MXPA06000403A - Use of soluble cd164 in inflammatory and/or autoimmune disorders - Google Patents

Use of soluble cd164 in inflammatory and/or autoimmune disorders

Info

Publication number
MXPA06000403A
MXPA06000403A MXPA/A/2006/000403A MXPA06000403A MXPA06000403A MX PA06000403 A MXPA06000403 A MX PA06000403A MX PA06000403 A MXPA06000403 A MX PA06000403A MX PA06000403 A MXPA06000403 A MX PA06000403A
Authority
MX
Mexico
Prior art keywords
seq
soluble
sequence
protein
use according
Prior art date
Application number
MXPA/A/2006/000403A
Other languages
Spanish (es)
Inventor
Chvatchko Yolande
Original Assignee
Applied Research Systems Ars Holding Nv
Chvatchko Yolande
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 Applied Research Systems Ars Holding Nv, Chvatchko Yolande filed Critical Applied Research Systems Ars Holding Nv
Publication of MXPA06000403A publication Critical patent/MXPA06000403A/en

Links

Abstract

The present invention relates to novel therapeutic uses of soluble proteins comprising the extracellular region of human CD164, in particular for treating inflammatory and/or autoimmune disorders.

Description

USE OF SOLUBLE CD164 IN INFLAMMATORY AND / OR AUTOIMMUNE DISORDERS FIELD OF THE INVENTION The present invention relates to the field of inflammation and autoimmune disorders, in particular the discovery of new proteins useful for preventing and / or treating inflammatory and / or autoimmune disorders. BACKGROUND OF THE INVENTION The following discussion is intended to facilitate the understanding of the invention, but it is not intended or admitted to be prior art to the invention. CD164 is a member of the superfamily of .glycoproteins of sialomucin or of the mucin-like receptor. Sialomucins are transmembrane glycoproteins in the size range of 50-3,000 kD that show limited similarity to cDNA and amino acid levels. Expressed proteins similar to mucin, share the common feature of carrying numerous O-glycosylations linked to serine and threonine residues, which infer multiple types of cell-cell or cell-extracellular matrix interactions. The dense array of 0-linked side chains is characterized by an extended outer structure that makes many of the molecules similar to mucin, long enough to protrude beyond Ref.:169206 of the polysaccharide glycocalyx that surrounds the cell, and also for optimal exposure and high multiplicity of terminal sugars. By virtue of the structural configuration as well as the negative charge, the mucin-like glycoproteins may act as a repulsive barrier, unless the cells possess mucin-specific receptors (adhesion). The functions of mucin receptors depend on the cell types and the activation states correlated with the core mucin peptide and with the cell-specific expression of glucosyl-transferases, which in turn regulate the structure and presentation of O-linked oligosaccharide side chains, membrane anchoring, signal transduction abilities and / or mucin trafficking to the correct cell domain. Human CD164 is an orthologous of murine MGC-24v (M. musculus) and rat endolin (R. norvegicus), a membranal protein found in the endosomal and lysosomal compartment of mammalian cells. The relationships between the different isoforms have been described, together with the functionally important domains of the subcellular distribution of CD16 / endolin (Chan YH et al., J Biol. Chem. 276: 2139-2152, 200.1). In its native state, human CD164 is a disulfide-linked homodimer of two subunits of 80-85 kDa. CD164 is highly glycosylated, containing 0- and N-linked glycans. The extracellular region is comprised of two mucin domains (I and II) linked by a non-mucin domain containing intra-catenary disulphide bridges as well as a cysteine-rich portion that resembles a consensus pattern previously found at the factor receptor receptors. growth and the cytokine. CD164 also contains a single pass transmembrane domain and an intracellular region of 13 amino acids, which includes a C-terminal portion (e.g., YHTL) capable of directing the protein to endosomes and lysosomes. Four species of human CD164 AR have been described that arise by alternative splicing of six exons of good faith from a unit of simple genomic transcription located on the human chromosome 6q21 (Zannettino A, J Biol Regul Homeost Agents, 15: 394-396 , 2001; Watt and Cham Leuk Lymph, 37 (: 1-25, 2000) There are probably 4 alternative promoters, two non-overlapping alternate late exons and an internal intron that is not always spliced, the predominant isoform of CD164 (El-6). ) represents a type I transmembrane glycoprotein of 178 amino acids.The other described isoforms are a CD164 sialomucin or delta 5 isoform of CD164 containing 178 amino acids, a delta 4 isoform of CD164 of 184 residues; and a mainly soluble isoform of 200 kD called MGC-24 (for the multiglucosylated core protein of 24 kD) lacking the transmembrane anchoring portion and having 189 residues. All isoforms are highly glycosylated proteins with 0- and N-linked glycosylation sites (Figure 1). The functions of CD164 include the mediation or regulation of the adhesion of hematopoietic progenitor cells and the negative regulation of their growth and / or differentiation. CD164 is usually expressed by hematopoietic stem cells or CD34 + _ and CD34Io / ~ hematopoietic stem cells and associated microenvironmental cells (Watt et al., Blood 92: 849-866, 1998). CD164 is also expressed by cells that form myeloid and erythroid colonies, which are involved, on the estro- mal and endothelial cells of the bone marrow, weakly on lymphocytes, and on mesenchymal stem cells. CD164 can play a key role in hematopoiesis by facilitating the adhesion of human CD34 + cells to the bone marrow stroma or by suppressing the proliferation of hematopoietic progenitor cells CD34 + CD38Io / - acting as a potent signaling molecule (Zannettino et al. , 92: 2613-2628, 1998). These effects involve the epitopes of class I and / or II of CD164 recognized by the monoclonal antibodies (mAbs) 105A5 and 103B2 / 9E10. The epitopes are carbohydrate dependent and are located on domain I of N-terminal mucin (Watt et al., Blood, 95, 3113-3124, 2000; Doyonnas et al., J Immunol, 165: 840-851, 2000) . The interaction of hematopoietic cells with stromal / endothelial cells in their immediate microenvironment is thought to be of greater importance in the regulation of self-renewal, latency, compromise and migration of the hematopoietic line. These interactions involve cooperation between adhesion receptors, their cognate ligands and cytokines. A range of cell adhesion molecules (CAMS) are involved in these processes, including Ig, integrin, cadherin, selectin and mucin-like protein families. In vitro, CD164 showed a role in biogenic differentiation (Lee et al., Mol Cell Biol, 21: 7696-7706, 2001). Overexpression of CD164 in myoblast cell lines accelerated the expression of the biochemical labeling of differentiation and increased the formation of multinucleated myotubes, while CD164 antisense or soluble extracellular regions of CD164 inhibited myogenesis. The link site to peanut agglutinin (PNA, for its acronym in English) of soluble MGC-24 represents a marker of carbohydrate associated with the tumor, expressed in many carcinomas. Total MCG-24 mRNA was found in lesser amount in human colorectal carcinomas compared to normal adjacent mucosal tissues (Matsui et al., J Biochem, 127: 1103-1107, 2000). The invasion of the lymphatic vessels by the carcinoma was correlated to the low levels of mRNA of MGC-24 in colon carcinomas, while the high levels did correlate with the lower venous invasion and less remote metastasis. Monoclonal antibodies specific for CD164 could prove useful for the diagnosis or therapy of cancer and the inhibition of hematopoiesis (EP889054, EP761814). Other proteins Similar to CD164 have been described (NOV25, WO 02/098917; SEQ ID No .: 1 7852, EP1033401; Figure 1), but their biological properties have not been analyzed. BRIEF DESCRIPTION OF THE INVENTION It has surprisingly been found that a soluble protein comprising the mature form of the extracellular domain of human CD164 has an inhibitory effect on expression in cytokines (namely interferon ?, IL2, IL-4, IL-5). , IL-10 and TNF-a) in cells that normally produce cytokines when these are stimulated with agents such as concavalin A. In addition, this soluble fragment of CD164 inhibits the relevant physiological responses (such as the migration of lymphocytes or macrophages) in animal models relevant to inflammatory and / or autoimmune diseases. Therefore, soluble proteins comprising a sequence having at least 85% homology to the mature form of the extracellular domain of human CD164 can be used for the manufacture of a medicament for the treatment and / or prevention of inflammatory disorders and / or autoinrpunes. Pharmaceutical compositions comprising any of these soluble proteins are suitable for the treatment and / or prevention of inflammatory and / or autoimmune disorders and can generally be administered to an individual to inhibit the expression of cytokines. Other features and advantages of the invention will become apparent from the following detailed description. BRIEF DESCRIPTION OF THE FIGURES Figure 1A amino acid alignment of full-length human CD164 (hCD164: NCBI Acc. No. NP_006007; SEQ ID No .: 3), CD164 human-delta4 (hCD164-DELTA4, NCBI Acc. No. AAG53908; SEQ ID No. 4), CD164-delta5 (hCD164-DELTA5; NCBI Acc. No. AAG53907; SEQ ID No. : 5), and MGC-24 (hMGC-24; NCBI Acc. No. Q04900; SEQ ID No .: 6). The sequences of signals are framed. The end of the extracellular region is indicated by an arrow. The glycosylation sites are indicated by an asterisk.
Figure IB: amino acid alignment of the mature form of the extracellular domains of CD164 (amino acids 1-40 of SEQ ID NO: 1, corresponding to amino acids 24-163 of SEQ ID NO: 3 and to amino acids__1_ - 140 of SEQ ID No .: 2), MGC-24 (amino acids 24-163 of SEQ ID NO: 6), CD164-delta4 (amino acids 24 .- 150 of SEQ ID No .: 4), CD164-deltad (amino acids 24-145 of SEQ ID No: 5 ), SEQ ID No .: 7852 (EP1033401, amino acids 24-163 of SEQ ID No .: 7), and NOV25 (WO 02/098917, amino acids 24-161 of SEQ ID No .: 8). Positions in NOV25 different from SEQ ID No .: 1 are underlined. Figures 2A-2B: effect of the administration of sf-CD164 for the mixture of human PBMC cells, stimulated with ConA, on the expression of IL-2 (Figure 2A) and TNF-a (Figure 2B). The X axis represents the concentration of sf-CD164 in μg / ml. The Y axis represents the percentage of cytokine released by secretion. Figures 3A-3B: effect of the administration of sf-CD164 for human CD4 T cells, stimulated with ConA, on the expression of IL-2 (Figure 3A) and TNF-a (Figure 3B). The X axis represents the concentration of sf-CD164 in μg / ml. The Y axis represents the percentage of cytokine release by secretion.
Figure 4: effect of the administration of sf-CD164 on the stimulation of TNF-a in the animal model for the release of TNF-a induced by LPS. The asterisks indicate the statistical significance. Figures 5A-5B: effect of the administration of sf-CD164 on cell migration in the animal model for the recruitment of cells induced by thioglycolate (Figure 5A) or by LPS (Figure 5B) in the peritoneum. The Y axis represents the concentration of cells per μl (macrophages in A, activated lymphocytes in B). The asterisks indicate the statistical significance. Figure 6: effect of the administration of sf-CD164 on the proliferation of MBP-specific, autoantigenic T cells. The X axis represents the radioactivity (CPM, counts per minute) related to the incorporation of radiolabeled nucleotides (3H-thymidine) by dividing cells. The asterisks indicate the statistical significance. Figures 7A-7C: effect of the administration of sf-CD164 for the animal model of ConA-induced hepatitis, on transaminase levels (ALAT; (Figure 7A)), IL-6 release (Figure 7B), and release of IFN-? (Figure 7C). Dexa means dexamethasone. The asterisks indicate the statistical significance.
DETAILED DESCRIPTION OF THE INVENTION In accordance with the present invention, it has been found that the mature form of the extracellular domain of CD164 (SEQ ID No .: 1) has an inhibitory effect on the cellular expression of various cytokines (namely interferon?, IL -2, IL-4, IL-5, IL-10 and TNF-a) 'after stimulation of these cells with agents such as concanavalin A (ConA). Subsequent confirmations of the therapeutic usefulness of this protein sequence were obtained in animal models for diseases, where the soluble protein demonstrated valuable biological properties in vivo, such as reduction of lymphocyte migration or inhibition of cell proliferation T-specific myelin basic protein (MBP, for its acronym in English). There is no indication in the prior art that the extracellular domain of human CD164, when isolated from the rest of the molecule as a soluble protein, has some effect on the expression of cytokines or on some other phenomenon related to autoimmune diseases and / or inflammatory The main objective of the present invention is the use of a soluble protein comprising a sequence having at least 85% homology with the mature form of the extracellular domain of human CD164 (SEQ ID No. 1) for the manufacture of a medicament for the treatment and / or prevention of inflammatory and / or autoimmune disorders. Among the soluble proteins that can be used according to the present invention, the most preferred soluble proteins are the mature form of the extracellular domain of human CD164 (SEQ ID No. 1), or the latter sequence fused to the signal sequence of human CD164. Other preferred soluble proteins that can be used according to the present invention are variants of SEQ ID NO: 1 in the form of active muteins or isoforms of SEQ ID NO: 1. The isoforms of human CD164 having the less 85% homology with the mature form of the extracellular domain of human CD164 (SEQ ID No.: 1) are known in the literature (Chan YH et al., J Biol Chem, 276: 2139-2152, 2001; Figure 1) . One of them called MGC-24 (SEQ ID No .: 6) is known to be soluble, whether it lacks a functional transmembrane domain, while two others called CD164-delta 4 (SEQ ID No .: 4) and CD164 -delta 5 (SEQ ID No .: 5) still retain a transmembranal domain. Therefore, the mature form of the extracellular domain of these latter isoforms bound to the membrane, can be considered useful according to this invention. As "soluble proteins", the present invention includes protein sequences that do not contain sequences that allow integration into a cell membrane, such as the transmembrane domain in full-length human CD164. These soluble proteins, when expressed by cells, are therefore expected to be located in the cells, or preferably, secreted in the extracellular space if they are fused to a signal sequence. Soluble proteins comprising a sequence having at least 85% homology to the mature form of the extracellular domain of human CD164 (SEQ ID No. 1) are known in the literature (Lee YN et al., Mol Cell Biol, 21: 7696-7706, 2001) but there is no indication of any utility for the treatment and / or prevention of inflammatory and / or autoimmune disorders. The soluble protein sequences defined in the present invention that are useful for the treatment and / or prevention of inflammatory and / or autoimmune disorders, are also clearly distinct from any other human sequence that has, or is assumed to have, similar properties. WO 02/098917 describes the NOV25 protein (SEQ ID NO: 8; Figure IB) comprising a sequence 80% homologous to the mature form of the extracellular domain of human CD164 (SEQ ID No. 1), and suggests that it may be useful in a variety of diseases including autoimmune disease. However, this use is merely speculative, and in addition the document fails to recognize the therapeutic utility of the soluble fragment that can be isolated from the potential extracellular domain of this protein. -Decide that -predicted being located _ on a cell membrane. EP1033401 describes a protein (SEQ ID No .: 7582) comprising a sequence identical to the mature form of the extracellular domain of human CD164 (SEQ ID No.: 7, FIG. 0 IB). Although a hypothetical therapeutic use of this protein in medicine is suggested herein, for some forms of diseases, this document also fails to recognize the therapeutic utility of the soluble fragment that can be isolated from the potential extracellular domain of this protein. As "active", the present invention defines any variant of the mature form of the extracellular domain of human CD164 (SEQ ID No .: 1) having at least 85% homology with this sequence which, according to any assay presented in the examples, it has a comparable or even increased activity, when compared to SEQ ID NO: 1, and must also be accepted for any of the uses and methods claimed. By the activity that is "comparable" it is understood that the activity measured in any of the tests described for the variant of the soluble protein is at least of the same order of magnitude, and preferably 75%, 80%, 85%, 90 %, 95%, 96%, 97%, 98%, 99% or 100%. and not more than 101%, 102%, 103%, 104%, 105%, 110%, 115%, 120% or 125% of the activity measured using a soluble protein as defined by SEQ_LD_No_: 1 ... By the activity that is "increased" it is understood that the activity measured in any of the assays described for the soluble protein variant is at least 125%, 130%, 135%, 140%, 145%, 150%, 155 %, 160%, 170%, 180%, 190%, 200%, 225%, 250%, 275%, 300%, 325%, 350%, 375%, 400%, 450% or 500% of the activity measured using a soluble protein as defined by SEQ ID No. 1. As used herein, the term "muteins" refers to any sequence that has at least 85% homology to the mature form of the extracellular domain of CD164 human (SEQ ID No.: 1) that can be generated by insertion, deletion and / or substitution of one or more amino acid residues in SEQ ID No.: 1. Similar active muteins can be natural, such as those corresponding to a protein orthologous (for example, encoded by a non-human gene that has evolved from the common ancestor for CD164) or from polymorphisms in the human genome. In cases where nucleotide substitutions result in one or more amino acid changes, the preferred soluble proteins include those that retain one or more of the activity related to anti-inflammation and / or anti-autoimmunity. Alternatively, these sequences are synthetic or artificial, which can be prepared by known chemical synthesis, recombinant DNA technology, site-directed mutagenesis, or any other suitable known technique thereof, which provides a finite group of corresponding peptides or polypeptides, substantially mutated or shortened which can be routinely obtained and tested by a person of ordinary skill in the art using the teachings presented in the prior art and in the examples of the present invention. Preferred changes in these active muteins are commonly known as "conservative" or "safe" substitutions. Conservative amino acid substitutions are those with amino acids that have sufficiently similar chemical properties, in order to preserve the structure and biological function of the molecule. It is clear that the insertions and deletions of amino acids can also be carried out in the previously defined sequences without altering their function, particularly if the insertions or deletions are only involved with a few amino acids, for example, below ten, and preferably below three, and do not eliminate or displace amino