MXPA97009744A - Enzima convertidora tnf-a - Google Patents

Enzima convertidora tnf-a

Info

Publication number
MXPA97009744A
MXPA97009744A MXPA/A/1997/009744A MX9709744A MXPA97009744A MX PA97009744 A MXPA97009744 A MX PA97009744A MX 9709744 A MX9709744 A MX 9709744A MX PA97009744 A MXPA97009744 A MX PA97009744A
Authority
MX
Mexico
Prior art keywords
tace
ser
leu
asp
lys
Prior art date
Application number
MXPA/A/1997/009744A
Other languages
Spanish (es)
Other versions
MX9709744A (en
Inventor
P Cerretti Douglas
J March Carl
A Black Roy
Rauch Charles
Original Assignee
Immunex Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US08/655,345 external-priority patent/US5830742A/en
Application filed by Immunex Corporation filed Critical Immunex Corporation
Publication of MX9709744A publication Critical patent/MX9709744A/en
Publication of MXPA97009744A publication Critical patent/MXPA97009744A/en

Links

Abstract

The present invention relates to a metalloprotease that converts TNF-alpha from the 26 KD cell form to the 17 KD cell form, the cDNA sequence has been isolated and purified and known. In particular, the protease has a molecular weight of about 80 kD. The isolated and purified protease is useful for designing an inhibitor thereof, and may find use as a therapeutic agent. Assays for detecting the protease inhibitory activity of a molecule are also an aspect of the invention.

