WO1998014573A1 - Mcp-4 and mcp-5: novel chemokines - Google Patents

Mcp-4 and mcp-5: novel chemokines Download PDF

Info

Publication number
WO1998014573A1
WO1998014573A1 PCT/US1997/017900 US9717900W WO9814573A1 WO 1998014573 A1 WO1998014573 A1 WO 1998014573A1 US 9717900 W US9717900 W US 9717900W WO 9814573 A1 WO9814573 A1 WO 9814573A1
Authority
WO
WIPO (PCT)
Prior art keywords
lys
leu
ala
ser
thr
Prior art date
Application number
PCT/US1997/017900
Other languages
French (fr)
Other versions
WO1998014573A9 (en
Inventor
Andrew D. Luster
Eduardo Garcia-Zepeda
Mindy Sarafi
Original Assignee
The General Hospital 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
Application filed by The General Hospital Corporation filed Critical The General Hospital Corporation
Publication of WO1998014573A1 publication Critical patent/WO1998014573A1/en
Publication of WO1998014573A9 publication Critical patent/WO1998014573A9/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7158Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for chemokines
    • 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
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • C12N2799/026Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from a baculovirus
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Cell Biology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

Substantially pure nucleic acid molecules encoding the monocyte chemotactic proteins MCP-4 and MCP-5. These molecules and the polypeptides they encode are useful in treating diseases or conditions that: (1) are exacerbated by a local immune response, (2) would benefit from a local immune response, or (3) are caused by infectious agents that gain entry to mammalian cells via the chemokine receptors bound by MCP-4 or MCP-5.

