CA2190466A1 - Monocyte chemotactic protein-4 - Google Patents

Monocyte chemotactic protein-4

Info

Publication number
CA2190466A1
CA2190466A1 CA002190466A CA2190466A CA2190466A1 CA 2190466 A1 CA2190466 A1 CA 2190466A1 CA 002190466 A CA002190466 A CA 002190466A CA 2190466 A CA2190466 A CA 2190466A CA 2190466 A1 CA2190466 A1 CA 2190466A1
Authority
CA
Canada
Prior art keywords
polypeptide
mcp
polynucleotide
dna
sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002190466A
Other languages
French (fr)
Inventor
Haodong Li
Steven M. Ruben
Granger G. Sutton, Iii
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Human Genome Sciences Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to CA002190466A priority Critical patent/CA2190466A1/en
Publication of CA2190466A1 publication Critical patent/CA2190466A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/521Chemokines
    • C07K14/523Beta-chemokines, e.g. RANTES, I-309/TCA-3, MIP-1alpha, MIP-1beta/ACT-2/LD78/SCIF, MCP-1/MCAF, MCP-2, MCP-3, LDCF-1, LDCF-2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • Genetics & Genomics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Biophysics (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Toxicology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Polynucleotide which encode the polypeptide MCP-4, as well as such polypeptides, antibodies and antagonist/inhibitors against the polypeptide and the use of the polypeptide and antagonist/inhibitors as pharmaceutical for treatment of tumors, wounds, parasitic infection, regulation of hematopolesis, inflammation, rheumatoid arthritis, lung inflammation, allergies, atherosclerosis and infectious diseases such as tuberculosis.

Description

WO 95~314G7 21 g 0 4 ~ 6 PCrlUS94/05384 .

IL. '~ _ ~"rIC PROTEIN--4 This invention relates to newly identified polynucleotides, polypeptides encoded by such polynucleotides, the use of such polynucleotides and polypeptides, as well as the production of such polynucleotides and polypeptides. More particularly, the polypeptide of the present invention is monocyte chemotactic protein-4 (MCP-4~. The invention also relates to inhibiting the action of such polypeptides.
There are three forms of monocyte chemotactic protein, namely, MCP-l, MCP-2 and MCP-3. All of these proteins have been structurally and functionally characterized and have also been cloned and expressed. MCP-l and MCP-2 have the ability to attr~ct leukocytes tmonocytes, and leukocytes ), while MCP-3 also attracts eosinophils and T lymphocytes (Dahinderi, E. et al., J. Exp. Med., 179:751-756 (1994)).
Initially, human monocyte-specific attracting factor, was purified from a glioma cell line and a monocytic cell line. Matsushima, K. et al, J. Exp. Med., 169:1485-1490 (1989). This factor was originally designated glioma-derived chemotactic factor (GDCF) and monocyte chemotactic and activating factor (MCAF) by Matsushima, et al. This f actor is now ref erred to as MCP-l . Subsequent cloning of the cD~A for MCP-1 showed it to be highly similar to the murine JE gene. The JE gene could be massively induced in SUBSTITUTE SHEET (RULE 261 WO 95131467 219 0 4 6 ~ PCT/US94/05384 murine fibroblasts by platèlè~derived growth factor.
Cochran, B.H., et al, Cell 33:939-947 (1983). Murine JE i8 highly similar to MCP-1. The MCP-1 protein is 629<~
identical to murine JE in a region of 68 shared N-tPrmin~l residues. It is widely accepted that JE and MCP-1 are species homologs. The polypeptide of the present invention, MCP-4, i5 both structurally and functionally related to MCP-1 and the murine JE protein, see Figure 2.
A method of = suppressing tumor formation in a vertebrate by administering JE/MCP-l has been disclosed in PCT application WO-92/20372, along with methods of treating localized complicatlons of malignancies and methods of combatting parasitic infection by admini6tering JE/MCP-1.
Expression of the JE/MCP-1 protein in r~ nAnt cells was found to suppress the cells ability to form tumor6 tn vivo.
Human MCP-l is a basic peptide of 76 amino acids with a predicted molecular mass o~ 8,700 daltons. MCP--1 is inducibly expre6sed mainly in monocytes, endothelial cells and fibroblasts. Leonard, E.J. and Yoshimura, T., Immunol.
Today, 11:97-101 (1990). The factors which induce this expression is IL-1, TNF or lipopolysaccharide treatment.
Other properties of MCP-1 include the ability to strongly activate mature human basophils in a pertussis toxin-sensitive manner. MCP-1 is a cytokine capable of directly ;n~ ;ng histamine release by basophils, (Bischoff, S.C. et al., J. Exp. Med., 175:1271-1275 (1992)). Fur~h- le, MCP-1 promotes the formation of leukotriene C4 by basophils pretreated with Interleukin 3, Interleukin 5, or granulocyte/macrophage colony-stimulating factor. MCP-l induced basophil mediator release may play an important role in allergic inflammation and other pathologies expressing MCP-l.
Clones having a nucleotide sequence Pncofl;n~ a human monocyte chemotactic and activating factor (MCAF) reveal the primary structure of the MCAF polypeptide to be composed of a putative signal peptide sequence of 23 amino acid residues and a mature MCAF sequence of 76 amino acid residues. Furutani, Y.H., et al, Biochem. Biophys. Res.
-2-SUBSTITUTE SHEET (RULE 261 W0 95l31467 2 1 ~ 0 4 6 ~ PCr/[lS94/05384 Commu., 159: 249-55 ( 1989 ) . The complete amino aeid 6e~uence c~f human glioma-derived monocyte chemotactic factor (GDCF-2 ) has also been determined. This peptide attraets human monocytes but not neutrophils. It was established that GDCF-2 comprises 76 amino acid residues.
The peptide chain contains 4 half-cysteines, at positions 11, 12, 36 and 52, which create a pair of loops, clustered at the disulfide bridge6. Further, the MCP-l gene has been designated to human chromosome 17. Mehrabian, M.R., et al, G~-n ;rs~ 9:200-3 tl991). Certain data suggests that a potential role for MCP-1 is mediating monocytic infiltration of the artery wall. Monocytes appear to be central to atherogenesis both as the progenitors of foam cells and as a potential source of growth factors mediating intimal hyperplasia. Nelken, N.A., et al, J. Clin.
Invest., 88:1121-7 ~1991). It has also been found that synovial production of MCP-1 may play an important role in the recruitment of mononuclear phagocytes during inflammation associated with rheumatoid arthritis and that synovial tissue macrophages are the dominant source of this cytokine. MCP-1 levels were found to be significantly higher in synovial fluid from rheumatoid arthritis patients compared to synovial fluid from osteoarthritis patients or from patients with other arthritides. ~och, A.E., et al, J. Clin . Invest ., 90: 772-9 ( 1992 ) .
MCP-2 and MCP-3 are classified in a subfamily of proinflainmatory proteins and are functionally related to MCP-l because they specifically attract monocytes, but not neutrophils. Van Damme, J., et al, J. Exp. Med., 176:59-65 (1992). MCP-3 shows 71% and 589~ amino aeid homology to MCP-1 and MCP-2 respectively. ~CP-3 is an ;nfli -tory eytokine that regulates macrophage functions.
In accordance with one aspect of the present invention, there is provided a novel mature polypeptide which is MCP-4, as well as fragments, analogs and derivatives thereof. The polypeptide of the present invention is of human origin.