Description

ENZIMA CONVERTIDORA TNF-a FIELD OF THE INVENTION The invention is directed to a purified and isolated TNF-α converting enzyme, the nucleic acids encoding such an enzyme, processes for the production of recombinant TNF-α convertases, pharmaceutical compositions containing such enzymes, and their use in various trials and therapies.
BACKGROUND OF THE INVENTION Tumor necrosis factor-a (TN F-a, also known as "cachectin") is a mammalian protein capable of inducing a variety of effects in numerous cell types. TNF-α was initially characterized by its ability to cause lysis of tumor cells and is produced by activated cells such as mononuclear phagocytes, T cells, B cells, mast cells and NK cells. There are two forms of TNF-a, a type I membrane protein with a relative molecular mass of 26,000 (26 kD) and a soluble 17kD form generated from the protein bound to the cell by proteolytic cleavage. TNF-a is a major mediator of the host response to a gram-negative bacterium. Lipopolysaccharide (LPS, also called endotoxin), derived from the cell wall of gram-negative bacteria, is a potent stimulator of TNF-a synthesis. Because the deleterious effects, which may result from overproduction or unregulated production of TNF-α, are extremely serious, considerable efforts have been made to control or regulate the serum level of TNF-α. An important part in the effort to effectively control TNF-a levels is in the understanding of the mechanism of TNF-a biosynthesis. The mechanism by which TNF-a is secreted has not been previously elucidated. Kriegler et al. Cell, 53:45 (1988) made the conjecture that the "secretion" of TNF-a is due to the conversion of the molecule bound to the 26 kD membrane by the then unknown proteolytic enzyme or protease. Scuderi et al. , J. Immunology, 143: 168 (1989), suggested that the release of TNF-α from human leukocyte cells is dependent on one or more serine proteases, for example, a leukocyte elastase or trypsin. It was found that a serine protease inhibitor, p-toluenesulfonyl-L-arginine methyl ester, suppresses the release of TNF-a from human leukocyte in a concentration-dependent manner. Scuderi et al. they suggested that an arginine methyl ester competes for the arginine ligation site at the reactive center of the enzyme and therefore blocks hydrolysis. The inhibitor's lysine and phenylalanine analogues reportedly failed to resemble arginine methyl ester. However, it was never shown that this compound acted by inhibiting a protease that cuts 26 kD TNF. More recently, it has been reported that metalloprotease inhibitors block the release of TNF from THP-1 cells. See Mohler et al. , Nature 370: 218 (1994); Gearing et al, Nature, 370: 555 (1994); and McGeehan et al. , Nature, 370: 568 (1 994). Most, but not all, proteases recognize a specific amino acid sequence. Some proteases mainly recognize residues located in the N-terminus of the cut ligature, some recognize residues located in terminal C of the cut ligature, and some proteases recognize residues on both sides of the cut ligature. The metalloprotease enzymes utilize a bound metal ion, generally Zn2 *, to catalyze the hydrolysis of the peptide ligation. The metalloproteases are involved in the destruction of the union (the metalloproteases matrices), the regulation of blood pressure (angiotensin converting enzyme) and the regulation of hormone-peptide levels (neutral endopeptidase 24. 1 1) SUMMARY OF THE INVENTION The invention pertains to the biologically active TNF-a converting enzyme "TACE" as an isolated and purified polypeptide. In addition, the invention is directed to isolated nucleic acids encoding TACE and to expression vectors comprising a cDNA encoding TACE. Within the scope of this invention are host cells that have been transfected or transformed with expression vectors comprising a cDNA encoding TACE, and the processes for producing TACE by culturing such host cells under favorable conditions for the expression of TACE. By virtue of the purification of TACE, the antibodies, and in particular, the monoclonal antibodies against TACE are in one aspect of the invention. In addition, assays using TACE to classify potential inhibitors thereof, and methods for using TACE as a therapeutic agent for the treatment of diseases mediated by TNF-a linked to the cell or other molecules, are encompassed by this invention. Additionally, methods using TACE in the design of inhibitors thereof are also an aspect of the invention. The isolated and purified metalloprotease of the invention is capable of converting TNF-from the 26 kD membrane bound form to the 17 kD form, and having a molecular weight between about 66 kD and about 97 kD. The TACE cDNA sequence is shown in SEQ ID NO: 1. The isolated and purified transforming enzyme TNF-α ("TACE") comprises amino acids 18-824 of SEQ I D NO: 2. Inhibition of TACE inhibits the release of TNF-a in serum and other extracellular spaces. TACE inhibitors would therefore have clinical utility in treating conditions characterized by overproduction or upregulated production of TNF-α. A TACE inhibitor particularly useful for certain pathological conditions would selectively inhibit TACE, without affecting the serum levels of TNF-β (also known as lymphotoxin). Overproduction or unregulated production of TNF-a has been implicated in certain conditions and diseases, for example, Systemic Inflammatory Response Syndrome, reperfusion injury, cardiovascular disease, infectious disease such as HIV infection and HIV neuropathy, disorders. gynecological or obstetric, self-immunity / inflammatory disease, atopic / allergic diseases, malignancy, transplants including organ transplant rejection or graft-versus-host disease, cachexia, congenital, dermatological, neurological, renal diseases, toxicity and diseases metabolic / idiopathic.
TACE inhibitors would prevent the cutting of cell-bound TNF-α, thereby reducing the level of TNF-α in serum and tissues. The present invention encompasses such an embodiment and comprises a method for inhibiting the TNF-α cut-off of cell membranes in a mammal comprising administering to said mammal an effective amount of a compound that inhibits the proteolytic activity of TNF-α from an enzyme comprising the amino acid sequence from 18 to 671 through 824 of SEQ ID NO: 2. In addition, the invention comprises a method for treating a mammal having a disease characterized by an overproduction or an overregulated production of TNF-α, which comprises administering to the mammal a composition comprising an effective amount of a compound that inhibits proteolytic activity. of TNF-a of an enzyme comprising the amino acid sequence 18-824 of SEQ ID NO: 2. For such inhibitors they would be of important clinical utility and could be potential therapeutics for treating the disorders related to TN F-a listed above. The isolation and purification of TACE would provide a significant advance in the effort to develop inhibitors of such an enzyme, and the treatment of TNF-associated diseases, and truly, would lead to the use of TACE by itself as a therapeutic agent for certain physiological disorders. . For example, in addition to TNF-α, other cytokines as well as cytokine receptors and various adhesion proteins can be released from the cell surface by TACE or related proteases. TACE can be administered to modulate or remove cell surface cytokines, cytokine receptors and adhesion proteins involved in the growth of tumor cells, inflammation or fertilization.
DETAILED DESCRIPTION OF THE INVENTION A human TNF-a converting enzyme ("TACE") encoding cDNA has been isolated and described in SEQ ID NO: 1. This discovery of human TACE encoding cDNA allows the construction of expression vectors comprising TACE encoding nucleic acid sequences; host cells transfected or transformed with the expression vectors; Human TACE biologically active as isolated and purified proteins; and antibodies immunoreactive with TACE. The TACE polypeptides isolated and purified according to the invention are useful for detecting the TACE inhibitory activity of a molecule. In such a method involving conventional and routine techniques, a molecule of unknown TACE inhibitory activity is mixed with a substrate and incubated with a TACE polypeptide. The degree of substrate cutting can then be determined chromatographically.
In addition, the TACE polypeptides according to the invention are useful for the design based on the structure of a TACE inhibitor. Such a design would comprise the steps of determining the three-dimensional structure of such TACE polypeptide, analyzing the three-dimensional structure for possible sites of ligation of the substrates, synthesizing a molecule incorporating a predictable reactive site, and determining the TACE inhibitory activity of the molecule.
Immunoreactive antibodies with TACE, and in particular, monoclonal antibodies against TACE, are now available through the invention. Such antibodies may be useful for inhibiting TACE activity in vivo and for detecting the presence of TACE in a sample. As used herein, the term "TACE" refers to a genus of polypeptides that are capable of converting the 26kD cell membrane bound form of TNF-α (which includes an intracellular region, a membrane region, and an extracellular region), in the soluble 17kD form comprising the 156 C-terminal residues of the TN protein Fa. TACE encompasses proteins having the sequence of amino acids 18 to 824 of SEQ ID NO: 2, as well as those proteins that have a high degree of similarity (at least 80%, and more preferably 90% homology) with the amino acid sequence 18 to 824 of SEQ I D NO: 2, and whose proteins are biologically active. In addition, TACE refers to the biologically active gene products of nucleotides 52-2472 of SEQ ID NO: 1. Also encompassed by the term "TACE" are the membrane-bound proteins (which include an intracellular region, a membrane region and an extracellular region), and soluble or truncated proteins, which mainly comprise the extracellular portion of the protein , they retain biological activity and are capable of being secreted. Specific examples of such soluble proteins are those comprising amino acid sequence 18-671 of SEQ ID NO: 2. Truncated versions are those that have less than the extracellular portion of the protein and comprise, for example, amino acids 18-477 of SEQ ID NO: 2, or that comprise substantially all of the catalytic domain, i.e., amino acids 21 to 477 of SEQ ID NO: 2. The isolated and purified TACE according to the invention has a molecular weight between about 66 kD and about 97 kD as determined by SDS-polyacrylamide gel electrophoresis (SDS-PAGE). More specifically, it was found that TACE has a molecular weight of approximately 80 kD as determined by SDS-PAGE.
The term "isolated and purified" as used herein, means that TACE is essentially free of association with other proteins or polypeptides, for example, as a purification product of a recombinant host cell culture or as a purified product of a non-recombinant source. The term "substantially purified" as used herein, refers to a mixture containing TACE and is essentially free of association with other proteins or polypeptides, but by the presence of known proteins that can be removed using a specific antibody, and that substantially purified TACE retains biological activity. The term "purified TACE" refers to both the "isolated and purified" form of TACE and the "substantially purified" form of TACE, as both are described herein. The term "biologically active" as it refers to TACE, means that TACE is capable of converting the 26kD cell form of TN F-a into the 17 kD form. A "nucleotide sequence" refers to a polynucleotide molecule in the form of a separate fragment or as a component of a larger nucleic acid construct, which has been derived from DNA or RNA isolated in at least one substantially pure form ( that is, free of endogenous contaminating materials), and in an amount or concentration that allows the identification, manipulation and recovery of their component nucleotide sequences by standard biochemical methods (such as those set forth in Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd ed. Cold Spring Harbor Laboratiry, Cold Spring Harbor, NY (1989)). Such sequences are preferably provided in the form of an open reading frame uninterrupted by internal non-translated sequences, or introns, which are typically present in eukaryotic genes. Untranslated DNA sequences may be present 5 'or 3' of an open reading frame, where they do not interfere with the manipulation or expression of the ciphering region. A "TACE variant" as referred to herein, means a polypeptide substantially homologous to natural TACE, but having an amino acid sequence different from that of natural TACE (human, mouse or other mammalian species) due to a or more deletions, insertions or substitutions. The variant amino acid sequence is preferably at least 80% identical to a natural TACE amino acid sequence, most preferably at least 90% identical. The percentage of identity can be determined, for example, by comparing the sequence information using the GAP computer program, version 6.0 described by Devereux et al. (Nucí, Acids Res.12: 387, 1984) and available from University of Wisconsin Genetics Computer Group (UWGCG).
The GAP program uses the alignment method of Needleman and Wunsch (J. Mol. Biol. 48: 443, 1970), as reviewed by Smith and Waterman (Adv. Appl. Math 2: 482, 1981). The preferred omission parameters for the GAP program include: (1) a unary comparison matrix (containing a value of 1 for identities and 0 for non-identities) for nucleotides, and the heavy comparison matrix of Gribskov and Burgess, Nucí. Acids Res. 14: 6745, 1986, as decribed by Schwartz and Dayhoff, eds. Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, p. 353-358, 1979; (2) a penalty of 3.0 for each gap and an additional penalty of 0.10 for each symbol in each gap; and (3) no penalty for final gaps. The variants may comprise conservatively substituted sequences, meaning that a given amino acid residue is replaced by a residue having similar physicochemical characteristics. Conservative substitutions are well known in the art and include the substitution of one aliphatic residue for another, such as He, Val, Leu or Ala for another, or substitutions of one polar residue for another, such as between Lys and Arg.; Glu and Asp; or Gln and Asn. Conventional methods and procedures can be used to make and use such variants. Other conservative substitutions, for example, substitutions of whole regions having similar hydrophobic characteristics, are well known and routinely performed. Naturally occurring variants of TACE are also encompassed by the invention. Examples of such variants are proteins that result from alternating mRNA splicing events or proteolytic cleavage of the TACE protein, where the proteolytic property of TACE is retained. The alternating splicing of mRNA can produce a truncated but biologically active TACE protein, such as soluble form that naturally occurs from the protein, for example, as shown in SEQ ID NO: 4. Variations attributable to proteolysis include, for example, differences in the N- or C- terminals on the expression in different types of host cells, due to the proteolytic removal of one or more terminal amino acids of the TACE protein (generally of terminal amino acids 1 - 5) . As stated above, the invention provides isolated and purified, or homogeneous, TACE polypeptides, both reccombinant and non-recombinant. Variants and derivatives of the natural TACE proteins that retain the desired biological activity can be obtained by mutations of nucleotide sequences that encode natural TACE polypeptides. Alterations of the natural amino acid sequence can be achieved by any of a number of conventional methods. Mutations can be introduced into particular sites by synthesizing oligonucleotides containing a mutant sequence, flanked by restriction sites that allow ligation to fragments of the natural sequence. After ligation, the resulting reconstructed sequence encodes an analog having the insertion, substitution or deletion of the desired amino acid. Alternatively, site-specific mutagenesis procedures of targeted oligonucleotides can be employed to provide an altered gene wherein the "codons" can be altered by substitution, deletion or insertion. Exemplary methods for making the previously exposed alterations are described by Walder et al. (Gene 42: 133, 1986); Bauer et al. (Gene 37:73, 1885); Craik (BioTechniques, January 1985, 12-19); Smith et al. (Genetic Engineering: Principles and Methods, Plenum Press, 1981); Kunkel (Proc. Nati, Acad. Sci. USA 82: 488, 1985); Kunkel et al. (Methods in Enzymol., 154: 367, 1987); and U.S. Patent Nos. 4,518,584 and 4,737,4642 all being incorporated by reference. TACE can be modified to create TACE derivatives by forming aggregated or covalent conjugates with other chemical moieties, such as glycosyl groups, polyethylene glycol (PEG) groups, lipids, phosphate, acetyl groups, and the like. The covalent derivatives of TACE can be prepared by linking the chemical moieties to the functional groups on the amino acid side chains of TACE or at the N-terminus or C-terminus of a TACE polypeptide or the extracellular domain thereof. Other TACE derivatives within the scope of this invention include added or covalent conjugates of TACE or its fragments with other proteins or polypeptides, such as by synthesis in recombinant culture as N-terminal or C-terminal fusions. For example, the conjugate may comprise a leader or signal polypeptide sequence (eg, the leader of Saccharomyces factor a) at the N-terminus of a TACE polypeptide. The signal or leader peptide co-translationally or post-translationally directs the transfer of the conjugate from its synthesis site inside or outside the cell membrane or cell wall.