Description

MCP-4 AND MCP-5: NOVEL CHEMOKINES Background of the Invention The invention relates to novel chemoattractants.
Six different types of white blood cells (leukocytes) are typically found in the blood. These are neutrophils, eosinophils, basophils, monocytes, lymphocytes, and plasma cells. The neutrophils and monocytes are primarily responsible for attacking and destroying invading bacteria, viruses, and other harmful agents. Neutrophils circulate within the bloodstream as mature, functional cells.
Monocytes, however, circulate as immature cells that have a limited ability to fight infectious agents. It is only when monocytes are stimulated by chemotactic agents to move through the capillary wall into surrounding tissue that they become fully active. Once monocytes enter the tissues they begin to swell and many lysosomes and mitochondria appear in their cytoplasm. At this point, monocytes become called macrophages, which are extremely effective phagocytes. Each macrophage can engulf as many as 100 bacterial cells, as well as large particles, including whole red blood cells, malarial parasites, and necrotic tissue.
Chemokines are chemotactic cytokines that contribute to various immune and inflammatory responses by regulating the movement of selected blood-borne leukocytes into the tissues (Baggiolini et al., Adv. Immunol. 55:97-179, 1994; Oppenheim et al., Ann. Rev. Immunol. 9:617-648, 1991). All known chemokines have been assigned to one of three families on the basis of the chromosomal location of the genes that encode them and on the motif formed by conserved cysteine residues in the mature proteins. Two of these families, designated and β, have many members, all of which have four conserved cysteine residues. The first two cysteines of α chemokines are separated by a single amino acid (CXC motif), and these proteins are encoded by genes clustered on human chromosome 4. In contrast, the first two cysteines of all β family chemokines are adjacent to one another (CC motif), and these proteins are encoded by genes clustered on human chromosome 17.
Chemokines having the CXC motif (α chemokines) primarily affect neutrophils and lymphocytes, and they can modulate angiogenesis. Chemokines having the CC motif (β chemokines) affect monocytes, lymphocytes, eosinophils, and basophils with variable selectivity. In addition, certain chemokines in the and β family can inhibit HIV replication in T cells and monocytes, respectively. The third chemokine family currently has only one member: the T cell- specific chemoattractant, lymphotactin (Kelner et al., Science 266:1395-1399, 1994). The gene encoding lymphotactin is located on human chromosome 1, and the protein sequence contains only the second and fourth cysteines found in the and β chemokine families (Kennedy et al., J. Immunol. 155:203-209, 1995).
The monocyte chemoattractant proteins (MCPs), which constitute a subfamily of the β chemokines described above, include the three human MCP proteins (MCP- 1 , MCP-2, and MCP-3). These proteins are -65% identical, and all have Gin as their N-terminal amino acid (Van Damme et al., J. Exp. Med. 176:59- 65, 1992; Yoshimura et al., J. Exp. Med. 169:1449-1459, 1989).
The chemokine eotaxin is also a β chemokine. It is related in sequence to the MCP proteins, but does not contain an amino-terminal Gin residue. Eotaxin appears to be unique among the chemokines in that it causes the selective infiltration of eosinophils when injected subcutaneously and when administered directly to the lungs of naive guinea pigs.
Five genes encoding human β chemokine receptors, referred to as CKR1 through CKR5, have been cloned. (Charo et al., Proc. Natl. Acad. Sci. USA 91 :2752-2756, 1994; Combadiere et al, DNA and Cell Biol. 14:673-380, 1995;
Combadiere et al., J. Leukocyte Biol. in press, 1996; Gao et al, J. Exp. Med. 177:1421-1427, 1993; Neote et al., Cell 72:415-425, 1993; Power et al., J. Biol. Chem. 270: 19495-19500, 1995; Samson et al., Biochem. 35:3362-3367, 1996). CKR2a and CKR2b are splice variants of the same gene. These receptors are not ubiquitously expressed: CKR1, CKR4, and CKR5 appear to be widely expressed on leukocytes, while expression of CKR2 and CKR3 is mainly restricted to monocytes and eosinophils, respectively. Summary of the Invention
The invention features substantially pure nucleic acid molecules that encode MCP-4 and MCP-5, two novel members of the β-chemokine family. The invention also includes polypeptides encoded by these nucleic acid molecules. The nucleic acid molecules may consist of genomic DNA, cDNA, or mRNA, and, due to the degenerate nature of the genetic code, may vary in sequence provided that the encoded polypeptides are MCP-4 or MCP-5, as shown, for example, in Figs. 1 A and IB, respectively.
Substantially pure MCP-4 and MCP-5 polypeptides, biologically active fragments of these polypeptides, including immunogenic fragments, are also considered within the scope of the invention. The biological activity of any given fragment of MCP-4 or MCP-5 may be readily determined by conducting an assay of, e.g., monocyte chemotaxis, as described herein. The preferred polypeptides of the invention are both substantially homologous to MCP-4 or MCP-5 and retain the biological activity of the relevant poiypeptide, as described herein. The nucleic acid molecules can encode a mammalian MCP-4 or MCP-5 poiypeptide, such as those from a human, mouse, rat, guinea pig, cow, sheep, horse, pig, rabbit, monkey, dog, or cat. Preferably, the nucleic acid molecule encoding MCP-4 encodes human MCP-4 and the nucleic acid molecule encoding MCP-5 encodes murine MCP-5 or, more preferably, human MCP-5. The nucleic acid molecules can be placed under the control of a promoter, which may be constitutively active or induced by one or more external agents. The promoter can provide the means to achieve tissue-specific or cell type-specific expression of the nucleic acid molecules of the invention. Alternatively, or in addition, the nucleic acid molecules can be operably linked to a DNA regulatory sequence. Skilled artisans will recognize that the nucleic acid molecules can be placed into a vector construct, such as a plasmid or viral vector, which may in turn be used to transduce living cells with the nucleic acid molecules of the invention. Cells can be transfected with plasmid vectors by standard methods including, but not limited to, liposome-, polybrene-, or DEAE dextran-mediated transfection (see, e.g., Feigner et al, Proc. Natl. Acad. Sci. USA 84:7413, 1987; Ono et al., Neurosci. Lett. 117:259, 1990; Brigham et al., Am. J. Med. Sci. 298:278, 1989), electroporation (Neumann et al, EMBO J. 7:841, 1980), calcium phosphate precipitation (Graham et al., Virology 52:456, 1973; Wigler et al., Cell 14:725,
1978; Feigner et al., supra) microinjection (Wolff et al., Science 247: 1465, 1990), or velocity driven microprojectiles ("biolistics"). Viruses known to be useful for gene transfer include adenoviruses, adeno associated virus, herpes virus, mumps virus, poliovirus, retroviruses, Sindbis virus, and vaccinia virus such as canary pox virus.
The nucleic acid constructs described above are useful in a variety of ways. For example, the constructs may be used as a source of recombinantly produced MCP-4 or MCP-5 polypeptides. Alternatively, the constructs may be administered themselves in a therapeutic approach, as described below. In addition, the constructs may be used to generate transgenic animals that overexpress, or fail to express, MCP-4 or MCP-5. The animal can be a mouse, a worm, or any other animal considered useful for research or drug development. Transgenesis has become routine in the art of molecular biology.
MCP-4 or MCP- 5 polypeptides expressed by transfected cells can be purified and injected into an animal, such as a rabbit, in order to generate polyclonal antibodies that specifically bind MCP-4 or MCP-5. Monoclonal antibodies may also be prepared using standard hybridoma technology (see, e.g., Kohler et al., Nature 256:495, 1975; Kohler et al., Eur. J. Immunol. 6:292 and 6:511, 1976; Hammerling et al., In Monoclonal Antibodies and T Cell Hybridomas, Elsevier, NY, 1981). Alternatively, in various embodiments, an immunologically-active antibody fragment, such as an Fab', (Fab')2, or genetically engineered Fv fragment (see U.S.S.N. 4,946,778, hereby incorporated by reference) may also be useful. Once generated, the antibodies may be purified according to standard methods and injected into a mammal, such as a human, in order to inhibit the inflammatory response caused by chemotaxis of various leukocytes, particularly monocytes.
Preferably, the nucleic acid molecules of the invention, cells that express these molecules, or a therapeutic composition containing the encoded polypeptides, are administered to a mammal in order to stimulate a local immune response. This approach would be particularly beneficial to mammals that are suffering from cancer. The cancer may be evident as, for example, a lymphoma (such as Hodgkin's lymphoma), a plasmacytoma, a melanoma, a sarcoma, or a tumor within the lung or gastrointestinal tract.
In the event that the desired result is inhibition (rather than stimulation) of a local immune response, a mutant MCP-4 or MCP-5 poiypeptide, antibodies or fragments thereof that specifically bind MCP-4 or MCP- 5, or antisense MCP-4 or MCP-5 oligonucleotides may be administered. The mutant MCP-4 or MCP-5 polypeptides may lack, for example, between one and ten of the amino acid residues present at the amino terminus of the mature polypeptides. Alternatively, the mutant polypeptides may contain additional amino acid residues, for example between 3 and 10 amino acid residues, at the amino-terminus of the mature polypeptides. Where amino acid residues are added, they may be random or they may be selected to have particular biological properties such as stability or hydrophilicity. Antisense MCP-4 or MCP-5 oligonucleotides preferably consist of at least eight nucleotides, and have a sequence that is the reverse and complement of that found within the genes encoding these polypeptides. Suppression or inhibition of the local immune response would be beneficial when, for example, the mammal has been diagnosed as having asthma, chronic obstructive pulmonary disease, cystic fibrosis, sinusitis, rhinitis, atherosclerosis, glomerulonephritis, multiple sclerosis, or an inflammatory bowel disease. The nucleic acid molecules of the invention are also useful in treating a mammal that is suffering from an infectious disease, such as acquired immune deficiency syndrome (AIDS) or malaria. Preferably, the method of treatment consists of administering to the infected mammal an MCP-4 or MCP- 5 poiypeptide, or a fragment thereof that is sufficient to bind the chemokine receptors by which these infectious agents gain entry to the cell.
The local immune response may also be modulated by administering a compound that modulates the chemotactic activity of MCP-4 or MCP-5. Examples of such modulators include interferon-gamma, tumor necrosis factor-α, interleukin-1, and interleukin-4. Additional modulatory compounds may be discovered by, for example, the method described herein, whereby a population of cells that express MCP-4 or MCP-5 is obtained, a subset of these cells is contacted with a candidate modulatory compound and the level of MCP-4 or MCP-5 expression in the subset of cells is compared with the level of expression in an equivalent number of cells in the initial population that were not contacted with the potential modulator. A difference in the level of MCP-4 or MCP-5 expression would indicate the presence of a compound that modulates MCP-4 or MCP-5 expression. Either gene expression or protein expression may be readily assessed by methods well known to skilled artisans. For example, mRNA expression could be assessed by Northern blot or RNAse protection analyses and protein expression could be assessed by Western blot analysis.
By "poiypeptide" is meant any chain of more than two amino acids, regardless of post-translational modification such as glycosylation or phosphorylation. By "substantially pure" is meant that the substance in question, be it a nucleic acid molecule, a poiypeptide, or an antibody, has been separated from the components that naturally accompany it. Typically, a poiypeptide is substantially pure when it is at least 60%, by weight, free from the proteins and naturally- occurring organic molecules with which it is naturally associated. Preferably, a preparation of MCP-4 or MCP-5 is at least 75%, more preferably at least 90%, and most preferably at least 99% pure, by weight. A substantially pure poiypeptide may be obtained, for example, by extraction from a natural source (e.g., a mammalian cell), by expression of a recombinant nucleic acid encoding the poiypeptide, or by chemically synthesizing the poiypeptide. Purity can be measured by any appropriate method, such as column chromatography, polyacrylamide gel electrophoresis, or by HPLC analysis. In reference to a DNA molecule, "substantially pure" means that the molecule is free of the genes that flank it in the naturally-occurring genome of the organism from which it is obtained. The term therefore includes, for example, a recombinant DNA molecule which is incorporated into a vector; into an autonomously replicating plasmid or virus; into the genomic DNA or a prokaryote or eukaryote; or which exists as a separate molecule (e.g., a cDNA or a genomic or cDNA fragment produced by the polymerase chain reaction (PCR) or by restriction endonuclease digestion). It also includes a recombinant DNA which is part of a hybrid gene encoding additional poiypeptide sequence.
By "substantially identical" to MCP-4 and MCP-5 is meant a sequence having at least 80% identity to the MCP-4 and MCP-5 sequences shown in Figs. 1 A and IB. Preferably, the identity is 90% and most preferably the identity is 95%.
By "operably linked" is meant that a gene or portion of a gene is connected to one or more regulatory elements, which are also a particular DNA sequences, in such a way as to permit expression of the gene when the appropriate molecules (e.g., transcriptional activator proteins) are bound to the regulatory element(s).
The term "transgenic" as used in reference to an animal or a cell, means an animal or cell into which a DNA sequence has been inserted by artifice and which becomes a part of the genome of the animal or cell.
By "biological activity" of MCP-4 or MCP-5 is meant the biological activity (preferably monocyte chemoattractant activity) which is at least 50% of that observed using MCP-4 or MCP-5, having the sequence shown in Figs. 1 A and IB respectively, in any of the assays describes herein. Preferably the activity is 80% of the activity observed with the MCP-4 and MCP-5 polypeptides of Figs. 1A and
IB, and most preferably, the activity is 95% of that observed with the MCP-4 and MCP-5 polypeptides of Figures 1A and IB.
By "modulatory compound" or "modulator" is meant any compound that is capable of increasing or decreasing the expression or activity of MCP-4 or MCP-5. Expression may be modified at the level of transcription, translation, or post- translation. Activity may be modified at the level of receptor binding or within the cytoplasm of a cell. Activity may be assessed as, for example, monocyte chemotaxis per unit of MCP-4 or MCP- 5 protein. Activity would be considered stimulated if, for example, the number of monocytes in a target tissue is at least 20% greater than the number of monocytes in a control, untreated tissue that is in all other substantive ways identical to the treated tissue. Conversely, activity would be considered inhibited if, for example, the number of monocytes in a target tissue is at least 20% less than the number of monocytes in a control, untreated tissue that is in all other substantive ways identical to the treated tissue. Other features and advantages of the invention will be apparent from the following description and from the claims. Brief Description of the Drawings Fig. 1A is a schematic representation of the nucleotide sequence (bottom line; SEQ ID NO: 1) and predicted amino acid sequence (top line; SEQ ID NO:2) of human MCP-4. The stop codon is indicated by asterisks; the predicted site for signal peptidase cleavage is indicated by an arrow; and a putative mRNA destabilizing sequence is underlined. The putative mature protein is numbered
Figure imgf000011_0001
Fig. IB is a schematic representation of the nucleotide sequence (bottom line; SEQ ID NO:3) and predicted amino acid sequence (top line; SEQ ID NO:4) of murine MCP-5. The intron/exon borders are indicated by triangles; the ATTTA sequence (which may decrease mRNA stability) is singly underlined; the predicted polyadenylation signal is doubly underlined; and the predicted site for a signal peptidase cleavage is marked with an arrow (just 5' to the +1 site). Fig. 2 A is a schematic representation of the human MCP-4 amino acid sequence (SEQ ID NO:2), aligned with other chemokines having the CC motif (indicated by bold face type; SEQ ID NOs:5-14). The +1 predicts the mature N-terminus after signal peptidase cleavage (arrow), and gaps to maximize alignment are indicated by (-). The last 48 amino acids of murine MCP-1 have been omitted.
Fig. 2B is a schematic representation of the amino acid sequence of human MCP-4 (SEQ ID NO:2) murine MCP-5 (SEQ ID NO:4), and other mouse and human CC chemokines (SEQ ID NOs:5-8 and 14-16). Conserved amino acids are boxed, and the position of leader sequence cleavage is indicated by an arrow. Figs. 3 A and 3B are photographs of a Southern blot probed at high stringency (Fig. 3 A) or low stringency (Fig. 3B) with a 32P-labeled human MCP-4 cDNA probe. Human genomic DNA was digested with PstI (lane 1), Bglll (lane 2), Hindlll (lane 3), or EcoRI (lane 4). Fig. 3C is a photograph of a Southern blot probed with a full-length MCP-5 cDNA probe. Mouse genomic DNA was digested with BamHI (lane 1), EcoRI (lane 2), Xbal (lane 3), or Hindlll (lane 4).
In each of Figs. 3A - 3C, the position of molecular weight markers are indicated (in kb) on the left.
Fig. 3D is a representation of the genomic organization of the mouse MCP- 5 gene, including selected restriction sites (top). The mature mRNA is also shown schematically (bottom), with the locations of the start (ATG) and stop codons (TGA) indicated. Fig. 3E is a photograph of PCR products obtained by amplifying genomic
DNA from C57BL/6J, Mus spretus, and heterozygous mice with primers in intron 2 of MCP-5.
Fig. 3F is a map depicting the location of the MCP-5 gene on mouse chromosome 11. Fig. 4A is a photograph of a Northern blot that was probed with a
32P-labeled human MCP-4 cDNA probe (upper blots) and a human β-actin cDNA probe (lower blots). Poly-A+ mRNA was isolated from the human tissues indicated. The position of molecular weight markers is shown (in kb) on the left. Fig. 4B is a photograph of a Northern blot depicting MCP-5 expression in normal mouse tissues. The blot was probed sequentially with cDNA probes for
MCP-5 and, as a control for RNA loading, rpL32 ribosomal protein. The positions of 18s and 28s RNA are indicated on the left.
Fig. 4C is a photograph of a Northern blot depicting MCP-5 expression in murine leukocytes and in tissues of mice that serve as models of pulmonary inflammation. Each lane represents an individual mouse from a representative experiment (n=3).
Fig. 4D is a photograph of a Northern blot depicting MCP-5 expression in activated macrophages (from the RAW 264.7 macrophage cell line) and endothelial cells (SVEC cells). The cells were untreated (Ctl) or treated with IL-4 IFNγ, IL-lβ, or LPS as indicated.
Fig. 5 A is a series of photographs of a Northern blot that was probed sequentially with MCP-4, MCP-1, VCAM-1 and β-Actin cDNA probes. Total RNA isolated from human umbilical vein endothelial cells was treated for the time shown with IFNγ, TNFα, IL-4, or IL-lα, or a combination of IFNγ/TNFα, IFNγ/IL-4, or TNFα/IL-4.
Fig. 5B is a pair of photographs of a Northern blot hybridized sequentially with MCP-4 and GADPH cDNA probes. The samples consisted of total RNA, which was isolated from cells of the human respiratory epithelial cell line A549.
The cells were either untreated (control) or treated with the cytokines listed above the corresponding lane of the blot (PMA, TNFα, IL-lβ, IFNγ, TGFα, and TGFβ).
Fig. 5C is a pair of photographs of a Northern blot that was hybridized sequentially with MCP-4 and GADPH cDNA probes. The samples consisted of total RNA, which was isolated from cells of the human respiratory epithelial cell line A549 at various times after treatment with 0.1 ng/ml IL-lβ.
Fig. 5D is a pair of photographs of a Northern blot that was hybridized sequentially with MCP-4 and β-actin cDNA probes. Total RNA was isolated from the human respiratory epithelial cell line BEAS-2B either before (control), 6 hours, or 18 hours after stimulation with the cytokines listed above the corresponding lanes.