SU~STITUTE SHEEI ~RULE 26 WO 9~31467 21 g O ~ 6 6 PCT/US94/05384 In accordance with another aspect of the present invention, there are provided polynucleotides (DNA or R
which encode such polypeptides.
In accordance with yet a fu~her aspect of the present invention, there i6 provided a proce66 for producing 6uch polypeptide by r~ i n~nt techniques .
In accordance with yet a further aspect of the present invention, there is provided a process for u~;l;7;nq such polypeptide, or polynucleotide encoding 6uch polypeptide for therapeutic purpo6e6, for example, to treat tumor6, to promote wound healing, to combat para6itic infection and to regulate hematopoie6i6.
In accordance with yet a further a6pect of the pre6ent invention, there i6 provided an antibody again6t 6uch po lypeptide 6 .
In accordance with yet another a6pect of the pre6ent invention, there are provided antagoni6t/inhibitor6 to 6uch polypeptides, which may be u6ed to inhibit the action of such polypeptide6 for therapeutic purpo6e6, for example, to treat rheumatoid arthriti6, lung inflammation, allergy, inf ectiou6 di6eases and to prevent inf lammation and atherosclerosis .
The8e and other aspect6 of the present invention should be apparent to those 6killed in the art from the teaching6 herein.
The following drawing6 are illu6trative of: ofl;~ Ls of the invention and are not meant to limit the scope of the invention as Isn~ 6ed by the claim6.
FIG. 1 depict6 the cDNA 6equence and corre6ponding deduced amino acid 6equence of MCP-4. The 119 amino acid 6equence 6hown i6 the full length protein, with approximately the first 22 amino acid6 representing a leader sequence such that the mature form of the protein is 97 amino acids in length. The standard one letter ab~reviation for amino acids is used.
FIG. 2 illustrates the cD~A sequence homology between MCP-4 and the murine JE protein. The top sequence in each SUB~TJ~UTE SHEET (RULE 26) W0 95/31467 21 9 ~ 4 6 ~ PCTIUS94/05384 three segments is MCP-4 and the bottom sequence i8 murine JE protein.
FIG. 3 shows the results of a Northern blot analysis of the mRNA transcript for MCP-4 in human cells.
Figure 4 shows the banding pattern of human MCP-4 following bacterial expression and purif ication .
Figure 5 is a 6chematic representation of the pQE-9 vector .
In accordance with an aspect of the present invention, there is provided an isolated nucleic acid (polynucleotide) which encodes for the mature polypeptide having the deduced amino acid sequence of Figure l or for the mature polypeptide encoded by the cDNA of the clone deposited as ATCC Deposit No. 75703 on ~arch lO, 1994.
The polynucleotide of this invention was discovered from an activated monocyte cDNA library. It contains an open reading frame encoding a protein of approximately 119 amino acids in length of which the f irst 22 amino residues comprise a putative leader sequence. The mature protein therefore is predicted to be 97 amino acids in length. It is structurally related to mouse monocyte chemotactic protein (MCP-l or JE), showing 27% identity, and 56%
similarity over the entire human MCP-l protein sequence.
The polypeptide contains all four cysteine residues that occur in all chemokines in a characteri6tic motif. The sp~cing between these cysteines is conserved compared with the murine MCP-l/JE which strongly suggests that the new gene i6 a rlll k; n~ .
The polynucleotide of the present invention may be in the form of RNA or in the form of DNA, which DNA includes cDNA, genomic DNA, and synthetic DNA. The DNA may be double-6tranded or single-stranded, and if single stranded may be the coding strand or non-coding (anti-sense) strand.
The coding sequence which encodes the mature polypeptide may be identical to the coding sequence shown in Figure l or that of the deposited clone or may be a different coding sequence which coding sequence, as a result of the redundancy or degeneracy of the genetic code, encodes the SUBSTITUTE StlEET ~RULE 26 Wo 95/31467 2 1 9 0 4 6 ~ 1~, 5 ~ PCr/US94/05384 same, mature polypeptide as the DNA of Figure 1 or the deposited cDNA.
The polynucleo~ide which encodes for the mature polypeptide of Figure 1 or for the mature polypeptide encoded by the deposited cDNA may include: only the coding sequence for the mature polypeptide; the coding sequence for the mature polypeptide and additional coding sequence such as a leader or secretory sequence or a proprotein aequence; the coding 6equence for the mature polypeptide (and optionally additional coding sequence) and non-coding sequence, such as introns or non-coding sequence 5 ' and/or
3 ' of the coding sequence for the mature polypeptide.
Thus, the term "polynucleotide encoding 8 polypeptide"
P~ ~~ses a polynucleotide which includes only coding sequence for the polypeptide as well as a polynucleotide which i n~ rlP~ additional coding and/or non-coding sequence .
The present invention f urther relates to variants of the hereinabove described polynucleotides which encode for fragments, analogs and derivatives of the polypeptide having the deduced amino acid sequence of Figure 1 or the polypeptide encoded by the cDNA of the deposited clone.
The variant of the polynucleotide may be a naturally occurring allelic variant of the polynucleotide or a non-naturally occurring variant of the polynucleotide.
Thus, the present invention includes polynucleotides Pncorl;n~ the same mature polypeptide as shown in Figure 1 or the same mature polypeptide encoded by the cDNA of the deposited clone as well as variants of such polynucleotides which variants encode for a fragment, derivative or analog of the polypeptide of Figure 1 or the polypeptide encoded by the cDNA of the deposited clone. Such nucleotide variants include deletion variants, substitution variants and addition or insertion variant6.
As hereinabove indicated, the polynucleotide may have a coding sequence which is a naturally occurring allelic variant of the coding sequence shown in Figure 1 or of the coding sequence of the deposited clone. As known in the SLIBSTITUTE SHEET (RULE 26) ~ WO 95l31467 2 1 ~ ~ ~ 6 6 PCrlUSg4/05384 art, an allelic variant is an alternate form of a polynucleotide sequence which may have a substitution, deletion or addition of one or more nucleotides, which does not substantially alter the function of the encoded polypeptide .
The present invention also includes polynucleotides, wherein the coding sequence for the mature polypeptide may be fused in the same reading frame to a polynucleotide sequence which aids in expres6ion and secretion of a polypeptide from a host cell, for example, a leader sequence which functions as a secretory sequence for controlling transport of a polypeptide from the cell.
The polypeptide having a leader sequence is a preprotein and may have the leader sequence cleaved by the host cell to form the mature form of the polypeptide. The polynucleotides may also encode for a proprotein which is the mature protein plus additional 5~ amino acid residues.
A mature protein having a prosequence is a proprotein and is an inactive form of the protein. Once the prosequence is cleaved an active mature protein remains.
Thus, for example, the polynucleotide of the present invention may encode for a mature protein, or for a protein having a prosequence or for a protein having both a prosequence and a presequence (leader sequence).
The polynucleotides of the present invention may also have the coding sequence fused in frame to a marker sequence which allows for purification of the polypeptide of the present invention. The marker sequence may be a hexa-histidine tag supplied by a pQE-9 vector to provide for purification of the mature polypeptide fused to the marker in the case of a bacterial host, or, for example, the marker sequence may be a hemagglutinin (HA) tag when a r 1 ;An host, e.g. COS-7 cells, is used. The HA tag corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson, I., et al., Cell, 37:767 ( 1984 ) ) .
The present invention further relate8 to polynucleotides which hybridize to the hereinabove-SUSSTITUTE SHEET (ilULE 26) Wo 95/31467 2 ~ ~ ~ 4 ~ ~ PCrlUS9~105384 described sequences if there is at least 50% and preferably7O9~ identity between the sequence8. The present invention particularly relates to polynucleotides which hybridize under stringent conditions to the herein~bove-described polynucleotides . As herein used, the term "stringent conditions" means hybridization will occur only if there is at least 95% and preferably at least 97% identity between the sequences. The polynucleotides which hybridize to the hereinabove described polynucleotides in a pref erred embodiment encode polypeptides which retain substantially the same biological f unction or activity as the mature polypeptide encoded by the cDNA of Figure 1 or the deposited cDNA .
The deposit(s) referred to herein will be maintained under the terms of the Budapest Treaty on the International Recognition of the Deposit of Micro-organisms for purposes of Patent Procedure. These deposit6 are provided merely as convenience to those of 6kill in the art and are not an admission that a deposit is required under 35 U.S.C. ~112.
The sequence of the polynucleotides contained in the deposited materials, as well as the amino acid sequence of the polypeptides encoded thereby, ~re incorpor~ted herein by reference and are controlling in the event of any conflict with any description of sequences herein. A
license may be required to make, use or sell the deposited materials, and no such license is hereby granted.
The present invention further relates to a MCP-4 polypeptide which has the deduced amino acid sequence of Figure l or which has the amino acid sequence encoded by the deposited cDNA, as welI as fragments, analogs and derivatives of such polypeptide.
The terms "fragment, " "derivative" and "analog" when referring to the polypeptide of Figure l or that encoded by the deposited cDNA, means a polypeptide which retains essentially the same biological function or activity as such polypeptide. Thus, an analog includes a proprotein which can be activated by cleavage of the proprotein portion to produce an active mature polypeptide.