acids that are critical for the functional conformation of a protein or a peptide. The literature provides many models in which selection of conservative amino acid substitutions can be made based on statistical and physicochemical studies on the sequence and / or structure of the natural protein (Rogov SI and Nekrasov AN, Protein Eng. 14: 459-463, 2001). Protein design experiments have shown that the use of specific subsets of amino acids can produce folding and active proteins, which aid in the classification of "synonymous" amino acid substitutions that can be more easily accommodated in the protein structure (Murphy LR et al. al., Protein Eng. 13: 149-52, 2000). The groups of synonymous amino acids and the most preferred synonyms are those defined in Table I. Alternatively, the amino acids in the soluble proteins of the invention that are essential for function can also be identified by methods known in the art, such as site-directed mutagenesis or alanine scanning mutagenesis (eg, Cunningham, et al. al., Science, 244: 1081-5, 1989). Of special interest are substitutions of amino acids loaded with other charged or neutral amino acids that can produce highly desirable enhanced characteristic proteins, such as less aggregation. Aggregation can not only reduce the activity but also the problem when preparing pharmaceutically or physiologically acceptable formulations, because the aggregates can be ipthonogenic (Cleland__et al., Crit Rey Ther Drug Carrier Syst, 10: 307-77, 1993). Other examples of production of amino acid substitutions in proteins, which can be used to obtain muteins of soluble proteins for the uses of the present invention, include any of the known method steps, such as those presented in the Patents, from the United States Nos. 4,959,314, 4,588,585, 4,737,462, 5,116,943, 4,965,195; 4,879,111, 5,017,691 and 4,904,584. Alternatively, the active mutein may result from sequential alterations that reduce the immunogenicity of the soluble protein, when administered to a mammal. The literature provides many examples of these sequential alterations, which can be designed and introduced in this scope or for other functional optimizations that allow a safe and effective administration of a therapeutic protein, especially when this is a non-human, not a mammalian, protein. or unnatural (Vasserot AP et al., Drug Disc Today, 8: 118-126, 2003; Marshall SA et al., Drug Disc Today, 8: 212-221, 2003; Schellekens H, Nat Rev Drug Disc, 1: 457-461, 2002; Gendel SM, Ann NY Acad SCI, 964: 87-98, 2002; Graddis TJ et al., Curr Pharm Biotechnol, 3: 285-97, 2002; WO 03/104263; WO 03/006047; WO 02/98454, WO 02/96454, WO 02/79415, WO 02/79232, WO 02/66514, WO 01/40281, WO 98/52976, WO 96/40792, WO 94/11028). It is clear that amino acid insertions and deletions can also be performed on the previously defined sequences without altering their function, particularly if the insertions or deletions involve only a few amino acids, for example, below thirty, and preferably below ten, and they do not eliminate or displace amino acids that are identical for a functional conformation, for example, cysteines or prolines. These alterations may appear at the amino or carboxyl termini or at any site between those terminal positions, interspersed either individually between residues in the sequence or in one or more contiguous groups within the sequence. As a practical matter, if any particular polypeptide has a percentage of homology to the mature form of the extracellular domain of human CD164 (SEQ ID No .: 1) this can be determined conventionally using known computer programs. Such algorithms and programs include, but are not limited to, TBLASTN, BLASTP, FASTA, TFASTA and CLUSTALW (Pearson and Lipman, (1988) Proc Nati Acad Sci USA 85 (8): 2444-8; Altschul et al., (1990 ) J Mol Biol 215 (3): 403-410; Thompson et al., (1994) Nucleic Acids Res 22 (2): 4673-4680; Higgins et al., (1996) Meth Enzymol 266: 383-402; Altschul et al., (1997) Nuc Acids Res 25: 3389-3402; Altschul et al., (1993) Nature Genetics 3: 266-272). In a particularly preferred embodiment, the homologies in the protein and nucleic acid sequence are evaluated using the basic local alignment research tool ("BLAST") which is well known in the art (See, for example, example, Karlin and Altschul (1990) Proc Nati Acad Sci USA 87 (6): 2264-8; Altschul et al., 1990, 1993, 1997 supra). BLAST programs identify homologous sequences by identifying similar segments, which are referred to herein as "highly qualified segment pairs," between an amino acid or nucleic acid search sequence, and a test sequence that is preferably obtained from of a database of protein or nucleic acid sequences. Highly qualified segment pairs are preferably identified (eg, aligned) by means of a rating matrix, many of which are known in the art. Preferably, the qualification matrix used is the BL0SUM62 matrix (see Gonnet et al., (1992) Science 256 (5062): 1443-5, Henikoff and Henikoff (1993) Proteins 17 (1): 49-61). Less preferably, the PAM or PAM250 matrices can also be used (see, for example, Schwartz and Dayhoff, eds. (1978) Matrices for Detecting Distance 'Relationships: Atlas of Protein Sequence and Structure, Washington: National Biomedical Research Foundation). BLAST programs evaluate the statistical significance of all the pairs of high-scoring segments identified, and preferably select those segments that satisfy a threshold of significance specified by the user, such as a percentage homology specified by the user. Preferably, the statistical significance of a pair of highly rated segments is evaluated using the Karlin statistical significance formula (See for example, Karlin and Altschul, (1990) Proc Nati Acad Sci USA 87 (6): 2264-8). BLAST programs can be used with the default parameters or with the modified parameters provided by the user. Preferably, the parameters are default parameters. A preferred method for determining the best complete match between a search sequence (a sequence of the present invention) and an objective sequence, also referred to as a global sequential alignment, can be determined using the FASTDB computer program based on the Brutlag algorithm. et al. (1990) Comp.
App. Biosci, 6: 237-245. In a sequential alignment the search and target sequence are both amino acid sequences. The result of the alignment of the global sequence is in the percentage identity. The preferred parameters used in an alignment of FASTDB amino acids are: Matrix = PAM 0, k-tupie = 2, penalty for mal-coupling = 1, penalty for union = 20, group for randomization = 25, length = 0, cut-off rating = 1, window size = length of the sequence, penalty for empty space = 5, penalty for empty space size = 0.05, window size = 247 or the length of the amino acid sequence, whichever is shorter. If the target sequence is shorter than the search sequence due to N- suppressions. or C-terminals, not due to internal deletions, the results in percentage of identity, must be manually corrected because the FASTDB program does not explain the N- and C-terminal truncations of the target sequence, when the global percentage identity is calculated . For the objective sequences truncated at the N- and C- ends, relative to the search sequence, the percentage identity is corrected by calculating the number of residues of the search sequence that are N-C-terminal of the target sequence , which are not coupled / aligned with a corresponding target residue, as a percentage of the total bases of the search sequences. If a residue is coupled / aligned, this is determined by the alignment results of the FASTDB sequence. This percentage is then subtracted from the percentage identity, calculated by the previous FASTDB program, using the specified parameters, to arrive at a final percentage identity grade. This final percentage identification is that which is used for the purposes of the present invention. Only the residues at the N- and C- ends of the target sequence, which are not coupled / aligned with the search sequence, are considered for the purpose of manually adjusting the percentage identity rating. That is, only search amino acid residues outside the N- and C-terminal residues furthest from the target sequence. For example, an objec- tive sequence of 90"amino acid residues is aligned with a search sequence of 100 residues to determine percent identity.The suppression occurs at the N- end of the target sequence and therefore the FASTDB alignment does not couple / align with the first residues at the N- end.The 10 unpaired residues represent 10% of the sequence (number of residues at the ends N and C-, not the coupled number / total residues in the sequence of search) so that 10% is subtracted from the percentage identity rating calculated by the FASTDB program.If the remaining 90 residues were perfectly coupled, the final percentage identity would be 90%.
In preferred embodiments, post-translationally modified forms of soluble proteins comprising a sequence having at least 85% homology to the mature form of the extracellular domain of human CD164 (SEQ ID No. 1) can be used for manufacture of a medicament for the treatment and / or prevention of inflammatory and / or autoimmune disorders. In particular, these proteins can be acetylated, amidated, glycosylated, phosphorylated and / or myristoylated. It is known that human CD164 is modified with such groups, and a series of specific positions can be indicated as described in SEQ ID NO: 1: a) Potential N-glycosylation sites are located at residues 3, 9, 18 , 49, 54, 71, 81, 98 and 123; b) Potential O-glycosylated sites are located at residues 11, 12, 17, 20, 21, 25, 26, 31, 32, 89, 90, 92, 96, 99, 100, 104, 108, 110, 111 , 112, 113, 115, 117, 118, 119, 121, 122, 125, 127, 129, 130, 136.. c) Potential protein kinase phosphorylation sites dependent on cAMP- and cGMP-, potential, are located in residues 134 to 137; d) Potential protein kinase C phosphorylation sites are located at residues 100 to 102 and 112 to 114; e) Potential phosphorylation sites of casein kinase II are located at residues 73 to 76 and 136 to 139; f) The potential N-myristoylation site on sf-CD164 is located at residue 119. It is evident that such modifications may be present at the corresponding positions of the homologous soluble proteins defined above, as identified by the sequential alignment (Fig. ). In a further preferred embodiment, the soluble protein comprising a sequence having at least 85% homology with the mature form of the extracellular domain of the Human CD164 (SEQ ID No.: 1) is a soluble fusion protein. These soluble fusion proteins can be obtained by cloning a polynucleotide encoding the soluble protein, comprising a sequence having at least 85% homology to the mature form of the extracellular domain of CD164 (SEQ ID NO: 1) intrastructurally to the coding sequences for a heterologous protein sequence. The term "heterologous" when used herein is intended to designate any polypeptide other than a human CD164 polypeptide. Examples of heterologous sequences which may be comprised in the soluble fusion proteins either at the N-terminus or at the C-terminus, are the following: the extracellular domains of the membrane-bound protein, the immunoglobulin constant regions ( Fe region), multimerization domains, extracellular protein domains, signal sequences, export sequences, or sequences that allow purification by affinity chromatography. Many of these heterologous sequences are commercially available in the expression plasmids, since these sequences are commonly included in the fusion proteins in order to provide additional properties without significantly impairing the specific biological activity of the protein fused thereto (Terpe K, Appl Microbiol Biotechnol, 60: 523-33, 2003). Examples of such additional properties are a longer half-life in body fluids, extracellular localization or an easier purification procedure as allowed by a stretch of histidines forming the so-called "histidine tag" (Gentz et al., Proc Nati Acad Sci USA, 86: 821-4, 1989) or by the "HA" tag, an epitope derived from the influenza hemagglutinin protein (Wilson et al., Cell, 37: 767-78, 1994). If necessary, the heterologous sequence can be removed by proteolytic cleavage, for example, by inserting a proteolytic cleavage site between the soluble protein and the heterologous sequence, and exposing the purified soluble fusion protein to the appropriate protease. These characteristics are of particular importance for soluble fusion proteins, since they facilitate their production and use in the preparation of pharmaceutical compositions. For example, the soluble protein used in the examples (sf-CD164; SEQ ID NO: 2) was purified by means of a hexa-histidine peptide fused at the C-terminus of soluble CD164. When the soluble fusion protein comprises an immunoglobulin region, the fusion may be direct, or by means of a short linker peptide which may be as short as 1 to 3 amino acid residues of length or longer, for example, 13 amino acid residues in length. Said linker may be a tripeptide of the sequence EFM (Glu-Phe-Met), for example, or a ligand sequence of 13 amino acids comprising Glu-Phe-Gly-Ala-Gly-Leu-Val-Leu- Gly-Gly- Gln-Phe-Met introduced between the sequence of the substances of the invention and the immunoglobulin sequence. The resulting fusion protein has improved properties, such as a prolonged residence time in body fluids (half-life), increased specific activity, increased expression level, or purification of the fusion protein is facilitated.
In a preferred embodiment, the fusion protein is fused to the constant region of an Ig molecule. Preferably, this is fused to the heavy chain regions, with the CH2 and CH3 domains of the human IgG1, eg, Other isoforms of the Ig molecules are also suitable for the generation of fusion proteins according to the present invention, such as the IgG2 or IgG isoforms, or other Ig classes, such as IgM or IgA, for example. The fusion proteins can be monomeric or multimeric, hetero- or homomultimeric. In an additional preferred embodiment, the functional derivative comprises at least one portion linked to one or more functional groups, which appear as one or more side chains on the amino acid residues. Preferably, the portion is a polyethylene (PEG) portion. PEGylation can be carried out by known methods, such as those described in W099 / 55377, for example. Soluble proteins and soluble fusion proteins comprising a sequence having at least 85% homology to the mature form of the extracellular domain of human CD164 (SEQ ID NO: 1) can be extracted and isolated from body fluids, cells or human or mammalian tissues that express them naturally. In particular, cells either directly isolated or cultured can express these soluble proteins (naturally or after exposure to an inducing agent) and secrete them. Methods for purifying proteins are known in the art, and include the use of detergents or chaotropic agents to disintegrate the particles, followed by differential extraction and separation of the polypeptides by ion exchange chromatography, affinity chromatography, sedimentation according to to density and gel electrophoresis. In general, soluble proteins and soluble fusion proteins can be prepared by any method known in the art, including technologies related to recombinant DNA and chemical synthesis technologies. The technologies related to recombinant DNA allow the production of soluble proteins and soluble fusion proteins, firstly through the generation of the polynucleotides that encode them. These nucleic acids can be obtained by PCR from genomic DNA or, more efficiently, from a vector containing the complete sequence of CD164 (SEQ ID NO: 3) or any other relevant homologous sequences. Oligonucleotide primers complementary to the desired sequence contain restriction endonuclease sequences that allow digestion by restriction endonucleases specific for subsequent cloning, taking care to ensure that the sequence encoding the soluble protein is properly positioned with respect to the signal polyA and the rest of the other sequences in the plasmid of _5__expression. Using common genetic engineering techniques, these polynucleotides can be cloned into a replicable expression vector of viral or plasmid origin, which are used to transform a prokaryotic host cell or eukaryotic, using episomal or non-homologously integrated vectors, as well as technologies based on transformation, infection, precipitation or transfection. These vectors must allow the expression of recombinant proteins in the prokaryotic host cell or Eukaryotic, under the control of its own regulatory sequences of transcription initiation and termination, which are chosen to be constitutively active or inducible in said cell. A cell line enriched substantially in such cells can then be isolated to provide a stable cell line expressing the protein of interest. Many books and journals provide teachings on how to clone and produce recombinant proteins using vectors and prokaryotic host cells or eukaryotes, such as some titles in the series "A Practical Approach" published by Oxford University Press ("DNA Cloning 2: Expression Systems", 1995, "DNA Cloning 4: Mammalian Systems", 1996, "Protein Expression", 1999; "Protein Purification Techniques", 2001). A typical expression vector must comprise: a) a DNA sequence encoding a soluble protein or a soluble fusion protein comprising a sequence that. has at least 85% homology with the mature form of the extracellular domain of human CD164 (SEQ ID No .: 1); and b) an expression cassette; wherein sequence (a) is operably associated with a tissue-specific or constitutive promoter, included in sequence (b). The expression vector is any of the mammalian, yeast, insect or bacterial expression systems known in the art. Commercially available vectors and expression systems are available from a variety of providers including Genetics Institute (Cambridge, MA), Stragene (La Jolla, California), Promega (Madison, Wisconsin), and Invitrogen (San Diego, California).