TACE polypeptide conjugates can comprise added peptides to facilitate the purification and identification of TACE. Such peptides include, for example, poly-His or the antigenic identification peptides described in U.S. Patent No. 5,011,912 and Hopp et al, Bio / Technology 6: 1204, 1988. The invention also includes TACE polypeptides with or without associated natural pattern glycosylation. TACE expressed in yeast or mammalian expression systems (e.g., COS-7 cells) may be similar to or significantly different from a natural TACE polypeptide in molecular weight and glycosylation pattern, depending on the choice of the expression system. TACE polypeptides in bacterial expression systems, such as E. coli, provide non-glycosylated molecules. The glycosyl groups can be removed by conventional methods, in particular those using glycopeptidase. In general, glycosylated TACE can be incubated with a molar excess of glycopeptidase (Boehringer Mannheim). Equivalent DNA constructs that encode various additions or substitutions of amino acid sequences or residues, or deletions of internal or terminal sequences or residues not necessary for biological activity, are encompassed by the invention. For example, N-glycosylation sites in the extracellular domain of TACE can be modified to prevent glycosylation, allowing expression of an analogous reduced carbohydrate in yeast and mammalian expression systems. N-glycosylation sites in eukaryotic polypeptides are characterized by a triple of amino acids Asn-X-Y, wherein X is an amino acid except Pro and Y is Ser or Thr. Appropriate substitutions, additions or deletions to the nucleotide sequence encoding these triples will result in the prevention of binding of carbohydrate residues to the Asn side chain. The alteration of a single nucleotide, chosen so that Asn is replaced by a different amino acid, for example, is sufficient to inactivate a glycosylation site N. Known procedures for inactivating N-glycosylation sites in proteins include those described in the patent U.S. 5,071, 972 and EP 276,846, incorporated herein by reference. In another example, sequences encoding Cys residues that are not essential for biological activity can be altered to cause the Cys residues to be suppressed or replaced with other amino acids, preventing the formation of incorrect intramolecular disulfide bridges upon renaturation. Other equivalents are prepared by modifying adjacent dibasic amino acid residues to enhance expression in yeast systems, in which the activity of KEX2 proteases is present. EP 212,914 describes the use of site-specific mutagenesis to inactivate KEX2 protease processor sites in a protein. The KEX2 protease processing sites are inactivated by deletion, addition or substitution of residues to alter the Arg-Arg, Arg-Lys and Lis-Arg pairs to eliminate the occurrence of these adjacent basic residues. Lys-Lys pairs are considerably less susceptible to KEX2 cleavage, and the conversion of Arg-Lys or Lys-Arg to Lys-Lys represents a preferred and conservative approach to inactivate KEX2 sites. Nucleic acid sequences within the scope of the invention include isolated RNA and DNA sequences that hybridize to the natural TACE nucleotide sequences described herein under conditions of high or moderate severity, and that encode biologically active TACE. The conditions of moderate severity, as is known to those having ordinary skill in the art, and as defined by Sambrook et al. Molecular Cloning: A Laboratory Manual, 2 ed. Vol 1, page 1 .101 -104, Cold Spring Harbor Laboratory Press, (1989), include the use of a prewashing solution of 5 X SSC, 0.5% SDS, 1.0 mM EDTA (pH 8.0) and hybridization conditions of approximately 50 ° C - 60 ° C, 5 X SSC, overnight, preferably 55 ° C. Conditions of high severity include higher temperatures of hybridization and washing. The skilled artisan will recognize that the temperature and concentration of salt in the wash solution can be adjusted as necessary according to factors such as the length of the test. Due to the known degeneracy of the genetic code wherein more than one codon can encode the same amino acid, a DNA sequence can vary from that shown in SEQ ID NO: 1 and still encode a TACE protein having the amino acid sequence of SEQ ID. NO: 2 Such variant DNA sequences can result from silent mutations (e.g., occurring during PCR amplification), or they can be the product of deliberate mutagenesis of a natural sequence. The invention thus provides isolated DNA sequences encoding biologically active TACE, selected from: (a) the coding region of a natural mammalian TACE gene; (b) cDNA comprising the nucleotide sequence presented in SEQ ID NO: 1; (c) DNA capable of hybridization to a DNA of (a) under moderately severe conditions and encoding biologically active TACE; and (d) DNA, which is degenerated as a result of the genetic code to a DNA defined in (a), (b) or (c) and which codes for biologically active TACE. The TACE proteins encoded by such equivalent DNA sequences are encompassed by the invention. DNAs that are equivalent to the DNA sequence of SEQ ID NO: 1 will hybridize under moderately severe or highly severe conditions to a natural double-stranded DNA sequence encoding polypeptides comprising 18-Xaa amino acid sequences of SEQ ID NO: 2, wherein Xaa is an amino acid from 671 to 824. Examples of the TACE proteins encoded by such DNA, include, but are not limited to, TACE fragments (soluble or membrane bound) and TACE proteins comprising site ( s) N-inactivated glycosylation (s), KEX2 protease processing site (s), or substitution (s) of conservative amino acid (s), as previously described. TACE proteins encoded by DNA derived from other mammalian species, wherein the DNA will hybridize under conditions of moderate or high severity to complement the cDNA of SEQ ID NO: 1 are also encompassed. Alternatively, proteins that bind TACE, such as the anti-TACE antibodies of the invention, can be ligated to a solid phase such as a column chromatography matrix or a similar substrate suitable for identifying, separating or purifying expressing cells. the TACE on its surface. The adhesion of proteins that bind TACE to a solid phase contact surface can be achieved by any means, for example, magnetic microspheres can be coated with proteins that bind TACE and held in the incubation vessel through a magnetic field. Suspensions of cell mixtures are contacted with the solid phase having proteins that bind TACE therein. Cells that have TACE on their surface bind to the fixed TACE binding protein and then unbound cells are washed out. The affinity ligation method is useful for purifying, classifying or separating such cells that express TACE from the solution. Methods of releasing positively selected cells from the solid phase are known in the art and encompass, for example, the use of enzymes. Such enzymes are preferably non-toxic and non-detrimental to the cells and are preferably directed to cleaving the bound partner to the cell surface. Alternatively, mixtures of cells suspected of containing cells that express TACE can first be incubated with a biotinylated TACE-binding protein. Incubation periods are typically at least one hour in duration to ensure sufficient binding to TACE. The resulting mixture is then passed through a column packed with beads coated with avidin, by means of which the high affinity of biotin for avidin provides the binding of the cells that bind TACE to the beads. The use of avidin-coated beads is known in the art. See Berenson, et al. , J. Cell. Biochem., 10D: 239 (1989). The washing of the unbound material and the release of the ligated cells is carried out using conventional methods. In the methods previously described, the proteins that bind TACE suitable are the anti-TACE antibodies, and other proteins that are capable of binding with high affinity of TACE. A preferred TACE binding protein is an anti-TACE monoclonal antibody obtained, for example, as described in Example 4. TACE polypeptides can exist as oligomers, such as covalently linked or non-covalently bonded trimers or dimers. The oligomers can be linked by disulfide bonds formed between cysteine residues in different TACE polypeptides. In an embodiment of the invention, a TACE dimer is created by fusing TACE to an Fe region of an antibody (e.g., IgG 1) in a manner that does not interfere with the biological activity of TACE. The Fe polypeptide is preferably fused to the C-terminus of a soluble TACE (comprising only the extracellular domain). The preparation of fusion proteins comprising heterologous polypeptides fused to various portions of polypeptides derived from antibodies (including the Fe domain) has been described, for example, by Ashkenazi et al. (PNAS USA 88: 1 0535, 1991) and Byrn et al. (Nature 344: 677, 1990), incorporated herein by reference. A fusion of gene encoding the fusion protein TACE: Fc is inserted into an appropriate expression vector. TACE: Fc fusion proteins are allowed to join together much like the antibody molecules, over which they exchange disulfide ligands of between polypeptides Fe, producing divalent TACE If the fusion proteins are made with both heavy and light chains of an antibody, it is possible to form a TACE oligomer with as many as four extracellular regions of TACE. Alternatively, one can bind two soluble TACE domains with a peptide linker. Recombinant expression vectors containing a nucleic acid sequence encoding TACE can be prepared using well-known methods. Expression vectors include an operably linked TACE DNA sequence. to suitable translational or trans-transcriptional regulatory nucleotide sequences, such as those derived from an insect, viral, microbial or mammalian gene Examples of regulatory sequences include promoters, trans-transcriptional promoters or operators, a ribosomal ligation site of mRNA, and appropriate sequences that they control the initiation and termination of transcription and translation. The nucleotide sequences are "operably linked" when the regulatory sequence is functionally related to the TACE DNA sequence. Thus, a promoter nucleotide sequence is operably linked to a TACE DNA sequence if the promoter nucleotide sequence controls the transcription of TACE. the TACE DNA sequence, if the promoter nucleotide sequence controls the transcription of the TACE DNA sequence. The ability to replicate in the desired host cells, usually conferred by an origin of replication, and a selection gene by which the transformants are identified, can be further incorporated into the expression vector. In addition, sequences encoding appropriate signal peptides that are not naturally associated with TACE can be incorporated into expression vectors. For example, a DNA sequence for a signal peptide (secretory leader) can be fused uin-frame "to the TACE sequence, such that TACE is initially translated as a fusion protein comprising the signal peptide. which is functional in the intended host cells, enhances the extracellular secretion of the TACE polypeptide.The signal peptide can be excised from the TACE polypeptide on the TACE secretion of the cell.
Suitable host cells for expression of TACE polypeptides include prokaryotes, yeast or larger eukaryotic cells. Appropriate cloning and expression vectors for use with bacterial, fungal, yeast and mammalian cell hosts are described, for example, in Powels et al. Cloning Vectors: A Laboratory Manual, Elsevier, New York, (1985). Cell-free translation systems can also be used to produce TACE polypeptides using RNAs derived from DNA constructs described herein. Prokaryotes include gram negative or gram positive organisms, for example E. coli or Bacilli. Prokaryotic host cells suitable for transformation include, for example, E. coli, Bacillus subtilis, Salmonella ryphimurium, and various other species within the genera Pseudomonas, Streptomyces, and Staphylococcus. In a prokaryotic host cell, such as E. coli, a TACE polypeptide may include an N-terminal methionine residue to facilitate expression of the recombinant polypeptide in the prokaryotic host cell. The N-terminal Met can be excised from the expressed recombinant TACE polypeptide.