SUBSTITUTE SHEET ~RULE 261 Wo 9~/31467 ~ 4 ~ 6 PCTIUS9410~384 The polypeptide of the present invention may be a recombinant polypeptide, a natural polypeptide or a synthetic polypeptide, preferably a recombinant polypeptide .
- The fragment, derivative or analog of the polypeptide of Figure 1 or that encoded by the deposited cDNA may be (i) one in which one or more of the amino acid residues are 6ubstituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code, or (ii) one in which one or more of the amino acid residues includes a substituent group, or (iii) one in which the mature polypeptide is fused with another compound, such as a compound to increase the half-life of the polypeptide (for example, polyethylene glycol), or (iv) one in which the additional amino acids are f used to the mature polypeptide, such as a leader or secretory sequence or a sequence which is employed for purif ication of the mature polypeptide or a proprotein se~uence. Such fragments, derivatives and analogs are deemed to be within the scope of those skilled in the art f rom the teachings herein .
The polypeptides and polynucleotides of the present invention are preferably provided in an isolated form, and preferably are purified to homogeneity.
The term "isolated" means that the material is removed f rom its original environment ( e . g ., the natural environment if it is naturally occurring). For example, a naturally-occurring polynucleotide or polypeptide present in a living animal is not isolated, but the same polynucleotide or polypeptide, separated from some or all of the coexisting materials in the natural system, is isolated. Such polynucleotides could be part of a vector and/or such polynucleotides or polypeptides could be part of a composition, and still be isolated in that such vector or composition is not part of its natural environment.
The present invention also relates to vectors which include polynucleotides of the present invention, host _g _ SUESTITUTE SHEEt (RULE 261 WO 95131~67 219 0 4 6 6 PCrNS9~105384 cells which are genetically ~113i n~F-red with vectors o~ the invention and the productlon of ~~ polypeptldes o~ the invention by re~ ; nAnt techn-i~ue6 .
Host cells are genetically ~n~ine-ored (tr~n~ ed or transformed or transfected) with the vectors of this invention which may be, for example, a cloning vector or an expression vector. The vector may be, for example, in the form of a plasmid, a viral particle, a phage, etc. The engineered host cells can be cultured in conventional nutrient media 'if;ed as appropriate for activating promoters, selecting transformants or amplifying the MCP-4 genes. The culture conditions, such as temperature, pH and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.
The polynucleotides of the present invention may be employed for producing polypeptides by r~: ` i n~nt techniques. Thus, for example, the polynucleotide may be included in any one of a variety of expression vectors _or expressing a polypeptide. Such vectors include chromosomal, nonchL, s~ 1 and synthetic DNA sequences, e . g ., derivatives of SV40 ; bacterial plasmids ; phage DNA;
baculovirus; yeast plasmids; vectors derived f rom combinations of plasmids and phage DNA, viral DNA such as vaccinia, adenovirus, fowl pox virus, and pseudorabies.
However, any other vector may be used as long as it is replicable and viable in the host.
The appropriate DNA sequence may be inserted into the vector by a variety of procedures. In general, the DNA
sequence is inserted into an appropriate restriction endonuclease site(s) by procedures known in the art. Such procedures and others are deemed to be within the scope of those skilled in the art.
'rhe DNA sequence in the expression vector is operatively linked to an appropriate expression control 6equence~s) (promoter) to direct mRNA synthesis. As representative examples of such promoters, there may be mentioned: LTR or SV40 promoter, the E. CQli. lac or tr~, SIJBSTITUTE SHEET (RUL E 261 ~ WO95/31467 21 9 ~ ~ 6 6 PCIIUS94/05384 the phage lambda P~ promoter and other promoters known to control expression o~ gene6 in prokaryotic or eukaryotic cells or their viruses. The expression vector also contains a ribosome binding site for translation initiation and a transcription terminator. The vector may also include appropriate sequences for amplifying expres8ion.
In addition, the expression vectors preferably contain one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells such as dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, or such as tetracycline or ampi-cillin resistance in E. coli.
The vector containing the appropriate DNA sequence as hereinabove described, as well as an appropriate promoter or control sequence, may be employed to transform an appropriate host to permit the host to express the protein.
As representative examples of appropriate hosts, there may be mentioned: bacterial cells, such as E. coli, Streptomyces, Salmonella typh; ~ m: fungal cells, such as yeast; insect cells such as Drosophila and Sf9; animal cells such as CHO, COS or Bowes melanoma; plant cells, etc.
The selection of an appropriate host is deemed to be within the scope of those skilled in the art from the te~-h;n~c herein .
~ ore particularly, the present invention also includes recombinant constructs comprising one or more of the sequences as broadly described above. The constructs comprise a vector, such as a plasmid or viral vector, into which a 8equence of the invention has been inserted, in a forward or reverse orientation. In a preferred aspect of this embodiment, the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the sequence. Large numbers of suitable vectors and promoters are known to those of skill in the art, and are commercially available. The following vectors are provided by way of example. Bacterial: pQE70, pQE60, pQE-9 (Qiagen), pbs, pDlO, phagescript, psiXl74, pbluescript SR, SU8STITUTE SHEET ~RULE Z6~