If desired, to increase expression and facilitate proper folding of the protein, the codon context and codon pairing of the sequence can be optimized for the particular expression organism into which the expression vector will be introduced ( U.S. Patent No. 5,082,767; Gustafsson C et al., Trends Biotechnol, 22: 346-53, 2004). Factors of importance in the selection of a particular plasmid of a viral vector include: the ease with which the recipient cells containing the vector can be recognized and selected from those recipient cells that do not contain the vector; the number of vector copies that are desired in a particular host; and if it is desirable to be able to "throw" the vector between the host cells of different species. A recumbent vector according to the invention comprises, but is not limited to, Yeast Artificial Chromosome (YAC), a Bacterial Artificial Chromosome (BAC), a phage, a plasmid, a cosmic, - a plasmid, a phagemid or even a linear DNA molecule which may consist of a chromosomal, non-chromosomal, semi-synthetic or synthetic DNA. In general, recombinant expression vectors will include origins of replication, selectable markers that allow the transformation of the host cell, and a promoter derived from a highly expressed gene, to direct the transcription of a downstream structural sequence. The heterologous structural sequence is assembled in the appropriate phase with the translation initiation and termination sequences, and preferably a guiding sequence capable of directing the secretion of the translated protein into the periplasmic space or the extracellular medium. In a specific modality, wherein the vector is adapted to transfect and express desired sequences in mammalian host cells, preferred vectors will comprise an origin of replication in the desired host, a suitable promoter and enhancer, and also any necessary ribosome binding sites, polyadenylation sites, donor sites and splice acceptors, transcription termination sequences, and non-transcribed flanking sequences of the 5 'end. DNA sequences derived from the SV40 viral genome, for example, SV40 origin sites, early promoter, enhancer, splice and polyadenylation, can be used to provide the required non-transcribed genetic elements. The suitable promoter regions used in the expression vectors of the present invention are chosen taking into account the host cell in which the heterologous gene is expressed. The particular promoter used to control the expression of a nucleic acid sequence of interest is not believed to be important, as long as it is capable of directing the expression of the nucleic acid in the target cells. Thus, where targeting a human cell, it is preferable to place the coding region of the nucleic acid adjacent to and under the control of a promoter that is capable of being expressed in a human cell, such as, for example, a human or viral. The promoter used can be a constitutive or inducible one. A suitable promoter can be heterologous with respect to the nucleic acid for which it controls expression alternatively it can be endogenous to the native polynucleotide containing the coding sequence to be expressed. In addition, the promoter is generally heterologous with respect to the recombinant vector sequences within which the promoter / coding sequence construct has been inserted. The promoter regions can be selected from any desired gene using, for example, CAT vectors (chloramphenicol transferase) and more preferably the vectors pKK232-8 and pCM7. Preferred bacterial promoters are the promoters of Lacl RNA polymerase, lacZ, or bacteriophage T3 or T7, the gpt, lambda PR, PL and trp promoters (EP 0036776), the polyhedrin promoter, or the protein promoter. baculovirus plO (Kit Novagen) (Smith et al., (1983) Mol Cell Biol 3 (12): 2156-65; O'Reilly et al., 1992), the lambda PR promoter or also the trc promoter. Eukaryotic promoters include immediate early CMV, HSV thymidine kinase, early and late SV40, LTRs from retroviruses, and mouse metallothionein L.
In addition, promoters specific for a particular cell type can be chosen, such as those that facilitate expression in liposus, muscle or liver tissue. The selection of a suitable vector and promoter is well within the level of ordinary skill in the art. Where a cDNA insert is employed, it will typically be desired to include a polyadenylation signal to effect appropriate polyadenylation of the gene transcript. The nature of the polyadenylation signal is not believed to be crucial to the successful practice of the invention, and any such sequence can be employed, such as human growth hormone and SV40 polyadenylation signals. Also contemplated as an expression cassette element is a terminator. These elements can serve to improve the message levels and to minimize the reading through the cassette to other sequences. The vectors may also contain additional non-coding sequences, including for example, but not limited to the 5 'coding sequences and 3 l vector sequence, the sequences used for purification, probing or priming. For example, heterologous sequences also include transcribed, n ot translated sequences that may play a role in transcription, and mRNA processing, e.g., ribosome binding and mRNA stability. The selected markers confer identifiable change to the cell, allowing easy identification of the cells containing the expression construct. Selectable marker genes for the selection of transformed host cells are preferably dihydrofolate reductase or neomycin resistance for the culture of eukaryotic cells, TRP1 for S. cerevisiae or resistance to tetracycline, rifampicin or ampicillin in E. coli, or sucrose sucrose for mycobacteria, the latter marker being a negative selection marker. As a representative but not limiting example, expression vectors useful for bacterial use may comprise a selectable marker and a bacterial origin of replication derived from commercially available plasmids comprising the genetic elements of pBR322 (ATCC 37017). Such commercial vectors include, but are not limited to, pKK223-3 (Pharmacia, Uppsala, Sweden) and pGEMl (Promega Biotec, Madison, WI, USA). Large numbers of other suitable vectors are known to those skilled in the art, and are commercially available, such as the following bacterial vectors: pTrc-His, pET30-His, pQE70, pQE-9 (Qiagen), pbs, pDlO, fagescript, psiX174, pbluescript SK, pbsks, pNH8A, pNH16A, pNH18A, pNH46A (Strategene); ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 (Pharmacia); pWLNEO, PSV2CAT, pOG44, pXTl, pSG (Stratagene), pSVK3, pBPV, pMSG, pSVL (Pharmacia); pQE-30 __ (QIAexpress). A suitable vector for the expression of the polypeptides is a baculoviral vector that can be propagated in insect cells and in insect cell lines. A suitable, specific host vector system is the baculovirus transfer vector pVL1392 / l393 (Pharmingen) which is used to transfect SF9 cell lines (ATCC No. CRL 1711) which is derived from Spodoptera frugiperda. Additional suitable baculovial vectors are known to those skilled in the art, for example, FastBacHT. Other suitable vectors for the expression of a globular head polypeptide APM1 in a baculoviral expression system include, but are not limited to, those described by Chai et al. (1993, Biotechnol Appl Biochem.Dec; 18 (Pt3); 259-73); Vlasak et al. (1983; Eur J Biochem Sep 1; 135 (1): 123-6), and Lenhard et al. (1996, Gene Mar 9; 169 (2): 187-90). Additional suitable vectors for the expression of the polypeptides are mammalian vectors. A number of suitable vector systems are known to those skilled in the art, for example, pcDNA4HisMax, pcDNA3, lHygro-His and pcDNA3. lHygro. Additional suitable vectors for the expression of the polypeptides are viral vectors, such as those derived from an adenovirus. Preferred adenoviral vectors according to the invention are those described by Feldman and Steg (1996; Semin Interv. Cardiol 1 (3): 203-8) or Ohno et al. (1994; Science 265 (5173): 781-4). Retroviral vectors and adeno-associated viral vectors are generally understood as the distribution systems of recombinant genes of choice, for the transfer of exogenous polynucleotides in vivo, particularly for mammals, including humans. These vectors provide efficient distribution of the genes to the cells, and the transferred nucleic acids are stably integrated into the chromosomal DNA of the host. Yet another possibility to express the polypeptides is to endogenously activate the genes by introducing the regulatory sequence into the correct locus of the genome by homologous recombination, thereby operably linking the regulatory sequence with the gene, the expression of which is required to be induced (WO91 / 09955; WO02 / 10372).
The host cells can be either prokaryotic or eukaryotic. Preferred are eukaryotic hosts, for example mammalian cells, such as human, monkey, mouse, and Chinese hamster's ovary (CHO) cells, because these provide post-translational modifications to the cells. protein molecules, including correct folding or glycosylation at the correct sites. Also the yeast cells. they can carry out post-translational peptide modifications including glycosylation. There are a number of recombinant DNA strategies that utilize strong promoter sequences and a high number of plasmid copies that can be used for the production of the desired proteins in yeast. Yeast recognizes the leader sequences in cloned mammalian gene products and secretes peptides that possess leader sequences (e.g., pre-peptides). Preferred host cells used as containers for expressing the soluble proteins are the following: a) prokaryotic host cells: strains of Escherichia coli (for example, strain DH5-a), Bacillus súbtilis, Salmonella typhimurium, and strains of species such as Pseudomonas, Streptomyces and Staphylococcus; b) eukaryotic host cells: HeLa cells (ATCC No. CCL2; No. CCL2.1; No. CCL2.2), Cv 1 cells (ATCC No. CCL70), COS cells (ATCC No. CRL1650; No CRL 1651) , Sf9 cells (ATCC No., CRL1711), C127 cells (ATCC No. CRL-1804), 3T3 (ATCC No. CRL-6361), CHO (ATCC No. CCL-61), 293 human kidney cells (ATCC No 45504; No. CRL-1573), BHK (ECACC No. 84100501; No. 84111301), PLC, HepG2 and Hep3B cells. For eukaryotic hosts (eg, yeast, insect or mammalian cells), the different regulatory sequences of transcription and translation may be employed, depending on the nature of the host. These can be derived from viral sources, such as adenovirus, bovine papilloma virus, simian virus or the like, where the regulatory signals are associated with a particular gene which has a high level of expression. Examples are the Herpes virus TK promoter, the SV40 early promoter, the yeast gal4 gene promoter, etc. The regulatory signals of transcription initiation can be selected which allow repression and activation, so that the expression of the genes can be modulated. Cells that have been stably transformed by the introduced DNA can also be selected by introducing one or more markers that allow the selection of the host cells containing the expression vector. The label can also provide phototrophy to an auxotropic host, resistance to biocides, for example, antibiotics, or heavy metals such as copper, or similar __. The selectable DNA marker can be either directly linked to the DNA gene sequences. which are to be expressed, or introduced into the same cell by co-transfection. Additional elements may also be necessary for the optimal synthesis of proteins of the invention. If the nucleic acid encoding the soluble protein lacks a methionine to serve as the starting site, a start methionine can be introduced next to the first codon of the nucleic acid using conventional techniques. Similarly, if the insert from the cDNA of the soluble CD164 polypeptide lacks a poly A signal, this sequence can be added to the construct by, for example, splicing the poly A signal from pSG5 (Stratagene) using the endonuclease enzymes of Bgll restriction and Salí and incorporating them into the mammalian expression vector pXTl (Stratagene). pXTl contains the LTRs and a portion of the gag gene from Moloney murine leukemia virus. The position of the LTRs in the construction allows stable and efficient transfection. The vector includes the herpes simplex thymidine kinase promoter and the selectable neomycin gene. Depending on the host used in a recombinant production procedure, the polypeptides of the present invention can be glycosylated or they can be non-glycosylated. In addition, the soluble proteins may also include an initial modified methionine residue, and in some cases as a result of the host-mediated processes. Thus, it is well known in the art that the N-terminal methionine encoded by the start codon of translation is generally removed with high efficiency from any protein after translation of all eukaryotic cells. While N-terminal methionine in most proteins is also efficiently removed in most prokaryotes, for some proteins, this process of prokaryotic elimination is inefficient, depending on the nature of the amino acid to which methionine N is covalently bonded. -terminal. The soluble proteins, given their limited length, can also be produced by chemical synthesis technologies, for example by synthesis in solid phase and synthesis in liquid phase. As a solid phase synthesis, for example, the amino acid corresponding to the C-terminus of the peptide to be synthesized is linked to a. support that is insoluble in organic solvents, and by alternating repetition of reactions, one where the amino acids with their amino groups and side chain functional groups protected with appropriate protective groups, are condensed __.not by. one in order from ^ 1 C-terminus to the N-terminus, and one where the amino acids linked to the resin or the amino acid protecting group of the peptides are released, the peptide chain is thus extended in this manner. Solid phase synthesis methods are classified to a large extent by the tBoc method and the Fmoc method, depending on the type of protective group used. Protective groups typically used include tBoc (t-butoxycarbonyl), Cl-Z (2-chlorobenzyloxycarbonyl), Br-Z (2-bromobenzyloxycarbonyl), Bzl (benzyl), Fmoc (9-fluorenylmethoxycarbonyl), Mbh (4.4%). -imethoxydibenzhydryl), Mtr (4-methoxy-2,3,6-trimethylbenzenesulfonyl), Trt (trityl), Cough (tosyl), Z (benzyloxycarbonyl) and Cl2-Bzl (2,6-dichlorobenzyl) for amino groups; N02 (nitro) and Pmc (2,2, 5, 7, 8-pentamethylchroman-6-sulfonyl) for the guanidino groups); and tBu (t-butyl) for the hydroxyl groups). After the synthesis of the desired peptide, it is subjected to the deprotection and cutting reaction of the solid support. Such a peptide cutting reaction can be carried out with hydrogen fluoride or trifluoromethanesulfonic acid for the Boc method, and with TFA for the Fmoc method. Fully synthetic proteins of a length comparable to that of the proteins of the invention are described in the literature (Brown A. et al., J Pept Sci 2: 40-46, 1996; Muir TW, Annu Rev Biochem, 72: 249-89, 2003; Casi G and Hilvert D, _ Curr Opin Struct Biol., 13: 589r94, 2003). _ The chemical synthesis of soluble proteins allows the expansion of the natural repertoire of protein structure and function making use of unnatural amino acids (Anthony-Cahill SJ and Magliery TJ, Curr Pharm Biotechnol, 3:, 285-97, 2002). These molecules can be designed on the sequence and / or the structure of the soluble proteins in order to select the residues that can be chemically modified at the level of the amino acid side chains, the amino acid chirality, and / or the chain main peptide, and then to improve relevant properties, such as potency, ease of purification, half-life. The alternative "synonym" groups, preferred for the amino acids that are to be included, are those defined in Table II. Techniques for the synthesis and development of these compounds are well known in the art (Hruby VJ and Balse PM, Curr Med Chem, 7: 945-70, 2000; Golebiowski A et al., Curr Opin Drug Discov Devel, 4: 428-34, 2001; Villain M et al., Chem Biol., 8: 673-9, 2001, WO02 / 10195). Various methodologies for the incorporation of non-natural amino acids within proteins are also described in the literature, using in vi tro and in vivo translation systems, to probe and / or improve the structure and function of the protein (Dougherty DA, Curr Opin Chem Bio, 4: 645-52, 2000). ___ ___ The_ purification of soluble synthetic or recombinant proteins, which can be used according to the invention, can be carried out by known methods for this purpose, for example, any conventional process involving precipitation, chromatography (exchange chromatography anionic or cationic, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxyapatite chromatography and lectin chromatography), electrophoresis, differential extraction, salt fractionation, centrifugation or the like. See, for example, Methods in Enzymology for a variety of methods to purify proteins. A purification method that can be used preferably is affinity chromatography using monoclonal antibodies, or any other chemical groups that bind to the target protein (CD164 directly soluble or, if this is a soluble fusion protein, the heterologous sequence such as a marker or histidine tag) with sufficient affinity and specificity. The linking groups are produced and immobilized on a gel matrix contained within a column. The impure preparations containing the protein are passed through the column. The soluble protein will be bound to the column by affinity while the impurities will pass through. After washing the remaining impurities, the soluble protein can be eluted from the gel by a change in pH or ionic strength. Alternatively, high performance liquid chromatography (HPLC) may be used. Elution can be carried out using a water-acetonitrile-based solvent, commonly used for protein purification. Alternatively, soluble proteins can be isolated from the milk of transgenic animals that "express soluble proteins, applying any of the large number of methods described in the literature (Protein Purification Applications, A Practical Approach (New Edition), Edited by Simón Roe, AEA Technology Products and Systems, Biosciences, Harwell; Clark (1998) J Mammary Gland Biol. Neoplasia 3: 337-50; U.S. Patent No. 6,140,552). The soluble protein comprising a sequence having at least 85% homology to the mature form of the extracellular domain of human CD164 (SEQ ID NO: 1) can be produced, formulated, administered or generically used for the manufacture of a medicament for the treatment and / or prevention of inflammatory and / or autoimmune disorders as an active derivative, a modified form resistant to proteolysis, a conjugate, a complex, a moiety, a precursor, and / or a salt._ _ _ _ __ The term "derivatives" as used herein, refers to derivatives that can be prepared from functional groups present on the side chains of the amino acid portions or on the N- or C-terminal groups according to known methods . Such derivatives include, for example, the aliphatic esters or amides of the carboxyl groups and the N-acyl derivatives of the free amino groups, or the 0-acyl derivatives of the free hydroxyl groups, and are formed with acyl groups, such as, for example, the alkanoyl or aroyl groups. The term "fraction" refers to any fragment of the polypeptide chain of the compound itself, alone or in combination with related molecules or residues linked thereto, for example residues of sugars or phosphates, or aggregates of the natural polypeptide or peptide. Such molecules can also result from other modifications that do not normally alter the primary sequence, for example the derivatization of chemical peptides in vivo or in vi tro (acetylation or carboxylation), those elaborated by modifying the phosphorylation pattern (introduction of phosphotyrosine residues). , phosphoserine or phosphothreonine) or glycosylation (by exposing the peptide to enzymes that affect glycosylation, eg, glycosylation or de-glycosylation enzymes of mammals), .. of a peptide during its synthesis and processing, or in processing steps later. "Precursors" are compounds that can be converted to the compounds of the present invention by metabolic and enzymatic processing before and after administration to cells or the body. The term "salts" refers herein to the salts of the carboxyl groups and to the acid addition salts of the amino groups of the peptides, polypeptides, or analogs thereof, of the present invention. The salts of a carboxyl group can be formed by means known in the art and include inorganic salts, for example, sodium, calcium, ammonium, ferric or zinc salts, and the like, and salts with organic bases such as those formed, for example , with amines, such as triethanolamine, arginine or lysine, piperidine, procaine and the like. Acid addition salts include, for example, salts with mineral acids such as, for example, hydrochloric acid and sulfuric acid, and salts with