Fig. 6A is a pair of photographs of a Northern blot that was hybridized sequentially with MCP-4 and β-actin cDNA probes. Total RNA was isolated from untreated U937 cells (control; lane 1), U937 cells stimulated for 18 hours with IFNγ (lane 2), untreated HL60 cells (lane 3), an eosinophilic subline of HL60 cells that were cultured with butyric acid (HL60-Eosinophilic; lane 4), human peripheral blood mononuclear cells (lane 5), and neutrophils (lane 6). Fig. 6B is a series of photographs of Northern blots that were hybridized with either MCP-4, RANTES, or β-actin probes. Total RNA was isolated from H9 T cells (lane 1), CD8+ HIV-specific cytolytic T cell clones that were unstimulated (lane 2) or stimulated with anti-CD3 (lane 3), and peripheral blood mononuclear cells cultured in the presence of PHA and IL-2 (lane 4).
Fig. 6C is a pair of photographs of a Northern blot that was hybridized sequentially with MCP-4 and β-actin cDNA probes. Total RNA was isolated from Jurkat Cells (lane 1), the RM3 subline of Raji cells (lane 2), freshly isolated purified peripheral blood eosinophils, either before (lane 3) or after (lane 4) culture with IL-3.
Fig. 7 is a photograph of an SDS-polyacrylamide gel (12.5%; Tris/Tricine) on which was electrophoresed recombinant MCP-4 His tagged protein (produced by E. coli) before factor Xa cleavage (lane 1; Coomassie stained), recombinant MCP-4 with the mature N-terminal Gin (PeproTech; lane 2; Coomassie stained), molecular weight standards (lane 3; values in kDa are indicated on the far left), and affinity purified MCP-4-FLAG protein (lane 4; Western blot).
Fig. 8 is a line graph depicting chemotaxis of human elutriated monocytes to recombinant MCP-4 (produced by E. coli) before (uncleaved; closed triangles) and after factor Xa cleavage (cleaved; open diamonds). The chemotactic response to MCP-1 is also shown (open circles). The results are expressed as the mean ±
SEM for replicate samples.
Fig. 9 A is a panel of line graphs depicting chemotaxis of monocytes to MCP-4. Monocytes were subjected to chemotaxis in a modified Boyden chamber, and the number of cells that migrated through the membrane were counted (Y axis). Control chemotactic stimuli are indicated.
Fig. 9B is a panel of line graphs depicting chemotaxis of eosinophils to MCP-4. Monocytes were subjected to chemotaxis in a modified Boyden chamber, and the number of cells that migrated through the membrane were counted.
Control chemotactic stimuli are indicated.
Fig. 9C is a panel of line graphs depicting chemotaxis of eosinophils.
Neutrophils were subjected to chemotaxis in the presence of MCP-4 in a modified Boyden chamber, and the number of cells that migrated through the membrane were counted (shown on left-hand graph). A control chemotactic stimulus, IL-8, is also shown (right-hand graph).
In each of Figs. 9 A, 9B, and 9C, the chemotaxis results are expressed as the mean ± SEM for replicate samples from one of five donors tested. Figs. 9D - 9G are line graphs depicting chemotaxis of human peripheral blood mononuclear cells (9D; n=4), mouse thiogylcolate elicited peritoneal macrophages (9E; n=l), mouse eosinophils (9F; n=5), and mouse neutrophils (9G; n=3) following exposure to increasing concentrations of the indicated chemokines in a modified Boyden chamber. In each of Figs. 9D - 9G, the results are expressed as the mean ± SEM for 8 hpf (high powered fields) counted in replicate wells.
Fig. 10A is a dark field photomicrograph of a section of tissue, obtained from a human patient with hypertrophic sinusitis, that was hybridized with a radiolabelled MCP-4 cRNA probe. "E" labels the epithelium. Fig. 1 OB is a bright field photomicrograph of a section of tissue, obtained from a human patient with hypertrophic sinusitis, that was hybridized with a digoxigenin-labeled MCP-4 cRNA probe. The arrows point to mononuclear cells. Fig. 11 A is a series of tracings representing calcium flux stimulated by
MCP-4 in mononuclear cells. Fura-2-loaded cells were exposed sequentially to the indicated chemokines with the concentration of each stimulus indicated in parenthesis below the name of the chemokine. Calcium flux is reported as ratio fluorescence of fura-2. Fig. 1 IB is a series of tracings representing calcium flux stimulated by MCP-4 in neutrophils. Fura-2-loaded cells were exposed to chemokines, as described for Fig. 11 A above, and changes in intracellular fluorescence were monitored. Figs. 11C - HE are a series of tracings representing calcium flux in human peripheral blood mononuclear cells (11C), eosinophils (1 ID), and neutrophils (1 IE) following sequential application of 100 nM of mMCP-5, mJE, hMCP-1, mMIP-lβ, mMIP-loc, eotaxin, and mKC, as indicated.
Fig. 1 IF is a line graph depicting the concentration dependence of MCP-4 induced calcium flux in human mononuclear cells. The magnitude of the calcium transient peak induced by MCP-4 is plotted as a percentage of the maximal response observed for MCP-5.
Fig. 12A is a series of tracings representing the selectivity of various chemokine receptors to MCP-4. Calcium flux was monitored by the relative fluorescence of fura-2-loaded HEK 293 cells stably expressing cloned CC chemokine receptors. Each row of tracings corresponds to the receptor indicated on the left. Arrows indicate the time of addition of the indicated chemokine at 50 nM (a = MCP-4, b = MlP-lα, c = MCP-1, and d = eotaxin).
Fig. 12B is a series of line graphs that identify CCR2 as a functional MCP-5 receptor. HEK-293 cells stably expressing human CCR2b (i, ii, and iii) or murine
CCR2 (iv, v) were loaded with indo-1 AM, and intracellular calcium concentrations were monitored by ratio fluorescence in response to the indicated concentrations of MCP-5 (i, iv), JE (ii, v), and MCP-1 (iii).
Fig. 12C is a series of line graphs that illustrate desensitization of CCR2b by MCP-5. HEK-293 cells stably expressing human CCR2b were exposed to
MCP-5 (i), MCP-1 (ii), or JE (iii) and subsequently challenged with 100 nM MCP- 5, 100 nM MCP-1, or 100 nM JE. Fig. 13 is a line graph in which the magnitude of the calcium transient peak induced by MCP-4 is plotted as a percentage of the maximal response observed for MCP-1 in the case of CKR2b (circles) and eotaxin in the case of CC CKR3 (squares). Fig. 14A is a series of tracings depicting desensitization of calcium transients in HEK 293 cells stably transfected with CKR2b. Relative fluorescence was monitored before and during sequential addition of chemokines at the times indicated by the arrows. The first stimulus added (SI) for each tracing is indicated at the left of the row in which it is found. The second stimulus added (S2) is indicated at the top of the column in which the tracing is found.
Fig. 14B is a series of tracings depicting desensitization of calcium transients in HEK 293 cells stably transfected with CKR3B. Fluorescence was monitored as described in the legend of Fig. 14A.
Detailed Description
Two new members of the β-chemokine family, MCP-4 and MCP-5, have been isolated and characterized. The expression pattern of the genes encoding these chemokines was examined in normal tissue and in a variety of clinically relevant circumstances, such as in cytokine stimulated endothelial, epithelial, and hematopoietic cells, in hypertrophic sinus tissue, and in two in vivo models of pulmonary inflammation. In addition, the chemotactic activity of MCP-4 and MCP-5 proteins on various leukocytes, and the receptors through which this activity is exerted, are described.
The following examples are meant to illustrate, not limit, the invention. Example 1 : Cloning of Human MCP-4 and MCP-5 cDNA
A. Isolation of the human MCP-4 gene
A human MCP-4 cDNA was isolated from a heart cDNA library using a human eotaxin genomic probe, as follows. A 1.1 kb genomic fragment of the human eotaxin gene containing exon 2 and the coding portion of exon 3 was 32P-labeled with the Klenow fragment of DNA polymerase and used to probe a λgtlO human heart cDNA library (Clontech, Palo Alto, CA). Approximately 106 phage were plated, transferred to GeneScreen Plus (DuPont, Wilmington, DE), and hybridized for 18 hours at 42 °C in a buffer containing 20 mM Tris (pH 7.5), 50% formamide, 10% dextran sulfate, 5X SSC, IX Denhardt's solution (0.0002% (w/v) polyvinylpyrrolidone, 0.0002% (w/v) BSA, 0.0002% (w/v) Ficoll 400), and 100 mg/ml of heat-denatured herring sperm DNA solution. The filters were washed for 30 minutes at room temperature with 2X SSC and 0.1% SDS followed by a 30 minute wash at 60°C with 0.2X SSC and 0.1% SDS. Eight hybridizing plaques were identified and purified. The inserts were amplified by the polymerase chain reaction (PCR) using a pair of λgtlO flanking primers. The PCR consisted of 30 thermocycles of 95 °C for 60 seconds (during which the DNA was denatured), 55 °C for 60 seconds (during which the primers annealed to the template), and 72 °C for 60 seconds (during which a complementary sequence was polymerized).
The amplified PCR products were subcloned into a pCRII vector (Invitrogen, San Diego, CA), and the sequence of both strands was obtained using SEQUENASE™ (USB-Amersham, Arlington Heights, IL). Alignment of the DNA sequences and homology analysis was performed by using the CLUSTAL analytical program and the GAP program (Genetics Computer Group at the University of Wisconsin,
Madison, WI).
The MCP-4 cDNA was 823 bp long, consisting of a 34 bp 5' untranslated region, a 297 bp coding region, and a 492 bp 3' untranslated region (Fig. 1 A). A single AUUUA sequence was identified in the 3' untranslated region (Fig 1 A). This sequence motif has been shown to decrease the stability of cytokine mRNA
(Shaw et al., Cell 46:659-667, 1986). The entire cDNA sequence of human MCP-4 was compared with the nucleotide sequence of the known human chemokines. MCP-4 was found to be 62%, 58%, and 57% identical to MCP-1, MCP-3, and eotaxin, respectively (Table 1). The nucleotide sequence of human MCP-4 was also compared to known mouse chemokines and found to be most homologous to eotaxin (57% identical), and 54% and 51% identical to MCP-3 and MCP-1, respectively. Table 1 Homology of Human MCP4 with other Chemokines
Chemokine Amino Acid Nucleic Acid Identity Similarity Identity
(%) (%) (%)
Hu MCP1 65 79 62
Hu MCP2 57 70 ND
Hu MCP3 65 77 58
HU EOT 66 76 57
Hu RANTES 32 57 41
Hu MlP-lα 39 54 45
Hu MIP-lβ 39 61 41
Hu-IL-8 28 53 27
Mu MCP1 54 69 51
Mu MCP3 53 70 54
Mu EOT 54 66 57
Mu RANTES 35 59 42
The human MCP-4 cDNA encodes a 99 amino acid protein containing a highly conserved 23 amino acid signal peptide (Fig 1 A). Signal peptidase cleavage is predicted to occur after the sequence, Pro-Gln-Gly-Leu-Ala, which is invariant in human MCP-1, -2, -3, and eotaxin (Fig. 2A). The resulting 76 amino acid sequence is predicted to have an N-terminal Gin, which is characteristic of the MCP family. The putative mature protein is predicted to have a molecular weight of 8,599 Da, a pi of 10, and appears to lack N-linked glycosylation sites. At the amino acid level, human MCP-4 exhibits 66%, 65%, 57%, and 65% identity with human eotaxin, MCP-1, MCP-2, and MCP-3, respectively, and has 54%, 54%, and 53% identity with mouse eotaxin, MCP-1 and MCP-3, respectively. Thus, human MCP-4 is as closely related to eotaxin as it is to proteins in the MCP family. However, in contrast to eotaxin, MCP-4 has an N-terminal Gin residue and it lacks the two amino acid N-terminal gap that is characteristic of the eotaxin proteins.
B. Isolation of the murine MCP- 5 gene
Human MCP-4 cDNA was radiolabelled and used as a probe to screen a 129SV mouse genomic library (Stratagene, La Jolla, CA). Approximately 106 phage were plated, transferred to GeneScreen Plus (DuPont, Wilmington, DE), hybridized for 18 hours at 50°C in a low stringency hybridization buffer (0.6M
NaCl, 80 mM Tris-HCl, 4 mM EDTA, 0.1% (w/v) sodium pyrophosphate, 0.1% (w/v) SDS, 10X Denhardt's solution, 100 μg/ml denatured herring sperm DNA) at 50°C and washed at 60°C in IX SSC/0.1% SDS. In order to exclude JE, the putative murine orthologue of human MCP-1, the filters were rehybridized as described with a full-length JE cDNA fragment and washed at 65 °C in
0.1X SSC/0.1% SDS. Fifteen plaques, which hybridized more strongly with the human MCP-4 probe than with the murine JE probe, were analyzed by PCR using two sets of degenerate oligonucleotides corresponding to highly conserved regions of murine and human CC chemokines and the MCP-eotaxin subfamily. The first set was designed to amplify Exon 1-2 and consisted of a 5' oligomer
(5'-CTTCTGKGYCTGCTGYTCA-3'; SEQ ID NO: 17) and a 3' oligomer (5*-ACAGCYTYYYDGGGACA-3*; SEQ ID NO: 18). The second set was designed to amplify Exon 2-3 and consisted of a 5' oligomer (5'-TGTCCCHRRRARGCTGT-3'; SEQ ID NO: 19) and a 3' oligomer (5'-GSKTCAGCRCAGAYYTC-3'; SEQ ID NO:20). The reactions were heated to
95 °C for 5 minutes and then subjected to 30 rounds of the following thermocycle: 94 °C for 1 minute, 45 °C for 1 minute and 72 °C for 1.5 minutes. The final round of extension at 72 °C was prolonged to 8 minutes. When the first set of primers was employed, five of the fifteen plaques were shown to contain DNA that was amplified as an 800 bp PCR product. These products were subcloned into the pCRJI vector (Invitrogen, San Diego, CA) and sequenced (Sequenase Kit, United States Biochemical, Cleveland, OH). All five subclones contained novel, overlapping sequence that appeared to be homologous to members of the MCP subfamily.
In order to determine the complete structure of the MCP- 5 cDNA, 5' and 3' RACE PCR was performed using RNA isolated from IFNγ-treated RAW 264.7 cells. The 5' and 3' ends of the cDNA for murine MCP-5 were isolated using a 3' oligonucleotide in exon 2 (5'-CTGGCTGCTTGTGATTCTCCTGT-3'; SEQ ID
NO:21), a 5' oligonucleotide at the end of exon 1
(5'-CAGTCCTCAGGTATTGGCTGG-3; SEQ ID NO:22), and the Marathon™ cDNA Amplification Kit (Clontech, Palo Alto, CA). The amplified products were cloned into the pCR2.1 vector (InVitrogen, San Diego, CA) and sequenced. Based on the sequence obtained, oligonucleotides capable of amplifying full-length cDNA from IFNγ-treated RAW cell poly A+ RNA were synthesized using the 5' oligomer 5'-AGCTTTCATTTCGAAGTCTTTG-3'(SEQ ID NO:23) and the 3' oligomer 5'-TAGATTCGGTTTAATTGGCCC-3'(SEQ ID NO:24). The products produced by PCR with these oligonucleotides were cloned into pCR2.1 and sequenced.
The cDNA was 514 bp long with an open reading from that encoded 104 amino acids. The 5' region of the cDNA encoded a 22 amino acid hydrophobic leader sequence whose cleavage site was predicted to occur at a position similar to the other MCPs and eotaxin (Figs. IB and 2B) resulting in a mature protein of 9.3 kDa with a pi of 9.4. The 3' untranslated region contained a single polyadenylation signal of a rare type also found in human eotaxin (ATTAAA) and four mRNA destabilization signals (ATTTA; Fig. 2B) that have been reported to decrease the mRNA stability of other cytokine mRNAs (Shaw et al., Cell 46:659-667, 1986). The sequence of murine MCP-5 has been deposited with GenBank and assigned the accession number U66670. The sequence comparison shown in Fig. 2B was generated using the program SeqVu (from The Garvin Institute of Medical Research, Sydney, Australia). Sequences were aligned for maximal homology, using the positions of the four conserved cysteine residues as fixed points.
The sequence of murine MCP-5 is most homologous to human MCP-1. The percent amino acid/nucleic acid identity is shown in Table 2. These figures depict comparison of the open reading frames and the full length cDNAs using the GAP program (supplied by the Genetics Computer Group at the University of Wisconsin Madison, WI), which uses the algorithm of Needleman and Wunsch to maximize the number of matches and minimize the number of gaps in two complete sequences. ND indicates that the percent identity was not determined. Like human MCP-1, murine MCP-5 does not contain the additional serine/threonine rich glycosylated 49 amino acid C-terminal extension that JE uniquely possesses (Fig. 2B). MCP-5 is unique among the MCP proteins in that its
N-terminal amino acid is predicted to by glycine, a feature that it shares with human eotaxin (Figs. IB and 2B).
TAB E 2 mMCP5 mJE mMCP3 mEot hMCPl hMCP2 hMCP3 hMCP hEot mMCP5 47/50 58/57 49/53 66/64 56/ND 60/62 56/58 58/59 mJE 47/50 49/54 49/51 52/63 53/ND 52/52 54/51 49/50 mMCP3 58/57 49/54 46/48 59/60 51/ND 61/67 53/67 54/54 mEot 49/53 49/51 46/48 53/54 52/ND 57/53 54/50 59/55 hMCPl 66/64 52/63 59/60 53/54 62/ND 74/66 65/61 71/63 hMCP2 56/ND 53 /ND 51/ND 52/ND 62 /ND 58/ND 57/ND 61/ND hMCP3 60/62 52/52 61/67 57/53 74/66 58/ND 64/58 70/63 hMCP4 56/58 54/51 53/67 54/50 65/61 57/ND 64/58 66/58 hEot 58/59 49/50 54/54 59/55 71/63 61/ND 70/63 66/58
Example 2: Gene Analysis and Chromosomal Location
A. Southern blot analysis of MCP-4
High molecular weight genomic DNA was isolated from human peripheral blood leukocytes (PBMCs), and separate samples were digested with one of four restriction endonucleases: PstI, Hindlll, Bglll, or EcoRI. Approximately 20 mg of digested DNA were electrophoresed on a 0.8% agarose gel, transferred to GeneScreen Plus, and hybridized for 18 hours at 42 °C with the MCP-4 cDNA probe described in Example 1. The blots were first washed at low stringency (2X SCC, 0.1% SDS, 50 °C) and then at high stringency (0.1X SSC, 0.1% SDS, 65°C) for 30 minutes each. This analysis revealed that human MCP-4 is a single copy gene (Fig. 3A). Furthermore, under highly stringent hybridization and washing conditions, the MCP-4 cDNA can be used as a probe to uniquely detect MCP-4. Low stringency Southern blot analysis (Fig. 3B) revealed multiple bands, which represent closely related β-chemokine genes.
In order to determine the chromosomal location of the MCP-4 gene, PCR-based screening of the National Institute of General Medical Sciences (NIGMS) somatic cell hybrid panel #2 (available from the A.T.C.C, Rockville, MD) was performed using MCP-4 specific oligonucleotide primers (5'-TCAGCCAGATGCACTCAACG-3' (SEQ ID NO.:25) and 5'-TGGAAAAGTCATTTCACGTA-3' (SEQ ID NO.:26)) from exons 2 and 3, respectively. The reaction was performed using 100 mg of template DNA, which was denatured initially at 94 °C for 2 minutes and then subjected to 30 repetitions of the following thermocycle: 94°C for 30 seconds, 55 °C for 30 seconds, and 72 °C for 60 seconds. The reaction products were separated by electrophoresis on a 1% agarose gel, and visualized by staining with ethidium bromide.
Oligonucleotide primers specific for human MCP-4 genomic sequences were also used to amplify human, mouse, and hamster genomic DNA. No product was seen when rodent DNA was used. However, an -850 bp product was observed in human genomic DNA and in a somatic cell hybrid line containing human chromosome 17 as its only human chromosome. The other rodent-human hybrid cell lines did not contain DNA that could be amplified with the designated primers. Thus, MCP-4 is located on human chromosome 17.
B. Southern blot analysis of MCP-5
Samples containing approximately 10 μg of mouse genomic DNA were digested separately with one of four restriction endonucleases: BamHI, EcoRI, Xbal, or Hindlll. The digests were then electrophoresed through an agarose gel (0.8%), transferred to GeneScreen Plus™, hybridized under conditions of high stringency (50% formamide,
10% dextran sulfate, 5X SSC, IX Denhardt's solution, 1% SDS, 100 μg/ml denatured herring sperm DNA, and 20 mM Tris at 42 °C) with a radiolabeled MCP-5 probe, and washed for 40 minutes at 55 °C in 0.2X SSC/0.1% SDS. The blot was then exposed to X- ray film. A photograph of the resulting autoradiogram is shown in Fig. 3C. A schematic representation of the genomic organization of the mouse MCP-5 gene, including the intron-exon structure, selected restriction sites, and the mature mRNA is shown in Fig. 3D.
C. The chromosomal location of MCP-5
The chromosomal location of MCP-5 was determined by single-stranded conformational polymorphism (SSCP) analysis using a pair of PCR primers in intron 2 that detected a polymorphism when using DNA from C57BL/6J and Mus spretus mice
(Fig. 3E). The 5' "sense" oligonucleotide used was
5'-TTACAGGTCAGGTCCCCTACT-3'(SEQ ID NO:27) and the 3' "anti-sense" oligonucleotide was 5'-CTCCTTATCCAGTATGGTCCTG-3' (SEQ ID NO:28). A panel of genomic DNA from 94 interspecific backcross animals between
(C57BL/6J X SPRET/Ei)F, X SPRET/Ei (Jackson BSS) was used to map the MCP-5 gene based on the SSCP polymorphism. The MCP-5 gene cosegregated with Scya7 (Fie) and Scyall (eotaxin) in this cross, placing it between Dl IMit markers 7 and 36 on Chromosome 11. A comparison with the consensus map from the Mouse Genome Database (Fig. 3F) revealed this to be the region of Chromosome 11 containing the CC chemokine gene cluster designated Scyal-11. The MCP-5 gene has been assigned the designation Scyal2 (Fig. 3F). In Fig. 3F, the right-hand panel shows a segment of the central part of the map of chromosome 11 from the Jackson BSS cross described, containing approximately 1500 loci. The data are: proximal-Nos2-l recombinant/94 animals (1.1 +/- 0.1 cM)-Scyal2-2 recombinants/94 animals (2.1 +/- 1.5 cM)-Mpmv4- distal. The left-hand panel of Fig. 3F shows the Mouse Genome Database consensus map, wherein the estimated relative positions of the loci are given at cM offset from the centromere. DllMit4 is offset at 37, Scyall is offset at 45.4, and DllBirl2 is offset at 53. Other CC chemokines are shown and referred to as Scyl-11. Example 3: MCP-4 and MCP-5 Gene Expression A. MCP-4 mRNA Expression in Normal Human Tissue