WO95131467 ~ 4 ~ 6 PCrlllS94/05384 pb6ks, pNH8A, pNH16a! pNH18A,~ E~NH46A (Stratagene); ptrc99a, pKK223--3, pKK233--3, pDR54~, pRI~ (ph;~r~^~-ii9) . I~ukaryotic:
pWLNEO, pSV2CAT, pOG44, pXT1, pSG (Stratagene) pSVK3, pBPV, pMSG, pSVL ~Pharmacia). However, any other plasmid or vector may be used as long as they are replicable and viable in the host.
Promoter regions can be selected from any desired gene using CAT (chloL h~on;col transferase) vectors or other vectors with selectable markers. Two appropriate vectors are PKR232-8 and PCM7. Particular named bacterial promoters include lacI, lacZ, T3, T7, gpt, lambda PR~ PL and trp. Eukaryotic promoters include CMV immediate early, HSV
thymidine kinase, early and late SV40, LTRs from retrovirus, and mouse metallothionein-I. Selection of the appropriate vector ,Ind promoter is well within the level of ordinary skill in the art.
In a further P~lhQri;--nt~ the present invention relates to host cell6 containing the above-described constructs.
The host cell can be a higher eukaryotic cell, such as a mammalian cell, or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell. Introduction of the construct in~o the host cell can be effected by calcium phosphate transfection, DEAE-Dextran mediated transfection, or electroporation. (Davis, L., Dibner, M., Battey, I., Basic Methods in Molecular Biology, (1986) ) .
The constructs in host cells can be used in a conventional manner to produce the gene product encoded by the re~ ;n~nt gequence. Alternatively, the polypeptide8 of the invention can be synthetically produced by conventional peptide synthesizers.
Mature proteins can be expressed in mammalian cells, yeast, bacteria, or other cells under the control of appropriate promoters. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention.
Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrook, SUBSTITUTE SHEET (RULE 261 ~ Wo 95/314G7 21 9 8 ~ 6 ~ PCTIUS94/05384 et al., Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring ~arbor, N.~., (1989), the di6closure of which is hereby incorporated by reference.
Transcription of the DNA encoding the polypeptides of the present invention by higher eukaryotes is increased by inserting an PnhAnc~r sequence into the vector. Enhancers are cis-acting elements of DNA, usually about from 10 to 300 bp that act on a promoter to increase its transcription. Examples including the SV40 F~nhAnc~r on the late side of the replication origin bp 100 to 270, a cyt~ virus early promoter enhancer, the polyoma ~nhAnc~r on the late 5ide of the replication origin, and adenovirus enhancers.
Generally, recombinant expression vectors will include origins of replication and selectable markers permitting transformation of the host cell, e.g., the ampicillin resistance gene of E. coli and S. cerevisiae TRP1 gene, and a promoter derived from a highly-expressed gene to direct transcription of a downstream structural sequence. Such promoters can be derived from operons encoding glycolytic enzymes such as 3-phosphoglycerate kinase (PG~ factor, acid phosphatase, or heat shock proteins, among others.
The heterologous structural sequence is assembled in ~p~Lu~liate phase with tran61ation initiation and termination sequences, and preferably, a leader sequence capable of directing secretion of translated protein into the periplasmic space or extrAcel 1111Ar medium. Optionally, the heterologous sequence can encode a fusion protein including an N-tPrm;nAl identification peptide imparting desired characteristics, e.g., st~hil;7ation or simplified purif ication of expressed recombinant product .
Useful expression vectors for bacterial use are constructed by inserting a structural DNA sequence encoding a desired protein together with suitable translation initiation and termination signals in operable reading phase with a functional promoter. The vector will comprise one or more phenotypic selectable markers and an origin of replication to ensure maintenance of the vector and to, if SUBSTITUTE SHEET (RULE 26) Wo 95/31467 21 g ~ ~ 6 6 rcr/uss4/o~384 desirable, provide amplification within the host. Suitable prokaryotic hosts for transformation include ~. coli, 3acillus subtilis, Salmonella typhimurium and various species within the genera Pse~ ~ -c, Streptomyces, and Staphylococcus, although others may also be employed as iY
matter of choice.
As a representative but nonlimiting example, useful expression vectors for bacterial use can comprise a selectable marker and bacterial origin of replication derived from commercially available plasmids comprising genetic elements of the well known cloning vector psR322 (ATCC 37017). Such commercial vectors include, for example, pKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and GEM1 (Promega Biotec, Madison, WI, USA). These pBR322 "backbone" sections are ' in~d with an appropriate promoter and the structural sequence to be expressed.
Following transformation of a suitable host strain and growth of the host strain to an eppropriate cell density, the selected promoter is induced by appropriate means (e.g., temperature shift or chemical inductionl and cells are cultured f or an additional period .
Cells are typically harvested by centrifugation, disrupted by physical or chemical means, and the resulting crude extract retained for further purification.
Microbial cells employed in expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents, such methods are well know to those skilled in the art.
Various l; An cell culture systems can also be employed to express re~ ' inAnt protein. Examples of r l iAn expresgion systems include the COS-7 lines of monkey kidney fibroblasts, described by Gluzman, Cell, 23:175 (1981), and other cell lines capable of expressing a compatible vector, for example, the C127, 3T3, CH0, HeLa and BHK cell lines . Mi l; An expression vectors will comprise an origin of replication, a suitable promoter and enhancer, and also any necessary ribosome binding sites, ~UBSrl~UTE SHEET (RULE 26 219~66 o95l31467 PCrlUSs4l05384 polyadenylation site, splice donor and acceptor sites, transcriptional termination sequences, and 5~ ~lanking nontranscribed sequences. DNA sequences derived from the SV40 splice, and polyadenylation sites may be used to provide the required nontranscribed genetic elements.
The polypeptide can be lecuv~:Led and purified from reL ;nAnt cell cultures by method6 including ~ illm sulfate or ethanol precipitation, acid extraction, anion or cation f.Yrh~n~e chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroYylapatite chromatography and lectin chromatography . It is pref erred to have low concentrations tapproximately 0.15-5 mM) of calcium ion present during purification. (Price et al., J. Biol.
Chem., 244:917 (1969) ) . Protein refolding steps can be used, as necessary, in completing configuration of the mature protein. Finally, high performance liquid chromatography (HPLC~ can be employed for final purif ication steps .
The polypeptides of the present invention may be a naturally purified product, or a product of chemical synthetic ~Luce~uLes, or produced by re ;nAnt techniques from a prokaryotic or eukaryotic host (for example, by bacterial, yeast, higher plant, insect and -1 iAn cells in culture). r~ep~n-lin~ upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosylated. Polypeptides of the invention may also include an initial meth;nn;n~ amino ~cid residue.
The polypeptides of the present invention, in particular MCP-4, may be used for the promotion of wound healing. Since MCP-4 is a rh lc; nQ~ it is a chemo-attractant for leukocytes (such as monocytes, T
lymphocytes, basophils, etc.); therefore, it causes infiltration of target immune cells to a wound area.
The ~CP-4 polypeptides may also be used as an anti-tumor treatment and for treating localized complications of a malignancy, such as pleural effusions or ascites.

SUBSIITUTE SHEE~ (RULE

Wo 95131-167 2 1~ d ~ 6 ~ PCTIUS94/053X4 Instilling MCP-4 into the involved anatomic space can lead to local monocyte accumulation and activation.
The presence of MCPs in vivo is ac -nied by a local increase in the presence of eosinophils which have the distinctive function of killing the larvae of parasites that invade tissues, as in schistosomiasis, trichinosis and ascariasis. Therefore, MCP-4 may be used for combatting parasitic infections.
MCP--4 polypeptides may also play a role in the regulation of hematopoiesis, by regulating various hematopoietic progenitor cell activation and dif f erentiation .
The polypeptides may also be employed in accordance with the present invention by expression of such polypeptides in vivo, which is often referred to as "gene therapy . "
For example, cells from a patient may be F~n~i nP~red with a polynucleotide (DNA or RNA) encoding an MCP-4 polypeptide ex vivo, with the engineered cells then being provided to a patient to be treated with the polypeptide.
Such methods are well-known in the art. For example, cells may be Pr~i ne~ored by procedures known in the art by use of a retroviral particle containing RNA encoding a polypeptide of the present invention.
Similarly, cells may be engineered in vivo for expression of an MCP-4 polypeptide in vivo by, for example, procedures known in the art. As known in the art, a producer cell for producing a retroviral particle containing RNA encoding the polypeptide of the present invention may be administered to a patient for f~n~; nf~rring cells in vivo and eYpression of the polypeptide in vivo.
These and other methods for administering a polypeptide of the present invention by such method should be apparent to those skilled in the art from the ~Arh;n~ of the present invention. For example, the expression vehicle for ~n~inf.Pring cells may be other than a retrovirus, for eYample, an adenovirus which may be used to ~n~inf~r cells in vivo after combination with a suitable delivery vehicle.