organic acids such as, for example, acetic acid or oxalic acid. Any such salts should have activity substantially similar to the peptides and polypeptides of the invention or their analogues. The conjugate or complex can be formed with a molecule chosen from radioactive labels, biotin, fluorescent labels, cytotoxic agents, drug delivery agents. These conjugates or complexes can be generated, using molecules and methods known in the art, for example to allow detection of the interaction with other proteins (radioactive or fluorescent markers, biotin), to improve the therapeutic efficacy (cytotoxic agents) or to improve the efficacy of drug distribution, using polymers such as polyethylene glycol and other natural or synthetic polymers (Pillai 0 and Panchagnula R, Curr Opin Chem Biol., 5: 447-451, 2001). The polymer can be of any molecular weight, and can be branched or unbranched. For polyethylene glycol, the preferred molecular weight is between about 1 kDa and about 100 kDa (the term "about") indicates that in polyethylene glycol preparations, some molecules will weigh more, some less, than the established molecular weight) for ease in handling and manufacturing . Other sizes may be used, depending on the desired therapeutic profile (for example, the duration of the desired sustained release, the effects, if any, on biological activity, ease of handling, degree or lack of antigenicity and other effects). known polyethylene glycol to a therapeutic protein or analog !. Polyethylene glycol molecules (or other chemical moieties) must be linked to the polypeptide in consideration of the effects on the functional or antigenic domains of the polypeptide There are a number of coupling methods available to those skilled in the art, for example, European Patent EP-0,401,384, incorporated by reference herein (coupling of PEG to GS-CSF), see also Malik et al. (1992) 'Ex Hematol 20 (8) : 1028-35, which reports the pegylation of GM-CSF using tresyl chloride). For example, polyethylene glycol can be covalently linked through amino acid residues via a reactive group, such as a free amino or carboxyl group. The reactive groups are those to which an activated polyethylene glycol molecule can be linked. The amino acid residues that have a free amino group can include lysine residues, and the N-terminal amino acid residues; those having a free carboxyl group may include aspartic acid residues, glutamic acid residues and C-terminal amino acid residues. The sulfhydryl groups can also be used as a reactive group for the linking of the polyethylene glycol molecules. Preferred for therapeutic purposes is coupling to an amino group, such as the linkage in the N-terminus or lysine group. A polypeptide resistant to proteolysis can be generated by the replacement of a peptide bond -CONH- with one or more of the following: a reduced bond (CH2NH); a reverse reverse link (NHCO); a methylene-oxy (CH2-0) bond; a thiomethylene bond (CH2-S); a carba bond (CH2CH2); a ketomethylene bond (CO-CH2); a hydroxyethylene bond (CHOH-CH2); a link (N-N); an E-alkene bond; o A link -CH = CH-. Thus, the invention also encompasses a soluble CD164 or a variant thereof, in which at least one peptide bond has been modified as described above. In addition, amino acids have chirality within the body either L or D. In some embodiments, it is preferable to alter the chirality of the amino acids in order to prolong the half-life within the body. Thus, in some embodiments, one or more of the polypeptides are preferably in the L configuration. In other embodiments, one or more of the amino acids are preferably in the D configuration. The therapeutic applications of the polypeptides of the invention and the Related reagents can be evaluated (in terms of safety, pharmacokinetics and efficacy) by means of in vivo or in vitro tests that make use of cells, tissues and animal models, which allow detecting an inhibition of the release and / or expression of cytokine, as well as in vivo or in vitro tests, such as the inhibition of cellular recruitment. The additional characterization of the biological and therapeutic activities described in the. present invention, can be obtained by the application of various molecular biology technologies, such as two-dimensional gel electrophoresis or RNA interference. A specific embodiment for a method for distributing a soluble protein into a vertebrate cell in vivo comprises the step of introducing a preparation comprising a physiologically acceptable carrier and a naked polynucleotide, which operably codes for the polypeptide of interest within the interstitial space of a tissue comprising the cell, whereby the naked polynucleotide is collected into the interior of the cell and has a physiological effect. This is particularly applicable for in vi tro transfer, but can also be applied live. A polynucleotide sequence encoding a soluble protein comprising a sequence having at least 85% homology to the mature form of the extracellular domain of human CD164 (SEQ ID NO: 1) can be used for the manufacture of a medicament for the treatment and / or prevention of inflammatory and / or autoimmune disorders. These polynucleotides can also be used for the generation of non-human animals and plants expressing recombinant CD164 polypeptides. The animals or plants can be transgenic, for example each of their cells contains a gene encoding the CD164 polypeptide, or alternatively, a polypeptide encoding the polypeptide can be introduced into the somatic cells of the animal or plant, by example within mammalian secretory epithelial cells. In preferred embodiments, the non-human animal is a mammal such as a cow, sheep, goat, pig or rabbit. Methods for making transgenic animals such as mammals are well known to those skilled in the art, and any such method can be used in the present invention. In addition, the transgenic mammals secreting the polypeptides of the recombinant soluble proteins can be generated in their milk. Typically, the encoded polypeptide will include a signal sequence to ensure the secretion of the protein within the milk. Compositions for in vi tro and in vivo use comprising a "naked" polynucleotide are described in the prior art (WO90 / 11092; WO95 / 11307; Tascon et al., Nature Medicine 2: 888-892, 1996). In still another embodiment of the invention, the transfer of a naked polynucleotide within the cells can be carried out with a bombardment of particles (biolistic), said microprojectile particles being coated with DNA, accelerated at a high speed that allows it to pierce the cell membranes and enter the cells without killing them, as described by Klein et al. ((1990) Curr Genet Feb; 17 (2): 97-103). In a further embodiment, the polynucleotide of the invention can be entrapped in a liposome (Ghosh and Bacchawat (1991) Targeted Diagn Ther 4: 87-103; Wong et al., (1980) Gene 10: 87-94; Nicolau et al. ., (1987) Methods Enzymol 149: 157-76). These liposomes may also be targeted to cells expressing LSR by incorporation of leptin, triglycerides, ACRP30 or other known LSR ligands, into the liposomal membrane. The amount of the vector to be injected into the desired host organism varies according to the injection site. As an indicative dose, between 0.1 and 100 μg of the vector will be injected into the body of an animal, preferably the body of a mammal, for example the mouse body. In yet another embodiment of the vector according to the invention, it can be introduced in vi tro into a host cell, preferably into a host cell previously harvested from the animal to be treated, and more preferably a somatic cell such as a muscle cell. . In a subsequent step, the cell that has been transformed with the vector that codes for it. The desired CD164 polypeptide or the desired fragment thereof is reintroduced into the body of the animal for the purpose of distributing the recombinant protein within the body, either locally or systemically. For in vivo administration, the polynucleotides can be administered in any formulation suitable, in any of a range of concentrations (eg 1-500 μg / ml, preferably 50-100 μg / ml), at any volume (eg 1-100 ml, preferably 1 to 20 ml), and can be administered in any number of times (for example, 1, 2, 3, 5 or 10 times), at any frequency (for example every 1, 2, 3, 5, 10 or any number of days). The right concentrations, the right frequencies, the right modes of administration, etc. they will depend on the particular polynucleotide, the vector, the animal, etc., and can be easily determined by a person skilled in the art. The soluble protein comprising - a sequence having at least 85% homology with the mature form of the extracellular domain of human CD164 (SEQ ID NO: 1) are capable of inhibiting, the expression of cytokine related to the proinflammatory and / or immune effect , and is thus believed to prevent and / or treat "inflammatory and / or autoimmune disorders".
The main function of the. immune system is to protect a. Individual against infection by foreign invaders such as microorganisms, but it may happen that the immune system attacks the individual's own tissues, leading to pathological states- known as autoimmune diseases, which are frequently associated with inflammatory processes. In particular, CD4 + T cells can be assigned to two different subgroups called T-helper type 1 (Thl) cells and T-helper type 2 (Th2) cells based on different, non-overlapping cytokine expression patterns. Thl is characterized by the secretion of IL-2, interferon- ?, IL-12 and TNF-a, and Th2 by the secretion of IL-4, IL-5, IL-9, IL-10 and IL-13. However, these are not strict subgroups, since IFN-? and IL-10 can suppress the effects associated with Thl, as well as Th2 responses, and IL-4 and IL-13 are also capable of promoting the production of IL-12, thereby promoting Thl and potentially inhibiting Th2 responses. Thl T cells are capable of mediating macrophage activation and delayed type hypersensitivity (DTH), giving rise to pro-inflammatory or cell-mediated immune responses, whereas Th2 T cells promote the secretion of IgGl and IgE, leading to immediate type hypersensitivity reactions (humoral immunity; stimulate antibody-mediated responses, activate mast cells, and promote tissue eosinophilia). Thl is a key feature in the pathogenesis of diseases such as rheumatoid arthritis,. sarcoidosis, and tuberculosis, while - that Th2 is involved in allergies, antiparasitic responses and in asthmatic airways (eg, role in fibrosis). A non-limiting list of disorders wherein a medicament or a pharmaceutical composition comprising a soluble protein comprising a sequence having at least 85% homology to the mature form of the extracellular domain of human CD164 (SEQ ID NO: 1) can be used , including: multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, juvenile idiopathic arthritis, psoriatic arthritis, osteoarthritis, spondyloarthropathies, inflammatory bowel disease, endotoxemia, Crohn's disease, Still's disease, uveitis, Wegener's granulomatosis, Behcet's disease, scleroderma , Sjogren's syndrome, sarcoidosis, gangrenous pyoderma, polymyositis, dermatomyositis, myocarditis, psoriasis, sclerosis; systemic, hepatitis C, allergies, allergic inflammation, allergic inflammation of the respiratory tract, chronic obstructive pulmonary disease (COPD), mesenteric infarction, stroke, ulcerative colitis, allergic asthma, bronchial asthma, mesenteric infarction, stroke, fibrosis, post-ischemic inflammation in muscle, kidney and heart, inflammation of the skin, glomerulonephritis, type I diabetes mellitus of juvenile onset , hypersensitivity diseases, viral or acute liver diseases, liver insufficiencies. alcoholic, tuberculosis, septic shock, HIV infection, graft versus host disease (GVHD) and atherosclerosis. Rheumatoid arthritis is a disease marked by signs and symptoms of joint inflammation. Systemic lupus erythematosus (SLE) is characterized by scaly and red marks on the skin, and by malfunction of the kidneys in the advanced stage of the disease, and is associated with inflammatory reactions triggered by the accumulation of immune complexes in the blood vessels, particularly in the kidneys. Multiple sclerosis is a human disease characterized by relapse, inflammatory conditions that can cause weakness, bodily tremors and, in extreme cases, paralysis, and is associated with the immune system attack of the protective myelin sheath surrounding the peripheral nerve cells. Allergic inflammation is consistent with an etiology based on Th2 cells, of the atopic disease. For example, defective preparation of Th2 cells in the absence of IL- resulted in a failure to generate allergic inflammatory responses after the subsequent challenge to the respiratory tract. IL-5 and IL-13 have shown that they are more directly responsible for the infiltrates of characteristic eosinophils, and mucus hypersecretion. __ _ In multiple sclerosis, __ Thl-mediated immune responses are thought to promote disease, whereas it is believed that Th2-mediated immune responses have an improving effect on the progression of the disease. T cells expressing IL-10 have been shown to suppress experimental autoimmune encephalomyelitis (EAE), a rat model for multiple sclerosis. It has been hypothesized that TNF-a is responsible for the induction of EAE (TNF-a can be secreted by the cultures of Thl and, Th2). It is considered that human systemic lupus erythematosus (SLE) is driven by a Th2 response. However, it has been shown that IFN-? it has a greater effect on the progression of the disease in a mouse model, while IL-4 is expected to mediate the maintenance of the disease. Myocarditis is defined by inflammation of the heart muscle and is thought to be mediated by an autoimmune response to a specific cardiac antigen after an acute infection of the upper respiratory tract. The severity of experimental autoimmune myocarditis (EAM) in the mouse model is reduced by the administration of anti-IL-4, indicating a role of IL-4 in the progression of the disease. A further embodiment of the invention is a method for inhibiting the expression of one or more cytokines in an individual, comprising administering a composition that includes a soluble protein comprising a sequence that has at least 85% homology to the mature form of the extracellular domain of human CD164 (SEQ ID NO: 1). The cytokine can be TNF-a, IFN- ?, IL-2-IL-4, IL-5 or IL-10. These methods comprise the provision or administration to individuals in need thereof, of a pharmaceutically or physiologically acceptable composition, as described below, and can be considered as methods for preventing and / or treating inflammation and / or autoimmune disorders. Yet another embodiment of the present invention is represented by pharmaceutical compositions comprising a soluble protein comprising a sequence having at least 85% homology to the mature form of the extracellular domain of human CD164 (SEQ ID NO: 1), in the presence of one or more pharmaceutically acceptable excipients, for the treatment of inflammatory and / or autoimmune disorders. These compositions may further comprise an additional immunosuppressive or anti-inflammatory substance. Alternatively, pharmaceutical compositions comprising the soluble form can be combined in a "cocktail" for use in various treatment regimens. The pharmaceutical compositions of the invention may also contain suitable pharmaceutically acceptable carriers, biologically compatible carriers and additives which are suitable for administration to an animal (eg, physiological saline) and which optionally comprise auxiliaries (as excipients, stabilizers). or diluents) that facilitate the processing of the active compounds into preparations that can be used pharmaceutically. The pharmaceutical compositions can be formulated in any acceptable manner to meet the needs of the. administration mode. For example, the use of biomaterials and other polymers for the administration of drugs, as well as different techniques and models to validate a specific mode of administration, are described in the literature (Cleland JL et al., Curr Opin Biotechnol, 12: 212- 9, 2001; Luo B and Prestwich GD, Exp Opin Ther Patents, 11: 1395-1410, 2001). It is understood that "pharmaceutically acceptable" encompasses any carrier, which does not interfere with the effectiveness of the biological activity of the active ingredient and that is not toxic to the host to which it is administered., E.g. for parenteral administration, the above active ingredients can be formulated in unit dose form for injection into vehicles such as saline, dextrose solution, serum albumin and Ringer's solution. ___. Any accepted mode of administration may be used, and determined by those skilled in the art to establish the desired blood levels of the active ingredients. For example, administration can be by various parenteral routes such as subcutaneous, intravenous, epidural, topical, intradermal, intrathecal, direct intraventricular, intraperitoneal, transdermal (eg, in delayed-release formulations), intramuscular, intraperitoneal, intranasal routes , intrapulmonary (inhaled), intraocular, oral or buccal. Parenteral administration can be by bolus injection or by gradual perfusion over time. Other particularly preferred routes of administration are the aerosol and the reservoir formulation. Sustained-release formulations, particularly the depot, of the medicaments of the invention are expressly contemplated. Preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions and emulsions, which may contain auxiliary agents or excipients known in the art, and may be prepared according to routine methods. In addition, suspension of the active compounds, as appropriate in suspensions for oily injection, can be administered. Suitable lipophilic solvents or vehicles include oils, for example, sesame oil, synthetic fatty acid esters, for example, sesame oil, or synthetic fatty acid esters, for example. , ethyl oleate or triglycerides. Suspensions for aqueous injection which may contain substances that increase the viscosity of the suspension include, for example, sodium carboxymethyl cellulose, sorbitol, and / or dextran. Optionally, the suspension may also contain stabilizers. The pharmaceutical compositions include solutions suitable for administration by injection, and contain from about 0.01 to 99 percent, preferably from about 20 to 75 percent of the active compound, together with the excipient. Compositions that can be administered rectally include suppositories. For parenteral administration (for example intravenous, subcutaneous, intramuscular), the active protein (s) can be formulated as a solution, suspension, emulsion or lyophilized powder, in association with a pharmaceutically acceptable parenteral vehicle (eg, water, saline, dextrose solution) and additives that maintain isotonicity (eg mannitol) or chemical stability (eg preservatives and buffers). The formulation is sterilized by commonly used techniques. For transmucosal administration, appropriate penetrants are used so that the barrier is permeated, in the formulation. Such penetrants are generally known in the art. Pharmaceutically or physiologically acceptable preparations that can be taken orally include hard capsules made of gelatin, as well as soft sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. Hard pressure adjusting capsules may contain the active ingredients in admixture with fillers such as lactose, binders such as