRI and HindlII and subsequently treated with phosphatase calf intestine The linear vector was fractionated on agarose gel and purified using glass beads The cDNA encoding a polypeptide of the present invention was amplified using the PCR primers corresponding to the 5 'and 3' end sequences. ', respectively. The 5 'primer containing an EcoRI site and the 3' primer containing a HindIII site. Equal amounts of the linear skeleton of murine sarcoma virus, Maloney, and the EcoRI and HindIII fragment were added together, in the presence of the T4 DNA ligase. The resulting mixture was maintained under conditions appropriate for the ligand of the two fragments. The ligation mixture was used to transform HB101 bacteria, which were placed on agar containing kanamycin in order to confirm that the vector has the gene of interest inserted properly. The amphotropic amphotropic pA317 or GP + aml2 packaging cells were grown in tissue culture at the confluent density in Dulbecco's Modified Eagles Medium (DMEM) with 10% 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 infectious viral particles that contain the gene (the packaging cells are now named as the producer cells). Fresh medium was added to the transduced producer cells, and then the medium was collected from a 10 cm plate of confluent producer cells. The spent medium, which contains the infectious viral particles, was filtered through a millipore filter to remove the detached production cells and this means was then used to infect the fibroblast cells. The medium was removed from the 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 fibroblasts will be infected and a selection is not required. If the title is very low, then it is necessary to use a retroviral vector that has a selectable marker, such as neo or his. The treated fibroblasts were then injected into the host, either alone or after they had grown to confluence in 3 microcarrier spheres of cytodex. The fibroblasts now produce the protein product. Numerous modifications and variants of the present invention are possible in the light of the foregoing teachings and, therefore, are within the scope of the appended claims, the invention can be practiced in a manner other than that particularly described.
LIST OF SEQUENCES (1) GENERAL INFORMATION (i) APPLICANT: HU AND COLLABORATORS. (Ü) TITLE OF THE INVENTION: GROWTH FACTOR 13 OF FIBROBLASTOS (Üi) NUMBER OF SEQUENCES: 8 (ÍV) CORRESPONDENCE DIRECTION: (A) RECIPIENT: CARELLA, BYRNE, BAIN, GILFILLAN, CECCI, STEWART & OLSTEIN (B) STREET: 6 BECKER FARM ROAD (C) CITY; ROSELAND (D) STATE: NEW JERSEY (E) COUNTRY: E.U.A. (F) POSTAL ZONE: 07068. (v) COMPUTER LEADABLE FORM: (A) TYPE OF MEDIA: 3.5-INCH (B) COMPUTER DISK: IBM PS / 2 (C) OPERATING SYSTEM: MS-DOS (D) SOFTWARE: WORD PERFECT 5.1 (vi) CURRENT DATA OF THE APPLICATION (A) NUMBER OF THE APPLICATION: (B) DEPOSIT DATE: Concurrently (C) CLASSIFICATION: (VÜ) PREVIOUS DATA OF THE APPLICATION. (A) NUMBER OF THE APPLICATION: 08 / 207,412 (B) DEPOSIT DATE: MARCH 8, 1994 (viii) INFORMATION OF THE APPORTER / AGENT: (A) NAME: FERRARO, GREGORY D. (B) REGISTRATION NUMBER: 36,134 (C) REFERENCE NUMBER / FILE: 325800-364 (ix) TELECOMMUNICATIONS INFORMATION: (A) TELEPHONE: 201-994-1700 (B) TELEFAX: 201-994-1744 (2) INFORMATION OF SEQ ID NO: l (i) CHARACTERISTICS OF THE SEQUENCE (A) LENGTH: 641 BASIC COUPLES (B) TYPE: NUCLEIC ACID (C) CORD CLASS: SIMPLE (D) TOPOLOGY: LINEAR (ii) TYPE OF MOLECULE: cDNA (xi) DESCRIPTION OF THE SEQUENCE: SEQ ID NO: l: CCCGCCTGCT GCCCAACCTC ACTCTGTGCT TACAGCTGCT «3ATTCTCTGC TGTCAAACTC 60 AGGGGGAGAA TCACCCGTCG