SUBSrITUTE SHEET (RULE 26) ~ Wo 951314G7 21 g O ~ 6 6 PCrlUss4/os384 The polypeptides of the present invention may be employed in combination with a suitable rhArr~ eutical carrier. Such compositions comprise a therapeutically effectiv~ amount of the polypeptide, and a pharmaceutically acceptable carrier or excipient. Such a carrier includes but is not limited to aaline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof. The formulation should suit the mode of administration.
The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the rhArr--eutical compositions of the invention. Associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of rh_rr-~-euticals or biological products, which notice ref lects approval by the agency of manufacture, use or sale for human administration. In addition, the polypeptides of the pre6ent invention may be employed in conjunction with other therapeutic compounds.
The pharmaceutical compositions may be administered in a convenient manner such as by the topical, intr~venous, intraperitoneal, intramuscular, subcutaneous, intranasal or intradermal routes. ~CP-4 is administered in an amount which is effective for treating and/or prophylaxis of the specif ic indication . The amounts and dosage regimens of MCP-4 administered to a subject will depend on a number of factors such as the mode of administration, the nature of the condition being treated ~nd the judgment of the prescribing physician. In general, the ~CP-4 will be administered in an amount of at least about lO ~lg/kg body weight and in most cases they will be administered in an amount not in excess of about 8 mg/Kg body weight per day.
n most cases, the dosage is from about lO yg/kg to about l mg/kg body weight daily, taking into account the routes of administr~tion, symptoms, etc.
The sequences of the present invention are also valuable for chromosome identification. The sequence is specif ically targeted to and can hybridize with a SUBSTtTLlTE SHEET (RULE 261 o 95~31467 ;~ 0 4 ~ 6 PCT/US94/05384 particular location on an individual human chromosome.
I1o~ v~:r, there i6 ~' current need ~or identi~ying particular sites on the chl~ ~ ~. Few chromosome marking reagents based on actual sequence data ( repeat polymorphisms ) are presently available for marking chromosomal location. The mapping of DNAs to chL~
according to the present invention is an important f irst step in correlating those sequences with genes associated with disease.
Briefly, sequences can be mapped to chL~ - - by preparing PCR primers (preferably l5-25 bp) from the cDNA.
Computer analysis o~ the cDNA is used to rapidly select primers that do not span more than one exon in the genomic DNA, thus complicating the amplification process. These primers are then used for PCR screening of somatic cell hybrids containing individual human ~IIL I { ~ - . Only those hybrids containing the human gene corresponding to the primer will yield an amplified fragment.
PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular DNA to a particular chromosome. Using the present invention with the same oligonucleotide primers, sublocalization can ~e achieved with panels of fragments from specific chL~ s~ -, or pools of large genomic clQnes in an analogous manner. Other mapping strategies thAt can similarly be used to map to its ch~ ~ - include ill situ hybridization, prescreening with labeled flow-sorted Cl1L~ _ ~ and preselection by hybridization to construct chromosome specif ic-cDNA
lib}aries .
Fluorescence in sltu hybridization (FISH) of a cDNA
clones to a metaphase chL, - 1 spread can be used to provide a precise chromosomal location in one step. This technique can be used with cDNA as short as 500 or 600 bases; however, clones larger than 2,000 bp have a higher 1 ;k~l ihos~d of binding to a unique chll 6~ '1 location with sufficient signal intensity for simple detection. FISH
requires use of the clones from which the EST was derived, and the longer the better. For example, 2, 000 bp is good, SUBS11TUTE SHEET (RUI~ 26 W095/314G7 f ~ PCrlUS94/05384
4,000 is better, and more than 4,000 i8 probably not neces6ary to get good results a reasonable percentage of the time. For a review of this technique, see Verma et al., Human Chromosomes: a Manual of Basic Techniques, Pergamon Press, New York ( I988 ) .
Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be correlated with genetic map data.
Such data are found, for example, in V. McKusick, M~nc~ n Inheritance in Man ~available on line through Johns Hopkins University Welch Medical Library). ~he relationship between genes and diseases that have been mapped to the same chromosomal region are then identified through linkage analysis ( coinheritance of physically adjacent genes ) .
Next, it is necessary to determine the differences in the cDNA or genomic sequence between af f ected and unaffected individuals. If a mutation is observed in some or all of the affected individuals but not in any normal individuals, then the mutation is likely to be the causative agent of the disease.
With current resolution of physical mapping and genetic mapping techniques, a cDNA precisely localized to a chromosomal region associated with the disease could be one of between 50 and 500 potential causative genes. tThis assumes 1 megabase mapping resolution and one gene per 20 kb) .
Comparison of affected and unaffected individuals generally involves first looking for structural alterations in the chromosomes, such as deletions or translocations that are visible from ch~ - spreads or detectable using PCR based on that cDNA sequence. Ultimately, complete sequencing of genes from several individuals is required to conf irm the presence of a mutation and to dist;n~li~h mutations from polymorphisms.
The polypeptides, their f ragments or other derivatives, or analogs thereof, or cells expressing them can be used as an; ~, ~~ to produce antibodies thereto.
These antibodies can be, for example, polyclonal or SUSSTITUTE SHEET ~RULE 261 Wo 9S131467 21~ 0 4 6 6 PCrNS94/OS384 monoclonal an~;ho~ s. The presen~imYention also includes chimeric, single chain, and h~ n;7ed antibodie6, as well Fab fragments, or the product of an Fab expression library. Various procedures known in the art may be used for the production of such antibodies and fragments.
Antibodies generated against the polypeptides corresponding to a sequence of the present invention can be obtained by direct injection of the polypeptides into an ~nimal or by administering the polypeptides to an animal, preferably a nonhuman. The antibody so obtained will then bind the polypeptides itself. In this manner, even a sequence Pncntl;n~ only a fragment of the polypeptides can be used to generate antibodies binding the whole native polypeptide6. Such antibodies can then be used to isolate the polypeptide from tissue expressing that polypeptide.
For preparation of monoclonal antibodies, any technique which provides antibodies produced by continuous cell line cultures can be used. Examples include the hybridoma technique (Rohler and Milstein, 1975, Nature, 256:495-497), the trioma technique, the human B-cell hybridoma technique (Kozbor et al., 1983, Immunology Today 4: 72 ), and the EBV-hybridoma technique to produce human monoclonal an~;ho~l;ec (Cole, et al., 1985, in Monoclonal An~; ho~ and Cancer Therapy, Alan R. Liss, Inc., pp . 77-96) .
Techniques described for the production of single chain antibodies (U.S. Patent 4,946,778) can be adapted to produce single chain antibodies to immunogenic polypeptide products of this invention.
The present invention also relates to a diagnostic assay for detectin~ the level of MCP-4 both quantitatively and qualitatively. Such assays are well known in the art and include an ELISA assay and the radio; ~ say. The levels of MCP-4 detected in the assay can be useful for the elucidation of the 6ignificance of MCP-4 in various diseases and for the diagnosis of disea8es in which MCP-4 may play a role.

SUBSTITUTE SHEET (RULE 26 ~ WO 9~/3l467 2 ~ ~ 4 ~6 PCT/US94/05384 This invention provides a method for identification of MCP--4 receptor6 . The gene ~n--o~l i n~ an MCP--4 receptor can be identified by expression cloning. Briefly, polyadenylated RNA i6 prepared from a cell responsive to MCP-4 and a cDNA library created from this RNA is divided into pools and used to transfect COS cells or other cells that are not responsive to MCP-4. Transfected cells which are grown on glass slides are exposed to labeled MCP-4.
MCP-4 can be labeled by a variety of means including iodidation or inclusion of a recognition site for a site-specif ic protein kinase . Following f ixation and incubation, the slides are subjected to autoradiographic analysis. Positive pools are identified and sub-pools are prepared and retransfected using an iterative sub-pooling and rescreening process, eventually yielding a single clone that encodes the putative receptor. As an alternative approach for receptor identification, labeled MCP-4 can be photoaffinity linked with cell membrane or extract preparations that express an MCP-4 receptor molecule.
Cross-linked material is resolved by PAGB and exposed to x-ray film. The labeled complex containing the MCP-4 receptor can be excised, resolved into peptide fragments, and subjected to protein microsequencing. The amino acid sequence obtained from microsequencing would be used to design a set of generate oligonucleotide probes to screen a cDNA library to identify a gene encoding the putative receptor.
This invention also provides a method of screening drugs to identify those which enhance (agonists) or block ( antagonists ) interaction of MCP-4 to its receptor . An agonist increases the biological fllncf it~nl: of MCP-4, while an antagonist reduces or eliminates such f unctions . As an example, a l; Rn cell or membrane preparation expressing an MCP-4 receptor would be incubated with labeled MCP-4 in the presence of drug. The ability of drug to enhance or block this interaction could then be measured. Alternatively, the response of a known second messenger system following interaction of MCP-4 and it8 receptor would be measured compared in the presence or SUBSTITIJT~ SHEET (RULE 26) Wo 95/31467 ~ 6 ~ PCT/US94/0538 absence of drug. Such second messenger systems include but ~re not limited to, cAMP guanylate cyclase, ion rhAnnt~l c or phosphoinositide hydrolysis.
The present invention is also directed to antagonist/inhibitor molecules to the polypeptides of the present invention. Antagonists include negative dominant mutants of MCP-4. MCP-4 is a tetrameric polypeptide wherein one mutated unit will cause the entire polypeptide to be non-functional. A negetive dominant mutant of MCP-4 binds to the MCP-4 receptor but fails to activate cells ( leukocytes and monocytes ) to which it binds . An assay to detect negative dominant mutants of MCP-4 is an in vitro chemotaxis assay wherein a multiwell chemotaxis chamber equipped with polyvinylpyrrolidone-free polycarbonate membranes is used to measure the chemoattractant ~bility of MCP-4 for leukocytes in the presence ~nd absence of potential antagonist/inhibitor or agonist molecules.
An example of an inhibitor is an antisense DNA or RNA
construct. Antisense technology can be used to control gene expression throuy-h triple-helix formation or antisense DNA or RNA, both of which methods Are based on binding of a polynucleotide to DNA or RNA. For example, the 5 ' coding portion of the polynucleotide sequence, which encodes for the mature polypeptides of the present invention, is used to design an antisense RNA oligonucleotide of from about 10 to 40 base pairs in length. A DNA ol; ~t~n~ leotide is designed to be complementary to a region of the gene involved in transcription (triple helix -see Lee et al., Nucl. Acids Res., 6:3073 (1979); Cooney et al, Science, 241:456 (1988); and Dervan et al., Science, 251: 1360 (1991~ ), thereby preventing transcription and the production of MCP-~. The antisense RNA oligonucleotide hybridizes to the mRNA i~ vivo and blocks translation of the mRNA molecule into the MCP-4 (antisense - Okano, J.
Neurochem., 56:560 (l991~; Oliyudev,~yllucleotides as Antisense Inhibitors of Gene Expression, CRC Press, Boca Raton, FL ( 1988 ) ) . Alternatively, antisense RNA and DNA