starches, and / or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds can. be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin or liquid polyethylene glycols. In addition, stabilizers can be added. All formulations for oral administration should be in doses suitable for such administration. For buccal administration, the compositions may take the form of tablets or lozenges formulated in a conventional manner. For administration by inhalation, the compounds for use according to the present invention are conveniently distributed in the form of an aerosol spray presentation from pressurized containers or a nebulizer, with the use of a. . gaseous propellant. "Suitable, for example, carbon dioxide. In the case of a pressurized aerosol the unit dose can be determined by providing a valve to distribute a measured quantity. Capsules and cartridges of, for example, gelatin, for use in an inhaler or insufflator, can be formulated containing a powder mixture of the compound and a suitable powder base such as lactose or starch. The compounds can be formulated for parenteral administration by injection, for example, by bolus injection or continuous infusion. The formulations for the injection may be presented in the form of two is units, for example, in ampoules or in multiple dose recipients, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and / or dispersing agents. Alternatively, the active ingredient may be in a powder or lyophilized form for constitution with a suitable vehicle, such as pyrogen-free water, before use. In addition to the formulations previously described, the compounds may also be formulated as a depot preparation. Such long-acting formulations can be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds can be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt. Additionally, the compounds can be distributed using a sustained release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various sustained release materials have been established and are well known to those skilled in the art. Sustained-release capsules can, depending on their chemical nature, release the compounds for a few weeks to more than 100 days. It is understood that the dose administered will be dependent on the age, sex, health and weight of the patient, the type of concurrent treatment if any, the frequency of treatment and the nature of the desired effect. The dose will be tailored to the individual subject, as understood and determined by a person skilled in the art. The total dose required for each treatment can be administered by _5_ dose _ multiple .or in .._ a,. = R-dQsis._ simple .. __. The ___ pharmaceutical composition of the present invention can be administered alone or in conjunction with other therapeutic products directed to the condition, or directed to other symptoms of the condition. Usually, a daily dose of the active ingredient is 0 comprised between 0.01 to 100 milligrams per kilogram of body weight or more. Ordinarily, 1 to 40 milligrams per kilogram per day are given in divided doses or in a sustained release form, which is effective to obtain the desired results. The second or subsequent administrations 5 may be performed at a dosage, which is the same as, less than, or greater than the initial or previous dose administered to the individual. An "effective amount" refers to an amount of the active ingredients that is sufficient to affect the course and the severity of the disease, leading to the reduction or remission of such pathology. The effective amount will depend on the route of administration and the condition of the patient. The dose ranges can also be determined using the value of the minimum effective concentration. The compounds should be administered using a regimen that maintains plasma levels above the minimum effective concentration by 10 to 90% "of time, preferably between 30 to 90%, and most preferably between 50 to 90%. local or selective uptake, the effective local concentration of the drug may not be related to the plasma concentration.The amount of composition administered, of course, will be dependent on the subject being treated, the subject's weight, the severity of the affliction, the manner of administration and judgment of the prescribing physician The administered dose, as a single or multiple dose, for an individual will vary depending on a variety of factors, including the pharmacokinetic properties, the route of administration, the conditions and characteristics of the patient ( sex, age, body weight, health, height), degree of symptoms, concurrent treatments, frequency of treatment and the desired effect. The substances of the invention can be administered daily or every third day, less frequently. Preferably, one or more of the substances of the invention are administered one, two or three times per week. The daily doses are usually administered in divided doses or in a sustained release form, effective to obtain the desired results. The second or subsequent administrations may be performed at a dose that is the same, less than or greater than the initial or previous dose administered to the individual. A second or subsequent administration may be administered during or before the onset of the disease. = _ .__ .-. According to the invention, the substances of the invention may be administered prophylactically or therapeutically to an individual before, simultaneously or sequentially with other regimens or therapeutic agents (e.g., multiple drug regimens), in a therapeutically effective amount. The active agents that are administered simultaneously with other therapeutic agents can be administered in the same or different compositions. For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. For example, a dose may be formulated in animal models to achieve a concentration range in circulation that includes or spans a concentration point or range, which decreases the expression of cytokines in an in vi tro system. Such information can be used to determine more precisely the useful doses in humans. A therapeutically effective dose refers to that amount of the compound that results in the improvement of symptoms in a patient. The toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or in experimental animals, for example, to determine LD50, the dose. lethal for 50% of the test population) and ED50 (the therapeutically effective dose in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and this can be expressed as the ratio between LD50 and ED50. Compounds that show high therapeutic indices are preferred. The data obtained from these cell culture assays and animal studies can be used in the formulation of a range of doses for human use. The dose of such compounds falls preferably within a range of circulating concentrations that include the ED50, with little or no toxicity. The dose may vary within this range depending on the dosage form employed and the route of administration used. The exact formulation, the route of administration and the dose can be chosen by the individual physician in view of the patient's condition. (See, for example, Fingí et al., 1975, in "The Pharmacological Basis of Therapeutics," Chapter 1). The present invention also provides novel screening assays and kits that include soluble proteins, which comprise a sequence having at least 85% homology to the mature form of the extracellular domain of human CD164 (SEQ ID NO: 1) which can - be used to identify and compare the properties of the compounds as inhibitors of cytokine secretion and expression. The kits and assays should comprise soluble proteins comprising a sequence having at least 85% homology to the mature form of the extracellular domain of human CD164 (SEQ ID NO: 1), optionally labeled or immobilized on a solid support. The following definitions are described to illustrate and define the meaning and scope of the terms used to describe the invention herein. As used interchangeably herein, the terms "oligonucleotides", and "polynucleotides", and nucleic acids include RNA, DNA sequences or hybrid RNA / DNA sequences of more than one nucleotide either in single chain or in of duplex. The terms encompass "modified nucleotides" which comprise at least one modification, including by way of example and not limitation: (a) an alternative linking group, (b) an analogous form of the purine, (c) an analogous form of pyrimidine, or (d) an analogous sugar. Examples of analogous linking groups, purines, pyrimidines and sugars are known in the prior art (WO95 / 04064). The polynucleotides encoding the soluble proteins can be prepared by any known method, including synthetic, recombinant, ex vivo, or a generation. combination thereof, as well as the use of any purification methods known in the art. The terms polynucleotide construction, recombinant polynucleotide and recombinant polypeptide are used herein consistently with their use in the art. The terms "in the 5 '" and "in the 3" direction "are also used herein consistently with their use in the art. The terms "base pairing" and "Watson base pairing"; Crick "are used interchangeably in the present and consistently with their use in the art." Similarly, the terms "complementary (a)", "complement thereof", "complement", "complementary polynucleotide", "complementary nucleic acid" and " "complementary nucleotide sequence" are used interchangeably herein and consistently with their use in the art. Similarly, the term "purified" is used herein to describe a soluble protein that has been separated from other compounds including, but are not limited to, nucleic acids, lipids, carbohydrates and other proteins In some preferred embodiments, a polypeptide is substantially pure when at least 50%, 60%, 75%, 85% and 90%, 95%, 96%, 97 %, 98%, 99% or 99.5% of the polypeptide molecules of a sample have a simple amino acid sequence In some preferred embodiments, a polypeptide "jsustancialmenee__ -puro_ _ comprises _ typically about 50%, 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 99.5% w / w of a protein sample. The purity or homogeneity of the polypeptide is indicated by a number of methods well known in the art, such as the agarose or polyacrylamide gel electrophoresis of a sample, followed by the visualization of a single polypeptide band after gel staining. For certain purposes, the highest resolution can be achieved by the use of HPLC or other methods well known in the art. In addition, as used herein, the term "purified" does not require absolute purity; rather, it is meant to be a relative definition. The purification of initial material or natural material for at least an order of 0 magnitude, preferably two or three orders, and more preferably four or five orders of magnitude, is expressly contemplated. Alternatively, the purification can be expressed as "at least" a percentage purity relative to the heterologous polynucleotides (DNA, RNA or both) or polypeptides. As a preferred embodiment, the CD164 polynucleotides or polypeptides are at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80% 90% 95%, 96%, 98%, 99% , 99.5% or 100% pure in relation to heterologous polynucleotides or polypeptides. As a further preferred embodiment, the polynucleotides or polypeptides have a purity "at least" in the range of any number, up to the thousandth position, between 90% and 100% (eg, at least 99.995% pure) relative to the heterologous polynucleotides or polypeptides. Additionally, the purity of the polynucleotides or polypeptides can be expressed as a percentage (as described above) in relation to all materials and compounds other than the carrier solution. Each number, up to the thousandths position, can be claimed as an individual species of purity. The term "isolated or isolated" requires that the material be removed from its original environment (for example, the natural environment if it is of natural origin). For example, a polynucleotide or polypeptide of natural origin present in a living animal is not isolated, but the same polynucleotide or DNA or polypeptide, separated from some or all of the materials coexisting in the natural system, is isolated. Such a polynucleotide could be part of a vector and / or such a polynucleotide or polypeptide could be part of a composition, and still be isolated, since the vector or composition is not part of its natural environment.
The term "primer" denotes a specific oligonucleotide sequence that is complementary to an objective nucleotide sequence, and used to hybridize to the target nucleotide sequence. A primer serves as a start for the polymerization of the. nucleotide catalyzed by DNA polymerase, RNA polymerase or reverse transcriptase. The terms "protein" or "polypeptide" refer to a polymer of amino acids without considering the length of the polymer. Thus, peptides, oligopeptides and proteins are included within the definition of polypeptide. This term also does not specify or exclude post-expression modifications of the polypeptides. For example, polypeptides that include the covalent bond of The glycosyl groups, acetyl groups, phosphate groups, lipidyl groups, myristoylated groups and the like, are expressly encompassed by the term polypeptide. Also included within the definition are the phosphorylated or dephosphorylated polypeptides. Also included within the definition are polypeptides containing one or more analogs of an amino acid (including, for example, amino acids of non-natural origin, amino acids that only appear naturally in an unrelated biological system, modified amino acids from mammalian systems, etc.), polypeptides with substituted bonds, as well as other modifications known in the art, both of natural origin and of non-natural origin. The terms "comprising", "consisting of" and "consisting essentially of" are defined according to their standard meaning. A defined meaning described in the M.P.E.P. it is of control over a meaning defined in the technique and a defined meaning described in the controls of the law of cases of the Federal Control Circuit, on a meaning described in M.P.E.P. With this in mind, the terms can be replaced by another one throughout the present application in order to link the specific meaning associated with each term. The term "treatment or treating" as used herein, refers to the administration of a compound after the onset of clinical symptoms. The term "prevent or prevent" as used herein, refers to the administration of a compound prior to the onset of clinical symptoms. The term "prevention" within the context of this invention refers not only to a complete prevention of the disease or one or more symptoms of the disease, but also to any prevention, attenuation, reduction, decrease or substantial or partial decrease in the effect , before or at the early onset of the disease. The term "treatment" within the context of this invention refers to any beneficial effect on the progression of the disease, including attenuation, reduction, decrease or decrease in pathological development after the onset of the disease. _ To aa the references cited in. The present, including magazine or excerpt articles, published or unpublished United States or foreign patent applications, US or foreign patents issued or any other references, are fully incorporated by reference herein, including all data, tables, figures and text presented in the cited references. Additionally, the complete contents of the references cited within the references cited herein are also fully incorporated by reference. Reference to steps of known methods, steps of conventional methods, known methods or conventional methods is in no way an admission that any aspect, description or embodiment of the present invention is described, taught or suggested in the relevant art. The invention will now be described by means of the following Examples, which should not be considered in any way as limiting of the present invention. The examples will refer to the specified figures.
The above description of the specific embodiments will thus completely reveal the general nature of the invention that others may, by applying knowledge within the experience of the technique i, including_the_contents of the references cited herein, modify and / or - easily adapting for various applications such as the specific modalities, without undue experimentation, and without departing from the general concept of the present invention. Therefore, such adaptations and modifications are intended to be within the meaning of a range of equivalents of the embodiments described, based on the teaching and guidance presented herein. It should be understood that the phraseology or terminology herein is for purposes of description and not limitation, such that the terminology or phraseology of the present specification is to be interpreted by the person skilled in the art, in light of the teachings and guidance presented herein, in combination with the knowledge of a person of ordinary skill in the art. EXAMPLES Example 1: Cloning, high-throughput expression, and purification in sF-CD164-tagged mammalian cells. The cDNA sequence coding for the complete extracellular region of human CD164 (residues 1-163 of NCBI Acc. No. NP_006007; SEQ ID NO: 3) was subcloned to generate an expression plasmid using the Gateway cloning technology "11 (Invitrogen)." This expression plasmid allows the expression and secretion of the mature form of the extracellular region of human CD164. (140 amino acids) ^ as a soluble fusion protein having a hexa-histidine tag or label fused to its C-terminus (146 amino acids; sf-CD164; SEQ ID NO: 2), then used for affinity purification. is driven by the natural CD164 signal sequence (residues 1-23 of NCBI Acc. No. NP_006007; SEQ ID NO: 3) .The mammalian cells chosen for high-throughput expression were 293 embryonic kidney cells. The human antigen expressing the Epstein-Barr virus nuclear antigen (HEK293-EBNA, Invitrogen). Cells were maintained in suspension in serum free VPRO Ex-cell medium (seed stock, maintenance member, JRH Biosciences). Sixteen to 20 hours before transfection (transfection on day -1), the cells were seeded (density of 2 × 10 cells / ml) in 2 × T225 flasks, each containing 50 ml of DMEM (Dulbeco's Modified Eagle Medium) / F12 (1: 1) with 2% fetal bovine serum (FBS) as planting medium (JRH Biosciences). The next day (day 0 transfection) the transfection took place using the JetPEIMR reagent (2 μl / μg of plasmid; PolyPlus-transfection). For each flask, 113 μg of the expression plasmid of sf-CD164 was co-transfected with 2.3 μg of a plasmid expressing the Fluorescent Protein V erele "(GFP poAsüs acronym in English.) The transfection mixture was then added to the flasks. 2xT225 and incubated at 37 ° C (5% C02) for 6 days Confirmation of positive transfection was performed by microscopy (Axiovert 10 Zeiss) on day 1 and on day 6 to qualitatively evaluate the fluorescence due to GFP. On day 6 (day of harvest) 100 ml of the supernatants from the two flasks were combined and centrifuged (4 ° C, 400 g), and placed in a container that had a unique identifier. starting from samples of 100 and 500 ml of the culture medium, from the cells expressing the C-terminal His-tagged recombinant protein, the samples were diluted with a volume of cold A buffer (Nah2P04 50 M; 600 mM NaCl; 8.7% (w / v) glycerol, pH 7.5) to final volumes of 200 and 1000 ml, respectively. The samples were filtered through a sterile 0.22 μm filter (Millipore, 500 ml filter unit) and kept at 4 ° C in the sterile medium bottle (Nalgene).