In order to examine the expression of human MCP -4 in different tissues, Northern blot analysis was performed as follows. Northern blots of multiple human tissue samples were obtained from Clontech and hybridized in the buffer described in Example 1 at 42°C for 18 hours. The blots were then washed with 0.2X SSC and 0.1% SDS at 65°C for 60 minutes. In order to control for potential variations in the quantity of RNA loaded, the blots were stripped by washing with 0. IX SSC and 0.1% SDS at 90 °C for 60 minutes and rehybridized with either a human β-Actin probe or a glyceraldehyde-3 phosphate dehydrogenase (GADPH) cDNA probe (Clontech).
This analysis revealed an -850 bp transcript that probably corresponds to the mature human MCP -4 mRNA. The highest levels of constitutive MCP-4 expression were observed in normal human small intestine, colon, and lung with lower but clearly detectable expression in heart, placenta, and thymus. Lower levels of expression were also seen in muscle, kidney, pancreas, prostate, liver, testis and ovary. However, no expression was detected in brain, spleen or peripheral blood leukocytes (Fig. 4A). B. Expression of MCP -4 in Inflamed Human Nasal Tissue
To examine the possibility that MCP -4 plays a role in allergic inflammation,
MCP-4 expression was analyzed by in situ hybridization of nasal tissue obtained from nine patients with sinusitis and from six normal controls. All patients were adults between the ages of 19-45, who were selected because the suffered from chronic sinusitis for more than one year, (Hamid et al., Proc. Natl. Acad. Sci. USA 84:6760-6764, 1987).
Patients were given an allergy skin test and, based on the result, placed in either an allergic or non-allergic subgroup. Biopsies of 2-3 mm were obtained from the maxillary sinuses of patients with chronic sinusitis and nasal mucosal biopsy specimens were taken from the inferior and middle turbinates of the six control subjects. The tissue was fixed by immersion in 4% paraformaldehyde for 2 hours, washed, embedded, and cut into sections (each 8 μm thick) before processing for in situ hybridization.
In situ hybridization was performed as previously described (Hamid et al., J. Clin. Invest. 87:1541-1546, 1991; Hamid et al., J. Clin. Invest. 94:870-876, 1994). Briefly,
35S-labelled RNA probes coding for MCP -4 mRNA were applied to permeabilized tissue sections, and the cRNA-mRNA hybrid was visualized using autoradiography. To characterize the MCP-4 mRNA-positive cells, particularly the inflammatory cells, digoxigenin-labeled probes were used. In a blind study, the MCP -4 mRNA signal was assessed in at least two sections from each subject. The hybridization signal in the epithelium was presented as the percentage of positive epithelial cells within the total epithelium. The mRNA positive cells in the subepithelium were expressed as the mean number of positive cells/high power field (0.20 mm2). As a negative control, sections were hybridized with sense probes or pretreated with RNase prior to hybridization with anti-sense probes.
The level of MCP -4 mRNA expression was significantly higher in the tissue obtained from patients with sinusitis (both in the epithelial and subepithelial tissue) than in the tissue obtained from unaffected individuals. A quantitative assessment of the MCP-4 mRNA-positive cells in these patients is shown in Table 3. There was not a significant difference between the expression of MCP-4 in the tissue obtained from individuals in the allergic and non-allergic groups. Most of the mRNA expression was -25- localized to the epithelium, particularly the basal layer. Most of the positive cells among the inflammatory infiltrate were mononuclear cells with abundant cytoplasm, a feature that is consistent with macrophage morphology. There were other positive cells, however, including small mononuclear cells, which could be lymphocytes, and granulocytes, which could be either eosinophils or neutrophils.
TABLE 3
Tissue studied No. of cases Mean % epithelium Mean no. of inflammatory studied +ve for MCP-4 mRNA cells/field +ve for MCP-4 mRNA in the subepithelium
Allergic sinusitis 4 71.8 ± 10.7 12 ± 1.5
Non-allergic sinusitis 5 77.5 ± 10.0 13.3 + 1.3
Normal control 6 14.6 ± 3.8 3.4 ± 0.3
C. MCP-5 Expression in Normal Murine Tissue and Murine Cell Lines
RNA was isolated from the organs of BALB/c mice by lysing the tissue in guanidinium isothiocyanate and pelleting the RNA through a 5.7 M CsCl2 cushion (Chirgwin et al., Biochem. 18:5294-5299, 1979). The polyA+ fraction was isolated from total RNA by oligo-dT cellulose chromatography (Pharmacia, Piscataway, NJ). RNA STAT-60 (Tel Test-B Inc., Friendswood, TX) was used to isolate RNA from mouse leukocytes and cell lines.
To generate a Northern blot, 10 μg of total RNA was fractionated on a 1.2% agarose gel containing 0.7% formaldehyde and transferred to a GeneScreen™ membrane. The membrane was then exposed to 32P-dCTP-labeled MCP-5 or JE cDNA probes under hybridization conditions. A probe for the ribosomal protein rpL32 (Shen et al., Proc. Natl. Acad. Sci. USA 89:8240-8244, 1992) was used as a control for the quantity of RNA loaded. Hybridization was performed under conditions of high stringency, and the membrane was washed at 55 °C in 0.2X SSC, 0.1% SDS for 40 minutes. SVEC cells, from an SV40 virus-immortalized murine endothelial cell line and RAW 264.7 cells (American Type Culture Collection, Rockville, MD) were cultured for 6 hours or 18 hours in DMEM supplemented with 10% iron fortified calf serum (without additions) that contained either 200 U/ml recombinant murine IFNγ (Genentech Inc., San Francisco, CA), 5 ng/ml recombinant murine IL-lβ (Genzyme, Cambridge, MA), or 10 ng/ml recombinant murine IL-4 (Genzyme, Cambridge, MA). Mouse bone marrow-derived mast cells were obtained by culturing mouse bone marrow from FVB mice in the presence of 50% WEHI-3 -conditioned medium for 4 weeks. Mast cells were sensitized with IgE anti-TNP at 50 μg/ml and either stimulated with TNP-BSA at 100 μg/ml for 4 hours at 37°C or treated with 2.5 mg/ml of concanavalin A (ConA) for 4 hours at 37°C. The procedures described above revealed an approximately 550 bp transcript that corresponds to the mature mouse MCP-5 mRNA. The highest levels of constitutive MCP-5 expression were observed in normal lymph nodes. MCP-5 mRNA was also seen in heart, lung, breast, salivary glands, thymus, colon, kidney, small intestine, and brain (the salivary gland and breast tissue contained lymph nodes). In contrast, no expression was detected in spleen, skeletal muscle, bone marrow, liver, or peripheral blood leukocytes (Fig. 4B). The expression of MCP-5 and JE were compared in the mouse tissues listed above because these proteins have similar biological profiles and bind the same receptor. The highest constitutive expression of JE was seen in the salivary gland with low levels detected in other organs, such as lymph nodes. The 550 and 800 bp JE transcripts correspond to known 3' splice variants (Kawahara et al., J. Biol. Chem. 264:679-682, 1989).
As shown in Fig. 4C, MCP-5, JE, and rpL32 expression was also examined in subsets of leukocytes. RNA was isolated from eosinophils purified from the spleens of CD5-IL5 transgenic mice (Eos), bone marrow-derived mast cells that were either untreated (Ctl) or stimulated with either IgE (anti-TNP and TNP-BSA (IgE)), or ConA (ConA). In addition, RNA was harvested from resident peritoneal macrophages (RM ), elicited peritoneal macrophages (EMΦ; 76% MNC, 24% PMN), and elicited peritoneal neutrophils (ENΦ; 95% PMN, 5% MNC). Blots were hybridized and washed sequentially under conditions of high stringency with MCP-5, JE and rpL32 cDNA probes and exposed for 14 days, 3 days, or 18 hours, respectively. No expression was detected in eosinophils, bone marrow-derived mast cells, or resident peritoneal macrophages. Furthermore, stimulation of these mast cells with either IgE or ConA did not induce MCP-5 expression. However, ConA induced the expression of JE (Fig. 4C) and both IgE and ConA treatment induced expression of murine CCR1 (Gao et al., Biochem. Biophys. Res. Comm. 223:679-684, 1996). Activated macrophages, which were elicited in the peritoneal cavity by treatment with thioglycolate, expressed significant levels of MCP-5 mRNA. Neutrophils, which were elicited in the peritoneal cavity by treatment with sodium casein, also appeared to express MCP-5 mRNA. MCP-5 mRNA was not detected in either an unstimulated or stimulated allogeneic cytolytic CD8+ T cell clone.
The ability of various stimuli to induce MCP-5 mRNA in the RAW264.7 macrophage cell line was evaluated because it appeared that activated macrophages were a primary source of MCP-5 mRNA. RNA was harvested from cells that were either untreated or treated for 18 hours with interferon-γ (IFNγ at 200 U/ml), or LPS (lipopolysaccharide) at 0.1 , 1.0, 10, or 100 ng/ml. The RNA was then analyzed on Northern blots that were hybridized with MCP-4, JE, and rpL32 cDNA probes, washed sequentially under conditions of high stringency, and exposed for 7 days, 3 days, and 18 hrs, respectively.
The expression of MCP-5 was also examined in cytokine-stimulated murine endothelial cells (SVEC cells). MCP-5 was not induced in SVEC cells by IFNγ, IL-4 (10 ng/ml), or IL-lβ (5 ng/ml), while JE was induced by IFNγ and IL-4 (Fig. 4D) and eotaxin was induced by IFNγ (Rothenberg et al. Proc. Natl. Acad. Sci. USA 92:8960-
8964, 1995). JE was similarly expressed in activated macrophages and induced in RAW cells by LPS and IFNγ (Fig. 4D).
D. MCP-5 mRNA Expression in Murine Models of Pulmonary Inflammation In order to determine whether MCP-5 is associated with the immune response in vivo, MCP-5 expression was examined in the lungs of mice that had been infected with Nippostronylus brasiliensis and in a murine model of OVA-induced pulmonary inflammation (Fig. 4C).
The N. brasiliensis model was generated as described by Coffman et al. (Science 245:308-310, 1989). Briefly, twelve week old female BALB/cJ mice were injected with
750 third stage N. brasiliensis larvae, their lungs were harvested 7, 10 or 14 days later, and RNA was extracted from the tissue and analyzed by Northern blotting. The level of MCP-5 mRNA was markedly increased by day 7. The expression level peaked on day 10 and returned to control levels by day 14. MCP-5 mRNA accumulation preceded the peak recruitment of pulmonary macrophages, neutrophils, and eosinophils that characterize the granulomatous immune response to this pathogen (Coffman et al., supra).
The aerosolized OVA model is generated as follows (see also MacLean et al., J. Exp. Med., in press, 1996). BALB/cJ mice that were between 5 and 10 weeks old were immunized with 10 μg of OVA (Sigma Chemical Co., St. Louis, MO) and 1 mg of aluminum hydroxide (via an intraperitoneal injection) on days 0, 7, and 14. Control mice were "sham-immunized" by treatment with aluminum hydroxide only. An aerosol challenge with OVA (50 mg/ml in sterile sale) was administered 7-10 days after the final immunization. The mice were sacrificed 3, 6, 24, or 48 hours after the aerosol challenge, their lungs were harvested, and RNA was extracted from the tissue. At least 3 mice were present in each group and at each time point of harvest. MCP-5 mRNA was detected in the OVA-immunized mice 3 hours after aerosol challenge (Fig. 4C; left-most lane labeled IC) and remained at an elevated level at the 48 hour time point (right-most lane labeled IC). In contrast, MCP-5 mRNA in challenged, sham-immunized mice (SC) remained at the same level as before the aerosol challenge. In this model, as following infection with N. brasiliensis, the accumulation of MCP-5 mRΝA preceeded the peak accumulation of pulmonary macrophages, lymphocytes and eosinophils that characterize the immune response.
Expression of JE was also elevated in both models of pulmonary inflammation.
However, in the OVA model, levels of JE mRΝA retumed to baseline by 48 hours but
MCP-5 levels remained elevated. Example 4: Cytokine Regulation of
MCP-4 in Endothelial and Epithelial Cells
RΝA was isolated from cultured cells using RΝA-STAT 60 (Tel Test-B Inc.,
Friendswood, TX), according to the manufacturer's recommendations. Total RΝA (20 mg) was fractionated on a 1.2% agarose gel containing 0.2 M formaldehyde, transferred to GeneScreen™, and hybridized with 32P-dCTP Klenow-labeled, random primed probes.
A human MCP-1 probe was generated from a full length human MCP-1 cDΝA isolated from a human lung library (Clontech), and the template for a human vascular cell adhesion molecule- 1 (VCAM-1) probe was as described in Cybulsky et al. (Am. J. Pathology, 138:815-820, 1991).
The capacity of human umbilical cord endothelial cells (HUVEC) to express MCP -4 mRNA was evaluated as follows. Human umbilical vein endothelial cells
(HUVEC) were obtained at passage two from Clonetics (San Diego, CA) and treated at passage three with the following cytokines: IL-lα (5 ng/ml), IL-4 (20 ng/ml), IFNγ (100 ng/ml), and TNFα (10 ng/ml; all cytokines were obtained from Genzyme, Cambridge, MA). These cells had no detectable constitutive MCP-4 expression, but IL-1 α, TNFα and, to a lesser extent, IFNγ stimulated the accumulation of MCP-4 mRNA within six hours. These levels decreased within 18 hours of stimulation (Fig. 5 A). In contrast to MCP-1 expression, the expression of human MCP-4 was not significantly increased in HUVECs by stimulation with IL-4 (Fig. 5A). TNFα and IFNγ acted synergistically, and IL-4 prolonged the TNFα-induced accumulation of eotaxin mRNA. Human MCP-1 and human vascular cell adhesion molecule (VCAM-1) were used as controls for HUVECs responsiveness to the cytokines tested, and β-actin was used as a control for RNA loading (Fig. 5A).
The ability of two respiratory epithelial cell lines, BEAS-2B and A549, were also tested for their ability to express MCP-4 mRNA in response to cytokine stimulation.
These cells were maintained in Dulbecco's Modified Eagle's Medium (DMEM)/F-12 (BEAS-2B) and F12K (A549) medium. Epithelial cells grown to confluence were stimulated with increasing doses of either IL-lβ, TNFα, or IFNγ, or with 10 nM phorbol 12-mysristate 13-acetate (PMA), transforming growth factor-α (TGFα; 10 ng/ml) or TGFβ (10 ng/ml). Human cells U937 and HL60 were obtained from the American Type
Culture Collection (A.T.C.C, Rockville, MD) and maintained in DMEM. MCP-4 mRNA expression was not detected by Northern analysis in A549 cells in the absence of cytokine stimulation. However, stimulation of the cells with 0.1 ng/ml of IL-lβ or 10 ng/ml of TNFα for 4 hours induced the accumulation of a distinct -850 bp MCP-4 mRNA species (Fig. 5B). The addition of 10 nM PMA to the medium was associated with the presence of a faint but detectable MCP -4 mRNA signal. However, MCP-4 mRNA was not detected after 4 hours of stimulation with 10 ng/ml IFNγ, 10 ng/ml
TGFα, or 5 ng/ml TGFβ (Fig. 5B). Treatment of A549 cells with 10 ng/ml of TNFα induced maximal MCP-4 mRNA expression at four hours, which declined over the subsequent 48 hours (Fig. 5C). Identical results were obtained for IL-lβ-induced accumulation of MCP -4 in A549 cells. Likewise, the addition of TNFα similarly increased MCP-4 mRNA expression in BEAS-2B cells six hours after stimulation, with decreasing levels after 18 hours of stimulation (Fig. 5D). IFNγ acted synergistically with
TNFα and IL-1 to induce MCP-4 mRNA accumulation in both BEAS-2B cells (Fig. 5D) and A549 cells. Furthermore, while IL-4 alone did not induce a detectable expression of MCP-4 in the BEAS-2B line, it clearly acted synergistically with TNF, IL-1 and IFNγ
(Fig 5D).
Example 5:
Expression of MCP -4 in Leukocytes
The expression of eotaxin mRNA was examined in U937 cells, a monocytic tumor line, in HL60 cells, a myelocytic tumor line, and in peripheral blood leukocyte subtypes by the method described in Example 4. U937 cells constitutively expressed MCP-4 mRNA, which was unaffected by IFNγ treatment (Fig. 6A). MCP-4 mRNA was undetectable by Northern blot analysis of total RNA in untreated HL60 cells. However, there was a detectable MCP-4 signal in an eosinophilic subline of HL60 (available from the A.T. CC, Rockville, MD). These cells were maintained in Iscove's Modification of
DMEM supplemented with 0.5 mM butryic acid (Sigma Chemical Co., St Louis, MO) as described by Tomonaga et al., 1986). The medium was supplemented with 10% iron-fortified calf serum (MediaTech, Hendon, VA), 50 U/ml penicillin, 50 mg/ml streptomycin, and 2 mM L-glutamine. Peripheral blood mononuclear cells, peripheral blood neutrophils, untreated peripheral blood eosinophils, peripheral blood eosinophils cultured in the presence of IL-3 for 3 days, Jurkat and H9 T cells, and RM3 B cells did not express detectable MCP-4 mRNA (Fig. 6A, 6B, and 6C). However, MCP-4 mRNA was detected in peripheral mononuclear cells cultured in the presence of PHA and IL-2 for 3 days (Fig 6B). While three HIV-specific cytolytic CD8+ T cell clones induced the expression of RANTES upon activation with an anti-CD3 monoclonal antibody, MCP-4 mRNA was not detected (data for one of the three clones tested is shown in Fig 6B). Example 6: Generation of Biologically Active Recombinant Human MCP -4 Protein
To examine the biological properties of human MCP-4, the protein was expressed and purified from COS cells and E. coli cells. A. Production of Recombinant MCP-4 in E. coli
DNA encoding recombinant human MCP -4, beginning with the putative mature amino-terminal glutamine at position 24 and ending with the carboxy terminal threonine at position 98, was amplified by PCR using the human MCP -4 cDNA as a template. The oligonucleotide primers used were designed to amplify a sequence that contained BamHI sites on both ends, and that would encode a protein with an amino-terminal factor Xa site
(IEGR). Amplified cDNA was subsequently sub-cloned using a TA cloning kit (Invitrogen) and sequenced. The construct was liberated by BamHI digestion, cloned into the BamHI sites of vector PQE8 (Qiagen Inc, Chatsworth, CA), and then transformed into the E. coli strain M15. Expression of MCP-4 in pQE8 resulted in a fusion protein containing an amino-terminal Met-Arg-Gly-Ser-(His)6 (SEQ ID NO.:29) tag, which was purified essentially as described (Rothenberg et al., Mol. Med. 2:334-348, 1996), with the following minor modifications. Recombinant Ml 5 E. coli containing the PQE8 plasmid engineered to express human MCP-4 were induced with isopropyl β-D-thiogalactopyranoside (IPTG) for 2 hours. Following centrifugation, the bacterial pellet was resuspended in 50 mM Tris (pH 8.0), 0.2 M NaCl, 2 mM EDTA, 0.1 mM
PMSF, 1 mM benzamidine, and 5 mM β-mercaptoethanol at 5 ml/gram of wet bacteria. Lysozyme was added to a final concentration of 0.2 mg/ml and the bacterial suspension was incubated on ice for 20 minutes. Triton-X-100 was then added to 1%, and the incubation was resumed for an additional 10 minutes. The suspension was sonicated on ice and centrifuged at 10,000 x g for 20 minutes at 4°C The pellet was resuspended 6 M guanidine HCl, 0.1 M sodium phosphate and 0.01 M Tris HCl (pH 8.0), and applied to a nickel-NTA chromatography column (ProBond", Invitrogen) as described (Rothenberg et al., Mol. Med. 2:334-348, 1996). Recombinant MCP-4 was eluted from the column with 8 M urea buffer (described above) at pH 8.0 containing 0.25 M imidazole. The protein was further purified by high pressure liquid chromatography (HPLC) on a C4 reverse phase semi-preparative column (#214TP510, Vydac, Hesperia CA). Buffer A consisted of Milli Q dH2O/0.1% trifluoroacetic acid (TFA), and buffer B consisted of 95% acetonitrile/0.1% TFA. The sample was loaded in 10% buffer B and run on a linear gradient on a Waters HPLC system. The gradient went from 10% to 20% buffer B over 5 minutes, and from 20% to 90% buffer B over 27 minutes. MCP-4 eluted at -40% buffer B. Fractions corresponding to peaks on the chromatogram were collected and analyzed by SDS-PAGE to ascertain which peak represented human MCP-4. Once this was established, the sample was lyophilized, resuspended in factor Xa buffer (20 mM Tris-HCl, 100 mM NaCl, 2 mM CaCl2, pH 8.0) containing 1 M Urea, and digested for 2 hours at room temperature with factor Xa (Boehringer Mannheim, Indianapolis, IN) at an enzyme/protein weight ratio of 1 : 10. The digested protein was repurified by reverse phase HPLC using the same conditions as above, lyophilized, and resuspended in H2O. The concentration of purified MCP-4 protein was determined by comparing the optical density (OD) at 550 nm to a dilution series of bovine serum albumen, following addition of the BCA reagent (Pierce, Rockford, IL). B. MCP-4 Expression in COS Cells
To express MCP-4 in COS cells, PCR primers were designed to amplify the coding region of human MCP-4 plus a 30 nucleotide sequence encoding a 2 amino acid spacer followed by the 10 amino acid FLAG epitope (GSDYKDDDDK; SEQ ID NO:30). The 30 nucleotide extension was located immediately upstream from the MCP-4 stop codon. The resulting PCR product was subcloned into the pCDNAl/AmpR vector, sequenced, and transfected into COS cells using DEAE-dextran, according to standard methods. Following transfection, cells were maintained in DMEM supplemented with 1% FCS for four days, after which time the supernatant was harvested and centrifuged at 1,000 x g for 15 minutes. MCP-4-FLAG was purified from the supernatant by affinity chromatography using an anti-FLAG monoclonal antibody (M2) column (Eastman Kodak