SU8~TITUTE ~HEET (RULE 26 Wo 95131467 21~ ~ 4 6 6 PCT/U~94/OS384 may be delivered to cell6 such that they are expressed i~
vivo to inhibit production of MCP-4.
Another example of an antagonist is a peptide derivative of MCP-4 which are naturally or synthetically modif ied analogs of MCP-4 that h~ve lost biological function yet still recognize and bind to receptors thereby effectively blocking the receptors.
The antagonist/inhibitors may be used to treat inf lammation by preventing the attraction of monocytes to a wound or a site of trauma, and to regulate normal plll Ary macrophage populations, since acute and chronic inflammatory p~ ry diseases are associated with sequestration of mononuclear phagocytes in the lung. They may also be used to treat rheumatoid arthritis, since MCP
levels were found to be significantly elevated in synovial f luid f rom rheumatoid arthritis patients which suggests that synovial production of MCP attracts monocytes whose influx and activation are important in the pathogenesis of both degenerative and inf lammatory arthropathies .
The antagonist/inhibitors may also be used for treating atherosclerosis, since MCPs mediate monocyte infiltration in the artery wall which infiltration leads to atherosclerosis, and to prevent allergies, since it has been shown that MCPs directly induce histamine release by basophi 18 .
Antagonist/inhibitors may also be used to treat infectious diseases such as tuberculosis, since tuberculosis targets cells, usually monocytes, causing the monocytes to release MCPs which attracts more monocytes to the lungs causing severe ; nf l; tion . The antagonist/inhibitors may be employed in a composition with a pharmaceutically acceptable carrier, e.g., as hereinabove described .
The present invention also relates to an assay for identifying potential antagonist/inhibitors specific to MCP-4 . An example of such an assay ~ i n~8 MCP-4 and a potential antagonist/inhibitor with membrane-bound MCP-4 receptors or recombinant MCP-4 receptors under ~ u~iate S118STITUTE SHEET (RULE 2~) W0 9~/31467 ~ PCTIUS94/0~384 condition6 for a competitive inhibition assay. MCP-4 can be la`oeled, such as by radioactivity, such that the number of MCP-4 molecules bound to the receptor can determine the ef f ectiveness of the potential antagonist/inhibitor .
The present invention will be further described with reference to the following ~ ; however, it is to be understood that the present invention is not limited to such examples. All parts or amounts, unless otherwise specified, are by weight.
In order to facilitate understanding of the following examples certain frequently occurring methods and/or terms will be described.
"Plasmids" are designated by a lower case p preceded ~nd/or f ollowed by capit~l letters ~nd/or numbers . The starting plasmids herein are either commercially available, publicly available on an unrestricted basis, or can be constructed from available pl~cm; rl~ in accord with published procedures. In addition, equivalent plasmids to those described are known in the art and will be apparent to the ordinarily skilled artisan.
"Digestion" of DNA refers to catalytic cleavage of the DNA with a restriction enzyme that acts only at certain sequences in the DNA. The various restriction enzymes used herein are commercially available and their reaction conditions, cofactors and other requirements were used as would be known to the ordinarily skilled artisan. For ~nalytical purposes, typically 1 yg of plasmid or DNA
fragment is used with about 2 units of enzyme in about 20 yl of buffer solution. For the purpose of isolating DNA
fragments for plasmid construction, typically 5 to 50 ~g of D~A are digested with 20 to 250 units of enzyme in a larger volume. Appropriate buffers and substrate amounts for particular restriction enzymes are specified by the manufacturer. Incubation times of about 1 hour at 37 C are ordinarily used, but may vary in accordance with the supplier~s instructions. After digestion the reaction is directly on a polyacrylamide gel to isolate345Xelectr the desired fragment.

SUBâTITUTE SHEET (RULE 26 W095l31467 ~ 5 Pcrlu~94105384 Size separation of the cleaved fragments is performed u~ing 8 percent polyacrylamide gel described by Goeddel, D.
et ql., Nucleic Acids Re6., 8:4057 (1980).
~ oligonucleotides~ refers to either a single stranded polydeoxynucleotide or two complementary polydeoxynucleotide strands which may be chemically synthesized. Such synthetic ol; gonllcleotides have no 5 ' phosphate and thus will not ligate to another ol ;gonllr~eotide without ~Idding a phosphate with an ATP in the presence of a kinase. A synthetic oligonucleotide will ligate to a fragment that has not been dephosphorylated.
"Ligation" refers to the process of forming phosphodiester bonds between two double stranded nucleic acid fragments (Maniatis, T., et al., Id., p. 146). Unless otherwise provided, ligation may be accomplished using known buffers and conditions with 10 units to T4 DNA ligase ( ligase ) per 0.5 ,ug of approximately equimolar amounts of the DNA f ragments to be ligated .
Unless otherwise stated, transformation was performed as described in the method of Graham, F. and Van der Eb, A., Virology, 52:456-457 (1973).
r le 1 Bacterial E~ression and p1lrification of MCP-4 The DNA sequence ~ncn~i;n~ for MCP-4, ATCC # 75703, is initially amplified using PCR oligonucleotide primers corresponding to the 5 ~ and 3 ' sequences of the processed MCP-4 protein (minus the signal peptide sequence) and the vector sequences 3 ~ to the MCP-4 gene. Additional nucleotides ~:u~ Le,,~ollding to MCP-4 were added to the 5 ' and 3 ' sequences respectively. The 5 ' oligonucleotide primer has the sequence 5 '-TCAGGATCCCCTACGGG~ L~ 3 ' contains a Bam H1 restriction enzyme site followed by 18 nucleotides of MCP-4 coding sequence starting f rom the presumed t~rm;nAl amino acid of the processed protein codon. The 3' sequence 3 '-CGCTCTAGAG~ rr-~rGGCCAGT-5 ' contains complementary sequences to the XbaI site and to a pBluescript SK- vector sequence located 3' to the MCP-4 DNA

SU8~TITUTE SHEET (RULE 261 4~
Wo 9~/31467 ~ PCr/Uss4/05384 insert. The restriction enzyme sites correspond to the restriction enzyme sites on th~ bacterial expression vector pQE-9. (Qiagen, Inc. 9259 Eton Avenue, Chatsworth, CA, 91311 ) . pQE-9 encodes antibiotic resistance (Ampr), a bacterial origin of replication ( ori ), an IPTG-regulatable promoter operator (P/O), a ribosome binding site (RBS), a 6-His tag and restriction enzyme site6. pQE-9 was then digested with Bam H1 and Xba I . The amplif ied sequences were ligated into pQE-9 and were inserted in frame with the 3equence encoding for the histidine tag and the RBS.
~igure 5 shows a schematic representation of this arrangement. The ligation mixture was then used to transform E. coli strain ml5/rep4 available from Qiagen under the trademark M15/rep 4 by the procedure described in Sambrook, J. et al, Molecular Cloning: A Laboratory Manual, Cold Spring Laboretory Press, 1989. M15/rep4 contains multiple ~copies of the plasmid pREP4, which expresses the lacI repressor and also confers kanamycin resistance (Ranr). Transformants are ;rl.ontif;~d by their ~bility to grow on LB plates and ampicillin/kanamycin resistant colonies were selected. Plasmid DNA was isolated and confirmed by restriction analysis. Clones ront~in;n~
the desired constructs were grown overnight (O/N) in liquid culture in LB media supplemented with both Amp (100 ug/ml) and Kan (25 ug/ml). The O/N culture is used to inoculate a large culture at a ratio of 1:100 to 1: 250 .
The cells were grown to an optical density 600 (O.D.a~) of between 0 . 4 and 0 . 6 . IPTG ( " Isopropyl-B-D-thiogalacto pyranoside" ) was then added to a f inal concentration of mM. IPTG induces by inactivating the lacI repressor, clearing the P/O leading to increased gene expression.
Cells were grown an extra 3 to 4 hours. Cells were then harvested by centrifugation. The cell pellet was solllhil;7r~rl in the chaotropic agent 6 Molar G1l~n;rl;nF- HCl.
After clarification, solubilized MCP-4 was purified from this solution by chromatography on a Nickel-Chelate column under conditions that allow for tight binding by proteins containing the 6-His tag . Hochuli , E . et al ., J .