The purification was performed at 4 ° C on a VISION workstation (Applied Biosystems) connected to an automatic sample loader (Labomatic). The purification procedure consisted of two sequential steps, affinity metal chromatography on a Poros 20 MC (Applied Biosystems) column loaded with Ni ions (4.6 x 50 mm, 0.83 ml), followed by gel filtration on a column with Sephadex G-25 medium (Amersham Pharmacia) (1.0 x 10 cm). For the first chromatography step, the metal affinity column (Ni column) was regenerated with 30 column volumes of EDTA solution (100 mM EDTA, 1 M sodium chloride, pH 8.0), recharged with Ni ions through of the wash with 15 column volumes of a 100 mM NiS04 solution, washed with 10 column volumes of buffer A, followed by 7 column volumes of buffer B (50 mM NaH2P04, 600 mM sodium chloride, 8.7% (p / v) glycerol, 400 mM; imidazole, pH 7.5), and finally equilibrated with 15 column volumes of buffer A containing 15 mM imidazole. The sample was transferred, by the Labomatic sample changer, to a 200 ml sampling cycle and subsequently loaded onto a Ni metal affinity column at a flow rate of 10 ml / minute. For the 1000 ml sample, the loading procedure was repeated 5 times. The Ni column was washed with 12 column volumes of buffer A, followed by 28 column volumes of buffer A containing 20 mM imidazole. During this wash, the loosely bound contaminating proteins were eluted from the column. The His-tagged recombinant protein was finely eluted from the Ni column with 10 column volumes of buffer B at a flow rate of 2 ml / minute, and the eluted protein was collected in a 1.6 ml fraction. For the second chromatography step, the Sephadex G-25 gel filtration column was regenerated with 2 ml of buffer D (1.137 M NaCl, 2.7 mM KCl, 1.5 mM KH2P04, 8 mM Na2HP04, pH 7.2), and subsequently equilibrated with 4 column volumes of buffer C (137 mM NaCl, 2.7 mM KCl, 1.5 mM KH2P04, 8 mM Na2HP04, 20% (w / v) glycerol, pH 7.4). The peak fraction eluted from the Ni column was automatically, through the integrated sample charger on the VISION, loaded onto the Sephadex G-25 column and the protein was eluted with the C buffer at a flow rate of 2 ml / minute. . The desalted sample was recovered in a 2.2 ml fraction. The fraction was filtered through a sterile centrifugation filter of 0.22 μm (Millipore), divided into aliquots, frozen and stored at -80 ° C. An aliquot of the sample was analyzed on SDS-PAGE (NuPAGE gel at 4-12%, Novex) by Coomassie staining and Western blotting with anti-His antibodies.
Coomassie staining was performed by incubating the NuPAGE gel in a Coomassie R250 0.1% blue stain solution (30% methanol, 10% acetic acid) at room temperature for 1 hour. The gel was subsequently destained in 20% methanol, 7.5% acetic acid until the background was clear and the protein bands were clearly visible. For the Western Blot assay, the proteins were electrotransferred from the NuPAGE gel to a nitrocellulose membrane at 290 mA for 1 hour at 4 ° C. The membrane was blocked with 5% milk powder in buffer E (137 mM NaCl, 2.7 mM KCl, 1.5 mM KH2P04, 8 mM Na2HP04, 0.1% Tween 20, pH 7.4) for 1 hour at room temperature, and subsequently incubated with a mixture of 2 rabbit anti-His polyclonal antibodies (G-18 and H-15, 0.2 μg / ml each, Santa Cruz) in 2.5% milk powder in buffer E overnight at 4 ° C. After an additional 1 hour of incubation at room temperature, the membrane was washed with buffer E (3 x 10 minutes), and then incubated with an anti-rabbit antibody, conjugated to HRP, secondary (DAKO, HRP 0399) diluted 1/3000 in buffer E containing 2.5% milk powder, for 2 hours at room temperature. After washing with buffer E (3 x 10 minutes), the membrane was developed with ECL equipment (Amersham Pharmacia) for 1 minute. The membrane was subsequently exposed to a Hyperfilm (Amersham Pharmacia), the revealed film and the Western blot image visually analyzed. The protein concentration in the samples was determined using the BCA protein assay kit (P_ierce) -con. bovine serum albumin as standard. Example 2: Effect of sf-CD164 on cytokine release, measured by cell-based assays The following cell-based assays, in vi tro, measure the effects of sf-CD164 on cytokine secretion induced by Concanavalin A (With A ) as measured by a cytokine spheres arrangement assay (CEA) for IL-2, IFN- ?, TNF-a, IL-5, IL-4 and IL-10. The following equipment and software were used: • 96 well microtiter plate photometer EX (Labsystem) Software Graph Pad (Prisma) Excel Software (Microsoft) Flow cytometer (Becton-Dickinson) • CBA Analysis Software Bell for Cell Culture Incubator for cell culture Centrifuge Pipettes The following materials and reagents were used: mononuclear cell culture DMEM (GIBCO) »Serum hümáñóXEipo AB (SIGMA) L-glutamine (GIBCO) penicillin-streptomycin (GIBCO) Ficoll (PHARMACIA) microtiter plate 96 wells for cell culture (COSTAR) Concanavalin A (SIGMA) CBA team of Thl / Th2 human cytokine (Becton-Dickinson)PBS (GIBCO) 50 ml Falcon sterile tubes (Becton-Dickinson) • Bovine serum albumin (BSA, SIGMA) Glycerol (MERCK) Dimethyl sulfoxide (DMSO, SIGMA) 96-well conical bottom microtiter plate (NUNC) • Separator autoMACS "11 and MACS cell isolation team (Miltenyl Biotec) Cells were isolated for cell-based assays as follows.
Mononuclear cells of human peripheral blood (PBMC for its acronym in English) were isolated from the mononuclear cell culture diluted with DMEM. 25 ml of the diluted blood were slowly added after this on a layer. from. 15. ml of Fic_oll_ in a 50 ml Falcon tube, and the tubes were centrifuged (2000 rpm, 20 minutes, at room temperature without brake). The interface (ring) was then collected and the cells were washed with 25 ml of DMEM, followed by a centrifugation step (1200 rpm, 5 minutes). This procedure was repeated three times. A mononuclear cell culture gave approximately 600 x 10 6 total cells. Sub-populations of leukocytes (T cells, B cells, monocytes) were prepared from the PBMC according to the instructions of the isolation equipment manufacturer (MACS; Miltenyl Biotec). The PBMC were isolated from the mononuclear cell culture as described above. Care was taken to ensure a simple cell suspension. For the preparation of CD4 + T cells, CD4 + T Cell Isolation Equipment II (Catalog Number 130-091-155, Miltenyl Biotec) was used. PBMC were counted, centrifuged for 10 minutes and resuspended in cold PBS buffer (phosphate buffered saline pH 7.2, supplemented with 0.5% BSA, and 2 mM EDTA) at a concentration of 2.5 x 108 cells per ml (40 μL of buffer by 107 cells). 10 μl of the biotin-antibody cocktail (supplied with the kit) was added per 107 total cells. The suspension was mixed thoroughly and incubated at 4-8 ° C for 10 minutes. 30 μl of buffer was added per 107 cells, followed by 20 μl of the anti-biotin microspheres per 107 total cells.The suspension was mixed thoroughly and incubated for an additional 15 minutes at 4-8 ° C. The cells were washed with buffer by adding 10 to 20x of the labeling volume, and centrifuged at 300xg for 10 minutes.The supernatant was completely removed and the cells resuspended up to 10 8 cells in 500 μl of buffer.The magnetic separation was carried out with an autoMACS1 separator. " The autoMACS ™ separator was prepared and prepared according to the manufacturer's instructions. The tube containing the magnetically labeled cells was placed in the autoMACS separator "and the" exhaust "program was chosen.The negative fraction was collected (exit gate," negl ") This fraction represents the enriched CD4 + T cells. required, the positive fraction was subsequently collected ("posl" exit gate.) This fraction represents the non-CD4 + magnetically labeled T cells.
The conditions applied for the cell-based assays were as follows: 100 000 cells / well in 96 well plates in 100 μl final in 2% glycerol. = 5 ng / ml of the -mitógeno-Concanavalin A (ConA). - --- - 48 hours for each trial. The cells were prepared in each well by mixing: 80. μl of 1.25 x 106 cells / ml were diluted in DEMEM + 2.5% Human Serum + 1% L-glutamine + 1% penicillin-streptomycin. 10 μl of the solution containing sf-CD164 that was diluted in PBS + 20% glycerol (the final dilution of the proteins is 1/10); - 10 μl of ConA. After 48 hours, the cell supernatants were harvested and the human cytokines were measured by the human cytokine CBA kit Th1 / th2 (Becton-Dickinson). Human Mixed Thl / Th2 Capture Spheres, in suspension, were prepared by vortexing vigorously for a few seconds, before mixing with samples from the microwell plate. For each test to be analyzed, a 10 μl aliquot of each capture sphere was added into a single tube labeled "mixed capture spheres". The mixture of spheres was perfectly stirred in whirlwind. The supernatants were diluted (1: 4) using the assay diluent (20 μl of -5- supernatants - + - 60 μl assay diluent). The sample dilution was then mixed before transferring the samples to a 96 well, conical bottom (Nunc) microtiter plate. _ The CBA assay of human Th1 / Th2 cytokine _ was performed by the addition of 50 μl of the diluted supernatants in a 96 well microtitre plate, conical bottom (Nunc). 50 μl of the mixed capture spheres were added followed by the addition of 50 μl of the human Thl / Th2 PE detection reagent. The plate was then incubated for 3 hours at room temperature and protected from direct exposure to light, followed by centrifugation at 1500 rpm for 5 minutes. The supernatant was then carefully discarded. In a subsequent step, 200 μl of the wash buffer was added twice in each well, centrifuged at 1500 rpm for 5 minutes and the supernatant was carefully discarded. 130 μl of the wash buffer were after this added to each well to resuspend the spheres button. The samples were finally analyzed on a flow cytometer. The data was analyzed using the CBA Application software, the Microsoft Software Activity Base and Excel. The effect of sf-CD164 on the release of cytokine from human PBMC cells (mixture) and the isolated cells, was measured. six -.cycins: TNF-a, IFN- ?, IL-2, IL-4- IL-5 and IL-10. The release of these cytokines was significantly decreased and in a dose-dependent manner by sf-CD164 in both cell-based assays (the IC50 are summarized in Table III). Two exemplary dose-dependent curves are shown for IL-2 and TNF-a for human PBMC cells and isolated CD4 + T cells, in Figures 2 and 3, respectively .. Example 3: Effect of sf-CD164 administration on the release of cytokine, measured in the animal model of TNF-a release induced by LPS The model for the release of TNF-a induced by lipopolysaccharide (LPS) in mice, was established according to the patent W098 / 38179. LPS (0111: B4; SIGMA) was injected (0.3 mg / kg, i.p.) in C3H / HeN mice (Charles River, France). Ninety minutes later blood was taken and the plasma TNF-α was determined using an ELISA kit (R &D). Sf-CD164 and dexamethasone were diluted in PBS and injected (sf-CD164 at 0.03, 0.1 and 0.3 mg / kg, iv, or dexamethasone at 0.1 mg / kg, subcutaneously) 15 minutes before the administration of LPS. Dexamethasone, the anti-inflammatory compound used as a positive control significantly inhibited (p <0.001) the release of TNF-a induced by LPS, by 72%. _.5. = Sf-CDl-64, at 0.3 mg / kg,. ^ Inhibited -significantly (p <0.01), the release of TNF-a induced by LPS by 38% (Figure 4).
The lower doses of 0.03 and 0.1 mg / kg were able to inhibit TNF-a but in a less statistically significant way. Example 4: effect of the administration of sf-CD164 on the recruitment of immune cells measured in two animal models The effect of sf-D164 on immune cell recruitment was first tested using the thioglycollate-induced peritoneal leukocyte recruitment assay (Figure 5). Mice (strain C3H, 8 weeks old, n = 6; Elevage Janvier, France) were injected with sf-CD164 (0.03, 0.1 and 0.3 mg / kg, i.v.) or dexamethasone (1 mg / kg, se) diluted in PBS containing 0.02% BSA. Thioglycolate (1.5%, 40 ml / kg, ip, SIGMA) was injected 15 minutes after. the administration of the test molecules. A second The administration of the test molecules was carried out 24 hours later. Forty-eight hours after challenge with thioglycollate, the animals were sacrificed and washing of the peritoneal cavity was conducted using 2 x 5 ml of 1 mM PBS-EDTA (+ 4 ° C). After centrifugation (10 _5 .... minutes _a_ 30.0.0.xrpm), _ the _button-_f.ue _._ r.esuspendi.do ... jen AL. jnl __ of PBS. The peritoneal cells were counted using a Beckman / Coulter counter. Dexamethasone significantly inhibited (p <0.001) the recruitment of macrophages by 69%. East The effect was in a dose-dependent manner. Sf-CD164 (0.03, 0.1 and 0.3 mg / kg) significantly inhibited the peritoneal recruitment of macrophages induced by thioglycollate, by 5%, 26% (p <0.05) and 43% (p <0.001), respectively, as well as the lymphocytes (in 14%, 18% and 34% respectively) and peritoneal recruitment of neutrophils (in 3%, 9% and 23% respectively). Similar results were obtained in the peritoneal recruitment of neutrophils and lymphocytes, induced by LPS (Figure 5). The same administration protocol described above was used with LPS (0111: B4, Sigma, 0.9 mg / kg, 40 ml / kg ip). Sf-CD164 (0.03, 0.1 and 0.3 mg / kg) significantly inhibited peritoneal recruitment of neutrophils induced by LPS in 9%, 35% (p <0.001) and 43% (p <0.001), 5 respectively. At the same doses, this also significantly inhibited the recruitment of activated lymphocytes in 8%, 26% (p <0.05) and 47% (p <0.001), respectively. Dexamethasone (0.1 mg / kg) significantly inhibited (p <0.001) the recruitment of activated lymphocytes. Example_5: effect of sf-CD164 in a cell-based assay for the processing and presentation of MBP-specific antigens An assay was developed to test the effect of sf-CD164 on the proliferation of myelin basic protein-specific T cells (MBP for its acronym in English), induced by the myelin basic protein peptide Acl-11 (MBP (Acl-11)). It has been shown that epicutaneous immunization (ECi) with the immunodominant peptide of the myelin basic protein (MBP), Acl-1, protects mice that are transgenic for an Acl-specific T cell receptor. ll, against the induced and spontaneous forms of experimental allergic encephalomyelitis (EAE). Spleens from transgenic BIO.PL and MBP mice were harvested and homogenized to obtain single cell suspensions. After lysis of the erythrocytes with Gay's solution, the splenocytes were resuspended in PBS, washed and counted. After the isolation procedures, cell viability was greater than 9 = 0% by trypan blue dye exclusion. The antigen-presenting cells (APCs) BIO.PL were then irradiated with 25 Gy of gamma radiation (stimulants), washed and resuspended in incomplete medium at 1.9x106 cells / ml. The population of responding cells was adjusted to 3.8.8 cells per ml in complete medium. 80 μl of each cell suspension per well were mixed in the 96-well plates. The antigen was then added in a volume of 20 μl: 10 μg / ml of murine MBP or 1 μg / ml of the MBP peptide of Acl-ll per well (suitable negative controls are BSA, MSA and a peptide derived from irrelevant MBP, respectively). Proteins or small molecules were added in a volume of 20 μl and then incubated at 37 ° C in a humidified atmosphere with 5% C02. After 3 days of culture, the supernatants were harvested and frozen at -80 ° C until the test for cytokine production, or 1 μCi of 3H_thymidine was added, and the incorporation of radioactivity was counted after 14-16 hours of incubation additional . Sf-CD164 (50 μg / ml) significantly inhibited the proliferation of MBP-specific T cells, induced by Acl-ll (1 μg / ml; Figure 6). In this way, sf-CD164 or soluble CD164 can be. useful in the treatment of multiple sclerosis.
Example 6: effect of the administration of sf-CD164 in an animal model of fulminant hepatic hepatitis The sf-CD164 protein has shown in vitro that it inhibits the secretion of certain cytokines by human blood mononuclear cells. ,, stimulated with ConA (PBMC). Since cytokines play a crucial role in hepatitis induced by ConA, induced pro T cells (Seino et al 2001, Annals of surgery 234, 681; Kusters S. Gastroenterology 111 (2): 0 462-71, 1996; Toyonaga et al. 1994, PNAS 91, 614-618), sf-CD164 was tested in this model. C57 / BL6 female mice (8 weeks old, IFFA CREDO) were used. In general, 7 animals were used per experimental group. The 5 mice were maintained under standard conditions under a 12-hour light-dark cycle, irradiated food and water ad l ibi tum were provided. Concanavalin A (ConA; Sigma ref C7275) was injected at 18 mg / kg intravenously, and blood samples were taken at 1.30 and 8 hours after injection. Sf-CD164 was injected 30 minutes before the injection of ConA. Positive controls were injected with dexamethasone (0.1 mg / kg), and the negative control was injected with PBS-5 BSA, 1.8% glycerol. At the time of sacrifice, the blood was taken from the heart. Cytokine levels IL-6 and IFN-? were measured using the TH1 / TH2 CBA assay 1.5 hours after the injection of ConA. The parameters were determined _._5. -sanguinees of transa inasa .using the. -Instrument COBAS (Hitachi). The experiment shows that sf-CD164 (1 mg / kg) protects against liver damage in a mouse model that mimics. fulminant hepatitis after the subcutaneous administration of sf-CD164, since it decreases the relevant parameters such as the levels of transaminases (ALAT), IFN-α, and the levels of cytokine IL-6 (Figure 7). The decrease in ALAT levels can be measured at. levels decreased IFN-? and IL-6. Different cytokines have been implicated in liver damage after the injection of ConA. For example, anti-TNF-alpha antibodies confer protection against the disease (Seino et al., 2001, Annals of surgery 234, ,681) and the inhibition of IL-4 production by NKT cells showed that it is hepatoprotective in hepatitis mediated by T cells, in mice (Ajuebor et al., 2003 J. Immunology 170, 5252-9).
TABLE 1 Amino Acid Group Synonym Groups Synonyms Most Preferred Ser Gly, Ala, Ser, Thr, Pro Thr, Ser ... Arg Asn, Lys, Gln, Arg, His Arg, Lys, His Leu Phe, He, Val, Leu, Met He , Val, Leu, Met Pro Gly, Wing, Ser, Thr, Pro Pro Thr Gly, Wing, Ser, Thr, Pro Thr, Ser Wing Gly, Thr, Pro, Wing, Ser Gly, Wing Val Met, Phe, Lie ,. Leu, Val Met, He, Val, Leu Gly Ala, Thr, Pro, Ser, < 51and Gly, Ala He Phe, He, Val, - Leu, Met He, Val, Leu, Met Phe Trp, Phe, Tyr Tyr, Phe Tyr Trp, Phe, Tyr Phe, Tyr Cys Ser, Thr, Cys Cys His Asn, Lys, Gln, Arg, His Arg, Lys, His Gln Glu, Asn, Asp, Gln Asn, Gln Asn Glu, Asn, Asp, Gln Asn, Gln Lys Asn, Lys, Gln, Arg, His Arg, Lys, His Asp Glu, Asn, Asp, Gln Asp, Glu Glu Glu, Asn, Asp, Gln Asp, Glu Met Phe, lie, Val, Leu, Met He, Val., Leu, Met Trp Trp, Phe, Tyr Trp TABLE II Phe D-Phe, Tyr, D-Thr, L-Dopa, His, D-His, Trp, D-Trp, Trans-3, 4 or 5-phenylproline, AdaA, AdaG, cis-3, 4, or 5- phenylproline, Bpa, D-Bpa Tyr D-Tyr, Phe, D-Phe, L-Dopa, His, D-His Cys D-Cys, S-Me-Cys, Met, D-Met, Thr, D-Thr Gln D-Gln, Asn, D-Asn, Glu, D-Glu, Asp, D-Asp As-n D-Asn, Asp, D-Asp, Glu, D-Glu, Gln, D-Gln Lys D-Lys, Arg, D-Arg, homo-Arg, D-homo-Arg, Met, D-Met, lie, D-Ile, Orn, D-Orn Asp D-Asp, D-Asn, Asn, Glu, D-Glu, Gln, D-Gln Glu D-Glu, D-Asp, Asp, Asn, D-Asn, Gln, D-Gln Met D-Met, S-e-Cys, Lie, D-Ile, Leu, D-Leu, Val, D-Val TABLE III It is noted that in relation to this date, the best method known to the applicant to carry out the aforementioned invention is that which is clear from the present description of the invention.