CCTAAGGTTA ACCAGTACGT GAGGGACCAG GGCGCCATGA 120 CCGACCAGCT GAGCAGGCGG CAGATCCGCG AGTACCAACT CTACAGCAGG ACCAGTGGCA 180 AGCACGTGCA GGTCCCCGGG CGTCGCATCT CCGCCACCGC CGAGGACGGC AACAAGTTTG 240 C AAGCTCAT AGTGGAGACG «3ACACGTTTG GCAGCCGGGT TCGCATCAAA GGGGCTGAGA 300 GTGAGAAGTA CATCTGTATG AACAAGAGGG GCAAGCTCAT CGGGAAGCCC AGCGGGAAGA 3 60 GCAAAGACTG CGTGTTCACG GAGATCGTGC TGGAGAACAA CTATACGGCC TTCCAGAACG 420 CCCGGCA GA «GGGCTGGTTC ATGGTCTTCA CGCGGCAGGG GCGGCCCCGC CAGGCTTCCC 4 80 GCAGCCGCCA GAACCAGCGC GAGGCCCACT TCATCAAGCG CCTCTACCAA GGCCAGCTGC 54 0 CCTTCCCCAA C ACGCCGAG AAGCAGAAGC AGTTCGAGTT TGTGGGCTCC GCCCCCACCC 600 GTCGGACCAA GCGCACACGG CGGCCCCAGC CCCTCACGTA G 6 1 (2) INFORMATION OF SEQ ID NO: 2 (i) CHARACTERISTICS OF THE SEQUENCE (A) LENGTH: 212 AMINO ACIDS (B) TYPE: AMINO ACIDS (C) LACE CLASS: (D) TOPOLOGY: LINEAR (Ü) TYPE OF MOLECULE : PROTEIN (Xi) DESCRIPTION OF THE SEQUENCE: SEQ ID NO: 2: Arg Leu Leu Pro Asn Leu Thr Leu -20 Cys Leu Gln Leu Leu He Leu Cys Cys -15 Gln Thr -10 -5 Gla Gly Glu Asn His ro Ser Pro Asn Phe Asn Gln Tyr 1 Val Arg Asp Gln 5 10 Gly Wing Met Thr 15 Asp Gln Leu Ser Arg Arg Gln He Arg Glu Tyr 20 25 Gln Leu 30 Tyr Ser Arg Thr Ser Gly Lys His Val Gln Val Pro Gly 35 40 Arg Arg Ala Thr Gly Asn 45 He Ser Wing Glu Asp Lys Phe Wing Lys 50 55 Leu lie Val Glu Thr Asp Thr Phe Gly Ser Arg 60 Val Arg He Lys .65 70 Gly Wing Glu Ser Glu Lys Tyr He CyS Met Asn 75 Lys Arg Gly Lys 80 85 Leu lie Gly Lys 90 Pro Ser Gly Lys Ser Lys Asp Cys Val 95 100 Phe Thr Glu He Val Leu Glu Asn Ala Phe 105 Asn Tyr Thr Gln Asn 110 115 Wing Arg His Glu Gl Trp Phe Met Val Phe Thr Arg Gln 120 Gly Arg 125 130 Phe Arg Gln Wing Ser Arg 135 Ser Arg Gln Asn Gln Arg Glu Ala Hiá 140 145 Phe lie Lys Arg 150 Leu Tyr Gln Gly Gln Leu Pro Phe Pro Asn His 155 160 Wing Glu Lys Gln Lys Gln Phe Glu Phe Val Gly Ser Wing 165 Pro Thr 170 175 Arg Arg Thr Lys Arg Thr Arg Arg L Leeuu TThhrr 180 1 1 «85c; Pro Gln Pro 190 (2) INFORMATION OF SEQ ID NO: 3 (i) CHARACTERISTICS OF THE SEQUENCE (A) LENGTH: 32 BASIC COUPLES (B) TYPE: NUCLEIC ACID (C) LACE CLASS: SINGLE (D) TOPOLOGY: LINEAR (ii) TYPE OF MOLECULE: Oligonucleotide (xi) DESCRIPTION OF THE SEQUENCE: SEQ ID NO: 3: ft GCCAGACCAT GGAGAATCAC CCGTCTCCTA AT 32 (2) INFORMATION OF SEQ ID NO: 4 (i) CHARACTERISTICS OF THE SEQUENCE (A) LENGTH: 30 BASIC COUPLES (B) TYPE: NUCLEIC ACID (C) CORD CLASS: SINGLE (D) TOPOLOGY: LINEAR (Ü) TYPE OF MOLECULE: OLIGONUCLEOTIDE (Xi) DESCRIPTION OF THE SEQUENCE: SEQ ID NO: 4: GATTTAAGAT CTCGTGAGGG GCTGGGGCCG 30 (2) INFORMATION OF SEQ ID NO: 5 (i) CHARACTERISTICS OF THE SEQUENCE (A) LENGTH: 30 BASIC COUPLES (B) TYPE: NUCLEIC ACID (C) LACE CLASS: SINGLE (D) TOPOLOGY: LINEAR (ii) TYPE OF MOLECULE: Oligonucleotide (Xi) DESCRIPTION OF THE SEQUENCE: SEQ ID NO: 5: CTAGTGGATC CCGAGAATCA CCCGTCTCCT 30 (2) INFORMATION OF SEQ ID NO: 6 (i) CHARACTERISTICS OF THE SEQUENCE (A) LENGTH: 30 BASIC COUPLES (B) TYPE: NUCLEIC ACID (C) LACE CLASS: SINGLE (D) TOPOLOGY: LINEAR (ii) TYPE OF MOLECULE: Oligonucleotide (xi) DESCRIPTION OF SEQUENCE: SEQ ID NO: 6: CGACTTCTAG AACCTCGGGG ATCTGGCTCC 30 (2) INFORMATION OF SEQ ID NO: 7 (i) CHARACTERISTICS OF THE SEQUENCE (A) LENGTH: BASIC COUPLES (B) TYPE: NUCLEIC ACID (C) LACE CLASS: SIMPLE (D) TOPOLOGY: LINEAR (ii) TYPE OF MOLECULE: Oligonucleotide (xi) DESCRIPTION OF THE SEQUENCE: SEQ ID NO: 7: (2) INFORMATION OF SEQ ID NO: 8 (i) CHARACTERISTICS OF THE SEQUENCE (A) LENGTH: 30 BASIC COUPLES (B) TYPE: NUCLEIC ACID (C) LACE CLASS: SINGLE (D) TOPOLOGY: LINEAR (ii) TYPE OF MOLECULE: Oligonucleotide (xi) DESCRIPTION OF SEQUENCE: SEQ ID NO: l: GATTTACTCG AGCGTGAGGG GCTGGGGCCG 30