Expression vectors for use in prokaryotic host cells generally comprise one or more phenotypic selectable marker genes. A phenotypic selectable marker gene is, for example, a gene that encodes a protein that confers antibiotic resistance or that provides an autotrophic requirement. Examples of useful expression vectors for prokaryotic host cells include those derived from commercially available plasmids, such as the cloning vector pBR322 (ATCC 37017). PBR322 contains genes for ampicillin and tetracycline resistance, and thus simple means to identify transformed cells. To construct an expression vector using pBR322, an appropriate promoter and a TACE DNA sequence are inserted into the vector pBR322. Other commercially available vectors include, for example, pKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and pGEM 1 (Promega Biotec, Madison, Wl, EU). Promoter sequences commonly used for recombinant prokaryotic host cell expression vectors include β-lactamase (penicillinase), lactose promoter system (Chang et al., Nature 275: 615, 1978, and Goeddel et al., Nature 281: 544, 1979), tryptophan (trp) promoter system (Goeddel et al., Nucí Acids Res. 8: 4057, 1980; and EP-A-36766) and tac promoter (Maniatis, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, p.412, 1982). A particularly useful prokaryotic host cell expression system employs a phage promoter? PL and a thermolabile repressor sequence cl857ts. Plasmid vectors available from the American Type Culture Collection, which incorporate derivatives of the promoter? PL include plasmids pHU B2 (resident in E. coli strain JM B9 (ATCC 37092)) and pPLc28 (resident in E.coli RR 1 (ATCC 53082)). TACE polypeptides can alternatively be expressed in yeast host cells, preferably of the genus Saccharomyces (e.g., S. cerevisiae). Other yeast genera, such as Pichia, K. lactis or Kluyveromyces, can also be employed. Yeast vectors will frequently contain a replication sequence origin of a 2μ yeast plasmid, an autonomously replicating sequence (ARS), a promoter region, sequences for polyadenylation, sequences for transcription termination, and a selectable marker gene . Suitable promoter sequences for yeast vectors include, among others, promoters for metallothionein, 3-phosphoglycerate kinase, (Hitzeman et al., J. Biol. Chem. 255: 2073, 1980) or other glycolytic enzymes (Hess et al. , J. Adv. Enzyme Reg. 7: 149, 1968, and Holland et al., Biochem. 17: 4900, 1978), such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6 -phosphate isomerase, and clucokinase. Other vectors and promoters suitable for use in the expression of yeast are described further in Hitzeman, EPA-73,657 or Fleer et al. , Gene, 107: 285-195 (1991); and van den Berg et al. , Bio / Technology, 8: 135-1 39 (1990). Another alternative is the repressible ADH2 glucose promoter described by Russell et al. (J. Biol. Chem. 258: 2674. 1982) and Beier et al. (Nature 300: 724, 1982). The "shuttle" vectors replicable in both yeast and E. coli can be constructed by inserting DNA sequences from pBR322 for selection and replication in E. coli (Ampr gene and origin of replication) in the yeast vectors described above. The leader sequence of yeast factor-a can be used to direct the secretion of a TACE polypeptide. The leading factor-a sequence is often inserted between the promoter sequence and the structural gene sequence. See, for example, Kurjan et al. , Cell 30: 933, 1982; Bítter et al. , Proc. Nati Acad. Sci USA 81: 5330, 1984; U.S. Patent 4, 546, 082; and EP 324,274. Other leader sequences suitable for facilitating the secretion of recombinant polypeptides from yeast hosts are known to those skilled in the art. A leader sequence can be modified near its 3 'end to contain one or more restriction sites. This will facilitate the fusion of the leader sequence to the structural gene. Yeast transformation protocols are known to those skilled in the art. One such protocol is described by Hinnen et al. , Proc. Nati Acad. Sci USA 75: 1929, 1978. The protocol of Hinnen et al. select for Trp + transformants in a selective medium, where the selective medium consists of 0.67% yeast nitrogen base, 0.5% casamino acids, 2% glucose, 10 μg / ml adenine and 20 μg / ml uracil. Yeast host cells transformed by vectors containing the ADH2 promoter sequence can be grown to induce expression in a "rich" medium. An example of a rich medium is one consisting of 1% yeast extract, 2% peptone, and 1% glucose supplemented with 80 μg / ml adenine and 80 μg / ml uracil. The derepression of the ADH2 promoter occurs when glucose is depleted from the medium. The insect or mammalian host cell culture systems can also be used to express recombinant TACE polypeptides. Baculovirus systems for the production of heterologous proteins in insect cells are reviewed by Luckow and Summers, Bio / Technology 6:47 (1988). Established cell lines of mammalian origin can also be employed. Examples of mammalian host cell lines include the COS-7 line of monkey kidney cells (ATCC CRL 1651) (Gluzman et al., Cell 23: 1 75, 1981), L cells, C 127 cells, 3T3 cells ( ATCC CCL 163), Chinese hamster ovary (CHO) cells, HeLa cells, and BHK cell lines (ATCC CRL 10), and the cell line CV-1 / EBNA-1 derived from the kidney cell line of African green monkey CVI (ATCC CCL 70) as described by McMahan et al. (EMBO J. 10: 2821, 1991). The translational and transcriptional control sequences for expression vectors of mammalian host cells can be cut from viral genomes. Intensifying sequences and promoter sequences commonly used are derived from Polyoma virus, Adenovirus 2, Simian Virus 40 (SV40) and human cytomegalovirus. DNA sequences derived from the SV40 viral genome, for example SV40 origin, early and late promoter, enhancer, splice and polyadenylation sites can be used to provide other genetic elements for the expression of a structural gene sequence in a mammalian host cell . Viral early and late promoters are particularly useful because both are readily obtained from a viral genome as a fragment, which may also contain a viral origin of replication (Fiers et al., Nature 273: 13, 1978). Longer or shorter SV40 fragments may also be used, provided that the sequence of approximately 250 base pairs extending from the HindIII site to the Bgl I site located in the SV40 viral origin of the replication site is included. Exemplary expression vectors for use in mammalian host cells can be constructed as described by Okayama and Berg (Mol Cell. Biol., 1983). A useful system for stable high level expression of mammalian cDNAs in murine C 127 mammary epithelial cells can be constructed substantially as described by Cosman et al. (Mol Immunol., 23: 935, 1986). A useful high expression vector, PMLSV N 1 / N4, described by Cosman et al. , Nature 312: 768. 1984) has been deposited as ATCC 39890. Additional useful mammalian expression vectors are described in EPA-A-0367566, and in U.S. Patent Application Serial No. 07/701, 41 5, filed on May 16, 1991, incorporated herein by reference. The vectors can be derived from retroviruses. Instead of the natural signal sequence, a heterologous signal sequence may be added, such as the signal sequence for IL-7 described in US Pat. No. 4,965, 195; the signal sequence for the IL-2 receptor appears in Cosman et al. , Nature 312: 768 (1984); the IL-4 signal peptide described in EP 367,566; the signal peptide of the IL-1 type I receptor described in US Pat. No. 4,968,607; and the signal peptide of receptor I L-1 type I I described in EP 460,846. A TACE protein isolated and purified according to the invention can be produced by recombinant expression systems as described above or purified from naturally occurring cells. TACE can be substantially purified, as indicated by a single protein band upon analysis by SDS-polyacrylamide gel electrophoresis (SDS-PAGE). A process for producing TACE comprises culturing a transformed host cell with an expression vector comprising a DNA sequence encoding TACE under conditions sufficient to promote TACE expression. TACE is then recovered from a culture medium or cell extracts, depending on the expression system employed. As is known to the skilled artisan, the methods for purifying a recombinant protein will vary according to such factors as the type of host cells employed and whether the recombinant protein is secreted or not in the culture medium. For example, when the expression systems that secrete the recombinant protein are employed, the culture medium can first be concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit. Following the concentration step, the concentrate can be applied to a purification matrix such as a gel filtration medium. Alternatively, an anion exchange resin may be employed, for example, a matrix or substrate having pendant diethylaminoethyl groups (DEAE). The matrices can be acrylamide, agarose, dextran, cellulose or other types commonly used in protein purification. Alternatively, a cation exchange step can be employed. Suitable cation exchangers include several insoluble matrices comprising sulfopropyl or carboxymethyl groups. Sulfopropyl groups are preferred. Finally, one or more reverse phase high performance liquid chromatography steps (RP-H PLC) employing hydrophobic PLC RP-H media (eg, silica gel having methyl groups or other outstanding aliphatic groups), can be used to purify TACE additionally. Some or all of the above steps of purification, in various combinations, are well known and can be employed to provide an isolated and purified recombinant protein. In addition to producing TACE recombinantly, TACE can be isolated and purified from an activated monocytic cell line, TH P-1. THP-1 cells typically produce more TNF-a than HL-60 cells, and are a preferred source for TACE. Other sources for TACE can be used, and TAC E can also be found in other cell types to produce TN F-a. Once a source for TACE has been identified, TACE can be isolated and purified first by optionally stimulating the source cells to produce TN F-a. Stimulation may not be necessary, however, it can be done using techniques that are well known in the art. The cells are then collected, washed and the plasma membranes are isolated according to conventional procedures. A particularly preferred method of isolating plasma membranes is method number three as described in Maeda et. to the. , Biiochim. et. Biophys. Acta, 731: 1 15 (1983); except that dithiothreitol should not be included in this method, since it was determined that dithiothreitol blocks the activity of TACE. The cell membrane proteins can then be solubilized by suspending the membrane preparation in a diluted solution of non-ionic detergent, followed by brief homogenization. The phospholipids can then be extracted using conventional methods. It is possible to use an affinity column comprising a protein that binds TACE to affinity-purified TACE polypeptides. The TACE polypeptides can be removed from an affinity column using conventional techniques, for example, in a high salt levigation buffer, and then dialyzed into a smaller salt buffer to be used or to change the pH or other components depending on the matrix of affinity used. Example 4 describes a method for using the TACE of the invention to generate monoclonal antibodies directed against TACE. The recombinant protein produced in the bacterial culture is usually isolated by initial dissolution of the host cells, centrifugation, extraction of cell pellets in the case of an insoluble polypeptide, or of the supernatant fluid in the case of a soluble polypeptide, followed by one or more steps of concentration, salt precipitation, ion exchange, affinity purification or size exclusion chromatography. Finally, the RP-H PLC can be used for final purification steps. The microbial cells can be dissolved by any conventional method, including freeze-thaw cycles, sonication, mechanical dissolution or use of cell lysis agents. The transformed yeast host cells are preferably used to express TACE as a secreted polypeptide in order to simplify purification. The recombinant polypeptide secreted from a yeast host cell fermentation can be purified by methods analogous to those described by Urdal et al. (J. Chromatog. 296: 171, 1984). Urdal et al. describes two sequential, reversed-phase HPLC steps for the purification of recombinant human I L-2 on a preparative H PLC column. The sense or anti-sense oligonucleotides comprising a single filament nucleic acid sequence (either R NA or DNA) capable of binding to a target TACE mRNA sequence (forming a duplex) or to a TACE sequence in the helix of double-stranded DNA (forming a triple helix) can be made according to the invention. Sense or anti-sense oligonucleotides, according to the present invention, comprise a fragment of the coding region of TACE cDNA. Such a fragment generally comprises at least about 14 nucleotides, preferably from about 14 to about 30 nucleotides. The ability to create a sense or anti-sense oligonucleotide, based on a cDNA sequence for a given protein is described in, for example, Stein and Cohen, Cancer Res. 48: 2659, 1988 and van der Krol et al. , BioTechniques 6: 958, 1988. Ligation of sense or anti-sense oligonucleotides to target nucleic acid sequences results in the formation of complexes that block translation (RNA) or transcription (DNA) by one of several means, including the intensified degradation of the duplos, the premature termination of transcription or translation or by other means. Anti-sense oligonucleotides can be used in this manner to block the expression of TACE proteins. The sense or anti-sense oligonucleotides further comprise oligonucleotides having phosphodiester-modified sugar backbones (or other sugar linkages, such as those described in WO91 / 06629) and wherein such sugar linkages are resistant to endogenous nucleases. Such oligonucleotides with resistant sugar linkages are stable in vivo (i.e., capable of resisting enzymatic degradation), but retain the specificity of the sequence to be able to ligate target nucleotide sequences. Other examples of sense or anti-sense oligonucleotides include those oligonucleotides, which are covalently bonded to organic portions, such as those described in WO 90/10448, and other portions that increase the affinity of the oligonucleotide for a target nucleic acid sequence, such as poly- (L-lysine). Still further, intercalators, such as ellipticine, and alkylating agents or metal complexes can be linked to sense or anti-sense oligonucleotides to modify binding specificities of the sense or antisense oligonucleotide for the target nucleotide sequence. The sense or anti-sense oligonucleotides can be introduced into a cell containing the target nucleic acid sequence by a gene transfer method, including, for example, CaPO4-mediated DNA transfection, electroporation, or using transfer vectors of genes such as Epstein-Barr virus. The sense or anti-sense oligonucleotides are preferably introduced into a cell containing the target nucleic acid sequence by insertion of the sense or anti-sense oligonucleotide into a suitable retroviral vector, then the cell is contacted with the vector retrovirus that contains the inserted sequence, either in vivo or ex vivo. Suitable retroviral vectors include, but are not limited to, the murine retrovirus M-MuLV, N2 (a retrovirus derived from M-MuLV), or the double copy vectors designated DCT5A, DCT5B and DCT5C (see Application of PCT US 90). / 02656). Sense or anti-sense oligonucleotides can also be introduced into a cell containing the target nucleotide sequence by forming a conjugate with a molecule that binds a ligand, as described in WO 91/04753. Suitable ligand-binding molecules include, but are not limited to, cell surface receptors, growth factors, other cytokines, or other ligands that bind cell surface receptors. Preferably, the conjugation of the ligand-binding molecule does not substantially interfere with the ability of the molecule that binds a ligand to bind to its receptor or corresponding molecule, or block the entry of the sense or anti-sense oligonucleotide, or its version. conjugated in the cell. Alternatively, a sense oligonucleotide or an antisense can be introduced into a cell containing the target nucleic acid sequence by the formation of a lipid-oligonucleotide complex, as described in WO 90/10448. The sense or antisense lipid oligonucleotide complex is preferably dissociated within the cell by an endogenous lipase. An isolated and purified TACE or a fragment thereof, and in particular, the extracellular domain of TACE, may also be useful as a therapeutic agent in regulating the levels of certain cell surface proteins. In addition to TNF-a, other cytokines as well as cytokine receptors and various adhesion proteins can be released from the cell surface by TACE or related proteases. TACE or a fragment thereof, in particular, the extracellular domain of TACE, can be administered to modulate or remove cell surface cytokines, cytokine receptors and adhesion proteins involved in the growth of tumor cells, inflammation or fertilization . When a therapeutic agent is used, TACE can be formulated into pharmaceutical compositions according to known methods. TACE can be combined in mixtures, either as the sole active material or with other known active materials, with pharmaceutically suitable diluents (e.g., Tris-HCl, acetate, phosphate), preservatives (e.g., Thimerosal, benzylalcohol, parabens) , emulsifiers, solubilizers, auxiliaries and / or carriers. Suitable carriers and their formulations are described in Remington's Pharmaceutical Sciences, 16th ed. 1980, Mack Publishing Co. In addition, such compositions