Claims (23)

CLAIMS Having described the invention as above, the content of the following claims is claimed as property:
1. The use of a soluble protein comprising a sequence having at least 85% homology with the mature form of the extracellular domain of human CD164 (SEQ ID NO: 1), for the manufacture of a medicament for the treatment and / or prevention of inflammatory and / or autoimmune disorders.
2. The use according to claim 1, wherein the soluble protein is chosen from: a) SEQ ID NO: 1; or b) SEQ ID NO: 1 fused to the signal sequence of human CD164.
3. The use according to claim 1, wherein the soluble protein is an active mutein or an isoform of SEQ ID NO: 1.
4. The use according to claim 3, wherein the soluble protein is chosen from: a) MGC-24 (SEQ ID NO: 6); or b) the mature form of the extracellular domain of any of the following isoforms of human CD164: CD164-delta 4 (SEQ ID NO: 4), CD164-delta 5 (SEQ ID NO: 5).
5. The use according to any of the preceding claims, wherein the soluble protein is glycosylated.
6. The use according to claim 5, wherein the soluble protein is glycosylated at any of the positions as described in SEQ ID NO: 1.
7. The use according to any of the preceding claims, wherein the soluble protein is phosphorylated.
8. The use according to claim 7, wherein the soluble protein is phosphorylated at any of the positions as described in SEQ ID NO: 1.
9. The use according to any of the preceding claims, wherein the soluble protein is myristoylated.
10. The use according to claim 9, wherein the soluble protein is myristoylated in any of the positions described in SEQ ID NO: 1.
11. The use according to any of the preceding claims, wherein the soluble protein is a soluble fusion protein.
12. The use according to claim 11, wherein the soluble fusion protein comprises a signal sequence.
13. The use according to claims 11 or 12, wherein the soluble fusion protein contains a Histidine tag.
14. The use according to claim 13, wherein the soluble fusion protein is SEQ ID NO: 2.
15. The use according to claims 11 or 12, wherein the soluble fusion protein comprises an Fe region of an immunoglobulin.
16. The use according to any of the preceding claims, wherein the soluble protein is an active derivative, a modified form resistant to proteolysis, a conjugate, a complex, a moiety, a precursor, and / or a salt.
17. The use of a polynucleotide sequence encoding a soluble protein comprising a sequence having at least 85% homology to the mature form of the extracellular domain of human CD164 (SEQ ID NO: 1) for the manufacture of a medicament for the treatment and / or prevention of inflammatory and / or autoimmune disorders.
18. The use according to any of the preceding claims, wherein the inflammatory and / or autoimmune disease is selected from the group consisting of: multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, juvenile idiopathic arthritis, psoriatic arthritis, osteoarthritis, spondyloarthropathies, disease inflammatory bowel, endotoxemia, Crohn's disease, Still's disease, uveitis, Wegener's granulomatosis, Behcet's disease, scleroderma, Sjogren's syndrome, sarcoidosis, gangrenous pyoderma, polymyositis, dermatomyositis, myocarditis, psoriasis, systemic sclerosis, hepatitis C, allergies, allergic inflammation, allergic inflammation of the respiratory tract, chronic obstructive pulmonary disease (COPD), mesenteric infarction, stroke, ulcerative colitis, allergic asthma, bronchial asthma, mesenteric infarction, apoplexy, fibrosis, post-ischemic inflammation in muscle, kidney and heart, skin inflammation, glo merulonephritis, type I diabetes mellitus of juvenile onset, hypersensitivity diseases, viral or acute liver diseases, alcoholic liver failure, tuberculosis, septic shock, HIV infection, graft versus host disease (GVHD) and atherosclerosis.
19. A method for inhibiting the expression of one or more cytokines in an individual, characterized in that it comprises administering to said individual a composition comprising a soluble protein that includes a sequence having at least 85% homology with the mature form of the extracellular domain of Human CD164 (SEQ ID NO: 1).
20. The method according to claim 19, characterized in that the cytokine is TNF-a, IFN- ?, IL-2, IL-4, IL-5 or IL-10.
21. A pharmaceutical composition, characterized in that it comprises a soluble protein that includes a sequence having at least 85% homology with the mature form of the extracellular domain of human CD164 (SEQ ID NO: 1), in the presence of one or more pharmaceutically acceptable excipients, for the treatment of inflammation and / or autoimmune disorders.
22. Selection tests, characterized in that they include a soluble protein comprising a sequence having at least 85% homology with the mature form of the extracellular domain of CD164 (SEQ ID NO: 1), to identify and compare the properties of the compounds as inhibitors of cytokine secretion and expression.
23. The equipment for identifying and comparing the properties of the compounds as inhibitors of cytokine secretion and expression thereof, characterized in that they comprise soluble proteins that include a sequence that has at least 85% homology with the mature form of the extracellular domain of Human CD164 (SEQ ID NO: 1).
MXPA/A/2006/000403A 2003-07-23 2006-01-10 Use of soluble cd164 in inflammatory and/or autoimmune disorders MXPA06000403A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
EP03077316.2 2003-07-23