Co., New Haven, CT) at 4°C. After washing the column with TBS (50 mM Tris-HCl, 15 mM NaCl, pH 7.4) the recombinant MCP4-FLAG protein was eluted with 0.1 M glycine hydrochloride, pH 3.5 and immediately neutralized with 1 M Tris, pH 8.0. The eluted protein was concentrated by ultrafiltration using Centricon 3 (Amicon, Danvers, MA) and dialyzed into phosphate buffered saline, pH 7.4. Recovered protein was quantified using the BCA Protein Assay Reagent (Pierce, Rockford, IL), and approximately 100 ng of protein was electrophoresed into a Tris/Tricine 12.5% SDS-polyacrylamide gel under reducing conditions. The gel was electrotransferred onto a PVDF membrane (Dupont, Boston, MA) and analyzed by Western blotting using an anti-FLAG M2 monoclonal antibody followed by a horseradish peroxidase-conjugated sheep anti-mouse antibody (Kirkegard & Perry, Gaithersburg, MD) and developed by chemiluminescence (Dupont,
Boston, MA).
The MCP-4 cDNA was engineered into the pCDNA-I/Amp expression vector, which bears a C-terminal FLAG tag, and used to transiently transfect COS cells. The supernatant of these cells provided a source of MCP-4 protein that was produced and processed by eukaryotic cells. The FLAG epitope was used to detect and purify MCP-4 from the COS cell supernatant (Fig 7A, lane 4). MCP-4 was expressed in coli as a fusion protein containing an N-terminal histidine tag separated from the predicted mature N-terminal Gin of MCP -4 by a factor Xa consensus site. This strategy was employed to generate MCP-4 protein with the predicted mature N-terminus; the presence of an unmodified amino-terminal amino acid is critical for the biological activity of MCP- 1 and eotaxin. Following nickel affinity chromatography and reverse phase HPLC chromatography, the purified MCP-4 protein (with the tag) migrated as a doublet with an apparent Mr of -10-12 kDa when electrophoresed in a denaturing polyacrylamide gel (Fig 7 A, lane 1). This material exhibited no activity in an assay for monocyte chemotaxis (Fig. 7B), even at 1000 ng/ml. Therefore, the purified histidine tagged-MCP-4 protein was subjected to factor Xa digestion. Analysis by SDS-PAGE of the factor Xa digested (his)ό-tagged MCP-4 protein revealed that the doublet had been converted to a smaller doublet migrating at -8-9 kDa. This change in size is consistent with removal of the six N-terminal histidines. Two additional digestion products were also seen migrating at -6 kDa and -4 kDa. The factor Xa digested MCP -4 protein was repurified by reverse phase
HPLC and tested on elutriated peripheral blood monocytes. The uncut MCP-4 protein had no chemotactic activity, while the factor Xa cut MCP-4 protein exhibited potent and dose-dependent monocyte chemotactic activity (Fig 7B). Due to the fact that additional cleavage of MCP-4 by factor Xa could not be suppressed, subsequent experiments (presented in the Examples below) utilized MCP-4 produced by PeproTech (Rocky Hill,
NJ). Sequence analysis confirmed that this material was a single molecular species containing the mature N-terminal Gin. However, in the Tris/Trice SDS-PAGE gel system, this material also migrated as a doublet at -8 kDa with a dimer apparent at -16 kDa (Fig. 7A, lane 2).
Translation of human MCP-4, beginning with the putative N-terminal Gin residue, predicts an 8,599 Da protein. MCP-4 produced by E. coli migrates at -8 kDa on a 12.5%
Tris/Tricine SDS-polyacrylamide gel, while MCP-4-FLAG migrates at -9 kDa. MCP-4 has no consensus sites for N-linked glycosylation, but it may contain O-linked carbohydrates and sialic residues. However, since the MCP-4-FLAG contains 10 more amino acids at the carboxy terminus than the MCP-4 produced by E. coli, it is likely that MCP-4 is not heavily glycosylated, and may not be glycosylated at all. In addition,
MCP-4 seems to aggregate, as evidenced by the dimer that appears on the Coomassie stained gel.
Example 7: Induction of Chemotaxis
A. MCP -4 Induced Monocyte and Eosinophil Chemotaxis The chemotactic activity of the recombinant human MCP-4 protein was tested on primary human monocytes, neutrophils and eosinophils as follows. Fifty ml of replicate cells at 2 x 106 cells/ml for eosinophils, 1.0 x 106 cells/ml for neutrophils, and 5 x 106 cells/ml for mononuclear cells and monocytes in HBSS buffer (MediaTech) supplemented with 0.05% low endotoxin BSA (Sigma Chemical Co., St Louis, MO), were placed in the top wells of a 48 well micro-chemotaxis chamber (NeuroProbe, Inc.,
Cabin John, MD; Falk et al., J. Immunol. Methods 33:239-247, 1980). A standard 5-μm pore polycarbonate filter (eosinophils and monocytes) and a polyvinylpyrrolidone-free 3-μm pore polycarbonate filter (neutrophils) separated the cells from buffer alone (30 ml), buffer containing recombinant chemokines, or affinity purified FLAG-tagged MCP-4 protein. Cells were incubated at 37 °C (60 minutes for eosinophils, 30 minutes for neutrophils, and 90 minutes for mononuclear cells and monocytes). The cells that migrated across the filter and adhered to the bottom side were stained with Diff-Quick (Baxter Scientific, McGaw Park, IL), and the number of cells per 400X field were counted. MCP-4 exhibited chemotactic activity at concentrations comparable to other chemotactic MCPs, and this activity was as robust as the peak chemotactic responses elicited by these MCPs. However, the other MCPs did begin to exhibit a reproducible chemotactic response at -5-10 ng/ml, while 50 ng/ml of MCP-4 was required to detect a consistent chemotactic response, and the peak MCP-4 effect was seen at 100-1000 ng/ml. This response was observed for both purified monocytes (Fig. 8) and monocytes recruited from a mononuclear cell preparation (Fig. 9A). MCP-4 was also chemotactic for human peripheral blood eosinophils in a dose-dependent manner (Fig. 9B). There was variability in the magnitude of the eosinophil chemotactic response from donor to donor when compared to eotaxin, and variability in the dose-response, but purified eosinophils from all donors tested (n=5) responded to MCP-4. In contrast, recombinant MCP-4 did not elicit chemotaxis from human neutrophils, which responded strongly to IL-8 (Fig. 9C).
Recombinant MCP-4 FLAG protein was also chemotactic for human peripheral blood monocytes and eosinophils, but not neutrophils. Therefore, potential post-translational modifications of MCP-4 and the carboxy-terminal FLAG epitope do not significantly affect the chemotactic activity of MCP -4. B. MCP-5 Induction of Leukocyte Chemotaxis
Murine eosinophils, macrophages, and neutrophils and human mononuclear cells were suspended in HBSS with 0.05% BSA at 2.5, 5, 1, and 5 X 106 cells/ml, respectively, and replicate cells were placed in the top well of a 48 well micro-chemotaxis chamber (Neuro Probe, Inc. Cabin John, MD). A polycarbonate filter with 5-μm pores (eosinophils, macrophages, and mononuclear cells) and a PVP-free filter with 3-μm pores
(neutrophils) separated the cells from buffer alone or buffer containing purified recombinant murine eotaxin, human MCP-1, human IL-8 (PeproTech, Rocky Hill, NJ) or murine MlPlα, murine MlP-lβ, murine JE, murine KC (R&D Systems, Minneapolis, MN). Murine MCP-5 was expressed and purified from E. coli by PeproTech, Inc. as the predicted mature 82 amino acid protein beginning with the N-terminal glycine. N- terminal sequence analysis of the purified recombinant MCP-5 preparation confirmed its homogeneity and the N-terminal glycine. Cells were incubated at 37 °C for 30 minutes (neutrophils), 60 minutes (eosinophils) or 90 minutes (macrophages and mononuclear cells) and the cells that migrated across the filter and adhered to the bottom side of the filter were stained with Diff-Quick (Baxter Scientific, McGaw Park, IL). The chemotactic activity of purified recombinant MCP-5 was evaluated on human peripheral blood monocytes, mouse thioglycolate elicited peritoneal macrophages, mouse eosinophils, and mouse neutrophils. MCP-5 was a potent chemoattractant for human peripheral blood monocytes as was mouse JE and human MCP-1 (Fig. 9D). MCP-5 had no activity on elicited mouse peritoneal macrophages, as was also absent for murine JE, although these macrophages did respond to murine MlP-lα as a positive control
(Fig. 9E). MCP-5 had minimal activity on murine eosinophils and only at doses 1000 ng/ml, while these cells were very responsive to the positive controls, eotaxin (peak chemotaxis at 50 ng/ml 94.5 nM) and MlP-lα, and only minimally responsive to the negative controls, JE and MIP-1 β (Fig. 9F). Murine neutrophils exhibited no response to
MCP-5 but responded strongly to the control agents, mouse KC and human IL-8 (Fig.
9G). These results demonstrate that purified MCP-5 is a potent dose-dependent chemotactic agent for peripheral blood monocytes.
Example 8: MCP-4 Induces the Release of Histamine from Basophils
A. Purification of Monocytes. Eosinophils and Neutrophils
Human mononuclear cells and granulocytes were obtained from whole peripheral blood of non-atopic and mildly atopic donors by passing the blood through a two step (1.077/1.119) Ficoll-Hypaque (Sigma Chemical Co., St. Louis, MO) density gradient.
Monocytes were separated from the mononuclear cell layer by counter current flow elutriation using a JE-6 Beckman Centrifuge (Mason et al., Scand. J. Haematol, 34:5, 1985). The resulting monocyte fraction was estimated by examining Diff-Quick stained cytospin preparations and found to be more than 90% pure. Contaminating cells were lymphocytes. Human eosinophils were purified from the granulocyte layer by negative selection with anti-CD 16-coated immunomagnetic microbeads and the MACS magnet (Hansel et al., J. Immunol. Methods 145:105-110, 1991; Miltenyi Biotech, Auburn, CA). The resulting eosinophil purity was more than 90%, as determined by microscopic examination of Diff-Quick-stained cytospin preparations. Contaminating cells were neutrophils. B. Basophil Histamine Release Assays
Histamine release was determined by a modification of previously reported methods (Bischoff et al., J. Exp. Med., 172:1577-1582, 1990). The peripheral blood of mildly-atopic human donors was subjected to centrifugation over Ficoll-Paque (Pharmacia, Piscataway, NJ) and the top layer of cells was resuspended at 3 X 106 cells/ml in HA buffer (125 mM NaCl, 5 mM KC1, 1 mM MgCl2, 1 mM CaCl2, 0.5 mM glucose, and 0.025% BSA). Aliquots of the cells were warmed to 37°C, exposed to HA buffer alone, or to HA buffer containing either human IL-3 or human IL-5 (Genzyme) at 10 ng/ml for 10 minutes, and then challenged with recombinant human MCP-1, MCP-3, or MCP-4 (PeproTech Inc., Rocky Hill, NJ). After 20 minutes, the cells were centrifuged, and the supernatant was analyzed for histamine content by an enzyme-immunoassay (Immunotech, Westbrook, ME). Histamine release was expressed as the percentage of the total content of the sample (determined after cell lysis by sonication). In order to determine whether MCP -4 is able to stimulate histamine release from basophils, histamine release was measured from basophils that were obtained from mildly atopic donors, either as freshly isolated or primed with IL-3. MCP-4 induced histamine release from primed basophils from 2 of the 4 donors tested (Table 4). MCP-3 had a similar effect in one of the MCP-4 responsive donor tested. In contrast, MCP-1 induced histamine release from IL-3-primed basophils from all four donors.
TABLE 4
Histamine Release (%)
Chemokine
Cytokine None MCP MCP MCP4
1 3
10 ng/ml - 500 500 100 250 500 (ng/ml)
1 0 3.5 1.0 4.2 ND 1.2 ND
IL-3 3.6 29.0 8.0 ND 2.4 ND
2 0 4.2 2.9 2.0 ND ND 0.2
IL-3 4.1 77.0 41.0 ND ND 88.0
3 0 22.0 27.0 ND 28.0 ND 50.0
IL-3 36.0 87.0 ND 77.0 ND 83.0
4 0 0.1 0.2 ND 0.1 ND 0.2
IL-3 0.1 26.0 ND 1.0 ND 4.0
Example 9: Analysis of Calcium Flux
A. Calcium Flux in Mononuclear Cells in Response to MCP-4
To determine whether MCP-4 induces a calcium flux in responding leukocytes, mononuclear cells were loaded with the calcium sensitive dye fura-2 and their response to human MCP-4 was monitored with a fluorimeter. Purified cells were loaded with 5.0 mM of the acetoxymethyl ester of fura-2 (fura-2 AM; Molecular Probes, Eugene, OR) for
60 minutes at 37 °C in the dark at 1 x 107/ml in DMEM supplemented with
1% heat-inactivated FBS. Loaded cells were washed twice and resuspended in a buffer containing 145 mM NaCl, 4 mM KCl, 1 mM NaHPO4, 0.8 mM MgCl2, 1.8 mM CaCl2, 25 mM Hepes and 22 mM glucose. Two ml of cells (2 x 106 cells/ml) were placed in a continuously stirring cuvette at 37° C in a dual-wavelength excitation source fluorimeter
(Photon Technology Inc, South Brunswick, NJ). Changes in the level of cytosolic free calcium were determined after addition of various human chemokines by monitoring the excitation fluorescence intensity emitted at 510 nm in response to sequential excitation at
340 nm and 380 nm. The data are presented as the relative ratio of fluorescence at
340 nm and 380 nm. Concordant with the chemotaxis data presented above, MCP-4 induced a dose- dependent calcium flux in mononuclear cells (Fig. 10A), but not in neutrophils, even though the neutrophils responded appropriately to IL-8 (Fig. IOC). The half-maximal effective concentration of the MCP-4 induced monocyte calcium transient was -35 nM, reaching saturation at -125 nM (Fig. 10A). Rapid successive exposure to the same ligand is known to desensitize the signaling capacity of the G protein-linked receptors that are bound by chemokines. Likewise, exposure of cells to different ligands that utilize the same receptor can result in desensitization. Therefore, other chemokines, such as MCP-1, MCP-3, and RANTES, which are known to induce a calcium flux in human monocytes, were tested for the ability to cross-desensitize the MCP-4 signal. MCP-4 had no apparent effect on subsequent stimulation with either MCP-1 (Fig. 10A) or RANTES (Fig. 10B), even when a maximal initial MCP-4 stimulus was followed by a 20-fold lower MCP-1 stimulus. However, MCP-1 given first at 5 nM was able to completely desensitize the cells to a subsequent 100 nM MCP-4 stimulation (Fig. 10A). MCP-4 and MCP-3 were able to cross-desensitize each other in a dose-dependent manner (Fig. 10B), but MCP-4 and RANTES did not cross-desensitize each other (Fig. 10B). B. Calcium Flux in Leukocytes in Response to MCP-5
Chemokines induce cell migration and activation by binding to specific G protein- coupled, seven transmembrane cell surface receptors on leukocytes. Signalling through these receptors results in a transient calcium flux. In order to determine whether MCP-5 induced a calcium flux in responding leukocytes, and thus examine the leukocyte specificity of MCP-5, purified subsets of leukocytes were loaded with the calcium sensitive dye fura 2, and their response to MCP-5 was monitored by fluorimetry.
Monocytes, macrophages, eosinophils and neutrophils were purified as follows. Peripheral blood mononuclear cells (PBMCs) were obtained from normal donors by density gradient centrifugation using 1.077 Histopaque™ (Sigma Chemical Co., St. Louis, MO). Murine eosinophils were isolated from the spleens of IL-5 transgenic mice by immuno-magnetic separation to remove the contaminating splenocytes, as described by Rothenberg et al., Mol. Med. 2:334-348, 1996). This process included subjecting splenocytes to hypotonic lysis in order to remove erythrocytes. The remaining leukocytes were labeled with anti-Thy-1 (M5/49), anti-B220 (6B2), or anti-Lyt-2 (53-6.7). The antibody labeled cells were treated with sheep anti-rat serum coated-magnetic microbeads, and eosinophils were enriched by negative selection through a MACS magnet (Miltenyi Biotech., Aubum, CA). The purity of the eosinophils was greater than 90%, according to microscopic examination of Diff-Quick-stained (Baxter Scientific, McGaw Park, IL) cytospin preparations. The contaminating cells were mononuclear.
Macrophages were isolated from the peritoneal cavity of mice that had been treated 2 days prior with 1 ml of 2.5% Brewers thioglycollate (by intraperitoneal injection; Difco, Detroit, CA; Luo et al, J. Immunol. 153:4616-4624, 1994). Neutrophils were isolated from the peritoneal cavity of mice that had been treated either 4 or 18 hours previously with 1 mg of 9% sodium casein (by intraperitoneal injection; Sigma Chemical Co.,
St. Louis, MO). Macrophages and neutrophils were further purified by centrifugation in self-forming Percoll gradients. The resulting preparations were typically more than 90% pure.
Purified cells were loaded with 5.0 μM of the acetoxymethyl ester of fura-2 (fura- 2 AM; Molecular Probes, Eugene, OR) for 60 minutes at 37° C in the dark at 1 x 107 cells/ml in DMEM supplemented with 1% heat-inactivated FBS. Loaded cells were washed twice and resuspended in a buffer containing 145 mM NaCl, 4 mM KCl, 1 mM NaHPO4, 0.8 mM MgCl2, 1.8 mM CaCl2, 25 mM HEPES and 22 mM glucose. Two mis of cells (5 x 106 cells/ml) were placed in a continuously stirring cuvette at 37°C in a dual- wavelength excitation source fluorimeter (Photon Technology Inc., South Brunswick,
NJ). Changes in cytosolic free calcium were determined after addition of the chemokines by monitoring the excitation fluorescence intensity emitted at 150 nm in response to sequential excitation at 340 nm and 380 nm. The data are presented as the relative ratio of fluorescence at 340 and 380 nm. MCP-5 induced a dose-dependent calcium flux in mononuclear cells (Fig. 1 IC) but not in eosinophils, even at 50 μg/ml or 5 μM, or in neutrophils. As controls, the purified eosinophils responded appropriately to eotaxin and MlP-lα (Fig. 1 ID) but did not respond to JE or MlP-lβ. Furthermore purified neutrophils responded appropriately to KC and IL-8 (Fig. 1 IE). The half-maximal effective concentration of the MCP-5- induced mononuclear cell calcium transient was approximately 7.5 ng/ml (75 nM) and reached saturation at approximately 500 ng/ml (50 nM; Fig. 1 IF). These results demonstrate that MCP-5 induces a calcium flux in mononuclear cells, but not in eosinophils or neutrophils.
Example 10: Receptor Analysis A. CKR2b and CKR3 are MCP-4 Receptors To determine whether MCP-4 utilizes one of the known CC chemokine receptors, a panel of HEK-293 cell lines selected for stable expression of CKRs was used to test the ability of MCP-4 to induce a calcium transient (Fig. 11 A). HEK293 cells were transfected with pREP9 expression plasmids encoding CC CKR2B, CC CKR3 and CC CKR5, and a pCEP4 expression plasmid encoding CC CKR1 as previously described. Individual colonies resistant to G418 (1 mg/ml) or hygromycin (250 μg/ml) were selected and expanded. The derivation of these cell lines and their selectivity for CC chemokines other than MCP-4 have been previously reported. Cells were grown to confluence in DMEM with 10% FBS and G418 or hygromycin, harvested in PBS, loaded with 2.5 mM of fura-2 AM at 37° C for 45 minutes, and then washed twice in PBS. Cells (at 1 x lOTml HBSS pH 7.4) were placed in a continuously stirred cuvette at 37 °C in an MSIII fluorimeter (Photon Technology Inc., South Brunswick, NJ). Fluorescence was monitored at excitation frequencies of 340 nm and 380 nm, and emission frequencies of 510 nm. The data were collected every 200 msec, and presented as the relative ratio of fluorescence excited at 340 nm and 380 nm. MCP-4 at 50 nM induced a calcium flux response in the CKR2B and CKR3 transfectants, but not in the CKRl or CKR5 transfectants. The CKR1 cell line responded to MlP-lα, RANTES, and MCP-3, and the CKR5 cell line responded to MlP-lα, MlP-lβ, and RANTES, confirming expression of the corresponding receptors in these MCP-4 unresponsive targets. Both the CKR2B and CKR3 cell lines responded to MCP-4 in a concentration-dependent manner with half-maximal effective concentrations of 15 and 20 nM, respectively, and reached saturation at 100 nM in each case (Fig. 1 IB). The EC50 values are -2-fold greater than those previously defined for eotaxin at CKR3 and for MCP-1 and MCP-3 at CKR2B using the calcium flux response in the same transfectants. Cross-desensitization experiments were also performed on the CKR2b and CKR3 cell lines. As in mononuclear cells MCP-1 desensitized the MCP-4 response in the
CKR2B cell line (Fig. 12A), although MCP-4 and MCP-3 did not extinguish the MCP-1 response. MCP-3 cross-desensitized the MCP-4 response but, in contrast to mononuclear cells, MCP-4 did not cross-desensitize the MCP-3 response. Using the CKR3 cell line (Fig. 12B) we were able to demonstrate that eotaxin and MCP-4 cross-desensitized each other using the same concentration of ligand. This discrepancy has been seen with other chemokine-induced calcium transients on leukocytes vs. a cell line-expressing a cloned chemokine receptor. The mechanism of homologous and heterologous desensitization for chemokine receptors may involve both physical sequestration as well as functional inactivation by phosphorylation. The ability of each chemokine to induce these processes could differ in different cell types, even for the same receptor, and could account for the asymmetric desensitization patterns observed in primary leukocytes and receptor transfectants.