may contain TACE in complex with polyethylene glycol (PEG), metal ions, or incorporated into polymeric compounds such as polyacetic acid, polyglycolic acid, hydrogels, etc. , or incorporated into liposomes, microemulsions, micelles, unilamellar or multilamellar vesicles, spheroplasts or traces of erythrocytes. Such compositions will influence the physical state, solubility, stability, release rate in vivo, and in vivo evacuation rate of TACE. TACE can be titrated using any of a variety of metalloprotease assays known in the art. In general, TACE can be assessed through the use of a peptide substrate representing the natural cut site of TNF-α. For example, in order to detect the cutting of a substrate by TACE, the substrate can be marked with a fluorescent group on one side of the cutting site and with a group that turns off the fluorescence on the opposite side of the cutting site. Upon cutting by TACE, the damper is eliminated, thereby providing a detectable signal. Alternatively, the substrate can be marked with a salient colorimetric group that absorbs more strongly on the cut. Alternatively, the substrate may have a thioester group synthesized at the substrate cutting site, so that upon cutting by TACE, the thiol group remains and can be easily detected using conventional methods. A particularly preferred method for detecting TACE activity in a sample was described in Example 1, infra. Other methods to detect the activity of TACE can be used without resorting to undue experimentation. As further described in Example 1, infra, a quantitative assay for TACE can also be used, the assay involves incubating the peptide substrate, at about 1 mM, with TACE at 37 ° C for a fixed period of time; Stop the reaction by adding an acid or a metal chelator, and determine the degree of cut by an H PLC analysis. Within one aspect of the invention, TACE, and peptides based on the amino acid sequence of TACE, can be used to prepare antibodies that specifically bind to TACE. A specific example of such an antibody preparation is described in Example 4 herein. The term "antibodies" is intended to include polyclonal antibodies, monoclonal antibodies, fragments thereof such as F (ab ') 2, and Fab fragments, as well as any recombinantly produced binding partner. Antibodies are defined to be specific binders if they bind TACE with a Ka greater than or equal to about 107 M "1. The affinities of binding partners or antibodies can be readily determined using conventional techniques, for example those described by Scatchard et al., Ann, NY Acad. Sci., 51: 660 (1949).
The antibodies can be rapidly generated from a variety of sources, for example, horses, cows, goats, sheep, dogs, chickens, rabbits, mice or rats, using methods that are well known in the art. In general, purified TACE, or a peptide based on the amino acid sequence of TACE that is properly conjugated, is administered to a host animal normally through a parenteral injection. The immunogenicity of TACE can be enhanced through the use of an adjuvant, for example a complete or incomplete Freund's aid. Following the booster immunizations, small serum samples are collected and tested for TACE or TACE peptides. Examples of various assays useful for such a determination include those described in: Antibodies: A Laboratory Manual, Harlow and Lane (eds), Cold Spring Harbor Laboratory Press, 1988; as well as also procedures such as counter-current immuno-electrophoresis (CIEP), radioimmunoassay, radioimmunoprecipitation, linked enzyme-linked immunosorbent assays (ELISA), "dot biot" assays, and "sandwich" assays, see US Pat. Nos. 4,376, 1 10 and 4,486, 530. Monoclonal antibodies can be easily prepared using well known methods, see for example, the procedures described in US Pat. Nos. RE 32,01 1, 4,902,614, 4,543,439 and 4,41 1, 993; Monoclonal Antibodies, Hybridomas: A New Dimension in Biological! Analyzes, Plenum Press, Kennett, McKearn, and Cechtol (eds.), 1980. Briefly, host animals, such as mice, are injected intraperitoneally at least once and preferably, at least twice in approximately 3-fold intervals. weeks, with isolated and purified TACE or conjugated TACE peptide, optionally in the presence of an auxiliary. Mouse sera are then titrated by dot biot or antibody capture (ABC) technique to determine which animal is best to fuse. Approximately two to three weeks later, the mice are given an intravenous TACE booster or a conjugated TACE peptide. The mice are subsequently sacrificed and the spleen cells are fused with commercially available myeloma cells, such as Ag8.653 (ATCC), following established protocols. Briefly, the myeloma cells are washed several times in the medium and fused to mouse spleen cells at a ratio of about three spleen cells to one myeloma cell. The fusion agent can be any suitable agent used in the art, for example, polyethylene glycol (PEG). The fusion is placed in plates containing the medium that allows the selective growth of the fused cells. The fused cells can then be allowed to grow for approximately eight days. The supernatants of the resulting hybridomas are collected and added to a plate that was first coated with goat anti-mouse Ig. Following the washings, an indication, such as, 125I-TACE is added to each well followed by the incubation. The positive cavities can be subsequently detected by autoradiography. The positive clones can be grown in a bulk culture and the supernatants are subsequently purified on a Protein A column (Pharmacia).
The monoclonal antibodies of the invention can be produced using alternative techniques, such as those described by Altin-Mees et al. , "Monoclonal Antibody Expression Libraries: A Rapid Alternative to Hybridomas", Strategies in Molecular Biology 3: 1-9 (1990), which is incorporated herein by reference. Similarly, ligation partners can be constructed using recombinant DNA techniques to incorporate the variable regions of a gene encoding a specific ligation antibody. Such a technique is described in Larrick et al. , Biotechnology, 7: 394 (1989). Other types of "antibodies" can be produced using the information provided herein in conjunction with the state of knowledge in the art. For example, humanized antibodies that are capable of specifically binding TACE are also encompassed by the invention. Once isolated and purified, antibodies against TACE can be used to detect the presence of TACE in a sample using established assay protocols. In addition, the antibodies of the invention can be used therapeutically to bind to TACE and inhibit its activity in vivo. TACE purified according to the invention will facilitate the discovery of TACE inhibitors, and thus, inhibitors of excessive TNF-α release. The use of a purified TACE polypeptide in the classification of potential inhibitors thereof is important and can virtually eliminate the possibility of interfering reactions with contaminants. Such a screening assay for detecting the TACE inhibitory activity of a molecule, would normally involve mixing the potential inhibitor molecule with an appropriate substrate, incubating TACE which is at least substantially purified with the mixture, and determining the degree of cut of the substrate as , for example, was described earlier. While several suitable substrates can be designed for use in the assay, preferably, a peptide substrate is used, and that substrate comprises the amino acid sequence Leu-Ala-GIn-Ala-Val-Arg-Ser-Ser (SEQ ID NO: 5). In addition, TACE polypeptides can also be used for the design based on the structure of TACE inhibitors. Such a structure-based design is also known as "rational drug design." The TACE polypeptides can be analyzed three-dimensionally by, for example, X-ray crystallography, nuclear magnetic resonance or homology modeling, all of which are well-known methods. The use of TACE structural information in molecular modeling parcel systems to assist in the design of the inhibitor and the TACE-inhibitor interaction is also encompassed by the invention. Such computer aided modeling and medically designed can use information such as chemical conformational analysis, electrostatic potential of molecules, protein folding, etc. For example, most of the class-specific inhibitor design of meta-proteases has focused on attempts to chelate or ligate the catalytic zinc atom. Synthetic inhibitors are usually designed to contain a negatively charged portion to which a series of other groups designed to fit the specificity pockets of the particular protease bind. A particular method of the invention comprises analyzing the three-dimensional structure of TACE for the possible sites of ligation of the substrates, synthesizing a new molecule incorporating a predictable reactive site, and testing the new molecule as described above. The following Examples provide an illustration of the embodiments of the invention and should not be considered to limit the scope of the invention, which is set forth in the appended claims. In the following Examples, all methods described are conventional, unless otherwise specified.
EXAMPLE 1 Purification of the TNF-a converting enzyme This example describes a method for purifying TACE. TACE was isolated and purified from the membranes of the human monocytic cell line, TH P-1, (ATCC No. TIB 202), which had been stimulated to produce TNF-a. TH P-1 cells were chosen because they produce more TN F-a than H L-60 cells, a most commonly used human monocytic cell line. Approximately 120 billion cells were stimulated using the procedure previously described by Kronheim et al. , Arch. Biochem. Biophys. 269: 698 (1992), incorporated herein by reference. Two hours after the stimulation, the cells were harvested by centrifugation. The harvested cells were washed at least twice with Hanks balanced salt solution, and the plasma membranes were isolated according to method number three as described by Maeda et al. , Biochim. et. Biophys. Acta 731: 15 (1983), except that dithiothreitol was not used, using 1.25 ml of homogenization buffer per ml of cell pellet. It was determined that the standard procedure of Maeda et al. , Id., Using dithiothreitol, failed to produce compounds having TACE activity (one assay for TACE activity is described below). The proteins were then solubilized by re-suspending the membrane preparation in a solution of 1% octylglucoside, 10 mM Tris-HCl (pH 8), 1 mM MgCl 2 and 30 mM NaCl and briefly homogenizing with a Homogenizer. Brinkman (twice, five seconds each time). The phospholipids were then extracted by adding four volumes of ice-cold acetone (0 ° C); After an incubation of thirty minutes at 4 ° C, the material extracted by acetone was centrifuged at 1500 rpm for 10 minutes in an H 1000B rotor.
Chromatography The pellet material was dissolved in 450 ml of Buffer A (Amorticuator A comprises 10 mM Tris-HCl (pH 7.5) and 1% octylglucoside (weight percent by volume)) and applied to 120 ml of DEAE column -Sepharose fast-flow (Pharmacia) at 4 ml per minute. The column was then washed with 360 ml of Buffer A at 6 ml per minute, and the protein was then levigated with an increasing gradient of NaCl (0-0.3 M) in Buffer A applied at 6 ml per minute for a period of 40 minutes. minutes TACE was levigated with a NaCl concentration of from about 50 to about 150 mM.
TACE was originally detected at this point by its ability to cut recombinant 26kD TNF-a fused to the "flag" sequence (T.P. Hopp, et al., Bio Technology, 6j_1204 (1988)) of eight amino acids at the amino terminus. The gene encoding human TNF-α was spliced to a DNA encoding the flag sequence, and this construct was placed in the vector pPL3 (C. Maliszewski et al., Molec.Immunol., 25: 429 (1987) The protein was then expressed in a protease-deficient species of E. coli (RT Líbby et al., DNA, 6: 221 (1987)), which was found necessary to prevent the degradation of the precursor by the bacterium. the removal of the growth medium, the bacteria were resuspended in 39 mM Tris-HCl (pH 8), 5 mM EDTA and the suspension was sonicated for approximately 30 seconds.The material was then centrifuged at 20,000 rpm in a rotor SS34 for 30 minutes, the supernatant fraction was discarded, and the pellet was resuspended with 8 M urea in 10 mM Tris-HCl (pH 8.) The material was homogenized with 25 beats in a "dounce" homogenizer and then was centrifuged at 20,000 rpm in a SS34 rotor for 30 minutes.The supernatant fraction, which contains the TNF-a precursor, was then dialyzed four times against 10 mM Tris-HCl (pH 8). This material was incubated at 37 ° C for at least 4 hours with the TACE levigated from DEAE-Sepharose, which had been treated with 1 mM of N-motoxysuccinyl-Ala-Ala-Val-chloro-methyl ketone, 10 μg / ml of leupeptipa, and 1 mg / ml of a1-protease inhibitor, all being commercially available. The N-terminus of the resulting 1 7 kD product was found to be that of authentic TNF-α. After the initial identification of the TACE in this manner, it was found that the enzyme also cuts a residue 8 peptide representing the segment Leu73-Ala7-Gln75-Ala76-i-Val77-Arg78-Ser79-Serd0 (SEQ ID NO: 5) of TNF-a. where the (J,) illustrates the cutting site. Based on this observation, a quantitative assay was established: the peptide, at 1 mM, was incubated with the enzyme at 37CC for a fixed period of time, in the presence of 0. 1 mM dichloroisocoumarin, 1 mM methoxysuccinyl-Ala Ala-Pro-Val-chloromethyl ketone, 10 μg / ml leupeptin, 10 μM bestatin, and 1 mg / ml a1-protease inhibitor (Sigma), all commercially available. The reaction was then stopped by the addition of acid or a metal chelator. The cutting gradation of this peptide, reflecting the amount of TACE present, was determined by applying the mixture to a Vydac C 18 column, and levigating with a 0 to 30% acetonitrile over a period of 15 minutes. The material that was levigó of the column DEAE with 0.05-0.25 M of NaCl had approximately one specific activity greater than 4-parts than the starting material. The levigated material was sonic and then stirred with wheat germ agglutinin agarose (Vector Laboratories) for two hours at 4 ° C. Before use, the wheat germ agglutininose was washed with 5 column volumes of Buffer B (Buffer B comprises 10 mM Tris-HCl (pH 7.5), 0.1 5 M NaCl, 0. 1 mM MnCl2 0. 