Publications (1)

Publication Number Publication Date
MXPA06000403A true MXPA06000403A (en) 2006-12-13

Family

ID=

Similar Documents

Publication Publication Date Title
CA2367750C (en) Tribonectins
JP2005218453A (en) 87 human secreted proteins
US8075894B2 (en) Use of soluble CD164 in inflammatory and/or autoimmune disorders
EP1015477B1 (en) 32 Human secreted proteins
JP2009131263A (en) 50 human secreted proteins
JP2001521383A (en) 20 human secretory proteins
SI9520118A (en) Compositions and treatment for multiple sclerosis
US6878687B1 (en) Protein HMAAD57
KR20070008510A (en) Therapeutic uses of chemokine variants
US7053190B2 (en) Secreted protein HRGDF73
US20030212000A1 (en) Immunoprotective methods for beta cell neogenesis
WO2000063227A1 (en) A splicing variant of human membrane-type matrix metalloproteinase-5 (mt-mmp5-l)
US7803910B2 (en) Soluble CD164 polypeptides
MXPA06000403A (en) Use of soluble cd164 in inflammatory and/or autoimmune disorders
AU2002249521B2 (en) Discriminative nucleic acid analysis using clone sequence signatures
JP2000512506A (en) New compound
JP2002502601A (en) Dendritic enriched secretory lymphocyte activating molecule
CA2263165A1 (en) Human induced tumor protein
US20070238664A1 (en) 70 Human Secreted Proteins
JP2002516573A (en) 207 human secreted proteins
EP1094832A1 (en) Therapeutic use of uncoupling protein-hhfcw60
EP1394252A2 (en) 70 human secreted proteins
EP1492557A2 (en) Xcrf polynucleotides and polypeptides and uses thereof
WO2002059369A2 (en) Compositions and methods for the treatment of diseases related to faulty cholesterol regulation
EP1333092A2 (en) 28 human secreted proteins