Claims (20)

  1. CLAIMS 1. An asylated polynucleotide, comprising a member selected from the group consisting of: (a) a polynucleotide, which encodes the polypeptide comprising amino acid -21 to amino acid 191, as set forth in SEQ ID NO: 2; (b) a polynucleotide, which encodes the polypeptide, comprising amino acid 1 to amino acid 191, as set forth in SEQ ID NO: 2; (c) a polynucleotide, capable of hybridising and which is at least 70% identical to the polynucleotide of (a) or (b); and (d) a polynucleotide fragment of the polynucleotides of (a), (b) or (c).
  2. 2. The polynucleotide of claim 1, wherein this polynucleotide is DNA.
  3. 3. The polynucleotide of claim 1, comprising from nucleotide 65 to nucleotide 641, as set forth in SEQ ID NO: 1.
  4. 4. An isolated polynucleotide, comprising a member selected from the group consisting of: (a) a polynucleotide, which encodes a mature polypeptide encoded by the DNA contained in Deposit No. 97147 of ATCC; (b) a polynucleotide, which encodes the polypeptide expressed by the DNA contained in Deposit No. 97147 of ATCC; (c) a polynucleotide, capable of hybridizing to, and which is at least 70% identical to the polynucleotide of (a) or (b); And (d) a polynucleotide fragment of the polynucleotides of (a), (b) or (c).
  5. 5. A vector containing the DNA of claim 2.
  6. 6. A host cell, genetically treated with the vector of claim 5.
  7. 7. A method for producing a polypeptide, which comprises: expressing the polypeptide encoded by this DNA from the host cell of claim 6.
  8. 8. A method for producing cells capable of expressing a polypeptide, which comprises genetically treating cells with the vector of claim 5.
  9. 9. A polypeptide, comprising a member selected from the group consisting of (i) a polypeptide, having the deduced amino acid sequence of SEQ ID NO: 2 and its fragments, analogs and derivatives; and (ii) a polypeptide encoding the cDNA of Deposit No. 97147 of ATCC and the fragments, analogs and derivatives of this polypeptide.
  10. 10. An antibody against the polypeptide of claim 9.
  11. 11. A compound, which inhibits the biological actions of the polypeptide of claim 9.
  12. 12. An agonist mimetic to the polypeptide of claim 9.
  13. 13. A method for the treatment of a patient in need of a FGF-13 polypeptide, this method comprises: administering to the patient a therapeutically effective amount of the polypeptide of claim 9.
  14. 14. A method for the treatment of a patient in need of inhibiting the FGF-13 polypeptide, this method comprises: administering to the patient a therapeutically effective amount of the compound of claim 11.
  15. 15. The method of claim 13, wherein the therapeutically effective amount of the polypeptide is administered by supplying the patient with the DNA encoding the polypeptide and expressing this polypeptide in vivo.
  16. 16. The method of claim 14, wherein the compound is a polypeptide and the therapeutically effective amount of the compound is administered by supplying the patient with the DNA encoding the antagonist and expressing this antagonist in vivo.
  17. 17. A method for identifying active compounds as agonists to the polypeptide of claim 9, this method comprises: (a) combining a compound to be classified to a cell-containing reaction mixture, under conditions where the cells are normally stimulated by the polypeptide , the reaction mixture contains a label incorporated in the cells, as they proliferate; and (b) determining the extent of proliferation of the cells, to identify whether the compound is an effective agonist.
  18. 18. A method for identifying active compounds as antagonists to the polypeptide of claim 9, this method comprises: (a) combining a compound to be classified, the polypeptide and the reaction mixture containing cells under conditions where the cells are normally stimulated by the polypeptide, this reaction mixture contains a label incorporated into the cells as they proliferate; and (b) determining the extent of proliferation of the cells, to identify whether the compound is an effective antagonist.
  19. 19. A method for diagnosing a disease or susceptibility to a disease related to a sub-expression of the polypeptide of claim 9, this method comprises: determining a mutation in the nucleic acid sequence encoding the polypeptide.
  20. 20. A diagnostic procedure, which comprises: analyzing the presence of the polypeptide of claim 9, in a sample derived from a host.
MXPA/A/1997/009217A 1997-11-27 Growth factor 13 of fibroblas MXPA97009217A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
MXPA/A/1997/009217A MXPA97009217A (en) 1997-11-27 Growth factor 13 of fibroblas

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
MXPA/A/1997/009217A MXPA97009217A (en) 1997-11-27 Growth factor 13 of fibroblas

Publications (2)

Publication Number Publication Date
MX9709217A MX9709217A (en) 1998-07-31
MXPA97009217A true MXPA97009217A (en) 1998-11-09

Family

ID=

Similar Documents

Publication Publication Date Title
US5773252A (en) Fibroblast growth factor 15
AU702131B2 (en) Fibroblast growth factor-14
US5763214A (en) Fibroblast growth factor 11
US5728546A (en) Fibroblast growth factor 13
AU702682B2 (en) Fibroblast growth factor 13
WO1996039509A1 (en) Fibroblast growth factor 15
US7208584B2 (en) Fibroblast growth factor-13
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
MXPA97009217A (en) Growth factor 13 of fibroblas
AU752592B2 (en) Fibroblast growth factor 15
MXPA97007897A (en) Factor-11 fibroblas growth
MXPA97006854A (en) Factor 14 of fibroblas growth
MXPA97009570A (en) Growth factor 3 endothelial, vascular, hum
JP2003047488A (en) Human hepatic cancer-derived growth factor-2
KR19980703870A (en) Fibroblast Growth Factor-11
MXPA97008493A (en) Factor 15 of fibroblas growth
CA2220988A1 (en) Fibroblast growth factor 13
KR19980703677A (en) Fibroblast Growth Factor-14