1 mM CaCl2, 1% octylglucoside and 10% glycerol); 1 ml of this resin was used for every 2 mg of protein in the sample, as determined by the BCA protein assay (Pierce). After two hours, the resin was washed with 7 volumes of Buffer B, and the material was then levigated with 5 column volumes of Buffer B plus 0.3 M of acitlglucosamine (Sigma), with intervals of 30 minutes between the application of each volume of column. Levigated fractions containing TACE activity had approximately a specific activity greater than ten parts than the starting material. These fractions were concentrated to approximately 5 ml with Centriprep-30 concentrators (Amicon) and then three-parts diluted with C cushion (C cushion comprises 10 mM Tris-HCl (pH 8), 1% octylglucoside and 10% glycerol ). The diluted material was sonic (three scans of 10 seconds) and then loaded onto a MonoQ Hr 5/5 column (Pharmacia) at 0.5 ml per minute. The column was then washed with 10 ml of Buffer C at 0.5 ml per minute, and the material was levigated with a gradient of 0 to 0.25 M NaCl in Buffer C at 0.5 ml per minute for a period of 30 minutes. TACE activity (detected at this stage and subsequently by incubation with the peptide substrate previously described in the absence of protease inhibitors) levigated with approximately 0.15 M NaCl. The concentration of NaCl in the MonoQ fractions containing activity was reduced by at least ten-parts by diluting the material in Shock absorber C, and then the material was applied to a hydroxyapatite column (American International Chemical, HS40 ceramic hydroxyapatite) to the speed of 0.5 ml per minute. After washing with three column volumes of Buffer C, the protein was levigated with a gradient of 0 to 50 mM sodium phosphate at 1 ml per minute for a period of 30 minutes. TACE levigated with approximately 15 mM sodium phosphate. The TACE levigated from the hydroxyapatite column was then concentrated to approximately 100 μl with Centricon-50 concentrators (Amicon), and applied to a column of Bio-Rad SEC-400 size (30 cm). The protein was levigated with cushion C run through the column at 0.5 ml per minute; the TACE levigó in approximately 28 minutes. TACE levigated from the size column was diluted three-fold in Buffer D (Buffer D comprises 20 mM MES (pH 6), 1% octylglucoside and 10% glycerol) and applied to 1 ml column of Red 120-agarose (Sigma) at 0.25 ml per minute. After the column was washed with 10 ml of Buffer D, the protein was levigated with a gradient of 0 to 1 M NaCl in Buffer D at 0.25 ml per minute for a period of 60 minutes. The TACE levigó with 0.2 to 0.3 M NaCl. Five percent of each levigated fraction was run on an SDS-polyacrylamide gel (10%), and a silver stain showed that the predominant protein in the fractions with activity ran approximately halfway between the 66 and 97 kD markers (Novex) in the gel, at approximately 80 kD. Trifluoroacetic acid (TFA) was added to 0.2% (volume by volume percentage) to a combination of the fractions containing the approximately 80 kD protein, and the mixture was then pumped to a C4 column from 2.1 x 5 cm to approximately 100 μl per Minute using a Shimadzu LC-10AD. The protein was levigated with a gradient of 0 to 100% acetonitrile in 0.1% TFA at 100 μl per minute for a period of 100 minutes. One-minute fractions were collected and 5 to 10% of each fraction was run on an SDS-polyacrylamide gel from Novex (10%). The fractions that were levigated with approximately 70% acetonitrile and containing a protein of approximately 80 kD were combined and evaporated to dryness.Generation of peptides and sequencing This combination of fractions was then dissolved in 200 μl of 50 mM Tris-HCl (pH 8), 1 mM EDTA, and an amount of endo-LYS-C (Promega) equal to about 1 was added. / 50 of the amount of protein in the sample. The material was incubated at 37 ° C overnight, and then a fresh aliquot of the same amount of endo-LYS-C was added for an additional 3 hours at 37 ° C. The resulting peptides were separated by applying the materail to a C18 capillary column at 20 μl per minute and levigating with an ascending gradient of acetonitrile (0.5% per minute) in 0.1% TFA over a period of 200 minutes. The peptides were sequenced with an ABI 476 automated sequencer or an ABI 494.
EXAMPLE 2 Preparation of Isolated and Purified TACE This Example describes a method for further purifying the TACE purified as obtained using the procedures described above. The purified TACE obtained from the THP-1 cells may contain small amounts of human lysosomal 85 kD sialoglycoprotein (Biochem Biophys, Res. Commun. 184: 604-61 1 (1992)) and human lysosomal alpha-mannosidase (Biochem. Biophys. Res. Commun. 200: 239-245 (1994)), which can be removed using standard immunosorbent procedures, as described in, for example, Robert K. Scopes, Protein Purification-Principles and Practice (Springer-Verlag, 2nd ed.) , p. 167-1. 72. Using the procedures described in this Example 2, isolated and purified TACE can be obtained.
EXAMPLE 3 Cloning of human TACE This example describes a method for isolating a DNA sequence encoding human TACE. A random primed cDNA library was generated from the commercially available cell line THP-1 (Amersham) using conventional methods. Polymerase chain reaction (PCR) amplifications (Mullis and Faloona, Meth. Enzymol 155: 35-350, 1987) were performed using the following detonators: Detonator (1): 5'-AARTAYGTNATHTAYCC-3 'SEQ I D NO: 6 Detonator (2): 5'-CCRCARTCRCAYTCYTC-3' SEQ I D NO: 7 The detonator (1) is based on the first five amino acids of the Peptide (2) with the addition of a triple that figures lysine at the 5% end The detonator (2) is the antisense to a conserved amino acid sequence Glu-Glu-Cys-Asp-Cys-Gly (EECDCG) SEQ ID NO: 8, which is found in a homologous metalloprotease, bovine reprolysin 1 (GenBank Access # Z21961). The single stranded cDNA was amplified using the mixed oligonucleotides described above under standard PCR conditions. The products of the PCR reaction were fractionated by gel electrophoresis and DNA bands of approximately 180 base pairs were isolated and subcloned into commercially available pBLUESCRIPT. Sequencing revealed a clone containing a nucleotide sequence that codes for the amino acids Ala-Val-Ser-Gly-Asp-His-Glu-Asn-Asn-Lys (SEQ ID NO: 9) and a nucleotide sequence that codes for the amino acids Glu-Glu-Cys-Asp-Cys-Gly (EECDCG) (SEQ ID NO: 8). This clone was called the "clone 30CD". The clone 30CD was sequenced and the detonators were generated based on this sequence. The detonators were then used to detect TACE cDNA in a phage library made from human KB cells. This library was classified under conventional conditions using a test based on the 30CD sequence. The positive hybridization plates were isolated and the DNA fragments of these clones were sequenced. Sequencing provided a full-length human TACE cDNA, which is shown in SEQ ID NO: 1. TACE human was found to be a type I transmembrane protein of 824 amino acids, including a signal peptide of 17 amino acids N- terminal. The signal peptide is followed by an extracellular domain of 654 amino acids, a transmembrane domain of 23 amino acids and a cytoplasmic domain of 130 amino acids. An alternate spliced variant was cloned and sequenced and found to contain the same amino acid sequence as TACE, except that a 50 base pair fragment is deleted at the 5 'end of the cytoplasmic domain, thereby changing the reading frame to encode a cytoplasmic domain of six amino acids. The amino acid sequence of this variant is shown in SEQ ID NO: 4c with the cDNA shown in SEQ ID NO: 3.
EXAMPLE 4 Preparation of antibodies against TACE This Example describes a method for generating monoclonal antibodies against TACE. Balb / c mice were injected intraperitoneally twice at 3 week intervals with 10ug of isolated and purified TACE of Example 1 or peptides based on the amino acid sequence of TACE in the presence of RIBI helper (RIBI Corp., Hamilton, Montana ). The mouse sera are then tested by conventional dot biot or antibody capture (ABC) technique to determine which animal is best to fuse. Three weeks later, the mice are given an intravenous booster of 3 ug of human TACE, or TACE peptide, suspended in sterile PBS. Three days later, the mice are sacrificed and the spleen cells are fused with Ag8.653 myeloma cells (ATCC) following established protocols. Briefly, Ag8.653 cells are washed several times in serum-free medium and fused to mouse spleen cells at a ratio of three spleen cells to one myeloma cell. The fusion agent is 50% PEG: 10% DMSO (Sigma). The fusion is placed in 96-well flat bottom plates (Corning) containing a DMEM medium supplemented with HAT and allowed to grow for eight days. Supernatants from the resulting hybridomas were collected and added to a 96-well plate for 60 minutes, which was first coated with goat anti-mouse Ig. Following the washings, 125 I-TACE was added to each cavity, incubated for 60 minutes at room temperature, and washed four times. Positive cavities can subsequently be detected by autoradiography at -70 ° C using Kodak X-Omat S film. Positive clones can be grown in bulk culture, and the supernatants are subsequently purified on a Protein A column (Pharmacia).
SEQUENCE LISTS (1) GENERAL INFORMATION: (i) APPLICANT: IMMUNEX CORPORATION (ii) TITLE OF THE INVENTION: CONVERTIBLE ENZYME TNF-a (iii) SEQUENCE NUMBER: 9 (iv) ADDRESS FOR CORRESPONDENCE: (A) DESTINY: IMMUNEX CORPORATION (B) STREET: 51 UNIVERSITY STREET (C) CITY: SEATTLE (D) STATE: WA (D) COUNTRY: EU (E) POSTAL CODE: 98101 (v) LEGIBLE COMPUTER FORM: (A) TYPE OF MEANS: FLEXIBLE DISK (B) COMPUTER: Apple Macintosh (C) OPERATING SYSTEM: Apple Operating System 7.5.2 (D) PACKAGE: Microsoft Word for Apple, version 6.0 (vi) CURRENT REQUEST DATA: (A) APPLICATION NUMBER: not yet assigned (B) SUBMISSION DATE: 3 JANUARY 1996 (C) CLASSI FICTION: (VN) PREVIOUS APPLICATION DATA: (A) APPLICATION NUMBER: to be assigned (B) DATE OF SUBMISSION; MAY 23, 1996 (vii) PREVIOUS APPLICATION DATA D: (A) APPLICATION NUMBER: 08 / 504,614 (B) SUBMISSION DATE: JULY 20, 1995 (vii) PREVIOUS APPLICATION DATA: (A) APPLICATION NUMBER : 08 / 428,458 (B) DATE OF SUBMISSION: JULY 8, 1995 (viii) ATTORNEY / AGENT INFORMATION: (A) NAME: Malaska, Stephen L. (B) N REGISTRATION NUMBER: 32, 655 (C) N REFERENCE NUMBER / DOCKET: 2507-WO (ix) TELECOMU INFORMATION NICATIONS : (A) TELEPHONE: (206) 5870430 (B) TELEFAX: (206) 2330644 (2) I NFORMATION FOR SEQ ID NO: 1: (i) CHARACTERISTICS OF THE SECU ENCIA: (A) LONGITU D: 2475 base pairs (B) TYPE: nucleic acid (C) FI LAMENT: simple (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: cDNA to mRNA (iii) HI POTETHICAL: NO (iv) ANTI-SENSE: NO (ix) CHARACTERISTICS: (A) NAME / KEY: CDS (B) U BICACON: 52..2472 (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 1 ATGAGGCAGT CTCTCCTATT CCTGACCAGC GTGGTTCCTT TCGTGCTGGC G CCG CGA 57 Pro Arg 1 CCT CCG GAT GAC CCG GGC TTC GGC CCC CAC CAG AGA CTC GAG AAG CTT 105 Pro Pro Asp Asp Pro Gly Phe Gly Pro His Gln Arg Leu Glu Lys Leu 5"10 15 GAT TCT TTG CTC TCA GAC TAC GAT ATT CTC TCT TTA TCT AAT ATC CAG 153 Asp Ser Leu Leu Ser Asp Tyr Asp He Leu Ser Leu Ser Asn He Gln 20 25 30 CAG CAT TCG GTA AGA AAA AGA GAT CTA CAG ACT TCA ACA CAT GTA GAA 201 Gln His Ser Val Arg Lys Arg Asp Leu Gln Thr Ser Thr His Val Glu "35 '40 45 50 ACA CTA CTA ACT TTT TCA GCT TTG AAA AGG CAT TTT AAA TTA TAC CTG 249 Thr Leu Leu Thr Phe Ser Ala Leu Lys Arg His Phe Lvs Leu Tyr Leu 55 60"65 ACÁ TCA AGT ACT GAA CGT TTT TCA CAAT AAT TTC AAG GTC GTG GTG GTG 297 Thr Ser Ser Thr Glu Arg Phe Ser Gln Asn Phe Lys Val Val Val Val 70 75 80 GAT GGT AAA AAC GAA AGC GAG TAC ACT GTA AAA TGG CAG GAC TTC TTC 345 Asp Gly Lys Asn Glu Ser Glu Tyr Thr Val Lys Trp Gln Asp Phe Phe 85 90 '95 ACT GGA CAC GTG GTT GGT GAG CCT GAC TCT AGG GTT CTA GCC CAC ATA 393 Thr Gly Kis Val Val Gly Glu Pro Asp Ser Arg Val Leu Ala His He 100 105 110 AGA GAT GAT GAT GTT ATA ATC AGA ATC AAC ACA GAT GGG GCC GAA TAT 441 Arg Asp Asp Asp Val He He Arg He Asn Thr As. Gly Ala Glu Tvr 115 '120 125"130 AAC ATA GAG CCA CTT TGG AGA TTT GTT AAT GAT ACC AAA GAC AAA AGA 489 Asn He Glu Pro Leu Trp Are Phe Val Asn Asp Thr Lvs ASD LVS Arg 135 140"?" 45 ATG TTA GTT TAT AAA TCT GAA GAT ATC AAG AAT GTT TCA CGT TTG CAC- 537 e_ Leu Val Tyr Lys Ser Glu Ace? He Lys Asn Val Ser Arg Leu Gln 150 '155 160 TCT CCA AAA GTG TGT GGT TA7 TTA AAA G7G GAT AAT GAA GAG TTG CTC 555 _er Prc lys Val Cys Gly Tyr Le- Lys Val Aep Asn Glu Glu Leu Leu 1-5" 17C 175 CCA AAA GGG TTA GTA AGA GAA CCA CCT GAA GAG CTT GTT CAT CGA 633 Pro Lys Gly Leu Val Asp Arg Glu Pro Pro Glu Glu Leu Val His Arg 180"185 190 GTG AAA AGA AGA GCT GAC CCA GAT CCC ATG AAG AAC ACG TGT AAA TTA 681 Val Lys Arg Arg Wing Asp Pro Asp Pro Met Lys Asn Thr Cys Lys Leu 195 200 205 210 TTG GTG GTA GCA GAT CAT CGC TTC TAC AGA TAC ATG GGC AGA GGG GAA 729 Leu Val Val Wing Asp His Arg Phe Tyr Arg Tyr Met Gly Arg Gly Glu 215 220 225 GAG AGT HERE ACT AAT TAC TTA ATA GAG CTA ATT GAC AGA GTT GAT 777 Glu Be Thr Thr Thr Asn Tyr Leu He Glu Leu He Asp Arg Val Asp 230 '235"240 GAC ATC TAT CGG AAC ACT TCA TGG GAT AAT GCA GGT TTT AAA GGC TAT 825 Asp He Tyr Arg Asn Thr Ser Tro Asp Asn Wing Gly Phe Lys Gly Tyr 245 250 255 GGA ATA CAG ATA GAG CAG ATT CGC ATT CTC AAG TCT CCA CA GAG GTA 873 Gly He Gln He Glu Gln He Arg He Leu Lys Ser Pro Gln Glu Val 260 265 270 AAA CCT GGT GAA AAG CAC TAC AAC ATG GCA AAA AGT TAC CCA AAT GAA 921 Lys Pro Gly Glu Lys His Tyr Asn Met Wing Lys Ser Tyr Pro Asn Glu 275"" 280"285 290 GAA AAG GAT GCT TGG GAT GTG AAG ATG TTG CTA GAG CA TTT AGC TTT 969 Glu Lys Asp Wing Trp Asp Val Lys Met Leu Leu Glu Gln Phe Ser Phe 295 '" 300 305 GAT ATA GCT GAG GAA GCA TCT AAA GTT TGC TTG GAC CAC CTT TTC ACÁ 1017 A = p He Wing Glu Glu Wing Ser Lys Val Cys Leu Wing His Leu Phe Thr 310 315 320 TAC CAAT GAT TTT GAT ATG GGA ACT CTT GGA TTA GCT TAT GTT GGC TCT 1065 Tyr Gln Asp Phe Asp Met Gly Thr Leu Gly Leu Wing Tyr Val Gly Ser 325 '' 330"335 CCC AGA GCA AAC AGC CAT GGA GGT GTT TGT CCA AAG GCT TAT TAT AGC 1113 Pro Arg Ala Asr. Ser His Gly Gly Val Cys Pro Lys Wing Tyr Tyr Ser 340 345 350 CCA GTT GGG AAG AAA AAT ATC TAT TTG AAT AGT GGT TTG ACG AGC HAL 11 he Pro Val Gly Lys Lys Asn He Tyr Leu Asn Ser Gly Leu Thr Ser Thr 355"'360" 365' 370 AAG AAT TAT GGT AAA ACC ATC CTT ACA AAG GAA GCT GAC CTG GTT ACA 1209 Lys Asr. Tyr Gly Lys Thr He Leu Tnr Lys Glu Wing Asp Leu Val Tnr 3 5 380 '385 ACT CAT GAA TTG GGA CAT AAT TTT GGA GCA GAA CAT GAT CCG GAT GGT 1257 r.r h_s Glu Leu Gly His Asr. Phe Giy Wing Glu His Asp Pro Asp Giv 390 395 '400 CTA C-CA GAA TGT GCC CCG AAT GAG GAC CAG GGA GGG AAA TAT GTC ATG 13 Or leu Wing Giu Cys Wing Pr: Asn Glu Asp Gln Gly Gly Lys Tyr Val Me . 405 41C "" 15"TAT CCC ATA GCT GTG AGT GGC GAT CAC GAG AAC AAT AAG ATG TTT TCA 1353 Tyr Pro He Wing Val Ser Gly Asp His Glu Asn Asn Lys Met Phe Ser 420 425 430 AAC TGC AGT AAA CAÁ TCA ATC TAT AAG ACC ATT GAA AGT AAG GCC CAG 1401 Asn Cys Ser Lys Gln Ser He Tyr Lys Thr He Glu Ser Lys Wing Gln 435 440 445 450 GAG TGT TTT CAA GAA CGC AGC AAT AAA GTT TGT GGG AAC TCG AGG GTG 1 49 Glu Cys Phe Gln Glu Arg Ser Asn Lys Val Cys Gly Asn Ser Arg Val 455 460 465 GAT GAA GGA GAA GAG TGT GAT CCT GGC ATC ATG TAT CTG AAC AAC GAC 1497 Asp Glu Glu Glu Glu Cys Asp Pro Gly He Met Tyr Leu Asn Asn Asp 470"" 475 480 ACC TGC TGC AAC AGC GAC TGC ACG TTG AAG GAA GGT GTC CAG TGC AGT 1545 Thr Cys Cys Asn Ser Asp Cys Thr Leu Lys Glu Gly Val Gln Cys Ser '485 490 495 GAC AGG AAC AGT CCT TGC TGT AAA AAC TGT CAG TTT GAG ACT GCC CAG 1593 Asp Arg Asn Ser Pro Cys Cys Lys Asn Cys Gln Phe Glu Thr Ala Gln 500 '505 510 AAG AAG TGC CAG GAG GCG ATT AAT GCT ACT TGC AAA GGC GTG TCC TAC 1641 Lys Lys Cys Gln Glu Ala He Asn Ala Thr Cys Lys Gly Val Ser Tyr 515 520 525 530 TGC ACA GGT AAT AGC AGT GAG TGC CCG CCT CCA GGA AAT GCT GAA GAT 1689 Cys Thr Gly Asn Ser Ser Glu Cys Pro Pro Pro Gly Asn Wing Glu Asp 535 540 545 GAC ACT GTT TGC TTG GAT CTT GGC AAG TGT AAG GATGGG AAA TGC ATC 1737 Asp Thr Val Cys Leu Asp Leu Gly Lys Cys Lys Asp Gly Lys Cys He 550 '555 560 CCT TTC TGC GAG AGG GAA CAG CAG CTG GAG TCC TGT GCA TGT AAT GAA 17e5 Pro Phe Cys Glu Arg Glu Gln Gln Leu Glu Ser Cys Ala Cys Asn Glu 565 570 575 ACT GAC AAC TCC TGC AAG GTG TGC TGG AGG GAC CTT TCC GGC CGC TGT ie 3 Thr Asp Asr. Be Cys Lys Val Cys Cys Arg Asp Leu Ser Glv Ars Cys 5S0 585 '590 GTG CCC TAT GTC GAT GAT GAA CAA AAG AAC TTA TTG AGG AAA GGA 18 SI Val Pro Tyr Val Asp Ala Glu Gln Lys Asn Leu Phe Leu Arg Lys Glv 595 600 605 '610 AAG CCC TGT ACA GTA GGA TTT TGT GAC ATG AAT GGC AAA TGT GAG AAA 1929 Lys Fre Cys Thr Val Gly Phe Cys Asp Met Asv Glv Lvs Cvs Glu Lys 615"620 - - - ^^^ CGA GTA CAG GAT GTA ATT GAA CGA TTT TGG GAT TTC ATT GAC CAG CTG 1977 Arg Val G Asp Val He Glu Arg Phe Trp Asp Phe He ASD Gln Leu 630 635"" 640 AGC ATC AAT ACT TTT GGA AAG TTT TTA GCA GAC AAC ATC GTT GGG TCT 2025 Ser H Asr. Thr Fhe Gly Lys Phe Leu Wing Asp Asr. He Val Gly Ser 645 650 '655 GTC CTG GTT TTC TCC TTG ATA TTT TGG ATT CCT TTC AGC ATT CTT GTC 2073 Val Leu Val Phe Ser Leu He Phe Trp He Pro Phe Ser He Leu Val 660 665 670 CAT TGT GTG GAT AAG AAA TTG GAT AAA CAG TAT GAA TCT CTG TCT CTG 2121 His Cys Val Asp Lys Lys Leu Asp Lys Gln Tyr Glu Ser Leu Ser Leu 675 '680 685 690 TTT CAC CCC AGT AAC GTC GAA ATG CTG AGC AGC ATG GAT TCT GCA TCG 2169 Phe His Pro Ser Asn Val Glu Met Leu Ser Ser Met Asp Ser Wing Ser 695 700 705 GTT CGC ATT ATC AAA CCC TTT CCT GCG CCC CAG ACT CCA GGC CGC CTG 2217 Val Arg He He Lys Pro Phe Pro Pro Wing Gln Thr Pro Gly Arg Leu 710 715 720 CAG CCT GCC CCT GTG ATC CCT TCG GCG CCA GCA GCT CCA AAA CTG GAC 2265 Gln Pro Pro Wing Val He Pro Wing Pro Wing Pro Wing Pro Lys Leu Asp 725 730 735 CAC CAG AGA ATG GAC ACC ATC CAG GAA GAC CCC AGC ACA GAC TCA CAT 2313 His Gln Arg Met Asp Thr He Gln Glu Asp Pro Ser Thr Asp Ser His 740 745 750 ATG GAC GAG GGG TTT GAG AAG GAC CCC TTC CCA AAT AGC AGC ACA 2361 Met Asp Glu Asp Gly Phe Glu Lys Asp Pro Phe Pro Asn Ser Ser Thr 755 760 765 770 GCT GCC AAG TCA TTT GAG GAT CTC ACG GAC CAT CCG GTC ACC AGA AGT 2409 Wing Wing Lys Ser Phe Glu Asp Leu Thr Asp His Pro Val Thr Arg Ser 775 780 785 GAA AAG GCT GCC TCC TTT AAA CTG CAG CGT CAG AAT CGT GTT GAC AGC 2457 Glu Lys Ala Ala Ser Phe Lys Leu Gln Arg Gln Asn Arg Val Asp Ser 790, 795 '800 AAA GAA ACÁ GAG TGC TAA 2.75 Lys Glu Thr Glu Cys 805 (2) IN TRAINING FOR SEQ ID NO: 2: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 807 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 2: Pro Arg Pro Pro Aso Asp Pro Gly Phe Gly Pro Kis Gln A.rg Leu Glu 1 5 10 15 Lvs Leu A.SD Ser Leu Leu Ser Asp Tyr Asp He Leu Ser Leu Ser Asn 20 25 '30 He Gln Gln His Ser Val Arg Lys Are Asp Leu Gin Thr Ser Thr His 35 40 45 Val Glu Thr Leu Leu Thr Phe Be Wing Leu Lys Arg Hia Phe Lys Leu 50 55 60 Tyr Leu Thr Ser Ser Thr Glu Arg Phe Ser Gln Asn Phe Lys Val Val 65 70 75 80 Val Val Aso Gly Lys Asn Glu Ser Glu Tyr Thr Val Lys Trp Gln Asp 85 90 95 Phe Phe Thr Gly His Val Val Gly Glu Pro Asp Ser Arg Val Leu Wing 100 105 110 His He Arg Asp Asp Asp Val He He Arg He Asn Thr Asp Gly Wing 115"'120 125 Glu Tyr Asn He Glu Pro Leu Trp Arg Phe Val Asn Asp Thr Lys Asp 130 135 140 Lys Arg Met Leu Val Tyr Lys Ser Glu Asp He Lys Asn Val Ser Arg 145 150"155 160 Leu Gln Ser Pro Lys Val Cys Gly Tyr Leu Lys Val Asp Asn Glu Glu 165"170 175 Leu Leu Pro Lys Gly Leu Val Asp Arg Glu Pro Pro Glu Glu Leu Val 180 185 190 His Ars Val Lvs Ars Arg Ala Asp Pro Asp Pro Met Lys Asn Thr Cys 195"200 205 Lvs Leu Leu Val Val Wing Asp His Arg Phe Tyr Arg Tyr Met Gly Arg 210 215 '220 Gly Glu Glu Be Thr Thr Thr Asn Tyr Leu He Glu Leu He Asp Arg 225 23C 235 240 Val Asp Asp He Tyr Arg A.sn Thr Ser Trp Asp Asn A.la Gly Phe Lys 245 250 255 Gly Tyr Gly He Gln He Glu Gln He Arg He Leu Lys Ser Pro Gln 260 265 270 Glu Val Lys Pro Gly Glu lys His Tyr Asn Met Wing Lys Ser Tyr Pro 275 280 285 Asn Glu Glu Lys Asp Wing Trp Asp Val Lys Met Leu Leu Glu Gln Phe 290 295 300 Ser Phe Asp He Wing Glu C-iu Wing Ser Lys Val Cys Leu Wing Kis Leu 305"310 315 320 Phe Thr Tyr Gl Asp Phe Asp Met Gly Thr Leu Gly Leu Ala Tyr Val 325 330 335 Gly Ser Pro Arg Ala Ai Ser Hrs Gly Gly Val Cys Pro Lys Wing Tyr 340 345 350 Tvr Ser Prc Val G (-l! V • Lvs Asr.: Ie Tvr Leu Asn Ser Glv Leu Thr 360 365 Be Thr Lys Asn Tyr Gly Lys Thr He Leu Thr Lys Glu Wing Asp Leu 370 375 380 Val Thr Thr His Glu Leu Giy His Asn Phe Gly Ala Glu His Asp Pro 385 390 395 400 Asp Gly Leu Wing Glu Cys Wing Pro Asn Glu Asp Gln Gly Gly Lys Tyr 405 410 '415 Val Met Tyr Pro He Wing Val Ser Gly Asp His Glu Asn Asn Lys Met 420 425 430 Phe Ser Asn Cys Ser Lys Gln Ser He Tyr Lys Thr He Glu Ser Lys 435 440 445 Wing Gln Glu Cys Phe Gln Glu Arg Ser Asn Lys Val Cys Gly Asn Ser 450 455 460 Arg Val Asp Glu Glu Glu Glu Cys Asp Pro Gly He Met Tyr Leu Asn 465 470 475 480 Asn Asp Thr Cys Cys Asn Ser Asp Cys Thr Leu Lys Glu Gly Val Gln 465 490 495 Cys Ser Asp Arg Asn Ser Pro Cys Cys Lys Asn Cys Gln Phe Glu Thr 500 505 510 Wing Gln Lys Lys Cys Gln Glu Wing He Asn Wing Thr Cys Lys Gly Val 515 520 525 Ser Tyr Cy3 Thr Gly Asn Ser Ser Giu Cys Pro Pro Pro Gly Asn Ala 530 535 540 Glu Asp Asp Thr Val Cys Leu Asp Leu Gly Lys Cys Lys Aso Glv Lys 545 550 '555' 560 Cys He Pro Phe Cys Glu A.rg Glu Gln Gln Leu Glu Ser Cys Ala Cys 565 570"575 Asn Glu Thr Asp Asn Ser Cys Lys Val Cys Cys Ara Asp Leu Ser Gly 580 585"590 Arg Cys Val Pro Tvr Val Asp Wing Glu Gln Lys Asn Leu Phe Leu Are 595 '" 600 605 Lys Glv Lys Pro Cys Thr Val Gly Phe Cys Asp Met Asn Gly Lys Cys 61Ó 615 620 Glu Lvs Arg Val Gln Asp Val He Glu Arg Phe Tr? Asp Phe He Asp 625"63C 635 640 Gln Leu Ser He Asn Thr Phe Gly Lys Phe Leu Wing Asp Asr. He Val 645 650 655 Gly Ser Val Leu Val Phe Ser Leu He Phe Trp He Pro Phe Ser He 660 665 _ - 670 Leu Val His Cvs Val Asp Lys Lys Leu Asp Lys Gin Tyr Glu Ser Leu 675"" 680 6E5 Being Leu Phe His Pro Being Asn Val Glu Met Leu Ser Being Met A = p Being 690 695 700 Wing Ser Val Arg He He Lys Pro Phe Pro Wing Pro Gln Thr Pro Glv 705 710 '715 720 Arg Leu Gln Pro Wing Pro Val He Pro Wing Pro Wing Pro Wing Pro Lys 725 730 735 Leu Asp His Gln Arg Met Asp Thr He Gln Glu Asp Pro Ser Thr Aso 740 745 750 Ser His Met Asp Glu Asp Gly Phe Glu Lys Asp Pro Phe Pro Asn Se- 755 760 765 Ser Thr Ala Ala Lys Ser Phe Glu Asp Leu Thr Asp His Pro Val Thr 770 775 780 Arg Ser Glu Lys Wing Wing Being Phe Lys Leu Gln Arg Gln Asn Arg Val 85 790 795 800 Asp Ser Lys Glu Thr Glu Cys 805 (2) INFORMATION FOR SEQ ID NO: 3: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 2097 base pairs (B) TYPE: nucleic acid (C) FILAMENTO: simple (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: cDNA to mRNA (iii) HYPOTHETICAL: NO (ix) CHARACTERISTICS: (A) NAME / KEY: CDS (B) LOCATION: 52..2094 (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 3: ATGAGGCAGT CTCTCCTATT CCTGACCAGC GTGGTTCCTT TCGTGCTGGC G CCG CGA 57 Pro Arg CCT CCG GAT GAC CCG GGC TTC GGC CCC CAC CAG AGA CTC GAG AAG CTT 105 Pro Pro Asp Asp Pro Gly Phe Gly Pro His Gln Arg Leu Giu Lys Leu 610"" 815 620 '825 GAT TCT TTG CTC TCA GAC TAC GAT ATT CTC TCT TTA TCT AAT ATC CAG 153 Asp Ser Leu Leu Ser Asp Tyr Asp He Leu Ser Leu Ser Asr. He Gln 830 835 840 CAG CAT TCG GTA AGA AAA AGA GAT CTA CAG ACT TCA ACA CAT GTA GAA 2C1 Gln His Ser Val Are Lys A.rg A = p Leu Gln Thr Ser Thr His Val Glu 845 850 855 ACA CTA CTA ACT TTT TCA GCT TTG AAA AGG CAT TTT AAA TTA TAC CTG 249 Thr Leu Leu Thr Phe Ser Wing Leu Lys Arg Kis Phe Lys Leu Tyr Leu 860 865 870 ACÁ TCA AGT ACT GAA CGT TTT TCA CAA AAT TTC AAG GTC GTG GTG GTG 297 Thr Ser Ser Thr Glu Arg Phe Ser Gln Asn Phe Lys Val Val Val Val 875 880 885 GAT GGT AAA AAC GAA AGC GAG TAC ACT GTA AAA TGG CAG GAC TTC TTC 345 Asp Gly Lys Asn Glu Ser Glu Tyr Thr Val Lys Trp Gln Asp Phe Phe 890 895 900"905 ACT GGA CAC GTG GTT GGT GAG CCT GAC TCT AGG GTT CTA GCC CAC ATA 393 Thr Gly His Val Val Gly Glu Pro Asp Ser Arg Val Leu Wing His He 910 915 920 AGA GAT GAT GAT GTT ATA ATC AGA ATC AAC ACA GAT GG GCC GAA TAT 441 Arg Asp Asp Asp Asp Val He He Arg He Asn Thr Asp Gly Wing Glu Tyr 925 930 935 AAC ATA GAG CCA CTG TGG AGA TTT GTT AAT GAT ACC AAA GAC AAA AGA 489 Asn He Glu Pro Leu Trp Arg Phe Val Asn Asp Thr Lys Asp Lys Arg 940 945 950 ATG TTA GTT TAT AAA TCT GAA GAT ATC AAG AAT GTT TCA CGT TTG CAG 537 Met Leu Val Tyr Lys Ser Glu Asp He Lys Asn Val Ser A rg Leu Gln 955"960 965 TCT CCA AAA GTG TGT GGT TAT AAA GTG GAT AAT GAA GAG TTG CTC 585 Ser Pro Lys Val Cys Gly Tyr Leu Lys Val Asp Asn Glu Glu Glu Leu Leu 970 975 980 980 985 CCA AAA GGG TTA GTA AGA GAA CGA CCT GAA GAT CTT GTT CAT CGA 633 Pro Lys Gly Leu Val Asp Arg Glu Pro Pro Glu Glu Leu Val His Arg 990 995 1000 GTG AAA AGA AGA GAC CAC GAT CCC ATG AAG AAC ACG TGT AAA TTA 681 Val Lys Arg Arg Wing Asp Pro Asp Pro Met Lys Asn Thr Cys Lvs Leu 1005 1010"1015 TTG GTG GTA GTA CAT CAT CGC TTC TAC AGA TAC ATG GGC AGA GGG GAA 729 Leu Val Val Ala Asp His Arg Phe Tyr Arg Tyr Met Gly Are Glv Giu 1020 '1025' * 1030 GAG AGT ACÁ ACT ACÁ AAT TAC TTA ATA GAC CTA ATT GAC AGA GTT GAT 777 Glu Be Thr Thr Thr Asn Tyr Leu He Glu Leu He Asp Are Val Aso 1035 1040 1045 GAC ATC TAT CGG AAC ACT TCA TCC- GA7 AAT GCA GGT TTT AAA GCC TAT 825 Asp He Tyr Arg Asn Thr Ser Crp Asp Asr. Wing Gly Phe Lys Glv Tyr 1050 1055 1060 '* "1065 GGA ATA CAG ATA GAG CAG ATT CCC ATT CTC AAG TCT CCA CA GAS GTA S73 Gly He Gln He Glu Gln He Arg He Leu Lys Ser Pro Glr. Glu Val 1070 1075"10SC AAA CCT GGT GAA AAG CAC TAC? -. C ATC- GCA AAA AGT TAC CCA AAT GAA 921 Lys Pro Gly Glu Lys Kis Tyr As- Met Wing Lys Ser Tyr Pro Asr. Gl_ 1085 109: IOS : GAA AAG GAT GCT TGG GAT GTG AAG ATG TTG CTA GAG CA TTT AGC TTT 969 Glu Lys Asp Wing Trp Asp Val Lys Met Leu Leu Glu Gln Phe Ser Phe 1100 1105 1110 GAT ATA GCT GAG GAA GCA TCT AAA GTT TGC TTG GCA CAC CTT TTC ACÁ 1017 Asp He Wing Glu Glu Wing Ser Lys Val Cys Leu Wing His Leu Phe Thr 1115 1120 '"1125 TAC CAAT GAT TTT GAT ATG GGA ACT CTT GGA TTA GCT TAT GTT GGC TCT 1065 Tyr Gln Asp Phe Asp Met Gly Thr Leu Gly Leu Ala Tyr Val Gly Ser 1130 1135 1140 1145 CCC AGA GCA AAC AGC CAT GGA GGT GTT TGT CCA AAG GCT TAT TAT AGC 1113 Pro Arg Ala Asn Ser His Gly Gly Val Cys Pro Lys Wing Tyr Tyr Ser 1150 1155 1160 CCA GTT GGG AAG AAA AAT ATC TAT TTG AAT AGT GGT TTG ACG AGC ACAL 1161 Pro Val Gly Lys Lys Asn He Tyr Leu Asn Ser Gly Leu Thr Ser Thr 1165 1170 1175 AAG AAT TAT GGT AAA ACC ATC CTT ACA AAG GAA GCT GAC CTG GTT ACA 1209 Lys Asn Tyr Gly Lys Thr He Leu Thr Lvs Glu Wing Asp Leu Val Thr 1180 '1185' 1190 ACT CAT GAA TTG GGA CAT AAT TTT GGA GCA GAA CAT GAT CCG GAT GGT 1257 Thr His Glu Leu G and His Asn Phe Gly Wing Glu His Asp Pro Asp Gly 1195 1200"1205 CTA GCA GAA TGT GCC CCG AAT GAG CAG GGA GGG AAA TAT GTC ATG 1305 Leu Wing Glu Cys Wing Pro Asn Giu Asp G n Gly Gly Lys Tyr Val Met 1210 1215 1220 1225 TAT CCC ATA GCT GTG AGT GGC GAT CAC GAG AAC AAT AAG ATG TTT TCA 1353 Tyr Pro He Wing Val Ser Gly Asp His Glu A.sn Asn Lys Met Phe Ser 1230 1235 1240 AAC TGC AGT AAA CAÁ TCA ATC TAT AAG ACC ATT GAA AGT AAG GCC CAG 1401 A = n Cys Ser Lys Gln Ser He Tyr Lys Thr He Giu Ser Lys Wing Gln 1245 1250 1255 GAG TGT TTT CAA GAA CGC AGC AAT AAA GTT TGT GGG AAC TCG AGG GTG 1449 Glu Cys Phe Gln- Glu Arg Ser Asr. Lys Val Cys Gly Asn Ser Arg Val 1260 1265"" 1270 GAT GAA GGA GAA GAG TGT GAT CCT GGC ATC ATG TAT CTG AAC AAC GAC 1497 Asp Giu Glu Glu Glu Cys Asp Pro Gly He Met Tyr Leu Asn Asn ASD 1275 128C 1285 ACC TGC TGC AAC AGC GAC TGC ACC TTG AAC- GAA GGT GTC CAG TGC AGT 1545 Thr Cys Cys Asn Ser Asp Cys T .-. R Leu Lvs Glu Giv Val Gln Cvs Ser 1290 1295 ~ 1300"'1305 GAC AGG AAC AGT CCT TGC TGT AAA AAC TCC CAG TTT GAG ACT GCC CAC-1593 Asp Arg Asn Ser Pro Cy = Cys lys Asn Cv; Gin Phe Glu Thr Ala Glr. 1310 131? 1320 í -_-_ f y-r-. ib- (_ -_ -no ___ • r..1.-_- .. G _. j -._. _ _- _ __-_-. G_ > C GT_ TCC TAC 16 - _ Lys Lys Cys Gln Glu Ala He As.-. Wing T .-. R Cys Lys Gly Val Ser Tvr 1325 1330"1335 TGC ACA GGT AAT AGC AGT GAG TGC CCG CCT CCA GGA AAT GCT GAA GAT 1689 Cys Thr Gly Asn Ser Ser Glu Cys Pro Pro Pro Gly Asn Ala Glu Asp 1340 1345 1350 GAC ACT GTT TGC TTG GAT CTT GGC AAG TGT AAG GAT GGG AAA TGC ATC 1737 Asp Thr Val Cvs Leu Asp Leu Gly Lys Cys Lys Asp Gly Lys Cys He 1355 '"1360 1365 CCT TTC TGC GAG AGG GAA CAG CAG CTG GAG TCC TGT GCA TGT AAT GAA 1785 Pro Phe Cys Glu Arg Glu Gl Gln Leu Glu Ser Cys Ala Cys Asn Glu 1370 1375 1380 1385 ACT GAC AAC TCC TGC AAG GTG TGC TGC AGG GAC CTT TCC GGC CGC TGT 1833 Thr Asp Asn Ser Cys Lys Val Cys Cys Arg Asp Leu Ser Gly Arg Cys 1390 1395"1400 GTG CCC TAT GTC GAT GCT GAA CAA AAG AAC TTA TTT TTG AGG AAA GGA 1881 Val Pro Tyr Val Asp Ala Glu Gln Lys Asn Leu Phe Leu Arg Lys Gly 1405 1410 1415 AAG CCC TGT ACÁ GTA GGA TTT TGT GAC ATG AAT GGC AAA TGT GAG AAA 1929 Lys Pro Cys Thr Val Gly Phe Cys Asp Met Asn Gly Lys Cys Glu Lys 1420"1425 CGA GTA CAG GAT GTA ATT GAA CGA TTT TGG GAT TTC ATT GAC CAG CTG 1977 Arg Val Gln Asp Val He Glu Arg Phe Trp Asp Phe He Asp Gln Leu 1435 1440 1445 AGC ATC AAT ACT TTT GGA AAG TTT TTA GCA GAC AAC ATC GTT GGG TCT 2025 Ser He Asn Thr Phe Gly Lys Phe Leu Wing Asp Asn He Val Gly Ser 1450 1455 1460 1465 GTC CTG GTT TTC TCC TTG ATA TTT TGG ATT CCT TTC AGC ATT CTT GTC 2073 Val Leu Val Phe Ser Leu He Phe Trp He Pro Phe Ser He Leu Val 1470 1475 1480 CAT TGT GTA ACG TCG AAA TGC TGA 2097 His Cys Val Thr Ser Lys Cys 1485 (2) INFORMATION FOR SEQ ID NO: 4: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LONGITU D: 681 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 4: Pro Arg Pro Prc Aso Asp Prc Glv? re Glv Prc Hrs Gin Are Le-- C ui 5 10 i- Lys Leu Asp Ser leu Leu Ser As .e. S e r Leu S e r As r i 20 3 C i e G r. C- ln K _ s S er Va l A rr S e - "- - u - - c 4 5 Val Glu Thr Leu Leu Thr Phe Ser Ala Leu Lys Arg His Phe Lys Leu 50 55 60 Tyr Leu Thr Ser Ser Tnr Glu Arg Phe Ser Gln Asn Phe Lys Val Val 65 70 75 80 Val Val Asp Gly Lys Asn Glu Ser Glu Tyr Thr Val Lys Trp Gln Asp 85 90 95 Phe Phe Thr Gly His Val Val Gly Glu Pro Asp Ser Arg Val Leu Wing 100 105 110 His He Arg Asp Asp A.sp Val He He Arg He Asn Thr Asp Gly Wing 115 '120 125 Glu Tyr Asn He Glu Pro Leu Trp Arg Phe Val Asn Asp Thr Lys Asp 130 135"140 Lys Arg Met Leu Val Tyr Lys Ser Glu Asp He Lys Asn Val Ser Arg 145 150" 155 160 Leu Gln Ser Pro Lys Val Cys Gly Tyr Leu Lys Val Asp Asn Glu Glu 165 170 '175 Leu Leu Pro Lys Gly Leu Val Asp Arg Glu Pro Pro Glu Glu Leu Val 180"185 190 His Arg Val Lys Arg Ars Wing Asp Pro Asp Pro Met Lys Asn Thr Cys 195 200" 205 Lys Le-u Leu Val Val Wing Asp His Arg Phe Tyr Arg Tyr Met Gly Arg 210 215 220 Gly Glu Glu Be Thr Thr Thr Asr. Tyr Leu He Glu Leu He ASD Arg 225 230 '235' 240 Val Asp Asp He Tyr Arg Asn Tr.r Ser Trp Asp Asn Wing Gly Phe Lys 245 250 * "255 Gly Tyr Gly He Gln He Glu Gin He Arg He Leu Lvs Ser Pro Gln 260 265"270 Glu Val Lys Pro Gly Giu Lys Hrs Tvr Asr. Met Wing Lvs Ser Tyr Pro 275 '2SC 285 Asn Glu Glu Lys Asp Wing Trp Asp Val Lys Met Leu Leu Glu Gln Phe 290 295"300 Ser Phe Asp He Wing Giu Giu Wing Being Lvs Val Cvs Leu Wing Kis Leu 305 31C 215"320 Phe Thr Tvr Gin Asp F: \ -. R Leu Glv Leu Ala Tvr Val 325 Giv Ser Prc Aro Ai, Gly Val Cys Pro Ly = A.la Tyr 340 350 Tyr Ser Prc Val G:: leu Asr. Ser Gly Leu Tr.r 355 3 £ 5 Ser Thr Lys Asn Tyr Gly Lys Thr He Leu Thr Lys Glu Wing Asp Leu 370 375 380 Val Thr Thr His Glu Leu Gly His Asn Phe Gly Ala Glu His Asp Pro 385 390 395 400 Asp Gly Leu Wing Glu Cys Wing Pro Asn Glu Asp Gln Gly Gly Lys Tyr 405 410 415 Val Met Tyr Pro He Wing Val Ser Gly Asp His Glu Asn Asn Lys Met 420 425 430 Phe Ser Asn Cys Ser Lys Gln Ser He Tyr Lys Thr He Glu Ser Lys 435 440 445 Wing Gln Glu Cys Phe Gln Glu Arg Ser Asn Lvs Val Cys Gly Asn Ser 450 * 455"460 Arg Val Asp Glu Gly Glu Glu Cys Asp Pro Gly He Met Tyr Leu Asn 465"470" 475 480 Asn Asp Thr Cys Cys Asn Ser Asp Cys Thr Leu Lys Glu Gly Val Gln 485"490" 495 Cys Ser Asp Arg Asn Ser Pro Cys Cys Lys Asn Cys Gln Phe Glu Thr 500 505 '"510 Wing Gln Lys Lys Cys Gln Glu Wing He Asn Wing Thr Cys Lys Gly Val ? 5 520 525 Ser Tyr Cys Thr Gly Asn Ser Glu Cys Pro Pro Pro Gly Asn Ala 530 535 540 Glu Asp Asp Thr Val Cys Leu Asp Leu Gly Lys Cys Lys Asp Glv Lvs 545"550 555 '560 Cys He Pro Phe Cys Glu Arg Glu Gln Gln Leu Glu Ser Cys Ala Cys 565 570 5 / _ Asr. Glu Thr Asp Asn Ser Cys Lys Val Cys Cys A.rg Asp Leu Ser Gly 580 585 590 A.rg Cys Val Prc Tyr Val Asp Ala Glu Gln Lys Asn Leu Phe Leu Are 595"600 '605 Lys Gly Lys Pro Cys Thr Val Gly Phe Cys Asp Met Asn Glv Lvs Cvs 610 615 620 Glu Lys A.rg Val Gln Asp Val He Glu Arg Phe Trp A.sp Phe He Asp € 15 630 635 640 Glr. Leu Ser He Asr. Thr Phe Giy Lys Phe Leu Wing Asp Asr. He Val 645 650"€ 55 Gly Ser Val Leu Val Phe Ser Leu lie Phe Trp He Pro Phe Ser lie 660 665"670 leu Val Hrs Cys Val Thr Ser Lys Cvs 675" 680 (2) IN TRAINING FOR SEQ ID NO: 5: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 8 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 5: Leu Ala Gln Ala Val Arg Ser Ser 1 5 (2) IN TRAINING FOR SEQ ID NO: 6: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LONGITU D: 17 base pairs (B) TYPE: nucleic acid (C) FI LAMENT: simple (D) TOPOLOGY: linear (iii) HI POTÉTICA: NO (xi) DESCRIPTION OF SEQUENCE: SEQ ID NO: 6: AARTAYGTNA TGTAYCC 17 (2) INFORMATION FOR SEQ ID NO: 7: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LONGITU D: 17 base pairs (B) TYPE: nucleic acid (C) FILAMENTO: simple (D) TOPOLOGY: linear (iii) ) HYPOTHETICAL: NO (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 7: CCRCARTCRC AYTCYTC 17 (2) INFORMATION FOR SEQ ID NO: 8: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 6 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEQ ID NO: 8: Glu Glu Cys Asp Cys Gly 1 5 (2) INFORMATION FOR SEQ ID NO: 9: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 11 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEQ ID NO: 9: lie Wing Val Ser Gly Asp His Glu Asn Asn Lys 1 5 10

Claims (1)

  1. CLAIMS A TACE polypeptide isolated and purified. An isolated and purified polypeptide according to claim 1, having a molecular weight of about 80 kD. An isolated and purified polypeptide according to claim 1, in a non-glycosylated form. An isolated and purified polypeptide according to claim 1, selected from the group consisting of a polypeptide comprising amino acids 18-Xaa of SEQ ID NO: 2, wherein Xaa is an amino acid selected from the group consisting of amino acids 671 a 824. The isolated and purified antibodies that bind to the polypeptide according to claim 1. The isolated and purified antibodies according to claim 5, wherein the antibodies are monoclonal antibodies. A method for detecting the TACE inhibitory activity of a molecule, comprising mixing said molecule with a substrate, incubating a polypeptide according to claim 1 with the mixture, and determining by means of chromatography the degree of cut of the substrate. A method for detecting the TACE inhibitory activity of a molecule according to claim 7, wherein the substrate comprises the amino acid sequence Leu-Ala-GIn-Ala-Val-Arg-Ser-Ser. 9. A method for using a polypeptide according to claim 1, in a design based on the structure of an inhibitor of said polypeptide, comprising the steps of determining the three-dimensional structure of such a polypeptide, analyzing the three-dimensional structure for possible binding sites of the substrates, synthesizing a molecule that incorporates a predictable reactive site, and determining the polypeptide inhibitory activity of the molecule. 10. A method for detecting the TNF cutting capacity of a molecule, comprising incubating said molecule with a substrate comprising the amino acid sequence Leu-Ala-GIn-Ala-Val-Arg-Ser-Ser, and determining the degree of cutting the substrate. eleven . An isolated nucleic acid selected from the group consisting of: (a) the coding region of a natural mammalian TACE gene; (b) cDNA comprising nucleotides 52-2472 of SEQ ID NO: 1; (c) the nucleic acid that is at least 80% identical to the nucleic acid of (a) or (b) and that encodes a polypeptide that converts TNF-a from the 26 kD form to the 17 kD form; and (d) the nucleic acid, which is degenerated as a result of the genetic code to a nucleic acid defined in (a), (b) or (c), and which codes for biologically active TACE. 12. An isolated nucleic acid according to claim 1, wherein the TACE is human TACE. 13. An isolated nucleic acid according to claim 1, which encodes a polypeptide comprising amino acids 18-671 of SEQ ID NO: 2. 14. An expression vector that directs the expression of a nucleic acid sequence according to claim 1. 15. A host cell transfected or transformed with the expression vector according to claim 1 1. 16. A process for producing a TACE polypeptide, which comprises culturing a host cell according to claim 15 under conditions that promote expression, and recovering the polypeptide from the culture medium. 17. A method for inhibiting the TNF-α cut-off of cell membranes in a mammal, comprising administering to said mammal an effective amount of a compound that inhibits the proteolytic activity of an enzyme comprising the amino acid sequence 18-671 of the SEQ ID NO: 2 18. A method for inhibiting the TNF-a cut of cell membranes comprising blocking the binding of TNF-α with an enzyme having amino acid sequence 18-674 of SEQ ID NO: 2. 19. A method for treating a mammal having a disease characterized by an overproduction or an overregulated production of TNF-a, which comprises administering to the mammal a composition comprising an amount of a compound that effectively inhibits the proteolytic activity of TNF-a from an enzyme comprising the amino acid sequence 18-671 of SEQ ID NO: 2.
MXPA/A/1997/009744A 1995-06-08 1997-12-05 Enzima convertidora tnf-a MXPA97009744A (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US428458 1995-04-26
US48245895A 1995-06-08 1995-06-08
US50461495A 1995-07-20 1995-07-20
US504614 1995-07-20
US08/655,345 US5830742A (en) 1995-06-08 1996-05-23 TNF-α converting enzyme
US08655345 1996-05-23

Publications (2)

Publication Number Publication Date
MX9709744A MX9709744A (en) 1998-07-31
MXPA97009744A true MXPA97009744A (en) 1998-11-09

Family

ID=

Similar Documents

Publication Publication Date Title
US7695948B2 (en) Antibodies that bind TNF-α converting enzyme
US5830742A (en) TNF-α converting enzyme
AU712759B2 (en) TNF-alpha converting enzyme
AU2315099A (en) Il-17rh dna and polypeptides
AU2015799A (en) V201 dna and polypeptides
US20090017492A1 (en) SVPH1-26 DNA and polypeptides
MXPA97009744A (en) Enzima convertidora tnf-a
AU752369B2 (en) TNF-alpha converting enzyme
US20030045683A1 (en) H14 DNA and polypeptides
AU745623B2 (en) V196 DNA and polypeptides
US7217553B2 (en) Nucleic acids encoding human adamalysin SVPH1-8
EP1027441A2 (en) Murine dna molecules encoding polypeptides having kinase functions
AU2015899A (en) V197 dna and polypeptides