SU9STITUTE SHEET (RULE 261 21~0~6~
WO 95131467 PCrlUS94/05384 Chromatography 411:177-184 (1984). ~CP-4 (954 pure) was eluted from the column in 6 molar guF~ni~7in~ HC1 pH 5.0 and for the purpose of renaturation adjusted to 3 molar g~,~n;.7inP HCl, lOOmM godium phosphate, 10 mmolar . glutathione (reduced) and 2 mmolar glutathione (oxidized).
After incubation in this solution for 12 hours the protein was dialyzed to 10 mmolar sodium phosphate. Figure 4.
Example 2 rnreS8ion Pattern of MCP-4 in human cells Northern blot analysis was carried out to examine the levels of expression of MCP-4 in human cells. Total cellular RNA samples were isolated with RNAzol~ B system (Biotecx Laboratories, Inc. 6023 South Loop East, Hou6ton, TX 77033 ) . About lO~Lg of total RNA isolated from each human tissue spec; f i f-d was separated on 1% agarose gel and blotted onto a nylon filter. (Sambrook, Fritsch, and Maniatis, Nolecular Cloning, Cold Spring Harbor Press, ( 1989 ) ) . The 7 ~h~l i n~ reaction was done according to the Stratagene Prime-It kit with 50ng DNA fragment. The labeled DNA was purified with a Select-G-50 column. (5 Prime - 3 Prime, Inc. 5603 Arapahoe Road, Boulder, C0 80303 ) . The filter was then hybridized with radioactive labeled full length MCP-4 gene at l,000,000 cpm/ml in 0.5 M NaP0~, pH 7.4 and 796 SDS overnight at 65 C. After wash twice at room temperature and twice at 60 C with 0.5 x SSC, 0.19~ SDS, the filter was then exposed at -70 C overnight with an intensifying screen. The message RNA for MCP-4 is abundant in activated and unactivated T cells, monocytes and T cell lines. Figure 3.
Numerous modifications and variations of the present invention are possible in light of the above t~arhin~ and, therefore, within the scope of the appended claims, the invention may be practiced otherwise than as particularly described .

SU~STITUTE SHEET (RULE 26) WO 95131467 219 ~ 4 6 ~ PCTIUS94/05384 ~, ~t;UU~ ; LISTING
( 1 ) GENERAL INFORMATION:
( i ) APPLICANT: LI, ET AL .
(ii) TITLE OF INVENTION: Monocyte Chemotactic Protein-4 ( iii ) NUMBER OF ~;uu~ ;S: 2 (iv) C:u~h'~ u~ ; ADDRESS:
(A) ADDRESSEE: CARELLA, BYRNE, BAIN, GILFILLAN, CECCHI, STEWART & OLSTEIN
( B ) STREET: 6 BECRER FARM ROAD
(C) CITY: Rn~T.PlNn ( D ) STATE: NEW ~ERSEY
( E ) COUNTRY: USA
(F) ZIP: 07068 (v) C~ ~ R~n~RTT FORM:
(A) MEDIUM TYPE: 3 . 5 INCH DISRETTE
( B ) Cu.~u ~: IBM PS/2 (C) OPERATING SYSTEM: MS--DOS
( D ) SOFTWARE: WORD PERFECT 5 .1 (vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER:
(B) FILING DATE: Submitted herewith (C) CLASSIFICATION:
(vii) PRIOR APPLICATION DATA
(A) APPLICATION NUMBER:
(B) FILING DATE:
(viii) ATTORNEY/AGENT INFORMATION:
( A ) NAME: FERRARO, GREGORY D .
(B) REGISTRATION NUMBER: 36,134 ( C ) k~ u~;/DOCRET NUMBER: 3 2 5 8 0 0 -16 0 (iX) T~T.~'nMMTTNICATION INFORMATION:
(A) TELEPHONE: 201--994--1700 (B) TELEFAX: 201--994--1744 ( 2 ) INFORMATION FOR SEQ ID NO :1:
;uu~;~u~; CHARACTERISTICS
(A) LENGTH: 360 BASE PAIRS

SUBS~lhrrE SHEET ~RULE 261 . `~ 21~4~
6EQU~NCE LISTING
( 1 ) GENERAL INFORMAlmION:
(i~ APPLICANT: numan Genome Science8, Inc.
9410 Key We~t Avenue Rockville, MD 20850 tJnited States of America APPLICANTS/INVENTORS: Li, Haodong Ruben, Steven M.
Sutton III, Granger G.
(ii) TITLE OF INVENTION: Monocyte Chemctactic Protein-4 (iii) NUMBER OF SEQUENCES: 5 (iV) mUKltl:;:~!'UN~r;N~ ADDRESS:
(A) ~nnR~qq~ Sterne, Ressler, Gcldstein & Fox, P.L.L.C.
(B) STREET: 1100 New York Ave, N.W., Suite 600 (C) CI~Y: Washington (D) STATE: D.C.
(E) COUNTRY: United States of America (F) ZIP: 20005-3934 (v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk (B) COMPUTER: IBM PC hl~
(C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: PatentIn Relea~e #1.0, Ver~ion #1.30 (vi) CURRENT APPLICATION DATA:
(A) APPLICATION NOMBER: PCT/US94/05384 (B) FILING DAT~: 16-MAY-1994 (C) CLASSIFICATION:
(viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: Gold~tein, Jorge A.
(B) REGISTRATION NUMBER: 29,021 (C) REFERENCE/DOCRET N[lMB~R: 1488.034PC00 (ix) TEL~i~._. mT~N INFoRMATIoN:
(A) TELEPHONE: 202-371-2600 (B) TEL13FAX: 202-371-2540 (2) INFORMATION FOR SEQ ID NO:1:
(i) Si;:QUENC~ R~-~TFRT.qTICS:
(A) LENGTH: 360 ba~e pairs (B) TYPE: nucleic acid (C) ~ ingle (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
,~ t (ix) FEAT~RE:
(A) NAME/REY: CDS
(B) LOCATION: 1..357 ( ix ) FBATURE:
(A~ NAME/RBY: Dig_peptide (B) LOCATION: 1..66 ( ix) FEATURE:
(A) NAME/KEY: mat_peptide (B) LOCATION: 67..357 (xi) SEQUBNCE DESCRIPTION: SEQ ID NO:1:

Met Ala Gly Leu Met Thr Ile Val Thr Ser Leu Leu Phe Leu Gly Val TGT GCC CAC CAC ATC ATC CCT ACG GGC TCT GTG GTC ATA CCC TCT CCC 96 _~
Cy3 Ala His His Ile Ile Pro Thr Gly Ser Val Val Ile Pro Ser Pro
-5 1 5 10 TGC TGC ATG TTC TTT GTT TCC A~G AGA ATT CCT GAG AAC CGA GTG GTC 144 Cys Cys Met Phe Phe Val Ser Lys Arg Ile Pro Glu Asn Arg Val Val Ser Tyr Gln Leu Ser Ser Arg Ser Thr Cys Leu Lys Gly Gly Val Ile TTC ACC ACC AAG AAG GGC CAG C~G TTC TGT GGC GAC CCC AAG CAG GAG 240 Phe Thr Thr Ly3 Lys Gly Gln Gln Phe Cys Gly Asp Pro Lys Gln Glu TGG GTC CAG AGG TAC ATG APG AAC CTG GAC GCC A~G CAG AAG AAG GCT 288 Trp Val Gln Arg Tyr Met Lys Asn Leu Asp Ala Lys Gln Lys Lys Ala TCC CCT AGG ~CC AGG GCA GTG GCT GTC APG GGC CCT GTC C~G AGA TAT 336 Ser Pro Arg Ala Arg Ala Val Ala Val Lys Gly Pro Val Gln Arg Tyr 75 80 85 9o Pro Gly Asn Gln Thr Thr Cys (2) INFORMATION FOR SEQ ID NO:2:
(i) SEQUENCE ~Tl~T:~'T~17T~:TICS:
(A) LENGTH: 119 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear . 219~fil~
(ii) MOLECULE TYPE: protein (xi) SEQVENCE ~ LUI~: SEQ ID NO:2:
Met Ala Gly Leu Met Thr Ile Val Thr Ser Leu Leu Phe Leu Gly Val Cys Ala Elis E~is Ile Ile Pro Thr Gly Ser Val Val Ile Pro Ser Pro ys Cya Met Phe Phe Val Ser Lys Arg Ile Pro Glu Asn Arg Val val er Tyr Gln Leu Ser Ser Arg Ser Thr Cys Leu Lys Gly Gly Val Ile he Thr Thr Lys Lys Gly Gln Gln Phe Cys Gly Asp Pro Lys Gln Glu Trp Val Gln Arg Tyr Met Lys Asn Leu Asp Ala Lys Gln Lys Lys Ala Ser Pro Arg Ala Arg Ala Val Ala Val Lys Gly Pro Val Gln Arg Tyr ro Gly Asn Gln Thr Thr Cys 2) INFORMATION FO~ SEQ ID NO:3:
(i) SEQUENCE ~7~ 'TR~TCTICS:
(A) LENGTEI: 148 amino acids (B) TYPE: amino acid (C) ST~NnRnNRq~: not relevant (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide (xi) 8EQUENC_ DESCRIPTION: SEQ ID NO:3:
Met Gln Val Pro Val Met Leu Leu Gly Leu Leu Phe Thr Val Ala Gly Trp Ser Ile E~i~ Val Leu Ala Gln Pro Asp Ala Val Asn Ala Pro Leu 20 2s 30 Thr Cys Cys Tyr Ser Phe Thr Ser Lys Met Ile Pro Met Ser Arg Leu Glu Ser Tyr Lys Arg Ile Thr Ser Ser Arg Cys Pro Lys Glu Ala Val so ss 60 =
~/