B. CCR2 is an MCP-5 Receptor
Rapid and successive exposure to the same ligand desensitizes the signalling capacity of G protein-linked receptors. Likewise, exposure of cells to different ligands that utilize the same receptor signalling pathway can also result in desensitization.
Therefore, the ability of other chemokines, such as mouse JE and human MCP-1, which are known to induce a calcium flux in human monocytes, were tested for their ability to cross-desensitize the MCP-5 signal. MCP-5 at 100 nM was able to completely desensitize the cells to subsequent stimulation with a 100 mM JE or human MCP-1 (Fig. 1 IC). This effect was dose-dependent. When the initial dose of MCP-5 was lowered to 1 nM, the mononuclear cells were no longer desensitized to subsequent stimulation with either 100 nM JE or MCP-1. In addition, 100 nM MCP-5 completely desensitized a subsequent cellular response to a 100 nM MCP-5 challenge. In contrast, mononuclear cells initially stimulated with 100 nM JE were not desensitized to a subsequent stimulation with 100 nM MCP-5 (Fig. 1 IC). Initial treatment of eosinophils and neutrophils with 100 nM MCP-5 had no effect on the ability of eosinophils to respond to 10 nM eotaxin or 10 mM MlP-l (Fig. 1 ID), or on the ability of neutrophils to respond to 10 nM KC (Fig. 1 IE). These results suggest that MCP-5 uses a receptor signalling pathway that is held in common by JE and MCP-1 and that is different from the receptor pathway utilized by MlP-lα, MlP-β, eotaxin, and KC. The studies of receptor desensitization described above suggested that MCP-5 shares a receptor with JE and MCP-1, and the signalling studies of eosinophils suggested that MCP-5 does not signal through the two receptors known to be expressed on eosinophils, CCR3 and CCR1. In order to directly test these suggestions, a panel of HEK-293 cell lines selected for stable expression of CCRs was used to examine the ability of MCP-5 to induce calcium flux.
HEK-293 cells stably expressing human CCR2b, CCR1, and CCR3, and murine CCR2 and CCR5 with the "FLAG" epitope at the extreme amino-terminus were prepared as described by Boring et al. (J. Biol. Chem. 271:7551-7558, 1996) and Monteclaro et al. (J. Biol. Chem. 271 : 19084-19092, 1996). The cells were grown in MEM with Eagle's salts supplemented with 10% fetal calf serum, 100 μg/ml penicillin, 100 μg/ml streptomycin, and 800 μg/ml G418 (Life Technologies Inc., Gaithersburg, MD). For calcium fluorimetry, cells were grown to log phase, loaded with the calcium-specific dye indo-1 AM (Molecular Probes, Eugene, OR), and assayed by spectro fluorimetry for changes in the concentration of intracellular calcium in response to addition of chemokines.
MCP-5 (100 nM) reproducibly induced a robust intracellular calcium flux in cells expressing CCR2 (Fig. 12B) but not CCR1, CCR3, or CCR5. In control experiments, functional expression of the cloned receptors in these MCP-5 unresponsive targets was confirmed: the CCR1- and CCR5-expressing cell lines responded to MlP-lα, and the
CCR3 -expressing line responded to eotaxin. Dose-response experiments demonstrated that MCP-5 was an excellent ligand for human CCR2b (EC50 < 5 nM) as was MCP-1, and both were more potent ligands than JE (Fig. 12B (i)). Since the MCP-5 described herein is a murine protein, it was compared directly to JE for activation of the murine MCP-1 receptor, CCR2. As shown in Fig. 12B (ii), JE induced a more robust intracellular calcium flux than MCP-5 in HEK-293 cells stably expressing murine CCR2.
Desensitization studies using cells transfected with human CCR2b revealed that MCP-5 blocked subsequent responses to MCP-5, MCP-1, and JE (Fig. 12C). Similar results were obtained when the initial agonist was MCP-1 or JE (Fig. 12C). However, MCP-1 did not completely desensitize the CCR2b transfectants to subsequent MCP-5 stimulation (Fig. 12C). These data are consistent with the desensitization responses observed on human PBMCs, and with the hypothesis that MCP-5 is a full agonist for the human MCP-1 receptor (CCR2b).
Other Embodiments The studies described herein, in which the novel chemokines MCP-4 and MCP-5 were discovered and characterized, provide the means to control the movement of inflammatory cells and, thereby, alter the immune response. For example, selectively suppressing these chemokines can be used to suppress the immune response in the event of an inflammatory condition such as asthma, atherosclerosis, arthritis, adult respiratory distress syndrome, multiple sclerosis, and glomerulonephritis. In addition, administration of MCP-4 or MCP-5, or the receptor binding domains of these proteins, may prevent infection from pathogens that utilize chemokine receptors to enter the cell. For example, the human immunodeficiency virus (HIV) is thought to be an infectious agent that uses the chemokine receptor binding mechanism. Therefore, blocking receptor binding could prevent viral entry into the cell. Conversely, administering MCP-4 or MCP-5 may enhance the immune response, e.g., in the event of an infection or tumor. MCP-4 and MCP-5 Protein Expression
In general, the polypeptides of the invention, MCP-4 and MCP-5, can be produced by transforming suitable host cells with all or part of the corresponding cDNA in a suitable expression vehicle. Those skilled in the art of molecular biology will understand that any of a wide variety of expression systems may be used to provide the recombinant poiypeptide. The precise host cell used is not critical to the invention. MCP-4 or MCP-5 polypeptides may be produced in a prokaryotic host, such as the bacterium E. coli, or in a eukaryotic host, such as an insect cell (e.g., Sf21 cells), or a mammalian cells (e.g., COS cells, NTH 3T3 cells, or HeLa cells). These cells are available from many sources, including the American Type Culture Collection (Rockville, MD; see also Ausubel et al., Current Protocols in Molecular Biology, John Wiley & Sons, New York, NY, 1994). For a detailed description of the production of MCP-4 by E. coli, see Example 6 above.
The method of transduction and the expression vehicle will depend on the host system selected. Transduction methods are described by Ausubel et al. (supra), and expression vehicles may be chosen from those widely known, such as those described in Cloning Vectors: A Laboratory Manual (P.H. Pouwels et al, 1985, Supp. 1987).
A preferred expression system is the baculovirus system, in which the expression vector pBacPAK9 (Clontech, Palo Alto, CA) is frequently used. If desired, this system may be used in conjunction with other protein expression techniques, for example, the myc tag technique described by Evan et al., (Mol. Cell. Biol. 5:3610-3616, 1985).
Alternatively, MCP-4 or MCP-5 may be produced by stably-transfected mammalian cell lines. Numerous vectors suitable for stable transfection of mammalian cells are available to the public (see Pouwels et al., supra). Methods for constructing stably-transfected mammalian cell lines are also publically available (e.g., in Ausubel et al., supra).
In addition to the example provided above, cDNA encoding all or part of the MCP-4 or MCP-5 poiypeptide can be cloned into an expression vector, such as pCVSEII- DHFR or pAdD26SV(A), which includes the dihydrofolate reductase (DHFR) gene. Integration of the plasmid containing the MCP-4- or MCP-5-encoding gene into the host cell chromosome is selected for by inclusion of 0.01-300 μM methotrexate in the cell culture medium (as described in Ausubel et al., supra). This type of selection can be accomplished in most cell types. Preferably, the cells will be DHFR-deficient, such as CHO DHFR* cells (A.T.C.C Accession No. CRL 9096). Recombinant protein expression can be increased by DHFR-mediated amplification of the transfected gene.
Methods for selecting cell lines bearing gene amplifications, which generally involve extended cell culture in medium containing increasing levels of methotrexate, are described in Ausubel et al. (supra).
Alternatively, the proteins of the invention, particularly fragments thereof, can be produced by chemical synthesis (e.g., using the methods described in Solid Phase Peptide
Synthesis, Second Edition, The Pierce Chemical Company, Rockford, IL, 1984).
Once the recombinant MCP-4 or MCP-5 proteins are expressed, they can be isolated by standard biochemical techniques, such as affinity chromatography, and purified further, e.g., by high performance liquid chromatography (see, e.g., Fisher, Laboratory Techniques in Biochemistry and Molecular Biology, Eds., Work and Burdon,
Elsevier, 1980). As described above, MCP-4 has an N-terminal glutamine residue, as does MCP-1, MCP-2, and MCP-3. The biological activity of the expressed poiypeptide may be substantially reduced either by addition of one or more amino acids prior to the N- terminal glutamine, or the deletion of one or more N-terminal residues. Addition of such amino acids may be particularly desirable for generating antiviral and anti-inflammatory poiypeptide compounds. Such mutants may be produced by standard methods of genetic engineering and used to suppress the immune response or block pathogen entry via chemokine receptors. It is likely that, when administered in excess, the
N-terminal mutant MCP-4 or MCP-5 poiypeptide will suppress the immune response by effectively competing for the limited number of cell surface receptors, thereby blocking the response of these cells to the wild-type chemokine signals. As discussed below, the gene encoding such mutant polypeptides can also be administered. Generation of Anti-MCP-4 or Anti-MCP-5 Antibodies In order to generate MCP-4 or MCP-5 -specific antibodies, the full-length MCP-4 or MCP-5 coding sequences or portions thereof can be expressed as C-terminal fusion molecules with glutathione S-transferase (GST; Smith et al., Gene 67:31-40, 1988). The coding sequence may encode either the full-length proteins, or fragments of these proteins that are likely to be antigenic, such as fragments that lie outside highly conserved regions or that contain a high proportion of charged residues. The fusion protein, which should conform to a predicted size, can then be purified with glutathione-Sepharose beads, eluted with glutathione, cleaved with thrombin (at an engineered cleavage site), and purified to the degree necessary for immunization of rabbits. Primary immunizations are typically carried out the Freund's complete adjuvant and subsequent immunizations with Freund's incomplete adjuvant. Antibody titres are monitored by Western blot and immunoprecipitation analyses using the thrombin-cleaved MCP-4 or MCP-5 protein fragment of the GST-MCP-4 or GST-MCP-5 fusion protein. Immune sera are affinity purified using CNBr-Sepharose-coupled MCP -4 or MCP-5 proteins. The specificity of the antiserum can be determined using a panel of unrelated GST fusion proteins (such as GST-p53, and GST-Rb) and GST-trypsin (which may be generated by PCR using published sequences). As an alternate or adjunct immunogen, peptides corresponding to relatively unique regions of MCP-4 or MCP-5 (i.e. peptides that can be used to generate antibodies that specifically bind MCP -4 or MCP-5) may be generated and coupled to keyhole limpet hemocyanin (KLH) through an introduced C-terminal lysine. Antiserum to each of these peptides is then similarly affinity purified on peptides conjugated to BSA, and specificity is tested by ELISA and Western blot analysis using peptide conjugates.
Alternatively, monoclonal antibodies may be prepared using the MCP -4 and MCP-5 proteins described herein and standard hybridoma technology (see, e.g., Kohler et al., Nature 256:495, 1975; Kohler et al, Eur. J. Immunol. 6:511, 1976; Kohler et al., Eur. J. Immunol. 6:292, 1976; Hammerline et al., In Monoclonal Antibodies and T Cell
Hybridomas, Elsevier, New York, NY, 1981). Once produced, monoclonal antibodies can be tested for their ability to specifically bind MCP-4 or MCP-5, as appropriate, by Western blot or immunoprecipitation analyses. Antibodies that specifically recognize MCP-4 or MCP-5 are considered useful in the invention; antibodies that can block the activity of MCP -4 or MCP-5 would be useful as agents to specifically block the accumulation of leukocytes (particularly monocytes) in unwanted instances of inflammation.
Generation of Transgenic Animals
Methods for generating transgenic animals, which incorporate an exogenous DNA molecule into their genome and pass it to their offspring, are well known to skilled artisans. Typically, the exogenous DNA is introduced into single-celled embryos that are obtained by mating male and female mice from a defined inbred genetic background (Brinster et al., Proc. Natl. Acad. Sci. USA 82:4438-4442, 1985). If the mating is successful, the female mouse is sacrificed and the fertilized eggs are removed from the reproductive system. At this time, pronuclei are visible under the light microscope.
Exogenous DNA, such as the MCP-4 or MCP-5 cDNA disclosed herein, is then introduced into the egg. Typically, the DNA is microinjected into a pronucleus, but other methods of transfection may also be used. A number of the microinjected eggs are then implanted into a pseudopregnant female mouse (pseudopregnancy is achieved by mating a normal female mouse with a vasectomized male mouse). These eggs will develop for the full period of gestation. The surrogate mother delivers the mice that develop from these eggs, which are then tested (when they are approximately 4 weeks old) for the presence of the exogenous DNA.
Identification and Administration of Compounds that Modulate Expression of MCP-4 and/or MCP-5 Isolation of the MCP-4 and MCP-5 cDNAs also facilitates the identification of molecules that increase or decrease the expression of these genes in vivo. Many quantitative assays for gene expression are known and may be utilized in this aspect of the invention. A few examples are provided below.
In order to identify molecules that modulate MCP-4 or MCP-5 gene expression, candidates are added at varying concentrations to the culture medium of cells expressing either MCP-4 or MCP-5 mRNA. MCP-4 or MCP-5 expression is then measured, for example, by Northern blot analysis using MCP -4 or MCP-5 cDNA hybridization probes. The level of MCP-4 or MCP-5 expression in the presence of the candidate molecule is compared with that measured in its absence. Alternatively, the effect of the candidate molecule may be assessed at the level of translation by measuring the level of MCP-4 or MCP-5 protein, for example, by Western blot analysis or immunoprecipitation with an MCP-4 or MCP-5 -specific antibody. If the modulatory compound is an inhibitory compound, it may function by competing with MCP-4 or MCP-5 for the limited number of chemokine receptors on the cell surface. These compounds may be identified by, for example, examining the binding of MCP-4 or MCP-5 to their respective receptors in the presence and absence of the candidate compound. A compound that blocks chemokine receptors may be especially useful as an antiviral agent because some viruses, including the human immunodeficiency virus that causes AIDS, gain entry to the cell via chemokine receptors. Candidate modulators may be purified, substantially purified, or may remain as one component of a mixture of compounds (e.g., as an extract or a supernatant obtained from a cell culture). In an assay of mixed compounds, MCP -4 or MCP-5 expression can be tested against progressively smaller subsets of the candidate compound pool. These subsets may be produced, e.g., by standard purification techniques such as HPLC or FPLC, until a single compound or a minimal group of compounds is demonstrated to modulate the expression of MCP-4 and/or MCP-5. Alternatively, or in addition, candidate modulatory compounds may be screened for those which modulate the chemotaxis of, e.g., monocytes, toward MCP-4 or MCP-5 producing cells. In this approach, chemotactic activity is simply compared in any standard assay of chemotaxis (including those described herein), under otherwise equivalent conditions, in the presence and absence of the candidate compound. Candidate modulatory compounds include peptide and non-peptide molecules, such as those found in cell extracts, mammalian serum, growth medium in which mammalian cells have been grown, or synthetic compounds. Particularly useful modulators of MCP-4 or MCP-5 expression include tumor necrosis factor-α, interleukin-1, interleukin-4, and interferon- gamma.
A molecule that decreases MCP-4 or MCP-5 expression or MCP-4 or MCP-5 chemotactic activity is considered particularly useful in the invention. This molecule may be used, for example, as a therapeutic to decrease the cellular levels of MCP-4 or MCP-5 expression, and thereby decrease the symptoms associated with MCP-4 or MCP-5 expression, including asthmatic reaction, chronic obstructive pulmonary diseases, bronchiectasis, cystic fibrosis, and inflammatory bowel diseases (i.e., Crohn's Disease and ulcerative colitis). Compounds that alleviate the inflammation associated with sinusitis are of particular interest. Infections, such as parasitic infections may also be treated with a molecule that inhibits MCP-4 or MCP-5 expression. Conversely, a molecule that enhances MCP-4 or MCP-5 expression may be used in the treatment of cancer. As described below, enhanced leukocyte chemotaxis to the site of the tumor is desirable.
Modulatory compounds may be administered with a pharmaceutically-acceptable diluent, carrier, or excipient, according to conventional pharmaceutical practice. Any appropriate route of administration may be employed. For example, administration can be parenteral, intravenous, subcutaneous, intramuscular, intracranial, intraorbital, opthalmic, intraventricular, intracapsular, intraspinal, intracistemal, intraperitoneal, transmucosal, or oral. The modulatory compound can be formulated in various ways, according to the corresponding route of administration. For example liquid solutions can be made for ingestion or injection; gels or powders can be made for ingestion, inhalation, or topical application. Methods for making such formulations are well known and can be found in, for example, "Remington's Pharmaceutical Sciences." MCP-4 or MCP-5-based Therapies
To enhance, for example, the infiltration of leukocytes into a tumor, a functional MCP-4 or MCP-5 gene or poiypeptide may be administered to the patient.
Retroviral vectors, adenoviral vectors, adeno-associated viral vectors, and other viral vectors with the appropriate tropism may be used as a gene transfer delivery system for a therapeutic eotaxin gene construct. Suitable vectors are generally known (see, e.g., Miller, Human Gene Therapy 15:14, 1990; Firedman, Science 244:1275-1281, 1989; Eglitis et al, BioTechniques 6:608-614, 1988; Tolstoshev et al., current Opinion in biotechnology 1:55-61, 1990). Retroviruses are particularly well developed and have been used in clinical settings (Rosenberg et al., N. Engl. J. Med 323:370, 1990; Anderson et al., U.S. Patent No. 5,399,346).
Gene therapies that are not mediated by viral vectors may also be employed. For example, nucleic acid molecules encoding all or part of MCP-4 or MCP-5 may be encapsulated in liposomes and administered by lipofection (Feigner et al., Proc. Natl. Acad. Sci. USA 84:7413, 1987; Ono et al, Neuroscience Lett. 117:259, 1990; Brigham et al., Am. J. Med. Sci. 298:278, 1989; Staubinger and Papahadjopoulos, Meth. Enzymol. 101 :512, 1983). Alternatively, the cells may be transfected by application of the nucleic acid molecules conjugated with asialorosonucoid-polylysine (Wu et al., J. Biol. Chem.
263:14621, 1988; Wu et al., J. Biol. Chem. 264:16985, 1989). Alternatively, MCP-4 or MCP-5 mRNA may be administered directly. The mRNA can be produced by any standard technique, but is most readily produced by in vitro transcription using an MCP-4 or MCP-5 cDNA under the control of a high efficiency promoter (e.g., the T7 promoter). Preferably, the therapeutic MCP-4 or MCP-5 DNA construct is applied to the site of the malignancy or inflammation, to the tissue in the larger vicinity of a malignancy or inflammation, or to the blood vessels supplying these areas. The DNA construct administered may contain any suitable promoter, from which MCP -4 or MCP-5 gene expression is directed. Examples of such promoters include the human cytomegalovirus promoter, the simian virus 40 promoter, and the metallothionein promoter. Alternatively, the promoter may be cell type-specific. For example, the tyrosine hydroxylase promoter may be used to direct gene expression within catecholinergic neurons.
Ideally, the production of MCP-4 or MCP-5 by any gene therapy approach described herein will result in a cellular level of MCP -4 or MCP-5 expression that is at least equivalent to the normal, cellular level of expression of these genes. Skilled artisans will recognize that these therapies may be used in combination with more traditional therapies, such as surgery, radiotherapy, or chemotherapy.
Additional embodiments are within the following claims.
What is claimed is:

Claims

SEQUENCE LISTING (1) GENERAL INFORMATION (i) APPLICANT: The General Hospital Corporation (ii) TITLE OF THE INVENTION: MCP-4 AND MCP-5 : NOVEL CHEMOKINES (iii) NUMBER OF SEQUENCES: 30(iv) CORRESPONDENCE ADDRESS:(A) ADDRESSEE: Clark & Elbing LLP(B) STREET: 176 Federal Street(C) CITY: Boston(D) STATE: MA(E) COUNTRY: USA(F) ZIP: 02110(v) COMPUTER READABLE FORM:(A) MEDIUM TYPE: Diskette(B) COMPUTER: IBM Compatible(C) OPERATING SYSTEM: DOS(D) SOFTWARE: FastSEQ for Windows Version 2.0(vi) CURRENT APPLICATION DATA:(A) APPLICATION NUMBER:(B) FILING DATE:(C) CLASSIFICATION:(vii) PRIOR APPLICATION DATA:(A) APPLICATION NUMBER: 60/027,128(B) FILING DATE: 30-SEP-1996(viii) ATTORNEY/ GENT INFORMATION:(A) NAME: Bieker-Brady, Kristina(B) REGISTRATION NUMBER: 39,109(C) REFERENCE/DOCKET NUMBER: 00786/293WO2(ix) TELECOMMUNICATION INFORMATION:(A) TELEPHONE: 617-428-0200(B) TELEFAX: 617-428-7045(C) TELEX:(2) INFORMATION FOR SEQ ID NO : 1 :(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 823 base pairs(B) TYPE: nucleic acid(C) STRANDEDNESS: single (D) TOPOLOGY: linear(ii) MOLECULE TYPE: Other(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 1 :AACATTGTGA AATCTCCAAC TCTTAACCTT CAACATGAAA GTCTCTGCAG TGCTTCTGTG 60CCTGCTGCTC ATGACAGCAG CTTTCAACCC CCAGGGACTT GCTCAGCCAG ATGCACTCAA 120CGTCCCATCT ACTTGCTGCT TCACATTTAG CAGTAAGAAG ATCTCCTTGC AGAGGCTGAA 180GAGCTATGTG ATCACCACCA GCAGGTGTCC CCAGAAGGCT GTCATCTTCA GAACCAAACT 240GGGCAAGGAG ATCTGTGCTG ACCCAAAGGA GAAGTGGGTC CAGAATTATA TGAAACACCT 300GGGCCGGAAA GCTCACACCC TGAAGACTTG AACTCTGCTA CCCCTACTGA AATCAAGCTG 360GAGTACGTGA AATGACTTTT CCATTCTCCT CTGGCCTCCT CTTCTATGCT TTGGAATACT 420TCTACCATAA TTTTCAAATA GGATGCATTC GGTTTTGTGA TTCAAAATGT ACTATGTGTT 480AAGTAATATT GGCTATTATT TGACTTGTTG CTGGTTTGGA GTTTATTTGA GTATTGCTGA 540TCTTTTCTAA AGCAAGGCCT TGAGCAAGTA GGTTGCTGTC TCTAAGCCCC CTTCCCTTCC 600ACTATGAGCT GCTGGCAGTG GGTTTGTATT CGGTTCCCAG GGGTTGAGAG CATGCCTGTG 660GGAGTCATGG ACATGAAGGG ATGCTGCAAT GTAGGAAGGA GAGCTCTTTG TGAATGTGAG 720GTGTTGCTAA ATATGTTATT GTGGAAAGAT GAATGCAATA GTAGGACTGC TGACATTTTG 780CAGAAAATAC ATTTTATTTA AAATCTCCTA AAAAAAAAAA AAA 823(2) INFORMATION FOR SEQ ID NO : 2 :(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 98 amino acids(B) TYPE: amino acid(C) STRANDEDNESS: unknown(D) TOPOLOGY: linear(ii) MOLECULE TYPE: protein(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 2 :Met Lys Val Ser Ala Val Leu Leu Cys Leu Leu Leu Met Thr Ala Ala1 5 10 15Phe Asn Pro Gin Gly Leu Ala Gin Pro Asp Ala Leu Asn Val Pro Ser20 25 30Thr Cys Cys Phe Thr Phe Ser Ser Lys Lys lie Ser Leu Gin Arg Leu35 40 45Lys Ser Tyr Val lie Thr Thr Ser Arg Cys Pro Gin Lys Ala Val lie50 55 60Phe Arg Thr Lys Leu Gly Lys Glu lie Cys Ala Asp Pro Lys Glu Lys 65 70 75 80Trp Val Gin Asn Tyr Met Lys His Leu Gly Arg Lys Ala His Thr Leu85 90 95Lys Thr(2) INFORMATION FOR SEQ ID NO : 3 ;(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 514 base pairs(B) TYPE: nucleic acid(C) STRANDEDNESS: single(D) TOPOLOGY: linear (ii) MOLECULE TYPE: Other(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 3 :CTAGCTTTCA TTTCGAAGTC TTTGACCTCA ACATGAAGAT TTCCACACTT CTATGCCTCC 60TGCTCATAGC TACCACCATC AGTCCTCAGG TATTGGCTGG ACCAGATGCG GTGAGCACCC 120CAGTCACGTG CTGTTATAAT GTTGTTAAGC AGAAGATTCA CGTCCGGAAG CTGAAGAGCT 180ACAGGAGAAT CACAAGCAGC CAGTGTCCCC GGGAAGCTGT GATCTTCAGG ACCATACTGG 240ATAAGGAGAT CTGTGCTGAC CCCAAGGAGA AGTGGGTTAA GAATTCCATA AACCACTTGG 300ATAAGACGTC TCAAACCTTC ATCCTTGAAC CTTCATGTCT AGGCTGAGAG TTCCAAAAAC 360TCTTACGTAT TTCCCCCTGA AGTTCCCCAC GGGCAGTGTG ATATTTATTA TGATATCTAA 420AAAGAGATGT TTTTAATAAT TTAAACAAAC TTGCTTAAAT AATATTTAAT GGTATTTAAG 480TAATATTTGG GCCAATTAAA CCGAATCTAA TTTA 514(2) INFORMATION FOR SEQ ID NO : 4 :(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 104 amino acids(B) TYPE: amino acid(C) STRANDEDNESS: unknown(D) TOPOLOGY: linear(ii) MOLECULE TYPE: protein(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 4 :Met Lys lie Ser Thr Leu Leu Cys Leu Leu Leu lie Ala Thr Thr lie1 5 10 15Ser Pro Gin Val Leu Ala Gly Pro Asp Ala Val Ser Thr Pro Val Thr20 25 30Cys Cys Tyr Asn Val Val Lys Gin Lys lie His Val Arg Lys Leu Lys35 40 45Ser Tyr Arg Arg lie Thr Ser Ser Gin Cys Pro Arg Glu Ala Val lie50 55 60Phe Arg Thr lie Leu Asp Lys Glu lie Cys Ala Asp Pro Lys Glu Lys 65 70 75 80Trp Val Lys Asn Ser lie Asn His Leu Asp Lys Thr Ser Gin Thr Phe85 90 95 lie Leu Glu Pro Ser Cys Leu Gly 100(2) INFORMATION FOR SEQ ID NO : 5 :(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 99 amino acids(B) TYPE: amino acid(C) STRANDEDNESS: unknown(D) TOPOLOGY: linear(ii) MOLECULE TYPE: protein(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 5 :Met Lys Val Ser Ala Ala Leu Leu Cys Leu Leu Leu lie Ala Ala Thr 1 5 10 15 Phe lie Pro Gin Gly Leu Ala Gin Pro Asp Ala lie Asn Ala Pro Val20 25 30Thr Cys Cys Tyr Asn Phe Thr Asn Arg Lys lie Ser Val Gin Arg Leu35 40 45Ala Ser Tyr Arg Arg lie Thr Ser Ser Lys Cys Pro Lys Glu Ala Val50 55 60 lie Phe Lys Thr lie Val Ala Lys Glu lie Cys Ala Asp Pro Lys Gin 65 70 75 80Lys Trp Val Gin Asp Ser Met Asp His Leu Asp Lys Gin Thr Gin Thr85 90 95Pro Lys Thr(2) INFORMATION FOR SEQ ID NO : 6 :(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 77 amino acids(B) TYPE: amino acid(C) STRANDEDNESS: unknown(D) TOPOLOGY: linear(ii) MOLECULE TYPE: protein(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 6 :Ala Gin Pro Asp Ser Val Ser lie Pro lie Thr Cys Cys Phe Asn Val1 5 10 15 lie Asn Arg Lys lie Pro lie Gin Arg Leu Glu Ser Tyr Thr Arg lie20 25 30Thr Asn lie Gin Cys Pro Lys Glu Ala Val lie Phe Lys Thr Lys Arg35 40 45Gly Lys Glu Val Cys Ala Asp Pro Lys Glu Arg Trp Val Arg Asp Ser50 55 60Met Lys His Leu Asp Gin lie Phe Gin Asn Leu Lys Pro 65 70 75(2) INFORMATION FOR SEQ ID NO : 7 :(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 99 amino acids(B) TYPE: amino acid(C) STRANDEDNESS: unknown(D) TOPOLOGY: linear(ii) MOLECULE TYPE: protein(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 7 :Met Lys Ala Ser Ala Ala Leu Leu Cys Leu Leu Leu Thr Ala Ala Ala1 5 10 15Phe Ser Pro Gin Gly Leu Ala Gin Pro Val Gly lie Asn Thr Ser Thr20 25 30Thr Cys Cys Tyr Arg Phe lie Asn Lys Lys lie Pro Lys Gin Arg Leu35 40 45Glu Ser Tyr Arg Arg Thr Thr Ser Ser His Cys Pro Arg Glu Ala Val 50 55 60 lie Phe Lys Thr Lys Leu Asp Lys Glu lie Cys Ala Asp Pro Thr Gin 65 70 75 80Lys Trp Val Gin Asp Phe Met Lys His Leu Asp Lys Lys Thr Gin Thr85 90 95Pro Lys Leu(2) INFORMATION FOR SEQ ID NO : 8 :(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 97 amino acids(B) TYPE: amino acid(C) STRANDEDNESS: unknown(D) TOPOLOGY: linear(ii) MOLECULE TYPE: protein(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 8 :Met Lys Val Ser Ala Ala Leu Leu Trp Leu Leu Leu lie Ala Ala Ala1 5 10 15Phe Ser Pro Gin Gly Leu Ala Gly Pro Ala Ser Val Pro Thr Thr Cys20 25 30Cys Phe Asn Leu Ala Asn Arg Lys lie Pro Leu Gin Arg Leu Glu Ser35 40 45Tyr Arg Arg lie Thr Ser Gly Lys Cys Pro Gin Lys Ala Val lie Phe50 55 60Lys Thr Lys Leu Ala Lys Asp lie Cys Ala Asp Pro Lys Lys Lys Trp 65 70 75 80Val Gin Asp Ser Met Lys Tyr Leu Asp Gin Lys Ser Pro Thr Pro Lys85 90 95Pro(2) INFORMATION FOR SEQ ID NO : 9 :(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 93 amino acids(B) TYPE: amino acid(C) STRANDEDNESS: unknown(D) TOPOLOGY: linear(ii) MOLECULE TYPE: protein(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 9 :Met Gin Val Ser Thr Ala Ala Leu Ala Val Leu Leu Cys Thr Met Ala1 5 10 15Leu Cys Asn Gin Val Leu Ser Ala Pro Leu Ala Ala Asp Thr Pro Thr20 25 30Ala Cys Cys Phe Ser Tyr Thr Ser Arg Gin lie Pro Gin Asn Phe lie35 40 45Ala Asp Tyr Phe Glu Thr Ser Ser Gin Cys Ser Lys Pro Ser Val lie 50 55 60 Phe Leu Thr Lys Arg Gly Arg Gin Val Cys Ala Asp Pro Ser Glu Glu65 70 75 80Trp Val Gin Lys Tyr Val Ser Asp Leu Glu Leu Ser Ala 85 90(2) INFORMATION FOR SEQ ID NO: 10:(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 92 amino acids(B) TYPE: amino acid(C) STRANDEDNESS: unknown(D) TOPOLOGY: linear(ii) MOLECULE TYPE: protein(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 10:Met Lys Leu Cys Val Thr Val Leu Ser Leu Leu Met Leu Val Ala Ala1 5 10 15Phe Cys Ser Pro Ala Leu Ser Ala Pro Met Gly Ser Asp Pro Pro Thr20 25 30Ala Cys Cys Phe Ser Tyr Thr Ala Arg Lys Leu Pro Arg Asn Phe Val35 40 45Val Asp Tyr Tyr Glu Thr Ser Ser Leu Cys Ser Gin Pro Ala Val Val50 55 60Phe Gin Thr Lys Arg Ser Lys Gin Val Cys Ala Asp Pro Ser Glu Ser 65 70 75 80Trp Val Gin Glu Tyr Val Tyr Asp Leu Glu Leu Asn 85 90(2) INFORMATION FOR SEQ ID NO: 11:(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 84 amino acids(B) TYPE: amino acid(C) STRANDEDNESS: unknown(D) TOPOLOGY: linear(ii) MOLECULE TYPE: protein(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 11:Met Lys Val Ser Ala Ala Arg Leu Ala Val lie Leu lie Ala Thr Ala1 5 10 15Leu Cys Ala Pro Ala Ser Ala Ser Pro Tyr Ser Ser Asp Thr Thr Pro20 25 30Cys Cys Phe Ala Tyr lie Ala Arg Pro Leu Pro Arg Ala His lie Lys35 40 45Glu Tyr Phe Tyr Thr Ser Gly Lys Cys Ser Asn Pro Ala Val Val Phe50 55 60Val Thr Arg Lys Asn Arg Gin Val Cys Ala Asn Pro Glu Lys Lys Trp 65 70 75 80Val Arg Met Ser (2) INFORMATION FOR SEQ ID NO : 12 :(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 148 amino acids(B) TYPE: amino acid(C) STRANDEDNESS: unknown(D) TOPOLOGY: linear(ii) MOLECULE TYPE: protein (v) FRAGMENT TYPE: N-terminal(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 12 :Met Gin Val Pro Val Met Leu Leu Gly Leu Leu Phe Thr Val Ala Gly1 5 10 15Trp Ser lie His Val Leu Ala Gin Pro Asp Ala Val Asn Ala Pro Leu20 25 30Thr Cys Cys Tyr Ser Phe Thr Ser Lys Met lie Pro Met Ser Arg Leu35 40 45Glu Ser Tyr Lys Arg lie Thr Ser Ser Arg Cys Pro Lys Glu Ala Val50 55 60Val Phe Val Thr Lys Leu Lys Arg Glu Val Cys Ala Asp Pro Lys Lys 65 70 75 80Glu Trp Val Gin Thr Tyr lie Lys Asn Leu Asp Arg Asn Gin Met Arg85 90 95Ser Glu Pro Thr Thr Leu Phe Lys Thr Ala Ser Ala Leu Arg Ser Ser100 105 110Ala Pro Leu Asn Val Lys Leu Thr Arg Lys Ser Glu Ala Asn Ala Ser115 120 125Thr Thr Phe Ser Thr Thr Thr Ser Ser Thr Ser Val Gly Val Thr Ser130 135 140Val Thr Val Asn 145(2) INFORMATION FOR SEQ ID NO: 13:(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 97 amino acids(B) TYPE: amino acid(C) STRANDEDNESS: unknown(D) TOPOLOGY: linear(ii) MOLECULE TYPE: protein(xi) SEQUENCE DESCRIPTION: SEQ ID NO:13:Met Arg lie Ser Ala Thr Leu Leu Cys Leu Leu Leu lie Ala Ala Ala1 5 10 15Phe Ser lie Gin Val Trp Ala Gin Pro Asp Gly Pro Asn Ala Ser Thr20 25 30Cys Cys Tyr Val Lys Lys Gin Lys lie Pro Lys Arg Asn Leu Lys Ser35 40 45Tyr Arg Arg lie Thr Ser Ser Arg Cys Pro Trp Glu Ala Val lie Phe50 55 60Lys Thr Lys Lys Gly Met Glu Val Cys Ala Glu Ala His Gin Lys Trp 65 70 75 80Val Glu Glu Ala lie Ala Tyr Leu Asp Met Lys Thr Pro Thr Pro Lys85 90 95Pro(2) INFORMATION FOR SEQ ID NO : 14 :(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 97 amino acids(B) TYPE: amino acid(C) STRANDEDNESS: unknown(D) TOPOLOGY: linear(ii) MOLECULE TYPE: protein(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 14 :Met Gin Ser Ser Thr Ala Leu Leu Phe Leu Leu Leu Thr Val Thr Ser1 5 10 15Phe Thr Ser Gly Val Leu Ala His Pro Gly Ser lie Pro Thr Ser Cys20 25 30Cys Phe lie Met Thr Ser Lys Lys lie Pro Asn Thr Leu Leu Lys Ser35 40 45Tyr Lys Arg lie Thr Asn Asn Arg Cys Thr Leu Lys Ala lie Val Phe50 55 60Lys Thr Arg Leu Gly Lys Glu lie Cys Ala Asp Pro Lys Lys Lys Trp 65 70 75 80Val Gin Asp Ala Thr Lys His Leu Asp Gin Lys Leu Gin Thr Pro Lys85 90 95Pro(2) INFORMATION FOR SEQ ID NO: 15:(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 148 amino acids(B) TYPE: amino acid(C) STRANDEDNESS: unknown(D) TOPOLOGY: linear(ii) MOLECULE TYPE: protein(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 15:Met Gin Val Pro Val Met Leu Leu Gly Leu Leu Phe Thr Val Ala Gly1 5 10 15Trp Ser lie His Val Leu Ala Gin Pro Asp Ala Val Asn Ala Pro Leu20 25 30Thr Cys Cys Tyr Ser Phe Thr Ser Lys Met lie Pro Met Ser Arg Leu35 40 45Glu Ser Tyr Lys Arg lie Thr Ser Ser Arg Cys Pro Lys Glu Ala Val50 55 60Val Phe Val Thr Lys Leu Lys Arg Glu Val Cys Ala Asp Pro Lys Lys 65 70 75 80 Glu Trp Val Gin Thr Tyr lie Lys Asn Leu Asp Arg Asn Gin Met Arg85 90 95Ser Glu Pro Thr Thr Leu Phe Lys Thr Ala Ser Ala Leu Arg Ser Ser100 105 110Ala Pro Leu Asn Val Lys Leu Thr Arg Lys Ser Glu Ala Asn Ala Ser115 120 125Thr Thr Phe Ser Thr Thr Thr Ser Ser Thr Ser Val Gly Val Thr Ser130 135 140Val Thr Val Asn 145(2) INFORMATION FOR SEQ ID NO: 16:(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 97 amino acids(B) TYPE: amino acid(C) STRANDEDNESS: unknown(D) TOPOLOGY: linear(ii) MOLECULE TYPE: protein(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 16:Met Arg lie Ser Ala Thr Leu Leu Cys Leu Leu Leu lie Ala Ala Ala1 5 10 15Phe Ser lie Gin Val Trp Ala Gin Pro Asp Gly Pro Asn Ala Ser Thr20 25 30Cys Cys Tyr Val Lys Lys Gin Lys lie Pro Lys Arg Asn Leu Lys Ser35 40 45Tyr Arg Arg lie Thr Ser Ser Arg Cys Pro Trp Glu Ala Val lie Phe50 55 60Lys Thr Lys Lys Gly Met Glu Val Cys Ala Glu Ala His Gin Lys Trp 65 70 75 80Val Glu Glu Ala lie Ala Tyr Leu Asp Met Lys Thr Pro Thr Pro Lys85 90 95Pro(2) INFORMATION FOR SEQ ID NO: 17:(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 19 base pairs(B) TYPE: nucleic acid(C) STRANDEDNESS: single(D) TOPOLOGY: linear(ii) MOLECULE TYPE: Other(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 17: CTTCTGKGYC TGCTGYTCA 19(2) INFORMATION FOR SEQ ID NO: 18: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 17 base pairs(B) TYPE: nucleic acid(C) STRANDEDNESS: single(D) TOPOLOGY: linear(ii) MOLECULE TYPE: Other(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 18 : ACAGCYTYYY DGGGACA 17(2) INFORMATION FOR SEQ ID NO: 19:(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 17 base pairs(B) TYPE: nucleic acid(C) STRANDEDNESS: single(D) TOPOLOGY: linear(ii) MOLECULE TYPE: Other(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 19: TGTCCCHRRR ARGCTGT 17(2) INFORMATION FOR SEQ ID NO: 20:(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 17 base pairs(B) TYPE: nucleic acid(C) STRANDEDNESS: single(D) TOPOLOGY: linear(ii) MOLECULE TYPE: Other(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 20: GSKTCAGCRC AGAYYTC 17(2) INFORMATION FOR SEQ ID NO: 21:(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 23 base pairs(B) TYPE: nucleic acid(C) STRANDEDNESS: single(D) TOPOLOGY: linear(ii) MOLECULE TYPE: Other(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 21: CTGGCTGCTT GTGATTCTCC TGT 23(2) INFORMATION FOR SEQ ID NO: 22: (i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 21 base pairs(B) TYPE: nucleic acid(C) STRANDEDNESS: single(D) TOPOLOGY: linear(ii) MOLECULE TYPE: Other(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 22: CAGTCCTCAG GTATTGGCTG G 21(2) INFORMATION FOR SEQ ID NO: 23:(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 22 base pairs(B) TYPE: nucleic acid(C) STRANDEDNESS: single(D) TOPOLOGY: linear(ii) MOLECULE TYPE: Other(xi) SEQUENCE DESCRIPTION: SEQ ID NO:23: AGCTTTCATT TCGAAGTCTT TG 22(2) INFORMATION FOR SEQ ID NO: 24:(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 21 base pairs(B) TYPE: nucleic acid(C) STRANDEDNESS: single(D) TOPOLOGY: linear(ii) MOLECULE TYPE: Other(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 24: TAGATTCGGT TTAATTGGCC C 21(2) INFORMATION FOR SEQ ID NO: 25:(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 20 base pairs(B) TYPE: nucleic acid(C) STRANDEDNESS: single(D) TOPOLOGY: linear(ii) MOLECULE TYPE: Other(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 25: TCAGCCAGAT GCACTCAACG 20(2) INFORMATION FOR SEQ ID NO: 26: (i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 20 base pairs(B) TYPE: nucleic acid(C) STRANDEDNESS: single(D) TOPOLOGY: linear(ii) MOLECULE TYPE: Other(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 26: TGGAAAAGTC ATTTCACGTA 20(2) INFORMATION FOR SEQ ID NO: 27:(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 21 base pairs(B) TYPE: nucleic acid(C) STRANDEDNESS: single(D) TOPOLOGY: linear(ii) MOLECULE TYPE: Other(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 27: TTACAGGTCA GGTCCCCTAC T 21(2) INFORMATION FOR SEQ ID NO: 8:(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 22 base pairs(B) TYPE: nucleic acid(C) STRANDEDNESS: single(D) TOPOLOGY: linear(ii) MOLECULE TYPE: Other(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 28: CTCCTTATCC AGTATGGTCC TG 22(2) INFORMATION FOR SEQ ID NO: 29:(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 89 amino acids(B) TYPE: amino acid(C) STRANDEDNESS: unknown(D) TOPOLOGY: unknown(ii) MOLECULE TYPE: protein(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 29:Met Arg Gly Ser His His His His His His lie Glu Gly Arg Gin Pro1 5 10 15Asp Ala Leu Asn Val Pro Ser Thr Cys Cys Phe Thr Phe Ser Ser Lys 20 25 30Lys lie Ser Leu Gin Arg Leu Lys Ser Tyr Val lie Thr Thr Ser Arg35 40 45Cys Pro Gin Lys Ala Val lie Phe Arg Thr Lys Leu Gly Lys Glu lie50 55 60Cys Ala Asp Pro Lys Glu Lys Trp Val Gin Asn Tyr Met Lys His Leu 65 70 75 80Gly Arg Lys Ala His Thr Leu Lys Thr 85(2) INFORMATION FOR SEQ ID NO: 30:(i) SEQUENCE CHARACTERISTICS:(A) LENGTH: 10 amino acids(B) TYPE: amino acid(C) STRANDEDNESS: unknown(D) TOPOLOGY: unknown(ii) MOLECULE TYPE: protein(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 30:Gly Ser Asp Tyr Lys Asp Asp Asp Asp Lys 1 5 10
1. A substantially pure nucleic acid molecule encoding MCP-4, or degenerate variants thereof.
2. The nucleic acid molecule of claim 1, wherein said MCP-4 is mammalian MCP-4.
3. The nucleic acid molecule of claim 1, wherein said molecule is operably linked to a regulatory sequence comprising a promoter.
4. A vector comprising the nucleic acid molecule of claim 1.
5. A cell comprising the vector of claim 4.
6. A transgenic animal generated from the cell of claim 5.
7. A substantially pure antibody that specifically binds a poiypeptide encoded by the nucleic acid of claim 1.
8. A substantially pure poiypeptide encoded by the nucleic acid of claim 1.
9. A therapeutic composition comprising as an active ingredient the poiypeptide of claim 8.
10. A method of stimulating local immune response in a mammal, said method comprising administering to said mammal the nucleic acid molecule of claim 1.
11. A method of stimulating local immune response in a mammal, said method comprising administering to said mammal the cell of claim 5.
12. A method of modulating local immune response in a mammal, said method comprising administering to said mammal an MCP-4 modulator.
13. A method of inhibiting a local immune response in a mammal, said method comprising administering to said mammal an antibody that specifically binds MCP-4.
14. A method of inhibiting a local immune response in a mammal, said method comprising administering to said mammal a nucleic acid molecule, said molecule (a) consisting of least 8 nucleotides, and (b) having a sequence which is the reverse and complement of the nucleic acid molecule of claim 1.
15. A method of inhibiting a local immune response in a mammal, said method comprising administering to said mammal a mutant MCP-4 poiypeptide.
16. A method of identifying a compound that is a modulator of MCP-4 expression, said method comprising
(a) obtaining a population of cells that express MCP-4;
(b) contacting a subset of said population of cells with a candidate modulator; and
(c) comparing the level of MCP-4 expression in said subset of cells with the level of MCP-4 expression in an equivalent number of cells from said population of cells, a difference in said level indicating the presence of a compound that modulates MCP-4 expression.
17. A method of treating a mammal that is suffering from an infectious disease, said method comprising administering to said mammal a poiypeptide encoded by the nucleic acid molecule of claim 1.
18. A substantially pure nucleic acid molecule encoding MCP-5, or degenerate variants thereof.
19. The nucleic acid molecule of claim 18, wherein said MCP-5 is mammalian MCP-5.
20. The nucleic acid molecule of claim 18, wherein said molecule is operably linked to a regulatory sequence comprising a promoter.
21. A vector comprising the nucleic acid molecule of claim 18.
22. A cell comprising the vector of claim 21.
23. A transgenic animal generated from the cell of claim 22.
24. A substantially pure antibody that specifically binds a poiypeptide encoded by the nucleic acid of claim 18.
25. A substantially pure poiypeptide encoded by the nucleic acid of claim 18.
26. A therapeutic composition comprising as an active ingredient the poiypeptide of claim 25.
27. A method of stimulating a local immune response in a mammal, said method comprising administering to said mammal the nucleic acid molecule of claim 18.
28. A method of stimulating a local immune response in a mammal, said method comprising administering to said mammal the cell of claim 22.
29. A method of modulating a local immune response in a mammal, said method comprising administering to said mammal an MCP-5 modulator.
30. A method of inhibiting a local immune response in a mammal, said method comprising administering to said mammal an antibody that specifically binds MCP-5.
31. A method of inhibiting a local immune response in a mammal, said method comprising administering to said mammal a nucleic acid molecule, the sequence of which is the reverse and complement of the nucleic acid molecule of claim 18.
32. A method of inhibiting a local immune response in a mammal, said method comprising administering to said mammal a mutant MCP-5 poiypeptide.
33. A method of identifying a compound that is a modulator of MCP-5 expression, said method comprising
(a) obtaining a population of cells that express MCP-5;
(b) contacting a subset of said population of cells with a candidate modulator; and
(c) comparing the level of MCP-5 expression in said subset of cells with the level of MCP- 5 expression in an equivalent number of cells from said population of cells, a difference in said level indicating the presence of a compound that modulates MCP-5 expression.
34. A method of treating a mammal that is suffering from an infectious disease, said method comprising administering to said mammal a poiypeptide encoded by the nucleic acid molecule of claim 18.
PCT/US1997/017900 1996-09-30 1997-09-30 Mcp-4 and mcp-5: novel chemokines WO1998014573A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US2712896P 1996-09-30 1996-09-30
US60/027,128 1996-09-30

Publications (2)

Publication Number Publication Date
WO1998014573A1 true WO1998014573A1 (en) 1998-04-09
WO1998014573A9 WO1998014573A9 (en) 1998-08-06

Family

ID=21835853

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1997/017900 WO1998014573A1 (en) 1996-09-30 1997-09-30 Mcp-4 and mcp-5: novel chemokines

Country Status (1)

Country Link
WO (1) WO1998014573A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0974357A1 (en) * 1998-07-16 2000-01-26 Schering-Plough Chemokines as adjuvants of immune response
US6174995B1 (en) 1994-08-23 2001-01-16 Haodong Li Human chemokines, CKβ4 and CKβ10/MCP-4
EP1100546A1 (en) * 1998-07-28 2001-05-23 Technion Research &amp; Development Foundation Ltd. Dna cytokine vaccines and use of same for protective immunity against multiple sclerosis
US6290948B1 (en) * 1996-05-14 2001-09-18 Smithkline Beecham Corporation Method of treating sepsis and ARDS using chamohine beta-10
US6391589B1 (en) 1994-08-23 2002-05-21 Human Genome Sciences, Inc. Human chemokine beta-10 mutant polypeptides
US6458349B1 (en) 1995-06-02 2002-10-01 Human Genome Sciences, Inc. Chemokine β-4 polypeptides
US7375192B2 (en) 2002-05-01 2008-05-20 Human Genome Sciences, Inc. Antibodies that specifically bind to chemokine beta-4
US9809647B2 (en) 2010-11-19 2017-11-07 Eisai R&D Management Co., Ltd. Neutralizing anti-CCL20 antibodies

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5413778A (en) * 1992-10-05 1995-05-09 The Regents Of The University Of Michigan Labelled monocyte chemoattractant protein material and medical uses thereof
US5459128A (en) * 1993-11-12 1995-10-17 Dana-Farber Cancer Institute Human monocyte chemoattractant protein-1 (MCP-1) derivatives

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5413778A (en) * 1992-10-05 1995-05-09 The Regents Of The University Of Michigan Labelled monocyte chemoattractant protein material and medical uses thereof
US5459128A (en) * 1993-11-12 1995-10-17 Dana-Farber Cancer Institute Human monocyte chemoattractant protein-1 (MCP-1) derivatives

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
J. EXP. MED., May 1996, Vol. 183, UGUCCIONI M. et al., "Monocyte Chemotactic Protein 4 (MCP-4), a Novel Structural and Functional Analoque of MCP-3 and Eotaxin", pages 2379-2384. *

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6673344B1 (en) 1994-08-23 2004-01-06 Human Genome Sciences, Inc. Antibodies to human CKβ-10/MCP-4
US7183081B2 (en) 1994-08-23 2007-02-27 Human Genome Sciences, Inc. Human Ckβ-10 polynucleotides
US6174995B1 (en) 1994-08-23 2001-01-16 Haodong Li Human chemokines, CKβ4 and CKβ10/MCP-4
US7138498B2 (en) 1994-08-23 2006-11-21 Human Genome Sciences, Inc. Antibodies to MCP-4
US6921645B2 (en) 1994-08-23 2005-07-26 Human Genome Sciences, Inc. Antibodies to chemokine β-4
US6391589B1 (en) 1994-08-23 2002-05-21 Human Genome Sciences, Inc. Human chemokine beta-10 mutant polypeptides
US6458349B1 (en) 1995-06-02 2002-10-01 Human Genome Sciences, Inc. Chemokine β-4 polypeptides
US6406688B1 (en) 1996-05-14 2002-06-18 Human Genome Sciences, Inc. Method of treating sepsis and ARDS with chemokine β-4
US6290948B1 (en) * 1996-05-14 2001-09-18 Smithkline Beecham Corporation Method of treating sepsis and ARDS using chamohine beta-10
EP0974357A1 (en) * 1998-07-16 2000-01-26 Schering-Plough Chemokines as adjuvants of immune response
WO2000003728A1 (en) * 1998-07-16 2000-01-27 Schering Corporation Chemokines as adjuvants of immune response
US7217700B2 (en) 1998-07-16 2007-05-15 Schering Corporation Chemokines as adjuvants of immune response
EP1100546A4 (en) * 1998-07-28 2002-08-14 Rappaport Family Inst For Res Dna cytokine vaccines and use of same for protective immunity against multiple sclerosis
EP1100546A1 (en) * 1998-07-28 2001-05-23 Technion Research &amp; Development Foundation Ltd. Dna cytokine vaccines and use of same for protective immunity against multiple sclerosis
US7375192B2 (en) 2002-05-01 2008-05-20 Human Genome Sciences, Inc. Antibodies that specifically bind to chemokine beta-4
US7943741B2 (en) 2002-05-01 2011-05-17 Human Genome Sciences, Inc. Antibodies that specifically bind to chemokine β-4
US9809647B2 (en) 2010-11-19 2017-11-07 Eisai R&D Management Co., Ltd. Neutralizing anti-CCL20 antibodies

Similar Documents

Publication Publication Date Title
US6111090A (en) Mammalian cell surface antigens; related reagents
Sarafi et al. Murine monocyte chemoattractant protein (MCP)-5: a novel CC chemokine that is a structural and functional homologue of human MCP-1
US5426181A (en) DNA encoding cytokine-induced protein, TSG-14
JPH11502420A (en) Mammalian chemokines CCF8 and chemokine receptor CCKR3
US6780973B1 (en) Eotaxin: an eosinophil chemoattractant
JP2002519062A (en) Mammalian genes; dendritic cell prostaglandin-like transporter (DC-PGT), HDTEA, HSLJD37R and RANKL, HCC5 chemokine, deubiquitination 11 and 12 (DUB11, DUB12), MD-1, MD-2 and cyclin E2 , Related reagents and methods
KR100502879B1 (en) Fusion Polypeptide Comprising an IgE-Binding Domain and a HSA Component, and Their Diagnostic and Therapeutic Uses
WO1992018641A1 (en) Interleukin-8 receptors and related molecules and methods
WO1998014573A1 (en) Mcp-4 and mcp-5: novel chemokines
US6013257A (en) Neurotactin and uses therefor
WO1998014573A9 (en) Mcp-4 and mcp-5: novel chemokines
US6096300A (en) Treatment of myeloproliferative disease with exodus chemokine
EP0948536A1 (en) A g protein-coupled receptor with an enlarged extracellular domain
US6673915B1 (en) Nucleic acid encoding monocyte chemotactic protein 4
US6485910B1 (en) Ras association domain containing protein
ES2281205T3 (en) ANTAGONIST ANTAGONISTS OF ATTRACTION CHEMIOCINE OF CUTANEOUS T CELLS (CTACK) OR VASOACTIVE INTESTINAL CONTRACTOR CHEMIOCINES (VIC).
US20100068763A1 (en) Mammalian chemokines; receptors; reagents; uses
US6057426A (en) Chemokine
US6500642B1 (en) Molecule associated with apoptosis
US20030162956A1 (en) Leukocyte regulatory factors 1 and 2
WO1999055864A1 (en) NOVEL POLYPEPTIDE, cDNA ENCODING THE SAME AND UTILIZATION THEREOF
KR100367213B1 (en) A cDNA ENCODING C6 β-CHEMOKINE LEUKOTACTIN-1(LKN-1) ISOLATED FROM HUMAN
WO1998024907A1 (en) Novel protein with chemokine activity
JPH11187882A (en) Novel polypeptide, its production, cdna coding for the same polypeptide, vector comprising the same cdna, host cell transformed by the same vector, antibody of the same polypeptide, and pharmaceutical composition containing the polypeptide of antibody
US6320023B1 (en) Macrophage derived chemokine

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): CA CN JP

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
COP Corrected version of pamphlet

Free format text: PAGES 1/32-32/32, DRAWINGS, REPLACED BY NEW PAGES 1/40-40/40; DUE TO LATE TRANSMITTAL BY THE RECEIVING OFFICE

121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: JP

Ref document number: 1998516916

Format of ref document f/p: F

NENP Non-entry into the national phase

Ref country code: CA

122 Ep: pct application non-entry in european phase