. ,,, 21g~466 Val Phe val Thr Lys Leu Lys Arg Glu Val Cy~ Ala Asp Pro Lys Lys Glu Trp Val Gln Thr Tyr Ile Lys Asn Leu Asp Arg Asn Gln Met Arg Ser Glu Pro Thr Thr Leu Phe Lys Thr Ala Ser Ala Leu Arg Ser Ser Ala Pro Leu Asn Val Lys Leu Thr Arg I.ys Ser Glu Ala Asn Ala Ser Thr Thr Phe Ser Thr Thr Thr Ser Ser Thr Ser Val Gly Val Thr Ser Val Thr Val A~n (2) INFORMATION FOR SEQ ID NO:4:
(i) SEQUENCE ~T~R~rT~Rr.qTICS
(A) LENGT~: 26 ba~e pairs (B) TYPE: nucleic acid (C) STR~ nN~qq: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: c~NA
(xi~ SEQUENCE DESCRIPTION: SEQ ID NO:4:

(2~ INFORMATION FOR SEQ ID NO:5:
(i~ SEQUENCE ~R~rT~RT~TIc8 (~ LENGTEI: 26 3~ase pairs (B~ TYPE: nucleic acid ( C ~ STR 7~ ~: 8 ingle (D~ TOPOLOGY: linear (ii~ MOL.ECUI,E TYPE: cDNA
(xi~ SEQI~ENCE DESCRIPTION: 8EQ ID NO:5:
CGCTCTAGAG TZ~ rr.~rt~ GCCAGT 26 0~ 2/

Claims (22)

WHAT IS CLAIMED IS:
1. An isolated polynucleotide selected from the group consisting of:
(a) a polynucleotide encoding an MCP-4 polypeptide having the deduced amino acid sequence of Figure 1 or a fragment, analog or derivative of said polypeptide;
(b) a polynucleotide encoding an MCP-4 polypeptide having the amino acid sequence encoded by the cDNA contained in ATCC Deposit No. 75703 or a fragment, analog or derivative of said polypeptide.
2. The polynucleotide of Claim 1 wherein the polynucleotide is DNA.
3. The polynucleotide of Claim 1 wherein the polynucleotide is DNA.
4. The polynucleotide of Claim 1 wherein the polynucleotide is genomic DNA.
5. The polynucleotide of Claim 2 wherein said polynucleotide encodes MCP-4 having the deduced amino acid sequence of Figure 1.
6. The polynucleotide of Claim 2 wherein said polynucleotide encodes the MCP-4 polypeptide encoded by the cDNA of ATCC Deposit No. 75703.
7. The polynucleotide of Claim 1 having the coding sequence of NCP-4 as shown in Figure 1.
8. The polynucleotide of Claim 2 having the coding sequence of NCP-4 deposited as ATCC Deposit No. 75703.
9. A vector containing the DNA of Claim 2.
10. A host cell genetically engineered with the vector of Claim 9.
11. A process for producing a polypeptide comprising: expressing from the host cell of Claim 10 the polypeptide encoded by said DNA.
12. A process for producing cells capable of expressing a polypeptide comprising genetically engineering cells with the vector of Claim 9.
13. An isolated DNA hybridizable to the DNA of Claim 2 and encoding a polypeptide having MCP-4 activity.
14. A polypeptide selected from the group consisting of (i) an MCP-4 polypeptide having the deduced amino acid sequence of Figure 1 and fragments, analogs and derivatives thereof and (ii) an MCP-4 polypeptide encoded by the cDNA of ATCC Deposit No. 75703 and fragments, analogs and derivatives of said polypeptide.
15. The polypeptide of Claim 14 wherein the polypeptide is MCP-4 having the deduced amino acid sequence of Figure 1.
16. An antibody against the polypeptide of claim 14.
17. An agonist to the polypeptide of claim 14.
18. An antagonist/inhibitor against the polypeptide of claim 14.
19. A method for the treatment of a patient having need of MCP-4 comprising: administering to the patient a therapeutically effective amount of the polypeptide of claim 14.
20. A method for the treatment of a patient having need to inhibit MCP-4 comprising: administering to the patient a therapeutically effective amount of the antagonist/inhibitor of Claim 18.
21. A pharmaceutical composition comprising the polypeptide of Claim 14 and a pharmaceutically acceptable carrier.
22. The method of Claim 19 wherein said therapeutically effective amount of the polypeptide is administered by providing to the patient DNA encoding said polypeptide and expressing said polypeptide in vivo.
CA002190466A 1994-05-16 1994-05-16 Monocyte chemotactic protein-4 Abandoned CA2190466A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CA002190466A CA2190466A1 (en) 1994-05-16 1994-05-16 Monocyte chemotactic protein-4

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CA002190466A CA2190466A1 (en) 1994-05-16 1994-05-16 Monocyte chemotactic protein-4

Publications (1)

Publication Number Publication Date
CA2190466A1 true CA2190466A1 (en) 1995-11-23

Family

ID=4159262

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002190466A Abandoned CA2190466A1 (en) 1994-05-16 1994-05-16 Monocyte chemotactic protein-4

Country Status (1)

Country Link
CA (1) CA2190466A1 (en)

Similar Documents

Publication Publication Date Title
EP0735818B1 (en) MACROPHAGE INFLAMMATORY PROTEINS MIP-3, MIP-4 AND MIP-1gamma
US5504003A (en) Macrophage inflammatory protein-3 and -4
US7198944B2 (en) Human chemokine β-9 viral vectors
AU708903B2 (en) Human chemokine polypeptides
AU723891B2 (en) Human chemokine beta-11 and human chemokine alpha-1
US6458349B1 (en) Chemokine β-4 polypeptides
EP0767796B1 (en) Chemotactic protein
EP0832233B1 (en) Human chemokine beta-13
EP0811059A1 (en) Human chemokine beta-11 and human chemokine alpha-1
US5958400A (en) Interleukin-6 splice variant
WO1998011138A1 (en) Chemokine alpha-4
US5866373A (en) Polynucleotide encoding a human chemotactic protein
WO1996040762A9 (en) Monocyte chemotactic protein-4
CA2190466A1 (en) Monocyte chemotactic protein-4
EP1125948A1 (en) Monocyte chemotactic protein-4
EP0777494B1 (en) Human chemokine polypeptides
EP1006188B1 (en) Human chemokine polypeptides
AU753088B2 (en) Human chemokine polypeptides
AU753730B2 (en) Human chemokine beta-13
AU767527B2 (en) Human chemokine polypeptides
CA2215383A1 (en) Human tumor necrosis factor receptor
AU4507900A (en) Human chemokine beta-11 and human chemokine alpha-1

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued