MXPA00009063A - Isolated mammalian membrane protein genes and related reagents - Google Patents

Isolated mammalian membrane protein genes and related reagents

Info

Publication number
MXPA00009063A
MXPA00009063A MXPA/A/2000/009063A MXPA00009063A MXPA00009063A MX PA00009063 A MXPA00009063 A MX PA00009063A MX PA00009063 A MXPA00009063 A MX PA00009063A MX PA00009063 A MXPA00009063 A MX PA00009063A
Authority
MX
Mexico
Prior art keywords
protein
cells
ser
binding
leu
Prior art date
Application number
MXPA/A/2000/009063A
Other languages
Spanish (es)
Inventor
Elizabeth Esther Mary Bates
Sem Saeland
Serge J E Lebecque
Lionel Chalus
Ahn B Quan
Daniel M Gorman
Joseph H Philipps Jr
Original Assignee
Schering Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Schering Corporation filed Critical Schering Corporation
Publication of MXPA00009063A publication Critical patent/MXPA00009063A/en

Links

Abstract

Nucleic acids encoding various dendritic cell membrane proteins, reagents related thereto, including specific antibodies, and purified proteins are described. Methods of using said reagents and related diagnostic kits are also provided.

Description

MEMBRANE PROTEIN GENES ISOLATED FROM MAMMALS. AND RELATED REAGENTS FIELD OF THE INVENTION The present invention contemplates compositions related to genes present in lymphocytes, for example, cells that function in the immune system. These genes function in the control of the development, differentiation and / or physiology of the mammalian immune system. In particular, the application provides nucleic acids, proteins, antibodies, and methods of using them.
BACKGROUND OF THE INVENTION The circulating component of the circulatory system of mammals comprises several types of cells, including white blood cells and red blood cells of erythroid and myeloid cell lineages. See, for example, Rapaport (1987) Introduction to Hematology (2nd ed.) Lippincott, Philadelphia, PA; Jandl (1987) Blood: Textbook of Hematoloqy, Little, Brown and Co., Boston, MA .; and Paul (ed. 1993) Fundamental Immunology (3rd ed.) Raven Press, N.Y. Dendritic cells (DC) are cells that present or process antigens, and are found in all tissues of the body. They can be classified into several categories, including: interstitial dendritic cells of the heart, kidney, intestine and lung; Langerhans cells in the skin and mucous membranes; interdigitating dendritic cells in the thymic medulla and secondary lymphoid tissue; and dendritic cells of the blood and lymph. Although the dendritic cells in each of these compartments are CD45 + leukocytes that apparently originate from the bone marrow, they may exhibit differences that are related to the microenvironment and its maturation state. These dendritic cells efficiently process and present antigens to, for example, T cells. They stimulate memory T cell responses and unaffected in the paracortical area of secondary lymphoid organs. There is some evidence of a role in the induction of tolerance. The primary and secondary B cell follicles contain follicular dendritic cells that trap and retain intact antigens as immune complexes for long periods. These dendritic cells present native antigens to B cells, and probably intervene in the maturation of antibodies by affinity, the generation of immune memory, and the maintenance of immune and humoral responses. Monocytes are phagocytic cells that belong to the mononuclear phagocyte system, and reside in the circulation. See Roitt (ed.) Encyclopedia of Immunoloqy, Academic Press, San Diego. These cells originate in the bone marrow and remain only for a short time in the marrow compartment once they differentiate. They can enter the circulation and can remain there for a relatively long period, for example, a few days. Monocytes can enter tissues and body cavities through the process designated as diapedesis, where they differentiate into macrophages and possibly dendritic cells. In an inflammatory response, the number of monocytes in the circulation can be doubled, and much of the increased number of monocytes enters the site of inflammation. The presentation of antigens refers to cellular events in which a proteinaceous antigen is absorbed, processed by antigen presenting cells (APC), and then recognized to initiate an immune response. The most active cells that present antigen have been characterized as macrophages, which are direct products of the development of monocytes, dendritic cells and certain B cells. Macrophages are present in most tissues, and are highly active in the incorporation of a wide variety of protein antigens and microorganisms. They have a highly developed endocytic activity, and they secrete many important products in the initiation of an immune response. For this reason, it is thought that many genes expressed by monocytes or induced by monocyte activation are probably important in uptake, processing, presentation or regulation of the resulting immune response due to the antigen. However, dendritic cells and monocytes are poorly characterized in terms of the proteins they express, as well as many of their functions and mechanisms of action, including their activated states. In particular, the processes and mechanisms related to the initiation of an immune response, including the presentation and processing of the antigen, remain uncertain. The lack of knowledge about the structural, biological and physiological properties of these cells limits their knowledge. In this way, medical conditions where the regulation, development or physiology of cells that present antigen is unusual, continue to be difficult to manage.
BRIEF DESCRIPTION OF THE INVENTION The present invention is based, in part, on the discovery of several mammalian Schering dendritic cell membrane protein (SDCMP) genes. The distribution data indicate a wider cellular distribution, and the structural data suggest a certain function and are exemplified by the specific SDCMP3 and SDCMP4 modalities. The SDCMP 3 and 4 modalities exhibit similarity to a class of asialoglycoprotein lectins and receptors (ASGPR). The invention encompasses agonists and antagonists of the gene products, for example, mutations (muteins) of the natural sequences, fusion proteins, chemical mimetics, antibodies and other structural or functional analogues. It is also directed to isolated genes that encode proteins of the invention. Various uses of these different protein or nucleic acid compositions are also provided. In particular embodiments, the invention provides a binding compound comprising an antibody binding site that specifically binds to a SDCMP3 or SDCMP4 protein. In preferred embodiments, in the binding compound, the antibody binding site is specifically immunoreactive with a protein of SEQ ID NO: 2, 4, 6 or 8; produced against a human or mouse SDCMP3 protein purified or produced recombinantly; produced against a human SDCMP4 protein purified or produced recombinantly; in a monoclonal antibody, Fab or F (ab) 2; or the binding compound is detectably labeled; sterile; or in a composition regulated in its pH. The invention encompasses methods of using said binding compounds and which comprise contacting the binding compound with a biological sample comprising an antigen to form a complex of binding compound: antigen. In certain embodiments, the biological sample is a human or rodent, and the binding compound is an antibody.
The invention also provides detection equipment comprising said binding compound and material with instructions for the use of said binding compound for detection; or a compartment that provides segregation of the binding compound. The invention also provides a substantially pure or isolated polypeptide, which binds specifically to said binding compounds.
In various embodiments, the polypeptide comprises at least one fragment of at least 14 amino acid residues of a primate or rodent SDCMP3 protein; comprises at least 14 amino acids of primate SDCMP4; it is a soluble polypeptide; it is marked in detectable form; it is in a sterile composition; it is in a composition regulated in its pH; it binds to a sialic acid residue; it is produced in recombinant form, or has a naturally occurring sequence of polypeptides. Nucleic acid modalities, including a nucleic acid encoding a prior polypeptide, are provided when it is purified. Frequently, the nucleic acid comprises at least 30 nucleotides of the coding portion of SEQ ID NO: 1 or 3; comprises at least 30 nucleotides of the coding portion of SEQ ID NO: 5 or 7; or may comprise an insert which hybridizes selectively with a nucleic acid encoding a polypeptide defined above. The invention also provides a cell transfected with said nucleic acid, for example, which consists of the portions of SEQ ID NO: 1, 3, 5 or 7 which code for protein. The invention also provides methods of using at least one strand of said nucleic acids to form a duplex nucleic acid, and which comprise the step of contacting said strand with a sample with a complementary strand capable of hybridizing in a specific manner. In preferred embodiments, the method allows detecting the duplex, or allows histological localization of the duplex.
Alternatively, the invention provides methods for using a described binding composition, which comprises the step of contacting the binding composition with a sample to form an antigen: binding composition complex. In preferred embodiments, the sample is a biological sample, including a body fluid; the antigen is in a cell; or the antigen is further purified. The invention further encompasses methods for using said polypeptides, which comprises contacting the polypeptide with a sample to form a polypeptide: binding composition complex. In preferred embodiments, the polypeptide is further purified. Another method provided is to modulate the physiology or function of the dendritic cells, which comprises the step of contacting the cell with a binding composition, as described; a SDCMP3 or SDCMP4 protein, as described; or a polypeptide, as described. The function can also result in the initiation or progression of an immune response. Typically, the contacting is in combination with an antigen, including a cell surface antigen, MHC class I antigen or MHC class II antigen.
DETAILED DESCRIPTION OF THE INVENTION All references cited herein are incorporated herein by reference to the same degree as indicating that each individual patent application or publication was specifically and individually incorporated as a reference in its entirety for all purposes.
I. General The present invention provides DNA sequences that encode mammalian proteins expressed in dendritic cells (DC). For a review of dendritic cells, see Steinman (1991) Annual Review of Immunology 9: 271-296; and Banchereau and Schmitt (eds. 1994) Dendritic Cells in Fundamental and Clinical Immunology, Plenum Press, NY. These proteins are designated as dendritic cell proteins because they are found in these cells, and they seem to exhibit a certain specific character in their expression. Specific modalities of these proteins in humans are provided below. The following descriptions are directed, for examples of purposes, to human DC genes, but are also applicable to structurally related modalities, eg, of sequence, from other sources or species of mammals, including individual or polymorphic variants. These will include, for example, proteins exhibiting relatively few sequence changes, for example, less than about 5%, and in number, for example, fewer than 20 substitutions of amino acid residues, typically less than 15, preferably less than 10. , and more preferably less than 5 substitutions, including 4, 3, 2 or 1. These will also include versions that are truncated of the total length, as described, and fusion proteins that contain substantial segments of these sequences.
II. Definitions The term "binding composition" refers to molecules that bind specifically to these DC proteins, for example, in an antibody-antigen interaction. Other compounds, e.g., proteins, can also be specifically associated with the respective protein. Typically, the specific association will be in a natural physiologically relevant protein-protein interaction, either covalent or non-covalent, and may include members of a multiple protein complex, including carrier compound or dimerization members. The molecule can be a polymer or chemical reagent. A functional analog can be a protein with structural modifications, or it can be a totally unrelated molecule, for example, which has a molecular form that interacts with the appropriate interaction determinants. The variants can function as protein agonists or antagonists; see, for example, Goodman, et al. (eds.) (1990) Goodman and Gilman's: The Pharmacological Bases of Therapeutics (8th ed.) Pergamon Press, Tarrytown, N.Y. The term "DC protein complex: binding agent", as used herein, refers to a complex of binding agent and DC protein. The specific binding of the binding agent means that the binding agent has a specific binding site that recognizes a site in the respective DC protein. For example, antibodies produced for the DC protein and which recognize an epitope in the DC protein are capable of forming a DC: antibody protein complex by specific binding. Typically, the formation of a complex of binding agent: DC protein allows the measurement of that DC protein in a mixture of other proteins and biological compounds. The term "DC protein complex: antibody" refers to a protein complex of DC: binding agent in which the binding agent is an antibody. The antibody can be monoclonal, polyclonal or even an antigen-binding fragment of an antibody, for example, including Fv, Fab or Fab2 fragments. The "homologous" nucleic acid sequences, when compared, exhibit significant similarity. Standards for nucleic acid homology are measurements for homology that are generally used in the art by comparison of sequences and / or phylogenetic relationship, or based on hybridization conditions. Hybridization conditions are described in greater detail below. An "isolated" nucleic acid is a nucleic acid, e.g., an RNA, DNA, or a mixed polymer, which is substantially separated from other components that naturally accompany a native sequence, e.g., proteins and flanking genomic sequences. the species from which they originate. The term encompasses a nucleic acid sequence that has been removed from its naturally occurring environment, and includes recombinant or cloned DNA isolates and chemically synthesized analogs or analogs synthesized biologically by heterologous systems. A substantially pure molecule includes isolated forms of the molecule. An isolated nucleic acid will generally be a homogeneous molecule composition but, in some embodiments, will contain minor heterogeneity. This heterogeneity is typically found at polymer ends or portions not critical for a desired biological function or activity. As used herein, the term "SDCMP3 protein" should encompass, when used in the context of a protein, a protein having amino acid sequences as shown in SEQ ID NO: 2 or 4, or a significant fragment of said protein. It refers to a polypeptide that interacts with the respective specific binding components of the SDCMP3 protein. These binding components, e.g., antibodies, typically bind to the SDCMP3 protein with high affinity, for example, at least about 100 nM, usually better than about 30 nM, preferably better than about 10 nM, and more preferably better of approximately 3 nM. Similarly, the use of the term SDCMP4 will be applied in relation to SEQ ID NO: 6 or 8. The term "polypeptide" or "protein", as used herein, includes a significant fragment or segment of said protein, and encompasses a stretch of amino acid residues of at least about 8 amino acids, usually at least 10 amino acids, more generally at least 12 amino acids, often at least 14 amino acids, more often at least 16 amino acids, typically at least 18 amino acids, more typically at least 20 amino acids, usually at least 22 amino acids, more usually at least 24 amino acids, preferably at least 26 amino acids, more preferably at least 28 amino acids, and, in particularly preferred embodiments , at least about 30 or more amino acids, for example, 35, 40, 45, 50, 60, 70, etc. A "recombinant" nucleic acid is typically defined by its structure. It can be a nucleic acid that is obtained by generating a sequence comprising the fusion of two fragments which are not naturally contiguous with each other, but it means that it excludes products of nature, for example, mutant forms that occur naturally. Certain forms are defined by a production method. In this regard, for example, a product obtained by a process, the method consists in the use of recombinant nucleic acid techniques, for example, involving human intervention in the nucleotide sequence, typically by selection or production. Thus, the invention encompasses, for example, nucleic acids comprising a sequence derived using an oligonucleotide synthesis method, and products obtained by transforming cells with a non-naturally occurring vector encoding these proteins. Frequently, this is done to replace a codon with a redundant codon that codes for the same amino acid or a conservative amino acid, while typically introducing or removing a site for recognition of the sequence, for example, for a restriction enzyme. Alternatively, it is carried out to join together nucleic acid segments of desired functions to generate an individual genetic entity comprising a desired combination of functions not present in the commonly available natural forms. Restriction enzyme recognition sites are often the target of such artificial manipulations, but other site-specific targets can be incorporated by design, for example, promoters, DNA replication sites, regulatory sequences, control sequences, or other useful features, for example, primer segments. A similar concept is used for a recombinant, for example, fusion polypeptide. Specifically included are synthetic nucleic acids which, by redundancy of the genetic code, code for polypeptides similar to fragments of these antigens, and fusions of sequences of several variants of different species. The "solubility" is reflected by the sedimentation measured in Svedberg units, which are a measure of the sedimentation rate of a molecule under particular conditions. The determination of the sedimentation rate was conventionally carried out in an analytical ultracentrifuge, but now it is typically carried out in a standard ultracentrifuge. See Freifelder (1982) Phvsical Biochemistry (2nd ed.) Freeman and Co., San Francisco, CA; and Cantor and Schimmel (1980) Biophysical Chemistry, parts 1-3, Freeman and Co., San Francisco, CA. As a general determination, a sample containing a putatively soluble polypeptide is rotated in a fully dimensioned standard ultracentrifuge at approximately 50K rpm for approximately 10 minutes, whereby the soluble molecules will remain in the supernatant. A soluble particle or polypeptide will typically be less than about 30S, more typically less than about 15S, usually less than about 10S, more usually less than about 6S, and, in particular embodiments, preferably less than about 4S, and more preferably less than about 3S. The solubility of a polypeptide or fragment depends on the environment and the polypeptide. Many parameters affect the solubility of the polypeptide, including temperature, electrolyte environment, size and molecular characteristics of the polypeptide, and the nature of the solvent. Typically, the temperature at which the polypeptide is used ranges from about 4 ° C to about 65 ° C. Usually, the temperature during use is greater than about 18 ° C, and more usually greater than about 22 ° C. For diagnostic purposes, the temperature will usually be almost room temperature or higher, but lower than the denaturing temperature of the components in the test. For therapeutic purposes, the temperature will usually be body temperature, typically about 37 ° C for humans, although under certain situations, the temperature may be higher or lower in situ or in vitro.
The size and structure of the polypeptide should generally be in a substantially stable and physiologically active state, and usually not in a denatured state. The polypeptide may be associated with other polypeptides in a quaternary structure, for example, to confer solubility, or associated with lipids or detergents in such a way as to approximate the natural interactions of the lipid bilayer. The solvent will usually be a biologically compatible pH regulator, of a type used for the preservation of biological activities, and will usually approximate a physiological solvent. Usually, the solvent will have a neutral pH, typically between about 5 and 10, and preferably about 7.5. In some instances, a detergent, typically a mild non-denaturing detergent, will be added, for example, CHS (cholesteryl hemisuccinate) or CHAPS (3 - ([3-colamidopropyl dimethylammonium] -1-propane) sulfonate, or a rather low detergent concentration to avoid significant disruption of the structural or physiological properties of the protein. "Substantially pure" typically means, for example, in the context of a protein, that the protein is isolated from other contaminating proteins, nucleic acids, or other biological compounds derived from the original organism. The purity, or "isolation", can be tested by standard methods, typically by weight, and will often be at least about 50% pure, more often at least about 60% pure, generally at least about 70% pure, more generally at least about 80% pure, often at least about 85% pure, more often at least about 90% pure, preferably at least about 95% pure, more preferably at least about 98% pure % and, in the most preferred embodiments, at least about 99% pure. Frequently, vehicles or excipients will be added, or the formulation may be sterile or may comprise pH regulator components. "Substantial similarity" in the context of comparing nucleic acid sequences means that the segments, or their complementary strands, when compared, are identical when they are optimally aligned, with insertions or deletions of appropriate nucleotides, in at least about 50% of nucleotides, usually at least 56%, more generally at least 59%, often at least 62%, more often at least 65%, often at least 68%, with more frequency at least 71%, typically at least 74%, more typically at least 77%, usually at least 80%, more usually at least about 85%, preferably at least about 90%, more preferably at least at least about 95 to 98% or more and, in particular embodiments, as high as about 99% or more of the nucleotides. Alternatively, there is substantial similarity when the segments hybridize under conditions of selective hybridization, with a chain, or its complement, typically using a sequence derived from SEQ ID No: 1, or appropriate parts of 3. Typically, selective hybridization will occur when there is one. at least about 55% similarity over a stretch of at least about 30 nucleotides, preferably at least about 65% over a stretch of at least about 25 nucleotides, more preferably at least about 75%, and most preferably at least about 90% over about 20 nucleotides. See Kanehisa (1984) Nucí. Acids Res. 12: 203-213. The length of the similarity comparison, as described, can be over longer stretches and, in certain embodiments, will be over a stretch of at least about 17 nucleotides, usually at least about 20 nucleotides, more usually at least about 24 nucleotides, typically at least about 28 nucleotides, more typically at least about 40 nucleotides, preferably at least about 50 nucleotides, and more preferably at least about 75 to 100 or more nucleotides. Comparison measurements for the SDCMP3 protein are not reflected in the comparison measures for the SDCMP4 protein. For sequence comparison, typically a sequence functions as a reference sequence with which test sequences are compared. When an algorithm is used for sequence comparison, the test and reference sequences are fed to a computer, the sequence coordinates are designated, if necessary, and the program parameters of the sequence algorithm are designated. The algorithm for sequence comparison allows the percentage of sequence identity for the test sequence to be calculated with respect to the reference sequence, based on the parameters of the designated program. The optical alignment of the sequences for comparison can be carried out, for example, by the local homology algorithm of Smith and Waterman (1981) Adv. Appl. Math. 2: 482, by the homology alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol. 48: 443, by the similarity search method of Pearson and Lipman (1988) Proc. Nati Acad. Sci. USA 85: 2444, through computational implementations of these algorithms (GAP, BESTFIT, FASTA and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wl), or by visual inspection (see generally Ausubel, et al., Cited above). An example of a useful algorithm is PILEUP. PILEUP creates an alignment of multiple sequences from a group of related sequences using progressive pairs alignments to show the relationship and percentage of sequence identity. This algorithm also allows to graph a tree or dendrogram that shows the grouping relationships used to create the alignment. PILEUP uses a simplification of the progressive alignment method of Feng and Doolittle (1987) J. Mol. Evol. 35: 351-360. The method used is similar to the method described by Higgins and Sharp (1989) CABIOS 5: 151-153. The program can align up to 300 sequences, each with a maximum length of 5000 nucleotides or amino acids. The multiple alignment procedure begins with the pairwise alignment of the two most similar sequences, producing a group of two aligned sequences. This group is then aligned with the most related sequence or group of aligned sequences. Two groups of sequences are aligned by a simple extension of the alignment in pairs of two individual sequences. The final alignment is achieved through a series of progressive alignments in pairs. The program is carried out by designating specific sequences and their amino acid or nucleotide coordinates for regions of sequence comparison, and designating the parameters of the program. For example, a reference sequence can be compared with other test sequences to determine the percent identity of sequences using the following parameters: weight of the missing space (3.00), weight of the missing space length, (0.10) and heavy terminal spaces. Another example of an algorithm that is suitable for determining the percentage of sequence identity and sequence similarity is the BLAST algorithm, which is described by Altschul, et al. (1990) J. Mol. Biol. 215: 403-410. Programs to conduct analyzes with the BLAST algorithm are publicly available through the National Center for Biotechnology Information (http: www.ncbi.nlm.nih.gov/). This algorithm involves first identifying high-scoring sequence pairs (HSPs) by identifying short words of length W in the sequence in question, which equals or satisfies 2 a certain threshold value T of positive value when it is aligned with a word of the same length in a database sequence. T is referred to as the score threshold for neighboring words (Altschul, et al., Cited above). These initial neighbor word hits work like seeds to initiate searches to find longer HSPs that contain them. The punches of words then extend in both directions along each sequence as long as the cumulative alignment score can be increased. The extent of the word hits in each direction stops when the cumulative alignment score decreases by the amount X from its maximum reached value; the cumulative score goes up to 0 or less, due to the accumulation of one or more alignments of negative scoring residues; or the end of any sequence is reached. The parameters of the BLAST algorithm, namely W, T and X, determine the sensitivity and speed of the alignment. The BLAST program uses as omissions a word length (W) of 11, the alignments of the BLOSUM62 scoring matrix (see Henikoff and Henikoff (1989) Proc. Nat'l Acad. Sci. USA 89: 10915) (B) 50, expectation (E) of 10, M = 5, N = 4, and a comparison of both chains. In addition to allowing to calculate the percentage of sequence identity, the BLAST algorithm allows carrying out a statistical analysis of the similarity between two sequences (see, for example, Karlin and Altschul (1993) Proc. Nat'l Acad. Sci. USA. 90: 5873-5787 A measure of the similarity provided by the BLAST algorithm is the smallest sum probability (P (N)), which provides an indication of the probability by which a match between two sequences of events would occur by chance. nucleotides or amino acids For example, a nucleic acid sequence is considered to be similar to a reference sequence if the probability of the smallest sum in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.1, more preferably less than about 0.01, and most preferably less than about 0.001 Another indication that two polypeptide nucleic acid sequences are substantially and identical, is that the polypeptide encoded by the first nucleic acid cross-reacts immunologically with the polypeptide encoded by the second nucleic acid, as described below. In this manner, a polypeptide is typically substantially identical to a second polypeptide, for example, wherein the two peptides differ only by conservative substitutions. Another indication that two nucleic acid sequences are substantially identical is that the two molecules hybridize to each other under stringent conditions, as described below. "Strict conditions", in relation to homology or substantial similarity in the hybridization context, will be strict conditions of combined salinity, temperature, organic solvents and other parameters, typically those controlled in hybridization reactions. The combination of parameters is more important than the measurement of any 2 individual parameter. See, for example, Wetmur and Davidson (1968) J. Mol. Biol. 31: 349-370. A nucleic acid probe that binds to a target nucleic acid under stringent conditions is specific for said target nucleic acid. Said probe is typically greater than 11 nucleotides in length, and is sufficiently identical or complementary to a target nucleic acid over the region specified by the sequence of the probe that binds to the target under stringent hybridization conditions. Generally, a positive signal will exhibit a signal at least 2 times on the background, preferably at least 5 times, and more preferably at least 15, 25 or even 50 times on the background. SDCMP proteins counterpart of other mammalian species, for example, primate or rodent, can be cloned and isolated by hybridization of crossed species of closely related species. See, for example, later. The similarity may be relatively low between distantly related species, and thus hybridization of relatively closely related species is advisable. Alternatively, the preparation of an antibody preparation exhibiting less species-specific character may be useful in cloning procedures for expression. The phrase "binds specifically to an antibody" or "specifically immunoreactive with", when referring to a protein or peptide, refers to a binding reaction which is determinative of the presence of the protein in the presence of a heterogeneous population of proteins and other biological components. In this manner, under designated immunoassay conditions, the specified antibodies bind to a particular protein and do not bind significantly to other proteins present in the sample. Specific binding to an antibody under such conditions may require an antibody that is selected for its specific character by a particular protein. For example, antibodies produced for the human immunogenic SDCMP3 protein can be selected with the amino acid sequence shown in SEQ ID NO: 2 to obtain antibodies specifically immunoreactive with the SDCMP protein, and not with other proteins. These antibodies recognize proteins highly similar to the human homologous SDCMP3 protein.
III. Nucleic acids These SDCMP genes are expressed selectively in dendritic cells. Preferred embodiments, as described, will be useful in standard procedures for isolating genes from other species, e.g., warm-blooded animals, such as mls and birds. Cross-hybridization will allow the isolation of related proteins from individuals, races or species. There are numerous different methods for successfully isolating a suitable nucleic acid clone based on the information provided herein. Southern blot hybridization studies should allow the identification of homologous genes in other species under suitable hybridization conditions.
Purified proteins or defined peptides are useful for generating antibodies by standard methods, as described below. Synthetic peptides or purified proteins can be presented to an immune system to generate polyclonal and monoclonal antibodies. See, for example, Coligan (1991) Current Protocols in Immunoloqy and Wiley / Greene, NY; and Harlow and Lane (1989) Antibodies: A Laboratory Manual Cold Spring Harbor Press, NY, citation incorporated herein by reference. Alternatively, an antigen-binding composition of SDCMP can be useful as a specific binding reagent, and one can take advantage of its specific binding character, for example, for the purification of a SDCMP protein. The specific binding composition can be used to select an expression library obtained from a cell line that expresses the SDCMP protein. There are many selection methods, for example, standard staining of the ligand expressed on its surface, or by panning. The selection of intracellular expression can also be carried out by various staining or immunofluorescence procedures. The binding compositions could be used for affinity purification or to select cells that express the antigen. Sequence analysis suggests that these SDCMPs are members of the lectin / asialoglycoprotein receptor superfamily. See also USSN 60 / 053,080, which is incorporated herein by reference.
Sequences encoding a primate SDCMP3, initially designated as lectin 73, isolated from a dendritic cell library, are shown in SEQ ID NO: 1 and 2. The open reading frame ranges from approximately 108 to 593. The comparison with SDCMP3 of rodent suggests that the sequence can be truncated at or near the C terminal. The sequence encoding the mouse counterpart is shown in SEQ ID NO: 3 and 4. Analysis of the human SDCMP3 protein suggests that the protein is a type II membrane protein, where the transmembrane segment is approximately ser22 to thr42. The cytoplasmic tail would be in the N terminal, from met to trp21. A type C lectin domain corresponds to approximately cys79 to arg162. The human protein has a predicted molecular weight of approximately 18, 500 daltons, with an isoelectric point of approximately 6, and a charge of approximately -2.6 to pH 7. Analysis of the hydrophilic character indicates significant sections of hydrophilic sequence of approximately 1-22, 42-63, 94-106 and 142- 162 These segments will probably be more antigenic. Similar analysis of the mouse SDCMP3 protein suggests that the protein is also a type II membrane protein, where the transmembrane segment ranges from about 20 to thr40. The cytoplasmic tail would also go from approximately met to trp19; and the type C lectin domain would correspond to approximately cys79 to at least arg162. Two putative N-glycosylation sites correspond to asn131-ser133 and asn 183-ser185. The particularly antigenic and computationally identified stretches for the human protein would encompass approximately met1-ser18; tyr43-arg53; Iys72-ser85; ser94-asn106; and ser135-arg162. See, for example, Beattie, et al. (1992) Eur. J. Biochem. 210: 59-66. The alignment of the primate SDCMP3 protein with the rodent SDCMP3 protein is shown in the following table: PICTURE pr MMQEQQPQST EKRGWLSLRL WSVAGISIAL LSACFIVSCV VTYHFTYGET MVQERQSQGK -GVCWT ro-LRL WSAAVIS LL LSTCFIASCV VTYQFI DQP pr GKRLSELHSY HSSLTCFSEG TKV-PAWGC CPASWKSFGS SCYFISSEEK ro SRRLYELHTY HSSLTCFSEG TMVSEKMWGC CPNHWKSFGS SCY ISTKEN pr VWSKSEQNCV EMGAHLWFN TEAEQNFIVQ QLNESFSYFL GLSDPQGNNN ro FWSTSEQNCV QMGAHLWIN TEAEQNFITQ QLNESLSYFL GLSDPQGNGK pr WQWIDKTPYE knvr ro WQWIDDTPFS QNVRFWHPHE PNLPEERCVS IVYWNPSKWG WNDVFCDSKH ro NSICEMKKIY L Analysis of the human SDCMP4 protein suggests that the protein is a type II membrane protein. There are two forms, the long form, designated initially as lectin 47, isolated from a dendritic cell library (SEQ ID NO: 5 and 6), and the short form (SEQ ID NO: 7 and 8), which corresponds to a deletion of nucleotides 362-499 of the long form, and which may result from an alternative splicing event. There are also differences in nucleotides 108 and 239. Mixed variations in the sequence may reflect sequencing errors or allelic variants. The predicted transmembrane segment of the long form ranges from approximately Ieu45 to met67. The proximal portion of the protein would be cytoplasmic. The particularly antigenic and computationally identified stretches for the human protein will encompass approximately met1-arg44; trp70-thr113; and asn139-cys220. A notable feature is the motif of incorporation (GYTQ, residues 14-17) in the intracytoplasmic domain. The CRD would range from about cys120 to met247 in the long form, and from about cys74 to met201 in the short form. The long shape would be predicted to have a molecular weight of approximately 27.6 kD, and the shape short approximately 22.5 kD with a calculated isoelectric point of approximately 4.6, and a charge of -7.8 to pH 7. The extracellular domain of the SDCMP4 proteins contains a carbohydrate recognition domain (CRD) - type C lectin (Ca ++ dependent), as indicated by significant sequence homology with other lectins. The prototype type C transmembrane type II lectins is the hepatic receptor of asyglycoprotein (ASGPR). The CRF of the hepatic ASGPR exhibits specific binding character for galactose. In addition, the intracellular domain of the ASGPR possesses a tyrosine-based motif that allows the incorporation of the ligand. Unlike the ASGPR or the macrophage mannose receptor, the SDCMP4 CRD sequence does not so emphatically suggest its specific character for sugars. This lack of suggestion is also a characteristic of type C lectins, as exemplified by NGK2 receptors in NK cells. The intracellular domains of both SDCMP4 modalities exhibit an incorporation sequence (YTQL) of the type YXX0, where 0 represents a hydrophobic amino acid. As a reference, the reason for incorporation of the ASGPRH1 chain in the liver is YQDL. Notably, several Type II transmembrane type C lectins (eg, NKG2 and human DC-IR, Ly49 and mouse NKRP1), are members of the immunoreceptor superfamily (IRS) system. Some forms of these receptors have the ability to deliver an inhibitory signal through an intracellular ITIM motif. In contrast, other forms lack an ITIM motif, and as such do not transmit a negative signal. One characteristic of such non-inhibitory IRS members is the presence of an amino acid with charge in the transmembrane region. Alternatively, the truncated forms may interact with transmembrane accessory molecules. See, for example, Lanier, et al. (1998) Nature 391: 703-7; and USSN 60 / 069,639, citations which are incorporated herein by reference. SDCMP4 neither exhibits an ITIM motif in its intracellular domain, nor a transmembrane residue with charge. On this basis, it seems unlikely that SDCMP4 will define a new family of type C lectin IRS genes.
Rather, it can be suggested that SDCMP4 is related to the system of ASGPR molecules that are involved in the incorporation of the ligand. Two forms of SDCMP4 have been identified, which differ by the presence of a membrane-proximal insertion of 46 amino acids in the extracellular domain. Insertions in this region also occur in the ASGPRs of macrophages and dendritic cells (ETA10). Finally, the expression of SDCMP4 has been observed by RT-PCR in myeloid cells (dendritic cells, monocytes and granulocytes). In contrast to SDCMP3, the expression of SDCMP4 is not down-regulated in DC after activation by PMA and ionomycin. The CRF of the hepatic ASGPR exhibits specific binding character for galactose. In addition, the intracellular domain of the ASGPR has a tyrosine-based motif that allows the incorporation of the ligand. Unlike the ASGPR or the macrophage mannose receptor, the SDCMP4 CRD sequence does not so emphatically suggest its specific character for sugars. This lack of suggestion is also a characteristic of type C lectins, as exemplified by NGK2 receptors in NK cells. The intracellular domain of SDCMP4 exhibits an incorporation sequence (YTQL) of the type YXX0, where 0 represents a hydrophobic amino acid. As a reference, the reason for incorporation of the ASGPRH1 chain in the liver is YQDL. Notably, several Type II transmembrane type C lectins (eg, NKG2 and human DC-IR, Ly49 and mouse NKRP1), are members of the immunoreceptor superfamily (IRS) system. Some forms of these receptors have the ability to deliver an inhibitory signal through an intracellular ITIM motif. In contrast, other forms lack an ITIM motif, and as such do not transmit a negative signal. One characteristic of such non-inhibitory IRS members is the presence of an amino acid with charge in the transmembrane region. SDCMP4 neither exhibits an ITIM motif in its intracellular domain, nor a transmembrane residue with charge. On this basis, it seems unlikely that SDCMP4 will define a new family of type C lectin IRS genes.
Rather, it can be suggested that SDCMP4 is related to the system of ASGPR molecules that are involved in the incorporation of the ligand. Two forms of SDCMP4 have been identified, which differ by the presence of a membrane proximal insertion of 46 amino acids in the extracellular domain. Insertions in this region also occur in the ASGPRs of macrophages and dendritic cells (ETA10). Finally, the expression of SDCMP4 has been observed by RT-PCR in myeloid cells (dendritic cells, monocytes and granulocytes). In contrast to SDCMP3, the expression of SDCMP4 is not down-regulated in DC after activation by PMA and ionomycin. The sequences next to these are the ETA10 sequences. See, for example, Suzuki, et al. (1996) J. Immunol. 156: 128-135; and Sato, et al. (1992) J. Biochem. 111: 331-336. The extracellular domain exhibits a number of characteristics indicative of a carbohydrate recognition domain (CRD) of type C (dependent on Ca ++). While the CRD of the human form appears truncated at its carboxylic end, the CRD of the mouse homologue (1469D4) is not truncated and clearly classifies the lectin as a novel member of the type C superfamily. The prototype of the lectins of Type II transmembrane type C is the hepatic receptor of asialoglycoprotein (ASGPR). However, the ASGPR possesses an intracytoplasmic sequence of incorporation of tyrosine-based ligand, which is not found in SDCMP3 neither of human nor of mouse. The gene coding for human SDCMP3 maps to chromosome 12 p12-13, for example, in the human NK receptor complex. Notably, this region includes the genes of NKG2 and the CD94 gene, which codes for Type II transmembrane type I iectins, and represents examples of the immunoreceptor superfamily (IRS) system. In this manner, the CD94-NKG2A / B heterodimers of killer cell inhibitor (KIR) receptors transduce a negative signal by virtue of a tyrosine-based intracellular ITIM motif in the NKG2 sequences. However, the other forms of NKG2 lack an ITIM motif, so the resulting heterodimers with CD94 are non-inhibitory. The intracellular domain of human SDCMP3 does not contain an ITIM motif. However, based on its chromosomal location, as well as its significant homology (36.2%) with the DC-IR of the IRS gene, it is predicted that it is a member of a novel family of type C lectin gene of the IRS. By analogy with other IRS genes, it is likely that SDCMP3 represents a family of genes that will comprise several members, with inhibitory (ITIM) or non-inhibitory function. By RT-PCR, the expression of SDCMP3 in primates is restricted to myeloid cells, being observed in dendritic cells (DC), monocytes and macrophages. The expression is selectively observed in DC derived from CD14, rather than in DC of the Langerhans type derived from CD1a. Finally, the expression of SDCMP3 is down-regulated by activation with PMA with ionomycin. Peptide segments can also be used to design and produce appropriate oligonucleotides to select a library for the presence of a similar gene, for example, an identical or polymorphic variant, or to identify a DC. The genetic code can be used to select suitable oligonucleotides useful as probes for selection. In combination with polymerase chain reaction (PCR) techniques, synthetic oligonucleotides will be useful for selecting desired clones from a library. Complementary sequences will also be used as probes or primers. Based on the identification probably of the terminal amino group, other peptides must be particularly useful, for example, coupled with anchor vector or complementary poly-A PCR techniques or with DNA complementary to other peptides. Techniques for nucleic acid manipulation of genes that code for these DC proteins, for example, subcloning of nucleic acid sequences encoding polypeptides into expression vectors, labeling probes, DNA hybridization, and the like, are generally described in Sambrook, et al. (1989) Molecular Cloning: A Laboratory Manual (2nd ed.) Vols. 1-3, Cold Spring Harbor Laboratory, Cold Spring Harbor Press, NY, citation incorporated herein by reference, and referred to in the sequential as "Sambrook, et al." See also Coligan, et al. (1987 and periodic supplements) Current Protocols in Molecular Biology Greene / Wiley, New York, NY, cited as "Coligan, et al": There are several methods to isolate the DNA sequences that code for these DC proteins. For example, DNA is isolated from a genomic or cDNA library using labeled oligonucleotide probes having sequences identical or complementary to the sequences described herein. Full length probes can be used, or oligonucleotide probes can be generated by comparing the described sequences with other proteins, and selecting specific primers. Such probes can be used directly in hybridization assays to isolate DNA encoding DC proteins, or probes can be designed for use in amplification techniques such as PCR, for the isolation of DNA encoding DC proteins. To prepare a cDNA library, messenger RNA is isolated from cells expressing the DC protein. CDNA is prepared from the messenger RNA, and ligated into a recombinant vector. The vector is transfected into a recombinant host for propagation, selection and cloning. Methods for obtaining and selecting cDNA libraries are well known. See Gubler and Hoffman (1983) Gene 25: 263-269; Sambrook, et al .; or Coligan, et al. For a genomic library, the cDNA can be extracted from the tissue, and mechanically cut or enzymatically digested to produce fragments of approximately 12-20 kb. The fragments are then separated by gradient centrifugation and cloned in lambda bacteriophage vectors. These vectors and phage are packaged in vitro, as described, for example, in Sambrook, et al. or Coligan, et al. The recombinant phages are analyzed by plaque hybridization, as described in Benton and Davis (1977) Science 196: 180-182. Colony hybridization is carried out as generally described, for example, in Grunstein, et al. (1975) Proc. Nati Acad. Sci. USA 72: 3961-3965. The DNA encoding a DC protein can be identified in genomic or cDNA libraries for its ability to hybridize with the nucleic acid probes described herein, for example, in colony or plate hybridization experiments. The corresponding DNA regions are isolated by standard methods well known to those skilled in the art. See Sambrook, et al. Various methods for amplifying target sequences, such as the polymerase chain reaction, can also be used to prepare DNA encoding DC proteins. Polymerase chain reaction (PCR) technology is used to amplify said nucleic acid sequences directly from messenger RNA, cDNA, and from genomic libraries or cDNA libraries. The isolated sequences encoding DC proteins can also be used as templates for PCR amplification.
In PCR techniques, oligonucleotide primers complementary to two 5 'regions are synthesized in the DNA region to be amplified. The polymerase chain reaction is then carried out using the two primers. See Innis, et al. (eds.) (1990) PCR Protocols: A Guide to Methods and Applications Academic Press, San Diego, CA. Initiators can be selected to amplify the entire regions encoding a full-length selected DC protein, or to amplify shorter DNA segments, as desired. In particular, the provided sequences provide primers, for example, from 15 to 30 nucleotides, which can be used to amplify the desired coding sequences, or fragments thereof. Once said regions are amplified by PCR, they can be sequenced and oligonucleotide probes can be prepared from the sequence obtained using standard techniques. These probes can then be used to isolate DNA molecules that code for other forms of the DC proteins. Oligonucleotides for use as probes are chemically synthesized according to the solid phase phosphoramide method, described first by Beaucage and Carruthers (1983) Tetrahedron Lett. 22 (20): 1859-1862, or using an automated synthesizer, as described in Needham-VanDevanter, et al. (1984) Nucleic Acids Res. 12: 6159-6168. The purification of the oligonucleotides is carried out, for example, by native acrylamide gel electrophoresis or by anionic exchange HPLC, as described in Pearson and Regnier (1983) J. Chrom. 255: 137-149. The sequence of the synthetic oligonucleotide can be verified using the chemical degradation method of Maxan and Gilbert in Grossman and Moldave (eds. 1980) Methods in Enzymology 65: 499-560 Academic Press, New York. This invention provides isolated DNA or fragments thereof that encode a DC protein, as described. In addition, this invention also provides isolated or recombinant DNA which encodes a biologically active protein or polypeptide which is capable of hybridizing under appropriate conditions, for example, high stringency, with the DNA sequences described herein. Said biologically active protein or polypeptide can be a naturally occurring form, or a recombinant protein or fragment, and has an amino acid sequence as described in SEQ ID NO: 2, 4, 6 or 8. Preferred embodiments will be natural isolates of total length, for example, of a primate. In the glycosylated form, proteins must exhibit larger sizes. In addition, this invention encompasses the use of isolated or recombinant DNA, or fragments thereof, which code for proteins that are homologous to each respective DC protein. The isolated DNA may have the respective regulatory sequences on the 5 'and 3' flanks, for example, promoters, enhancers, poly-A addition signals, and others.
IV. Obtaining DC gene products DNA molecules that code for these DC proteins, or fragments thereof, can be obtained by chemical synthesis, selection of cDNA libraries, or by selecting genomic libraries prepared from a wide variety of cell lines or tissue samples. These DNA molecules can be expressed in a wide variety of host cells for the synthesis of a full-length protein or fragments which, for example, can be used to generate monoclonal or polyclonal antibodies; for studies of union; for construction and expression of modified molecules, and for structure and function studies. Each of these DC proteins or fragments thereof can be expressed in host cells that are transformed or transfected with appropriate expression vectors. These molecules can be substantially purified to be free of protein or cellular contaminants, apart from those derived from the recombinant host and, therefore, are particularly useful in pharmaceutical compositions when combined with a pharmaceutically acceptable carrier and / or diluent. The antigen or portions thereof can be expressed as fusions with other proteins. Expression vectors are typically self-replicating DNA or RNA constructs containing the desired DC gene or fragments, usually operably linked to suitable genetic control elements that are recognized in a suitable host cell. These control elements are capable of effecting expression within a suitable host. The specific type of control elements necessary to effect the expression will depend on the final host cell used. In general, the genetic control elements may include a prokaryotic promoter system or a control system for eukaryotic promoter expression, and typically include a transcription promoter, an optional operator to control the initiation of transcription, enhancers of transcription to raise the level of expression of messenger RNA, a sequence encoding a suitable ribosome binding site, and sequences that terminate transcription and translation. Expression vectors also usually contain an origin of replication that allows the vector to replicate independently of the host cell. The vectors of this invention contain DNA molecules that code for several DC proteins, or a fragment thereof, typically encoding, for example, a biologically active protein or polypeptide. The DNA can be under the control of a viral promoter, and can code for a selection marker. This invention further contemplates the use of said expression vectors which are capable of expressing eukaryotic cDNA encoding a DC protein in a prokaryotic or eukaryotic host, wherein the vector is compatible with the host and wherein the eukaryotic cDNA encoding for the protein is inserted into the vector in such a way that the growth of the host containing the vector expresses the cDNA in question. Typically, the expression vectors are designed for stable replication in their host cells or for amplification in order to increase the total copy number of the desired gene per cell. It is not always necessary for an expression vector to replicate in a host cell, for example, it is possible to effect transient expression of the protein or its fragments in various hosts using vectors that do not contain a replication origin that is recognized by the host cell. It is also possible to use vectors that cause integration of a DC gene or its fragments into the host DNA by recombination, or integrate a promoter that controls the expression of an endogenous gene. The vectors, as used herein, comprise plasmids, viruses, bacteriophages, integrable DNA fragments, and other vehicles that allow the integration of DNA fragments into the host's genome. Expression vectors are specialized vectors that contain elements of genetic control that effect the expression of operably linked genes. Plasmids are the most commonly used form of vectors but all other forms of vectors serving in an equivalent function are suitable for use herein. See, for example, Pouweis, et al. (1985 and Supplements) Cloning Vectors: A Laboratorv Manual Elsevier, N.Y.; and Rodríguez, et al. Vectors: A Survev of Molecular Cloning Vectors and Their Uses Buttersworth, Boston, MA. Suitable host cells include prokaryotic, lower eukaryotic and higher eukaryotic organisms. Prokaryotes include both gram negative and gram positive organisms, for example, E. coli and B. subtilis. Lower eukaryotes include yeasts, for example, S. cerevisiae and Pichia, and species of the genus Dictyostelium. Higher eukaryotes include tissue culture cell lines established from animal cells, both of non-mammalian origin, eg, insect and bird cells, and of mammalian origin, eg, humans, primates and rodents. The prokaryotic host-vector systems include a wide variety of vectors for different species. As used herein, E. coli and its vectors will be used generically to include equivalent vectors used in other prokaryotes. A representative vector for amplifying DNA is pBR322 or its derivatives. Vectors that can be used to express DC proteins or fragments include, but are not limited to, vectors such as those containing the lac promoter (pUC series); trp promoter (pBR322-trp); Ipp promoter (pIN series); lambda-pP or pR promoters (pOTS); or hybrid promoters such as ptac (pDR540). See Brosius, et al. (1988) "Expression Vectors Employing Lambda-, trp-, lac-, and Ipp-derived Promoters", in Rodriguez and Denhardt (eds.) Vectors: A Survev of Molecular Cloning Vectors and Their Uses 10: 205-236 Buttersworth, Boston , MA. Lower eukaryotes, for example, yeasts and Dictyostelium, can be transformed with DC gene sequence containing vectors. For purposes of the invention, the most common lower eukaryotic host is baker's yeast, Saccharomyces cerevisiae. It will be used generically to represent lower eukaryotes although a number of other strains and species are also available. Yeast vectors typically consist of an origin of replication (less of the integrating type), a selection gene, a promoter, DNA encoding the desired protein or its fragments, and sequences for translation termination, polyadenylation, and termination of transcription. Suitable expression vectors for yeast include such constitutive promoters as 3-phosphoglycerate kinase and various other glycolytic enzyme gene promoters or said promoters such as the promoter alcohol dehydrogenase 2 or metallothionine promoter. Suitable vectors include derivatives of the following types: low copy number of auto-replication (such as the YRp file), high copy number of auto-replication (such as YEp series); integrating types (such as Ylp series), or mini-chromosomes (such as the YCp series). Higher eukaryotic tissue culture cells are the preferred host cells for expression of the DC protein. In principle, any higher eukaryotic tissue culture cell line can be used, for example, insect baculovirus expression systems, either from a strong vertebrate or invertebrate one. However, mammalian cells are preferred to achieve proper processing, both during translation and after translation. The transformation or transfection and propagation of said cells is routine. Useful cell lines include HeLa cells, Chinese hamster ovary (CHO) cell lines, rat breeding kidney (BRK) cell lines, insect cell lines, bird cell lines and simian cell lines (COS). Expression vectors for said cell lines usually include an origin of replication, a promoter, a translation initiation site, RNA splice sites (eg, if genomic DNA is used), a polyadenylation site, and a site of transcription termination. These vectors may also comprise a selection gene or amplification gene. Suitable expression vectors can be plasmids, viruses, or retroviruses carrying promoters derived, for example, from such sources such as adenovirus, SV40, parvovirus, vaccinia virus or cytomegalovirus. Representative examples of suitable expression vectors include pCDNAl; pCD, see Okayama, et al. (1985) Mol. Cell Biol. 5: 1136-1142: pMCIneo Poly-A, see Thomas, et al. (1987) Cell 51: 503-512; and a baculovirus vector such as pAC 373 or pAC 610. In certain cases, the DC proteins do not need to be glycosylated to promote biological responses in certain assays. However, it will often be desirable to express a DC polypeptide in a system that provides a defined or specific glycosylation pattern. In this case, the normal pattern will be provided naturally by the expression system. However, the standard may be modified by exposing the polypeptide, for example, in non-glycosylated form, to appropriate glycosylation proteins introduced into a heterologous expression system. For example, a DC gene can be co-transformed with one or more genes encoding mammalian enzymes or other glycosylation enzymes. It is further understood that overglycosylation can be negative for the biological activity of DC protein, and that one skilled in the art can carry out routine tests to optimize the degree of glycosylation that confers optimum biological activity. A DC protein, or a fragment thereof, can be genetically engineered to be linked by phosphatidyl inositol (Pl) to a cell membrane, but can be removed from the membranes by treatment with a phosphatidyl inositol cutting enzyme, for example, phosphatidyl inositol phospholipase-C. This releases the antigen in a biologically active form, and allows purification by standard protein chemistry procedures. See, for example, Low (1989) Biochem. Biophys. Acta 988: 427-454; Tse, et al. (1985) Science 230: 1003-1008; Brunner, et al. (1991) J. Cell Biol. 114: 1275-1283; and Coligan, et al. (eds.) (1996 and periodic supplements) Current Protocols in Protein Science, John Wiley and Sons, New York, NY. Now that these SDCMP proteins have been characterized, fragments or derivatives thereof can be prepared by conventional methods to synthesize peptides. These include methods such as those described in Stewart and Young (1984) Solid Phase Peptide Synthesis Pierce Chemical Co., Rockford, IL; Bodanszky and Bodanszky (1984) The Practice of Peptide Svnthesis Springer-Verlag, New York, NY; and Bodanszky (1984) The Principies of Peptide Svnthesis Springer-Verlag, New York, NY. See also Merrifield (1986) Science 232: 341-347; and Dawson, et al. (1994) Science 266: 776-779. For example, a process with azide, a process with acid chloride, a process with acid anhydride, a process with mixed anhydride, an active ester (for example, p-nitrophenyl ester, n-hydroxysuccinimide ester, or cyanomethyl ester) can be used, a process with carbodiimidazole, an oxidative-reducing process, or a process with dicyclohexylcarbodiimide (DCCD) / additive. The solid phase and solution phase syntheses can be applied to the above procedures. The prepared protein and fragments thereof can be isolated and purified from the reaction mixture by separation of peptides, for example, by extraction, precipitation, electrophoresis and various forms of chromatography, and the like. The DC proteins of this invention can be obtained in various degrees of purity depending on the desired use. Purification can be achieved by the use of known protein purification techniques or by the use of antibodies or binding members described herein, for example, in immunoabsorbent affinity chromatography. This immunoabsorbent affinity chromatography is performed by first binding the antibodies to a solid support and contacting the bound antibodies with used solubilized from appropriate source cells, used from other cells that express the protein, or used or supernatants of cells that produce the proteins as a result of DNA techniques, see below. Multiple cell lines can be selected for one that expresses said protein at a high level compared to other cells.
Several cell lines, for example, a mouse thymic stromal cell line TA4 is selected for its favorable handling properties. The proteins of natural DC cells can be isolated from natural sources, or by expression of a transformed cell using an appropriate expression vector. The purification of the expressed protein is achieved by standard procedures, or it can be combined with genetic manipulation means for effective purification at high efficiency from used or cell supernatants. The FLAG or HIS6 segments can be used for said purification characteristics.
V. Antibodies Antibodies can be generated to different DC proteins, including individual, polymorphic, allelic, strains or variants of species, and fragments thereof, both in their natural forms (total length) and in their recombining forms. Additionally, the antibodies can be cultured in DC proteins in their active forms or in their inactive forms. Anti-idiotypic antibodies can also be used. to. Production of antibodies A number of immunogens can be used to produce antibodies specifically reactive with these DC proteins. The recombinant protein is the preferred immunogen for the production of monoclonal or polyclonal antibodies. The naturally occurring protein can also be used in its pure or impure form. Synthetic peptides made using the human DC protein sequences described herein may also be used as an immunogen for the production of antibodies to the DC protein. The recombinant protein can be expressed in eukaryotic or prokaryotic cells as described herein, and purified as described. The product is injected into an animal capable of producing antibodies. Monoclonal or polyclonal antibodies can be generated for subsequent use in immunoassays to measure the protein. Methods for producing polyclonal antibodies are known to those skilled in the art. In brief, an immunogen, preferably a purified protein, is mixed with an adjuvant and the animals are immunized with the mixture. The immune response of the animal to the immunogenic preparation is monitored by taking test bleeds and determining the titre of reactivity to the DC protein of interest. When appropriate high titers of antibodies are obtained to the immunogen, the animal's blood is collected and antisera are prepared. If desired, additional antiserum fractionation can be performed to enrich the protein reactive antibodies. See, for example, Harlow and Lane. Monoclonal antibodies can be obtained by various techniques known to those skilled in the art. Briefly, the spleen cells of an animal immunized with a desired antigen are immortalized, commonly by fusion with a gelloid cell. See, for example, Kohler and Milstein (1976) Eur. J. Immunol. 6: 511-519, which is incorporated herein by reference. Alternative methods of immortalization cf include transformation with Epstein Barr Virus, oncogenes or retroviruses, or other methods known in the art. Colonies arising from immortalized single cells are selected for production of antibodies of the desired specificity and affinity for the antigen, and the yield of the monoclonal antibodies produced by said cells can be improved by various techniques, including injection into the peritoneal cavity of a host. vertebrate. Alternatively, DNA sequences encoding a monoclonal antibody or a binding fragment thereof can be isolated by selecting a human B cell DNA library according to the general protocol mentioned by Huse, et al. (1989) Science 246: 1275-1281. The antibodies, including binding fragments and individual chain versions, against predetermined fragments of these DC proteins can be increased by immunization of animals with conjugates of the fragments with carrier proteins as described above. The monoclonal antibodies are prepared from cells that secrete the desired antibody. These antibodies can be selected for binding to normal or defective DC proteins, or be selected for agonistic or antagonistic activity. These monoclonal antibodies will normally bind with at least one KD of about 1 mM, frequently at least 300 μM, typically at least about 10 μM, preferably at least about 30 μM, preferably at least 10 μM , and still most preferably at least about 3 μM or more. In some cases, it is convenient to prepare monoclonal antibodies from several mammalian hosts, such as mice, rodents, primates, humans, etc. The description of techniques for preparing said monoclonal antibodies can be found in, for example, Stites, et al. (eds.) Basic and Clinical Immunology (4th ed.) Lange Medical Publications, Los Altos, CA, and references cited therein; Harlow and Lane (1988) Antibodies: A Laboratorv Manual CSH Press; Goding (1986) Monoclonal Antibodies: Principies and Practices (2nd ed.) Academic Press, New York, NY; and particularly in Kohler and Milstein (1975) Nature 256: 495-497, which discusses a method for generating monoclonal antibodies. In summary, this method involves injecting an animal with an immunogen to initiate a humoral immune response. Then the animal is sacrificed and the cells taken from its spleen, which are then fused with myeloid cells. The result is a hybrid cell or "hybridoma" that is capable of reproducing in vitro. The hybridoma population is selected to isolate individual clones, each of which secretes a single antibody species to the immunogen. In this way, the individual antibody species obtained is the product of individual B cells cloned and immortalized from the immune animal generated in response to a specific site recognized in the immunogenic substance. 4 Other suitable techniques involve the selection of antibody libraries in phages or similar vectors. See Huse, et al. (1989) "Generation of a Large Combinatorial Library of the Immunoglobulin Repertoire in Phage Lambda," Science 246: 1275-1281; and Ward, et al. (1989) Nature 341: 544-546. The polypeptides and antibodies of the present invention can be used with or without modifications, including chimeric or humanized antibodies. Frequently, polypeptides and antibodies will be labeled binding, either covalently or non-covalently, to a substance that provides a detectable signal. A wide variety of brands and conjugation techniques are known and reported extensively in both the scientific literature and the patent literature. Suitable labels include radionuclides, enzymes, substrates, cofactors, inhibitors, fluorescent moieties, chemiluminescent portions, magnetic particles and the like. Patents, which teach the use of said trademarks, include U.S. Patent Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241. In addition, recombinant immunoglobulins can be produced. See, Cabilly, Patent of E.U.A. Do not. 4,816,567; and Queen, et al. (1989) Proc. Nat'l Acad. Sci. USA 86: 10029-10033. The antibodies of the invention can be used for affinity chromatography by isolating each DC protein. Columns can be prepared in which the antibodies are bound to a solid support, for example, particles, such as agarose, SEPHADEX, or the like, wherein a used cell can pass through the column, the column is washed, followed by concentrations increased from a mild denaturant, after which the purified DC protein will be released. The antibodies can also be used to select expression libraries for particular expression products. Normally the antibodies used in said procedure will be marked with a portion that allows easy detection of the presence of antigen by antibody binding. Antibodies to SCMP proteins can be used for the analysis or identification of population components of specific cells expressing the respective protein. By evaluating the expression products of DC proteins expressing cells it is possible to diagnose diseases, for example, immune-compromised conditions, depleted DC conditions, or overproduction of DC. The antibodies generated against each DC will also be useful for creating the anti-idiotypic antibodies. These will be useful for detecting or diagnosing various immunological conditions related to the expression of the respective antigens. b. Immunoassays A particular protein can be measured by a variety of immunoassay methods. For a review of immunological and immunoassay procedures in general, see Stites and Terr (eds.) 1991 Basic and Clinical Immunology (7th ed.). In addition, the immunoassays of the present invention can be carried out in any of the various configurations, which are described in detail in Maggio (ed.180) Enzvme Immunoassav CRC Press, Boca Raton, Florida; Tijan (1985) "Practice and Theory of Enzyme Immunoaassays," Laboratorv Techniques in Biochemistry and Molecular Biology, Elsevier Science Publisher B.V., Amsterdam; and Hariow and Lane Antibodies, A Laboratory Manual, supra, each of which is incorporated herein by reference. See also Chan (ed.) (1987) Immunoassay: A Practical Guide Academic Press, Orlando, FL; Price and Newman (eds.) (1991) Principles and Practice of Immunoassavs Stockton Press, NY; and Ngo (ed. 1988) Non-isotopic Immunoassays Plenum Press, NY. Immunoassays for measuring DC proteins can be made by a variety of methods known to those skilled in the art. In brief, the immunoassays for measuring the protein can be competitive or non-competitive binding assays. In competitive binding assays, the sample to be analyzed competes with a labeled analyte for specific binding sites on a capture agent bound to a solid surface. Preferably the capture agent is an antibody specifically reactive with the DC protein produced as described above. The concentration of labeled analyte bound to the capture agent is inversely proportional to the amount of free analyte present in the sample. In a competitive binding immunoassay, the DC protein present in the sample competes with labeled protein for binding to a specific binding agent, for example, an antibody specifically reactive with the DC protein. The binding agent can be attached to a solid surface to effect separation of the bound labeled protein and the unbound labeled protein. Alternatively, the competitive binding assay can be conducted in the liquid phase, and any of a variety of techniques known in the art can be used to separate the bound labeled protein from the unbound labeled protein. After separation, the amount of bound labeled protein is determined. The amount of protein present in the sample is inversely proportional to the amount of labeled protein binding. Alternatively, a homogeneous immunoassay can be carried out where a separation step is not necessary. In these immunoassays, the label on the protein is altered by the binding of the protein to its specific binding agent. This alteration in the marked protein results in an increase or decrease in the signal emitted by the brand, so that the measurement of the mark at the end of the immunoassay allows the detection or quantification of the protein. These DC proteins can also be determined quantitatively by a variety of non-competitive immunoassay methods. For example, a two-site solid phase sandwich immunoassay can be used. In this type of assay, a binding agent for the protein, for example an antibody, is fixed to a solid support. A second protein binding agent, which can also be an antibody, and which binds the protein at a different site, is labeled. After binding on both sides over the protein, the unbound labeled binding agent is removed and the amount of binding of labeled binding agent to the solid phase is measured. The binding amount of the labeled binding agent is directly proportional to the amount of protein in the sample. Western blot analysis can be used to determine the presence of DC proteins in a sample. Electrophoresis is performed, for example, on a tissue sample that is thought to contain the protein. After electrophoresis to separate the proteins, and to transfer the proteins to a suitable solid support such as nitrocellulose filter, the solid support is incubated with an antibody reagent with the denatured protein. This antibody can be labeled, or alternatively detected by subsequent incubation with a labeled second antibody that binds the primary antibody. The immunoassay formats described above employ labeled test components. The brand can be in a variety of ways. The tag may be coupled directly or indirectly to the desired component of the assay according to methods well known in the art. A wide variety of brands can be used. The component can be marked by any of the different methods. Traditionally, a radioactive label incorporating 3H, 125I, 35S, 14c, or 32p is used. Non-radioactive labels include ligands that bind labeled antibodies, fluorophores, chemiluminescent agents, enzymes, and antibodies that can serve as specific binding pair members for a labeled protein. The brand option depends on the required sensitivity, ease of conjugation with the compound, stability requirements, and available instrumentation. For a review of several marking or signal production systems that can be used, see the patent of E.U.A. No. 4,391, 904, which is incorporated herein by reference. Antibodies reactive with a particular protein can also be measured by a variety of immunoassay methods. For reviews of immunological procedures and immunoassays applicable to the measurement of antibodies by immunoassay techniques, see, for example, Stites and Terr (eds.) Basic and Clinical Immunology (7th ed.) Supra.; Maggio (ed.) Enzvme Immunoasav. supra; and Harlow and Lane Antibodies, A Laboratorv Manual, supra. A variety of different immunoassay formats, separation techniques and markings similar to those described above can be used for the measurement of specific proteins.
SAW. Purified SDCMP proteins The nucleotide and amino acid sequences SDCMP3 of primates, eg, humans, are provided in SEQ ID NO: 1 and 2; Rodents, for example, SDCMP3 mouse sequences are provided in SEQ ID NO: 3 and 4.
The SDCMP4 amino acid and nucleotide sequences of primates, eg, humans, are provided in SEQ ID NO: 5, 6, 7 and 8. The peptide sequences allow the preparation of peptides to generate antibodies and recognize said segments and allow the preparation of oligonucleotides encoding said sequences. Standard purification methods are available, and purification can be followed by the use of specific antibodies.
Vile. Physical variants The invention also comprises proteins or peptides having substantial amino acid sequence similarity to an amino acid sequence of a SEQ ID NO: 2, 4, 6 or 8. The variants exhibiting substitutions, for example, 20 or less, preferably 10 or less, and most preferably 5 or fewer substitutions, are also activated. Although the substitutions are conservative substitutions, the variants will share immunogenic or antigenic similarity or cross-reactivity with a corresponding natural sequence protein. Natural variants include individual, allelic, polymorphic, strain or species variants. The amino acid sequence similarity, or sequence identity, is determined by optimizing the residue pairs, if necessary, by entering spaces as required. This changes when conservative substitutions are considered as pairs. Conservative substitutions typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid; asparagine, glutamine; serine, theonin; lysine, arginine; and phenylalanine, tyrosine. The homologous amino acid sequences include interspecific and allelic natural variations in each respective protein sequence. Typical homologous proteins or peptides will have 50-100% similarity (if spaces can be introduced), at 75-100% similarity (if conservative substitutions are included) with the amino acid sequence of the relevant DC protein. The identity measurements will be at least 50%, generally at least 60%, very generally at least 65%, usually at least 70%, often at least 75%, preferably at least 80%, and most preferably at least 80%, and in particularly preferred embodiments, at least 85% or more. See also Needleham, et al. (1970) J. Mol. Biol. 48: 443-453; Sankoff, et al. (1983) Time Warps, String Edits, and Macromolecules: The Theorv and Practice of Sequence Comparison chapter one, Addison-Wesley, Readíng, MA; and NCBI program packages, at the NIH; and University of Wisconsin Genetics Computer Group (GCG), Madison, Wl. The nucleic acids encoding the corresponding mammalian DC proteins are typically hybridized to encode portions of SEQ ID NO: 1, 3, 5, or 7 under stringent conditions. For example, the nucleic acids encoding the respective DC proteins will typically hybridize to the nucleic acid of SEQ ID NO: 1, 3, 5, or 7, under stringent hybridization conditions, for example, providing a background signal of less 2X, preferably 5X, 15X, or 25X, while providing few false positive hybridization signals. In general, stringent conditions are selected at about 10 ° C less than the thermal melting point (Tm) for the sequence that is being hybridized to a defined ionic strength and defined pH. The Tm is the temperature (under defined ionic strength and pH) where 50% of the target sequence is hybridized to a perfectly aligned probe. Typically, the stringent conditions will be those where the salt concentration in the wash is about 0.02 molar at a pH of 7 and the temperature is at least 50 ° C. Other factors can significantly affect the stringency of hybridization, including, among others, the base composition and the size of the complementary chains, the presence of organic solvents such as formamide, and the degree of base misalignment. A preferred embodiment will include nucleic acids that will bind to sequences described in 50% formamide and 20-50 mM NaCl at 42 ° C. An isolated DC gene DNA can be easily modified by nucleotide substitutions, nucleotide deletions, nucleotide insertions, and inversions of nucleotide stretches. These modifications result in novel DNA sequences coding for these DC antigens, their derivatives or proteins having physiologically, immunogenic or highly similar antigenic activity. The modified sequences can be used to produce mutant antigens or increase expression. Increased expression may involve gene amplification, increased transcription, increased translation and other mechanisms. Said DC mutant protein derivatives include predetermined or site-specific mutations of the respective protein or its fragments. "DC mutant protein" comprises a polypeptide that falls within the definition of homology of the DC protein as set forth above, but having an amino acid sequence that differs from the sequence of the DC protein as found in nature , either by deletion, substitution or insertion. In particular, "site-specific mutant DC protein" generally includes proteins that have important similarity to a protein having a sequence, for example, SEQ ID NO: 2. In general, the variant will share many physical-chemical and biological activities, for example, antigenic or immunogenic, with those sequences, and in preferred embodiments will comprise most or all of the sequence described. Similar concepts apply to several DC proteins, particularly those found in various warm-blooded animals, for example primates and mammals. Although site-specific mutation sites are predetermined, mutani need not be specific in themselves. The DC protein mufagenesis can be carried out by making insertions or deletions of amino acids. Substitutions, deletions, insertions or any combination can be generated to arrive at a final construction. Inserts include amino- or carboxyl-terminal fusions. Random mutagenesis can be performed on an objective codon and the expressed mutants can be selected for the desired activity. Methods for making substitution mutations in predefermined sites in DNA having a known sequence are well known in the art, for example, by M13 primer mutagenesis techniques or polymerase chain reaction (PCR). See also, Sambrook, et al. (1989) and Ausubel, et al (1987 and supplements). Mutations in DNA should not normally place the coding sequences outside of the reading frames and preferably will not create complementary regions that can hybridize to produce secondary secondary mRNA sequences as loops or pins. The present invention also provides recombinant proteins, for example, heterologous fusion proteins that use segments from these proteins. A heterologous fusion protein is a fusion of proteins or segments that are natural but not normally fused in the same way. Therefore, the fusion production of an immunoglobulin with a respective DC polypeptide is a coninein-propylane molecule having sequences fused to a typical peptide bond, typically made as an individual translation product and exhibiting properties derived from each peptide. of font. A similar concept applies to heroeropic nucleic acid sequences. In addition, new constructs can be made by combining similar functional domains of other proteins. For exampledomains or other segments can be "exchanged" between different new polypeptides and fusion fragments, typically with proieins related, for example, to the families of lecfine or asialoglycoproteins. Preferably the inky acidic domains will be used, for example, intact Ig portions. See, for example, Cunningham, et al. (1989) Science 243: 1330-1336; and O'Dowd, ef al. (1988) J. Biol. Chem. 263.15985-15992. In this manner, new chimeric polypeptides that exhibit new combinations of specificities will result from the functional binding of proinin-binding specificities and other functional domains. In addition, mulangenesis can be applied by scanning alanine, preferably in residues that structurally exclude secondary growth, which will avoid most of the critical residues that generally interrupt tertiary growth. "Derivatives" of these DC antigens include mutants of amino acid sequences, glycosylation variants, and conjugates covalenides or aggregates with more chemical portions. Covalent derivatives can be prepared by linking functionalities to groups found in these DC protein amino acid side chains or at the N or C termini, by means that are well known in the art. These derivatives may include, without limiting, aliphatic esters or amides of the carboxyl terminus, or residues conferring carboxyl side chains, O-acyl derivatives of residues conferring hydroxyl group, and N-acyl derivatives of residues conining amino groups or amino terminal amino acids, for example, lysine or arginine. The acyl groups are selected from the group of alkyl portions including normal alkyl of C3 to C18, thus forming alkanoyl aroyl species. Covalent binding to carrier proteins is important when the immunogenic portions are haphen. In particular, glycosylation alterations are included, for example, made by modifying the glycosylation pads of a polypeptide during its synthesis and processing, or in additional processing steps. The particularly preferred means to achieve this is the exposure of the polypeptide to glycosylation enzymes derived from cells that normally provide such processing, for example, mammalian glycosylation enzymes. Deglycosylation enzymes are also present. Also included are versions of the same primary amino acid sequence having other lower modifications, including phosphorylated amino acid residues, for example, phosphotyrosine, phosphoserine or phosphofreonine, or other portions, including ribosyl group or crosslinking reagent. In addition, proteins are included comprising substrates, which must retain substantial immunogenicity, to produce antibodies recognizing a protein, for example, of SEQ ID NO: 2. Typically, the proteins will comprise less than 20 residue subscripts of the sequence described, Typically less than 10 subscriptions, preferably less than 5, and most preferably less than Fres. Similarly, proieins that initiate and end in esoteric domains will normally retain anigenicity and cross immunogenicity.
A major group of derivatives are the conjugates covalent of the DC proteins or fragments thereof with more proieins or polypeptides. These derivatives can be synthesized in recombinant culture such as N- or C-terminal fusions or by the use of agents known in the art for their use in crosslinking proteins through secondary reactive groups. Preferred propionic derivative sphrases with cross-linking agents will be found in free amino groups, carbohydrate moieties and cis-feine residues. Fusion polypeptides are also provided in the DC proiephins and other homologous or heirologous proieins. The polypeptide heirologists can be fusions against different surface markers, resulting in, for example, a hybrid proiein. Likewise, heterologous mergers can be constructed, which will present a combination of derived properties or activities of the proleins. Typical examples are fusions of a reporter polypeptide, e.g., luciferase, with a segment or domain of a proiein, e.g., a receptor binding segment, so that the presence or location of the fused protein can be readily determined. See, for example, Dull, ef al., Patent of E.U.A. DO NOT. 4,859,609. Other members of the gene fusion include bacterial ß-galaciosidase, ipep, Proiein A, β-lacmamase, alpha amylase, alcohol dehydrogenase, and yeast alpha mating factor. See, for example, Godowski, et al. (1988) Science 241: 812-816. Said polypeptides will also have amino acid residues that have been chemically modified by phosphorylation, sulphonation, bioillinylation, or the addition or removal of other portions, particularly those that have molecular forms similar to phosphate groups. In some embodiments, the modifications will be useful marker reactants, or serve as purification targets, e.g., affinity ligands. This invention also contemplates the use of derivatives of proiein domains of DC different than variations in amino acid sequence or glycosylation. Said derivatives include covalent or aggregation association with chemical portions. These derivatives are generally caffeinated in fres classes; (1) salts, (2) covalenid modifications of terminal and side chain residues, and (3) adsorption complexes, for example with cell membranes. Said covalenite or aggregation derivatives are useful as immunogens, as reactants in immunoassays or in pure purification methods such as affinity purification of binding ligands or other ligands. For example, a DC prolein protein can be immobilized by covalent attachment to a solid support such as Sepharose acylated by cyanogen bromide, by methods that are known in the art, or adsorbed on polyolefin surfaces, with or without entanglement of glutaraldehyde, for use in the assay or purification of amphibo-DC prophelin antibodies. DC proteins can also be labeled with a deficient group, for example, radioiodinated by the chloramine T procedure, covalently linked to chelates of rare metals, or conjugated to another fluorescent portion for use in diagnostic assays. The purification of these SDCMP profins can be effected by immobilized antibodies. The isolated DC prolein genes allow the transformation of cells lacking the expression of a corresponding DC proiein, for example, whether they are species or cells that lack corresponding proieins and have negative background activity. The expression of transformed genes will allow the isolation of pure antigenic cell lines, with variants of individual or defined species. This approach will allow a more sensitive detection and discrimination of the physiological effects of these DC proteins. Subcellular fragments, for example, cytoplasts or membrane fragments, can be isolated and used.
HIV Binding agent: DC protein complexes A DC protein that specifically binds to or that is specifically immunoreactive with a generated antibody confers a defined immunogen, for example, an immunogen consisting of an amino acid sequence of SEQ ID NO: 2, it is determined in an immunoassay. The immunoassay utilizes a polyclonal antiserum that was generated for the SEQ ID NO: 2 protein. This antiserum is selected because it has low cross reactivity with other members of the related families, and any cross-reactivity is removed by immunosorption prior to use in the immunoassay. To produce amps for use in an immunoassay, for For example, the procedure of SEQ ID NO: 2 is isolated as described herein. For example, a recombinant protein can be produced in a mammalian cell line. A mouse inbred strain such as BALB / c is immunized with the appropriate protein using a standard helper, such as the Freund's helper, and a spherical mouse immunization protocol (see Harlow and Lane, supra). In an alignant manner, a syngenpeptide derived from the sequences described in the present invention and conjugated to a carrier protein can be used as an immunogen. The polyclonal serum is collected and immunized with the immunogenic protein in a immunoassay, for example, a solid phase immunoassay with the immunogen immobilized on a solid support. The polyclonal antiserum with a titer of 104 or greater is selected and evaluated for its cross-reactivity with other related proteins, using a competitive binding immunoassay as described in Harlow and Lane, supra, pages 570-573. Preferably two different related proteins are used in this determination in conjunction with a given DC protein. For example, with the lecphin prophecy, at least two other family members are used to absorb the shared epílopes. In conjunction with the member of the SDCMP3 family, two other members of the family are used. These other members can be produced as recombinant proteins and isolated using standard molecular biology techniques and protein chemistry techniques as described herein. Immunoassays in the competitive binding format can be used for cross-reactivity determinations. For example, the protein of SEQ ID NO. 2 can be immobilized on a solid support. The propheins added to the trial compete with the binding of the amphi- suer to the immobilized anigen. The ability of the above proteins to compete with the binding of the ampicerous to the immobilized protein is compared to the protein of SEQ ID NO: 2. The percentage of cross-reactivity is calculated for the previous proteins, using standardized calculations. Those antisera with less than 10% cross-reactivity with each of the previously disabled proieins are selected and pooled. Cross-reactive antibodies are removed from the pooled enzyme by immunoabsorption with the propheins listed above. The immunoabsorbed and pooled aniiser is used in a competing binding immunoassay as described above for comparing a second protein to the immunogen protein (eg, the SDCMP3 protein of SEQ ID NO: 2). To make this comparison, both proteins are evaluated at a wide scale of concentrations and the amount of each protein required to inhibit 50% of the binding of the ampisuer to the immobilized protein is determined. If the amount of the second required propin is less than twice the amount of the SEQ ID NO: 2 protein that is required, then the second protein is said to specifically bind to an antibody generated for the immunogen. It is understood that DC proteins are a family of homologous proteins comprising two or more genes. For a particular gene product, it is the same as the family member of the Ig family of human, the invention comprises not only the amino acid sequences described herein, but also other proteins that are allelic, polymorphic, non-allelic or species variants. It is also understood that the term "human DC protein" includes unnatural mutations introduced by deliberate mulation using conventional recombinant technology as a mutation in a single site, or by cutting small sections of DNA encoding proiephs or splice variants of the gene, or by suspending or adding small numbers of new amino acids. These minor alterations must sustain the immunoidentity of the original molecule and / or its biological activity. Thus, such alterations include proteins that are specifically immunoreactive with a designated SDCMP protein that occurs naturally, for example, the human SDCMP4 protein that presents SEQ ID NO: 6 or 8. Particular protein modifications considered minor include substitution Amino acid preservative with similar chemical properties, as described above for each protein family as a whole. If a protein is optimally aligned with the protein of SEQ ID NO: 2, and using the conventional immunoassays described herein to determine immunoidentity, the protein compositions of the invention can be determined.
IX. USES The present invention provides reactants that will be used in diagnostic applications as described elsewhere herein, for example, in the general description for developmental abnormalities, or later in the description of diagnostic equipment. DC genes, for example DNA or RNA can be used as a component in a forensic assay. For example, the nucleotide sequences provided can be labeled using, for example, 32P or biotin and used to probe polymorphism graphs of standard resynchronizing fragments, providing a measurable character to assist in dislinging individuals. These probes can be used in well-known forensic techniques such as genetic fingerprinting. In addition, nucleotide probes made from the DC sequences can be used in in situ assays to detect chromosomal abnormalities. Antibodies and other binding agents directed towards DC proteins or nucleic acids can be used to purify the corresponding DC proiein molecule. As described in the following examples, the purification of antibodies from DC proteins is possible and easy to practice. Antibodies and other binding agents can be used in a diagnostic modality to defer whether the DC components are present in a tissue sample or cell population using the well-known techniques described herein. The ability to bind a binding agent to a DC protein provides a means to diagnose disorders associated with poor expression regulation. Antibodies and other DC binding agents are also useful as histological markers, or purification reagents. As described in the following examples, the expression of each of these proteins is limited to specific tissue types. By conducting a probe, such as an antibody or nucleic acid to the respective DC protein, it is possible to use the probe to distinguish tissue and cell type in situ or in vitro. In addition, the purified antigen can be used to deplete an antiserum preparation of these antibodies that bind with antigen selectivity. Thus, for example, mouse SDCMP3 can be used to deplete an anisomer generated for human SDCMP4 from components that can cross-react with mouse SDCMP3. Alternatively, SDCMP3 can be used to purify those components of an antiserum that bind with affinity to the respective antigen. SDCMP4 shares a number of features with hepatic ASGPR, the best known example of type II transmembrane I-type lectins. The hepatic ASGPR presents binding specificity for galactose residues, and its intracellular domain possesses a lirosine mRNA for ligand internment. These features allow liver ASGPR to bind to desialylated plasma glycoprofeins that express galactose residues, and subsequently provide for the removal of those proteins from the plasma.
The ligand specificity of SDCMP4 can not be inferred absolutely from its CRD sequence. However, the expression of SDCMP4 in DC is an indication that potentially amphigenic constituents, as they are found in microorganisms, could represent naphural ligands of SDCMP4. In this context, the mannose receptor, another type C lectin found in DC and macrophages, has the ability to bind and infer, for example, yeast particles following the recognition of the manly portions of its cell wall. The presence of a tyrosine-based hospitalization moiety in SDCMP4 predicts that the molecule plays a role in receptor-mediated endocytosis by DC. It is suggested that the functions of SDCMP4 as a "receptor-antigen" in DC, to intern ligands that will subsequently be directed in an intracellular processing path resulting in the presentation and initiation of antigens or promotion of an immune response. Such inpatient function mediated by SDCMP4 makes this receptor a potential target for directing antigens in DC, for example, to increase the presentation in T cells and subsequent immunization of specific immunity. Therefore, SDCMP4 can represent a receptor for antigen delivery in vaccination protocol, thus obtaining the target of the antigen in the appropriate cells for initiation of a vaccine response. The therapeutic significance of such a strategy may be of particular relevance in cancer immunotherapy, where the antigens associated with tumor (TAA) may be coupled to reagents that specifically recognize SDCMP4 for selective delivery to DC. This invention also provides reactants that may exhibit significant therapeutic value. The DC propheins (as they occur nafurally or recombinan) segments thereof, and antibodies thereto, together with the compounds identified because they have binding affinity to the DC protein, may be useful in the treatment of conditions associated with physiology or abnormal development, including abnormal proliferation, eg, carcinogenic conditions or degenerative conditions. Abnormal proliferation, regeneration, degeneration and atrophy can be modulated by appropriate therapeutic treatment using the compositions provided herein. For example, a disease or disorder associated with abnormal expression or abnormal signaling by a DC, for example, as a cell presenting an antigen, is a target for a pro-agonist or antagonist. Proteins also play a role in the regulation or development of hematopoietic cells, for example, lymphoid cells, that affect the immunological responses, for example, presentation of antigens and the resulting effector functions. It is believed that blockade of the SDCMP interaction can block signaling. In this way, for example, the use of selected polyclonal or monoclonal antibodies confers proteins may affect immune responses, for example, MLR. Alternately, soluble extracellular fragments can block interaction with a counterreceptor, thereby blocking such reaction. Because MLR is the diagnosis of initiation or maintenance of an immune response, these reagents may be useful for modulating the initiation and maintenance of immune responses. Other conditions of abnormal development are known in the type of cells that demonstrate to possess DC protein mRNA by northern blot analysis. See Berkow (ed.) The Merck Manual of Diagnosis and Therapy, Merck and Co., Rahway, NJ; and Thorn, et al. Harrison's Principies of Infernal Medicine, McGraw-Hill, NY. Developmental or functional abnormalities, for example, of the immune system, cause important medical abnormalities and decisions that may be susceptible to prevention or treatment by using compositions provided herein. Recombinant DC antibodies or antibodies can be purified and then administered to a patient. These reagents may be combined for therapeutic use with additional active or inert ingredients, for example, in vehicles or conventional pharmaceutically acceptable diluents, for example, immunogenic adjuvants, together with physiologically harmless stabilizers and excipients. In particular, these may be useful in a vaccine context, where the antigen is combined with one of these therapeutic versions of agonists or antagonists. These combinations can be filtered for sterility and placed in dosage form as by lyophilization in dose or storage containers in stabilized aqueous preparations. This invention also contemplates the use of antibodies or binding fragments thereof, including forms that are not binding complements. The selection of drugs using antibodies or receptors or fragments thereof can identify compounds that have binding affinity to those DC proteins, including the isolation of associated components. Subsequent biological assays can be used to determine whether the compound has intrinsic stimulating activity and is therefore a blocker or antagonist since it blocks the activity of the protein. Likewise, a compound that has an unmistakeable anti-viral activity can activate the cell through the protein and is therefore an agonist since it simulates the cell. This invention also contemplates the therapeutic use of antibodies to the prophets as an anonymity. The amounts of reagents necessary to effect therapy will depend on different factors, including means of administration, target site, physiological condition of the patient, and other medications administered. Therefore, doses of radiation should be titrated to optimize safety and efficacy. Typically, the doses used in vitro may provide useful guidance in amounts useful for in situ administration of these reagents. The evaluation in animals of effective doses for the treatment of particular ostomas will provide an indication of additional dose prediction in humans. Several considerations are described, for example, in Gilman, et al. (eds.) (1990) Goodman and Gilman's: The Pharmacological Bases of Therapeutics (8th ed.) Pergamon Press; and (1990) Remington's Pharmaceutical Sciences (17th ed.) Mack Publishing Co., Easton, PA. Methods for administration are discussed therein and subsequently herein, for example for oral, iniravenous, intraperitoneal or inframuscular administration, transdermal and oral diffusion. Pharmaceutically acceptable carriers include water, saline, regulators and other compounds described in, for example, Merck Index, Merck and Co., Rahway, NJ. The expected dose scales are presented in smaller quantities of concentrations of 1 mM, typically less than around concentrations of 10 μM, usually less than about 100 nM, preferably less than about 10 pM (picomolar), and still preferably less than about 1 fM (femtomolar), with an appropriate vehicle. Slow release formulations or a slow release device will often be used for continuous administration. The DC proteins, fragments of the same and antibodies for them or their fragments, anyagonisías and agonisfas, can be administered directly to the host that will be irritated, depending on the size of the compounds, it would be convenient to conjugate them in carriers carriers asal ovalbumin or serum albumin before his admiñisíración. Therapeutic formulations can be administered in many conventional dose formulations. Although it is possible that the active ingredients are administered alone, it is preferable to present them as a pharmaceutical formulation. The formulations typically comprise at least one active ingredient, as defined above, June with one or more pharmaceutically acceptable vehicles. Each vehicle will be pharmaceutically as well as physiologically acceptable in the sense of being compatible with other ingredients and not causing harm to the patient. The formulations include those suitable for oral, rectal, nasal or parenteral administration (including subcutaneous, intramuscular, infravenose and intradermal). The formulations can conveniently be presented in unit dosage form and can be prepared by methods known in the pharmaceutical art. See, for example, Gilman, et al. (eds.) (1990) Goodman and Gilman's: The Pharmacological Bases of Therapeutics (8th ed.) Pergamon Press; and (1990) Remington's Pharmaceutical Sciences (17th ed.) Mack Publishing Co., Easton, PA; Avis, et al. (eds.) (1993) Pharmaceutical Dosage Forms: Parenteral Medícations Dekker, NY; Lieberman, et al. (eds) (1990) Pharmaceuíical Dosage Forms: Tablets Dekker, NY, and Lieberman, ei al. (eds) (1990) Pharmaceuíical Dosage Forms: Disperse Svsfems Dekker, NY. The therapy of this invention can be combined with or used in association with other chemotherapeutic or chemopreventive agents. Both the natural form and the recombinant form of the DC proteins of this invention are particularly useful in equipment and test methods, which are capable of selecting compounds for protein binding activity. In recent years, several methods have been developed to test tests in order to allow the selection of tens of thousands of compounds in a short period. See, for example, Fodor, ei al. (1991) Science 251: 767-773, and other descriptions of chemical diversity libraries which describe means for testing binding affinity across a plurality of compounds. The development of suitable tests can be greatly facilitated by the availability of large amounts of purified proteins, for example, soluble versions of DC proteins, as provided by this invention. For example, antagonisms can often be found once the pro fi le is defined roughly. The testing of potential proiein analogs is now possible with the development of highly automated test methods that use a purified surface protein. In particular, new agonies and antagonists will be discovered by using selection techniques described herein. Of particular importance are compounds that have a combined binding affinity for related cell-surface multiply antigens, e.g., compounds that can serve as antagonists for species variants of a DC protein. This invention is particularly useful for screening compounds by the use of recombinant DC protein in a variety of drug selection techniques. The advantages of using a recombinant propine in the selection of specific ligands include: (a) an improved renewable source of the protein from a specific source; (b) a potentially larger number of amphigens per cell that gives a better signal to the noise ratio in tests; and (c) specificity of species variants (which theoretically gives a greater biological and disease specificity).
A method of drug selection utilizes eukaryotic or prokaryotic host cells, which are stably transformed with recombinant DNA molecules that express a DC protein. The cells can be isolated, which expresses that prophein in isolation from any other. Such cells, either in a viable or fixed form, can be used for standard surface protein binding assays. See also, Parce, et al. (1989) Science 246: 243-247; and Owicki, et al. (1990) Proc. Naí'l Acad. Sci. USA 87: 4007-4011, which describe sensitive methods for detecting cellular responses. Competitive tests are particularly useful when the cells (DC protein source) are in contact and are incubated with an antibody which has known binding affinity to the antigen, such as 125 I antibody, and a test sample whose binding affinity to the bonding composition is being measured. The bound and free labeled binding compositions are then separated to assess the degree of protein binding. The amount of bound test compound is inversely proportional to the amount of labeled antibody that binds to the known source. Many techniques can be used to separate the bound reactant from the free reactant to assess the degree of binding. This separation step would normally involve a standard procedure such as adhesion to filters followed by washing, adhesion to plasmics followed by washing or sterilization of the cell membranes. Viable cells can also be used to analyze the effects of drugs on these DC protein-mediated functions, eg, presentation of ananine or helper function. Another method uses the membranes of transformed eukaryotic or prokaryonic host cells as the source of a DC protein. These cells are stably transformed with DNA vectors that direct the expression of the appropriate protein, for example, a genetically engineered membrane bound form. Essentially, the membranes would be prepared from the cells and used in binding tests, such as the competitive test discussed above. Another method is to use dissolved, unpurified or solubilized DC protein, purified from host cells transformed eukaryotic or prokaryotic. This allows a "molecular" binding test with the advantages of increased specificity, the ability to automate and improve drug test performance. Another technique for analyzing drug involves a method which provides high-throughput screening for compounds that have adequate binding affinity to the respective DC protein and is described in detail in Geysen, European Patent Application 84/03564, published on September 13 of 1984. First, large numbers of different small peptide test compounds are synthesized on a solid substrate, for example, plasmid pins or some other suitable surface, see Fodor, et al, supra. Then, all the pins are reacted with solubilized, purified, purified or solubilized DC protein, and washed. The next step involves the defection of bound reagent, e.g., antibody.
A means of determining which sites interact with more specific proieins is a determination of physical structure, for example, x-ray crystallography or two-dimensional NMR technique. These techniques will provide a guideline as to which amino acid residues form molecular contact regions. For a detailed description of structural product de fi ning, see for example, Blundell and Johnson (1976) Proinin Crvsialloqraphv Academic Press, NY.
X. Equipment This invention also contemplates the use of prole DC, fragments thereof, peptides and their fusion products in a variety of equipment and diagnostic methods to detect the presence of a DC protein or message. Typically, the kit will have a compartment containing either a defined DC peptide or gene segment or a reagent which recognizes one or the other, for example, antibodies. A kit for determining the binding affinity of a test compound to the respective DC protein would normally consist of a test compound.; a labeled compound, for example an antibody having known binding affinity to the protein; a source of the DC protein (natural or recombinanfe); and a means for removing the bound labeled compound from the free compound, such as a solid phase to immobilize the DC profinine. Once the compounds are selected, those that have an adequate binding affinity to the prophecy in 8 can be evaluated. suitable biological tests, as are known in the art, to determine whether they acted as agonists or antagonists to regulate DC fusion. The availability of recombinant DC polypepides also provides well-defined standards for calibrating such tests. A preferred equipment for determining the concentration of, for example, a DC profine in a sample would normally consist of a labeled compound, eg, anannibody, which has known binding affinity to the DC protein, a source of DC protein ( natural or recombinant) and a means for removing the bound labeled compound from the free compound, for example, a solid phase to immobilize the DC protein. Typically, compartments containing reagents and instructions will be provided. The antibodies, which include antigen binding fragments, specific for the respective DC or its fragments, are useful in diagnostic applications to detect the presence of high levels of the protein and / or its fragments. Such diagnostic tests may employ used, living cells, fixed cells, immunofluorescence, cell cultures, body fluids and may further involve the detection of antigens in serum or the like. Diagnostic tests can be homogeneous (without a separation stage between the free reactive and the DC antigen / proiein complex) or heterogeneous (with a separation stage). There are different commercial tests, such as radioimmunoassay (RIA), enzyme-linked immunosorbent assay (ELISA), enzyme immunoassay (EIA), multiply enzyme enzyme immunoassay (EMIT), fluorescent immunoassay of their labeled surfactant (SLFIA) and the like . For example, unlabeled antibodies can be employed by using a second antibody which is labeled and which recognizes the antibody to the DC protein or a particular fragment thereof. Similar tests have been widely disseminated in the literature. See, for example, Harlow and Lane (1988) An ibodíes: A Laboratorv Manual, CSH Press, NY; Chan (ed. 1987) Immunoassay: A Practical Guide Academic Press, Orlando, FL; Prece and Newman (eds. 1991) Principies and Praciice of Immunoassav Síockfon Press, NY; and Ngo (ed. 1988) Nonisoíopic Immunoassav Plenum Press, NY. In particular, reagents can be useful to diagnose DC populations in biological samples, either to detect an excess or deficiency of DC in a sample. The test can be directed to histological analysis of a biopsy, or evaluation of DC numbers in a blood or tissue sample. Amphiidiological antibodies may have similar use to diagnose the presence of antibodies against a DC protein, such as the diagnosis of different abnormal states. For example, overproduction of the DC protein can result in different immunological reactions that can be diagnostic of abnormal physiological states, particularly in prolific cell conditions, such as cancer or abnormal differentiation. Frequently, the reagents for diagnostic tests are supplied in teams, in order to optimize the sensitivity of the test. For the subject invention, depending on the nature of the test, the protocol and the label are provided, whether a labeled or unmarked antibody or receptor, or labeled DC prolein. Normally, this is binding with other additives, such as pH regulators, stabilizers, materials necessary for signal production such as enzymes for enzymes and the like. Preferably, the equipment will also contain instructions for proper use and disposal of the content after use. Normally, the equipment has compartments for each useful reagent. Conveniently, the reagents are provided as a dry lyophilized powder, wherein the reagents can be reconsti- tuted in an aqueous medium that provides adequate concentrations of reagents to perform the test. Many of the aforementioned concerns of drug selection and diagnostic testing can be used without modification or can be modified in a variety of ways. For example, the marking can be achieved by the covalenle or non-covalent union of a portion which directly or indirectly provides a detectable signal. In many of these tests, the protein, the test compound, the DC protein, or the antibodies thereof can be labeled either directly or indirectly. The possibilities of direct labeling include labeling groups: radiolabels such as 125 |, enzymes (US Patent No. 3,645,090) such as peroxidase and alkaline phosphatase and fluorescent labels (US Patent No. 3,940.47) capable of monitoring the change in fluorescence intensity, wavelength shift, or fluorescence polarization. The possibilities of indirect labeling include the biotinylation of a constituent followed by avidin binding coupled to one of the aforementioned labeling groups. There are also numerous methods for separating the bound protein from the free protein, or alternatively the bound test compound from free test compound. The DC protein can be immobilized in different matrices followed by washing. Suitable matrices include plasmid, such as an ELISA plate, filaments and beads. Methods for immobilizing the DC protein to a matrix include, without limitation, direct adhesion to plastic, use of a capture antibody, chemical coupling and biotin-avidin. The last step in this method involves the precipitation of proiein / antibody complex by one of different methods including those that use, for example, an organic solvent such as polyethylene glycol or a salt, such as ammonium sulfate. Other suitable separation techniques include, without limitation, the method of magnetizable particles by fluorescein antibody described in Ralíle, et al., (1984) Clin. Chem, 30: 1457-1461, and the double separation of magnetic particles by antibody, as described in the patent of E.U.A. No. 4,659,678. Methods for linking proteins or their fragments to different brands have been widely reported in the literature and do not require detailed discussion in the present. Many of the techniques involve the use of activated carboxyl groups, either through the use of carbodiimide or active esters to form peptide bonds, the formation of thioethers by the reaction of a mercapto group with an activated halogen such as chloroacetyl, or an olefin activated such as maleimide, for binding, or the like. The fusion proteins will also find use in these applications. Another diagnostic aspect of this invention involves the use of oligonucleotide or polynucleotide sequences taken from the sequence of a specific DC protein. These sequences can be used as probes to detect message levels in samples from patients presumed to have an abnormal condition, for example, cancer or an immune problem. The preparation of nucleophil sequences of RNA and DNA, the labeling of the sequences and the preferred size of the sequences has received a wide description and discussion in the literature. Normally, an oligonucleotide probe must be at least about 14 nucleotides, normally at least about 18 nucleotides, and the polynucleotide probes can be greater than several kilobases. Several labels can be used, most commonly radionuclides, particularly 32p. However, other techniques may also be employed, such as the use of biotin-modified nucleotides for introduction into a polynucleotide. Biotin then serves as the site for binding to avidin or antibodies, which can be labeled with a wide variety of labels, such as radionuclides, fluorophores, enzymes, or the like. Alternatively, antibodies can be used which can recognize specific duplexes, including DNA duplexes, RNA duplexes, hybrid DNA-RNA duplexes or DNA-protein duplexes. The antibodies in turn, they can be marked and the test can be carried out when the duplex is bound to a surface, so that in the formation of the duplex on the surface, the presence of antibody bound to the duplex can be detected. The use of probes for the novel antisense RNA can be carried out in any conventional technique, such as hybridization. of nucleic acid, selection plus and minus, treatment with recombinational probes, reanalyzed translation by hybridization (HRT), and interrupted translation by hybridization (HART). This also includes amplification techniques, such as polymerase chain reaction (PCR). Diagnostic equipment is also included, which also tests the qualitative or quantitative presence of other markers. The diagnosis or prognosis may depend on the combination of multiple indicators used as markers. In this way, teams can try the combinations of markers. See, for example, Viallet, et al. (1989) Proqress in Growth Factor Res. 1: 89-97.
XI. Isolation of the binding member When having an element of a binding member of a specific interaction isolated, there are methods for isolating the member. See, Gearing, et al. (1989) EMBO J. 8: 3667-3676. For example, means for labeling a DC surface protein without interfering with binding to its receptor can be determined. For example, an affinity tag can be fused to either the amino or carboxyl terminus of the ligand. An expression library can be selected for specific binding to the DC protein, for example, by cell sorting, or other screening to detect subpopulations that express such a binding partner. See, for example, Ho, et al., (1993) Proc. Nat'1 Acad. Scí. USA 90: 11267-11271. In an alimative way, a panning method can be used. See, for example, Seed and Aruffo (1987) Proc. Nat'1 Acad. Sci. USA. 84: 3365-3369. A two-hybrid selection system can also be applied by making suitable constructions with the available DC protein sequences. See for example, Fields and Song (1989) Nature 340: 245-246. Propylene-branched enylacement techniques can also be applied to isolate binding members of a DC protein. This would allow the idenification of proteins which interact specifically with the appropriate DC protein. The broad scope of this invention is best understood with reference to the following examples, which are not intended to limit the invention to specific embodiments.
EXAMPLES I. General Methods Many of the standard methods below are described or referred to, for example, in Maniatis, et al., (1982) Molecular Cloning, A Laboraorv Manual Cold Spring Harbor Laboralory, Cold Spring Harbor Press, NY; Sambrook, et al. (1989) Molecular Cloning: A Laboratorv Manual (2nd ed.) Vols. 1-3, CSH Press, NY; Ausubel, et al., Biology Greene Publishing Associates, Brooklyn, NY; o Ausubel, et al. (1987 and supplements) Currení Proíocols in Molecular Biology Wiley / Greene, NY; Innis, e to al. (eds.) (1990) PCR Proíocols: A Guide ío Meihods and Applications Academic Press, NY. Methods for protein purification include methods such as ammonium sulfate precipitation, column chromatography, electrophoresis, centrifugation, crystallization and others. See for example, Ausubel et al. (1987 and periodic supplements); Deuischer (19909"Guide to Protein Purification," Methods in Enzymology vol.182, and other volumes in this series; Coligan, et al. (1996 and periodic supplements) Curreni Protocols in Protein Science Wiley / Greene, NY; and manufacturer's guide on the use of protein purification products, for example, Pharmacia, Piscataway, NJ or Bio-Rad, Richmond, CA. Combination with recombinant techniques allows fusion to suitable segments, for example, to a FLAG sequence or an equivalent which can be fused through a removable protease sequence See, for example, Hochuli (1989) Chemische Industrie 12: 69-70; Hochuli (1990) "Purification of Recombinant Proteins Wiih Metal Chelaie Absorbent" in Setlow (ed.) Geneííc Engineering, Principie and Meíhods 12: 87-98, Plenum Press, NY and Crowe, et al. (1992) OlAexpress: The High Level Expression and Proiein Purificaction System QUIAGEN, Inc., Chatsworth, CA. Methods for determining function Nm unological are described, for example in Hertzenberg, et al. (eds 19969 Weir's Handbook of Experimental Immunology vols 1-4, Blackwell Science; Coligan et al. (1992 and periodic supplements) Current Protocols in Immunology Wiley / Greene, NY; and Meyhods in Enzymology volumes 70, 73, 74, 84 , 92, 93, 108, 116, 121, 132, 150, 162, and 163. See also for example, Paul (ed.) (1993) Fundamental Immunology (3rd ed.) Raven Press, NY. The particularly useful functions of Dendritic cells are described for example, in Steinman (1991) Annual Review of Immunology 9: 271-296, and Banchereau and Schmitt (eds. 1994) Dentritic Cells in Fundamental and Clinical Immunology Plenum Press, NY. FACS analyzes are described in Melamed , et al. (1990) Flow Cvtometrv and Storing Wiley-Liss, Inc., New York, NY; Shapiro (1998) Practical Flow Cytomtrv Liss, New York, NY; and Robinson, et al. (1993) Handbook of Flow Cvtometrv Methods Wiley-Liss, New York, NY.
II. Generation of dendritic cells Human CD34 + cells were obtained in the following manner. See, for example, Caux et al. (1995) pages 1-5 in Benchereau and Schmitt Dentriíic Cells in Fundamental and Clinical Immunology Plenum Press, NY. Peripheral or umbilical blood cells, sometimes selected CD34 +, were cultured in the presence of Stem Cell Factor (SCF), GM-CSF; and FNT-a in an endotoxin-free RPMI 1640 medium (GIBCO, Grand Island, NY) supplemented with 10% (v / v) inactivated bovine fetal serum (FBS, Flow Laboratories, Irvine, CA), HEPES a 10mM, L-glutamine at 2mM, 2-mercaptoethanol at 5 X 10"5 M, penicillin (100 μg / ml) This is referred to as a complete medium CD34 + cells were seeded for expansion in flasks of 25 to 75 cm2 ( Corning, NY) at 2 X 104 cells / ml Optimal conditions were maintained by dividing these cultures on day 5 and 10 with a medium containing GM-CSF and fresh TNF-a (cell concentration: 1-3 x 105 In certain cases, the cells were classified by FACS for CD1a expression at approximately day 6. In certain situations, the cells were routinely collected after 12 days of culture, even if the adherenid cells were recovered using a solution of EDTA at 5 mM In other situations, CD1a + cells were activated by resuspensi n in complete medium at 5 x 10 6 cells / ml and activated for the appropriate time (for example, 1 or 6 hours) with 1 μg / ml of 12-mristal-13-phorbol acelaide (PMA, Sigma) and 100 ng / ml of onomycin (Calbiochem, La Jolla, CA). These cells expanded for another 6 days and the RNA was isolated for DNA library preparation.
III. RNA Isolation and Gene Library Construction Total RNA is isolated using, for example, the guanidine thiocyanalum / Cscl gradient procedure, as described by Chirgwin, et al. (1978) Biochem. 18.5294-5299. In a positive way, poly (A) + RNA is isolated using the OLIGOTEX mRNA isolation kit (QIAGEN). The double-stranded cDNA is generated using, for example, the SUPERSCRIPT plasmid system (Gibco BRL, Gaifhersburg, MD) for cDNA synthesis and plasmid cloning. The resulting double-stranded cDNA is cloned unidirectionally, for example, in pSportl and transfected by eleclroporation in ELECTROMAX DH10B ™ cells (Gibco BRL, Gaiíhersburg, MD).
IV. Sequencing DNA isolated from clones picked up randomly, or after subtractive hybridization using inactive cells, was subjected to nucleotide sequence analysis using standard techniques. A Taq Dideoxy Terminator cycle sequencing computer (Applied Biosystems, Fosíer Cify, CA) can be used. The labeled DNA fragments are separated using a DNA sequencing gel from a suitable automated sequencer. Alternatively, the isolated clone is sequenced as described for example, in Maniatis, et al. (1982) Molecular Cloninq. A Laboratorv Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor Press; Sambrook, et al. (1989) Molecular Cloninq: A Laboratorv Manual. (2nd ed.), Vols. 1-3 CSH Press, NY; Ausubel, et al., Biology, Greene Publishing Associates, Brookiyn, NY; o Ausubel, et al. (1987 and supplements) Current Protocols in Molecular Biology, Greene / wiley, New York. Chemical sequencing methods are also available, for example, using Maxam and Gilberf sequencing techniques.
V. Recombinant DC Gene Construction Poly (A) + RNA is isolated from suitable cell populations, for example, using the FastTrack mRNA kit (Invitrogen, San Diego, CA). The samples are subjected to electrophoresis, for example, on 1% agarose gel containing formaldehyde and transferred to a GeneScreen membrane (NEN Research Products, Boston, MA). Hybridization is performed, for example, at 65 ° C in 0.5 M NAHPO4 pH 7.2, 7% SDS, 1 mM EDTA, and 1% BSA (fraction V) with DC gene cDNA labeled 32p-dCTP at 107 cpm / ml. After hybridization, the filters are washed three times at 50 ° C in 0.2x SSc, 0.1% SDS, for example, for 30 minutes and exposed to film for 24 hours. Normally, a posiive signal will be 2X on background, preferably 5-25X. The construction of recombinant genes can be used to generate a probe to detect the message. The insert can be cut and used in the detection methods described above. Different standard methods for washing and cross-species hybridization are known in the art. See, for example, Sambrook, et al. and Ausubel.
SAW. Proiein expression of DC gene in E. coli PCR is used to make a construct comprising the open reading frame, preferably in operable association with the appropriate promoter, selection and regulatory sequences. The resulting reaction plasmid is transformed into a suitable E. coli strain, for example, Topp5, (Straiagene, La Jolla, CA). Ampicillin-resistant transformants (50 μg / ml) are grown in Luria Broth (Gibco) at 37 ° C until the optical density at 550 nm is 0.7. The recombinant protein is induced with isopropyl-β-D-thiogalacofopyranoside at 0.4 mm (Sigma, St. Louis, MO) and incubation of the cells continues at 20 ° C for an additional 18 hours. One-liter culture cells are harvested by centrifugation and resuspended, for example, in 200 ml of 30% ice-cold sucrose, 50 mM Tris, HCL pH 8.0, 1 mM ethylenediamine-tetraacetic acid. After 10 minutes on ice, water cooled on ice is added to a total volume of 2 liters. After 20 minutes on ice, the cells are removed by centrifugation and the supernatant is clarified by filtration through a 60 to 5 μM Millipack (Millipore Corp., Bedford, MA). The recombinant protein is purified through different purification methods, for example, different methods of ion exchange chromatography. Immunoaffinity methods using antibodies described below can also be used. Affinity methods can be used when an epitope tag is genetically manipulated in an expression construct. Similar methods are used to prepare cells and expression constructs in eukaryotic cells. Eukaryotic promoters and expression vectors can be produced, as described above.
VIL Mapping of human DC genes Isolation of DNA, digestion of resynchronizing enzymes, agarose gel electroshock, Southern blot blotting and hybridization are performed according to the standard techniques. See Jenkins, et al. (1982) JL Vitrol. 43: 26-36. The blocks can be prepared with Hybond-N nylon membrane (Amersham). The probe is labeled with dCTP 32p; washing is performed at a final asynchrony, for example, 0.1X SSC, 0.1% SCS, 65 ° C. Alternatively, a hybrid panel of mouse somatic cell BIOS Laboratories (New Haven, CT) can be combined with PCR methods. See Fan, et al. (1996) Immunogenetics 44: 97-103. The human SDCMP3 gene is located on chromosome 12 p12-13 (human NK receptor complex), as determined by hybrid radiation mapping with PCR primers.
VIII. Analysis of individual variation From the distribution data, a type of easily accessible abundant cell is selected to sample individuals. When using PCR techniques, a large population of individuals is analyzed for this gene. CDNA or other PCR methods are used to sequence the corresponding gene in different individuals, for example, exogamous mouse races and their sequences are compared. This indicates the point of divergence between racial populations or other populations, as well as the determination of which residues are likely to be modifiable without dramatic effects in function.
IX. Preparation of antibodies The recombinant DC proteins are generated by expression in E. coli as shown above and are tested for biological activity. Alternatively, natural protein sources can be used with available purification methods. The antibody reagents can be used in immunopurification or to screen separation methods. The active or denatured proteins can be used for immunization of suitable mammals, either for production of polyclonal serum or for the production of monoclonal antibodies.
X. Isolation of DC gene counterparts Human cDNA clones encoding these genes are used as probes or to designate PCR primers, to find counterparts in several primate species, eg, chimpanzees. Others can be identified from other animals, for example, domesticated farm species or pets.
XI. Use of reagents to analyze cell populations Defeiting the level of dendritic cells present in a sample is important in diagnosing abnormal disease conditions. For example, an increase in the number of dendritic cells in a tissue or in the lymphatic system may indicate the presence of DC hyperplasia, or rejection of tissue or graft. A low DC population may indicate an abnormal reaction to a bacterial or viral infection, which may require appropriate treatment to normalize the DC response. FACS analysis using a specific labeled binding agent for a cell surface DC protein, see, for example, Melamed et al. (1990) Flow Cvtometrv and Sorting Wiley-Liss, Inc., New York, NY; Shapiro (1988) Practical Flow Cvtometrv Liss, New York, NY; and Robinson, I went to. (1993) Handbook of Flow Cvtomeírv Meíhods Wiley-Líss, New York, NY, is used to determine the number of DCs present in a cell mixture, eg, PBMC, adherenid cells, eic. The binding agent is also used for histological analysis of tissue samples, either fresh or fixed, to analyze the infiltration of DC. Various cell populations can also be evaluated, either in a destructive cell test, or in certain tests where the cells retain viability. Alternatively, cell or tissue fixation methods can be used. The levels of DC transcripts are quantified, for example, using semi-quantitative PCR as described in Murphy, et al. (1993) jL Immunol. Meihods 162: 211-223. The primers or other methods are designed so that the genomic DNA is not detected.
XII. Preparation of immunoselective binding preparations The polyclonal anisomer is prepared, for example, as described above. The other asialoglucoprolein receptors are used to deplete components which bind specifically to them, leaving components which will bind to the desired SDCMP3 or SDCMP4. Such depleted sera can be attached to a solid substrate, for example, and can be used to immunoselect the animal from an impure source. The immunoselected antigen can be subjected to further purification by standard protein purification procedures, for example, ammonium sulfate precipitation, ion exchange or other methods of chromatography, HPLC, etc. The specific serum can be used to follow the purification, for example, by determining which fractions divide the desired protein.
XIII. Dissection distribution Two transcripts for human SDCMP3 have been detected by PCR analysis. The shortest form corresponds to a form in which a deletion corresponds to nucleotides 376-513 (269-406 of ORF), which retains the open reading frame. The cellular dissipation of the two forms seems to be similar. The distribution of primate SDCMP3 is detected in DC prepared from CD34 + progenitors cultured 12 days in GM-CSF and FNTa, activated 1-6 hours with PMA, ionomycin; TF1 (early myeloid cell line); and U937 (melanomonocytic cell line) activated with PMA and ionomycin. No signal was detected in non-activated cell lines Jurkat, CHA, MRD5, and JY. The subgroup evaluation of DC: the CD34 + progenitors were cultured 6 days with GM-CSF and FNTa, and classified by FACS in populations CD1a + and CD14 +. The classified subgroups were cultured 6 more days in GM-CSF and TNFa and activated with PMA and ionomycin for 1 hour or 6 hours. Expression was detected in DC derived from CD14, but not in DC derived from CD1a, and the expression was sub-regulated by activation by Pl. A much lower signal was detected in monocytes activated with PMA and ionomycin; and very weak signals were detected in PBL, both non-activated and activated by PMA, ionomycin. No signal was detected in several cells activated with PMA, onomycin: T cells, granulocytes, or B cells.
DC analysis: CD34 + progenitors cultured 12 in GM-CSF and FNTa, not activated or activated with PMA, ionomyin either for 1 or 6 hours. Signals were detected in the subgroups activated by CD14, but not in subgroups activated by CD1a. However, the expression was sub-regulated by activation by PMA, ionomycin. Macrophages were evaluated and signals were detected in DC (sub-regulated by activation with PMA, ionomycin); monocytes activated with PMA, ionomycin; and PBL (not activated or activated with PMA, ionomycin). The expression of SDCMP3 was not detected by RT-PCR in the following cell types: Langerhans cells, DC11c + or CD11c negative of peripheral blood and amygdala (with or without activation by PMA and ionomycin, or IL3 and anti-CD40) , B cells (with or without activation by PMA and ionomycin, or mBA aníi-CD40), T cells (with or without activation by PMA and ionomycin, or mABs aníi-CD3 and anti-CD28). By expressing sequences in cDNA sequence databases, the sequence has been detected in DC gene libraries; Activated monocytes and testosterone immunity. The murine homologue (1469D4) of SDCMP3 includes a mafia recognition motif (EPN) in its CRD. In addition, the mouse lectin has the consensus WND sequence characteristic of proteins that bind to sugars. Consequently, it can be expected that 1469D4 will have the ability to join crafty. Since the cell walls of microorganisms are rich in mannose, it is possible that the cells that present anígen (DC) can use the lecfine to trap and subsequently degrade microbial antigens through ex-cellular cell activity. By analogy to other type C lectins which exist in closely related forms, it can be predicted that a mannose binding form of SDCMP3 will be identified from human cells. Said binding activity in dendritic cells would represent an objective to upregulate the potential benefit in the treatment of infectious diseases. Another possible function of SDCMP3 could serve as an adhesion molecule between DC and other cell types that express a ligand, e.g., T cells, thus modulating the immune response. The sequence homology and chromosomal location of SDCMP3 strongly suggest that it is an element of a novel type C lectin family of IRS genes. The sequence of SDCMP3 will be useful to identify other elements of the family, through bioinformatics and PCR technology. By analogy to other IRS molecules, SDCMP3 is predicted to associate on the cell surface in a signaling receptor complex. Based on its resistance expression in DC and monocytic cells, SDCMP3 would represent a selective target for therapeutic intervention to modulate the activation of DC. Depending on the association demonstrated with an IRS signaling pathway by inhibition (ITIM) or activation (ITAM), the mobilization of SDCMP3 can either suppress or elevate immune responses.
In addition, the restricted expression of SDCMP3 suggests the possibility of selective drug delivery for dendritic cells and cells of the monocyte / macrophage series. The distribution of mouse SDCMP3 was evaluated by Soulhem blots from cDNA genes from different sources. The DNA (5 μg) of a primary amplified cDNA library was digested with suitable restriction enzymes to release the inserts, run on 1% agarose gel and transferred to a nylon membrane (Schleicher and Schuell, Keene, NH) . Samples for isolation of mouse mRNA include: mouse fibroblast L cell line at rest (C200); transfected cells Braf: ER (Fusion of Braf to esoteric receptor), confrol (C201); T cells, TH1 polarized (bright Mel14 spleen cells, CD4 +, polarized for 7 days with IFN-α and anti IL-4, T200); polarized TH2 T cells (bright Mel14, CD4 + spleen cells, polarized for 7 days with IL-4 and anti-IFN-?; T201); T cells, highly polarized with TH1 (see Openshaw, et al., (1995) J. Exp. Med .. 182: 1357-1367; activated with anti-CD3 during 2, 6, 16 hours, clustered, T202; T cells highly polarized with TH2 (see Openshaw, et al (1995) J. Exp. Med. 182: 1357-1367; activated with anti-CD3 for 2, 6, 16 hours, pooled; T203); pre T cells CD44-CD25 + , classified as thymus (T204), T cell clone TH1 D1.1, at rest for 3 weeks after the last stimulation with antigen (T205), T cell clone TH1 D1.1, stimulated for 15 hours with 10 μg / ml ConA (T206); T cell clone TH2 CDC35, at rest for 3 weeks after the last stimulation with antigen (T207); TH2 CDC35 cell clone, stimulated for 15 hours with 10 μg / ml ConA (T208); spleen T cells affected by Mel14 +, at rest (T209); Mel14 + T cells, polarized for Th1 with IFN -? / IL-12 / anti-IL-4 during 6, 12, 24 hours, pooled (T210); Mel14 + T cells, polarized for Th2 with IL-4 / anl-IFN-? last 6, 13, 24 hours, grouped (T211); A20 cell line of mature non-stimulated B-cell leukemia (B200); line of non-stimulated CH12 B cells (B201); large unstimulated B spleen cells (B202); total spleen B cells, activated by LPS (B203); spleen dendritic cells enriched with mefrizamide, at rest (D200); bone marrow dendritic cells, at rest (D201); line of monocytes RAW 264.7 activated with LPS last 4 hours (M200); bone marrow macrophages derived with GM and M-CSF (M201); macrophage line J774, at rest (M202); line of macrophages J774 + LPS + anti-IL-10 at 0.5, 1, 3, 6, 12 hours, grouped (M203); line of macrophages J774 + LPS + IL-10 at 0.5, 1, 3, 5, 12 hours, grouped (M204); TH2 initiators of aerosol-challenged mouse lung tissue, challenge with aerosolized OVA, pooled for 7, 14, 23 hours (see Garlísí, et al. (1995) Clinical Immunology and Immunopaihology 75: 75-83; X206); lung tissue infected by Nippostrongulus (see Coffman, et al (1989) Science 245: 308-310; X200); total adult lung cells, normal (O200); total lung rag-1 cells (see Schwarz, et al. (1993) Immunodefíciency 4: 249-252; O205); spleen cells IL-10 K.O. (see Kuhn, et al. (1991) Cell 75: 263-274; X201); total, normal adult spleen cells (0201); total spleen rag-1 cells (O207); patches of Peyer IL-10 K.O. (O202); total, normal Peyer patches (0210); mesenteric lymph nodes IL-10 K.O. (X203); total mesenteric lymph nodes, normal (0211); colon IL-10 K.O. (X203); total, normal colon cells (0212); NOD mouse pancreas cells (see Makino, et al (1980) Jikken Dobutsu 29: 1-13, X205); total frog rag-1 cells (O208); total kidney rag-1 cells (O209); total heart rag-1 cells (O202); total brain rag-1 cells (O203); total rag-1 testis cells (O204); total liver rag-1 cells (O206); normal articulation tissue of rafa (O300) and rat arthritic joint tissue (X300). Strong positive signals were detected in: bone marrow dendritic cells, at rest (D201); bone marrow macrophages derived with GM and M-CSF (M201). Low signals were detected in total thymus rag-1 cells (O208); and total spleen rag-1 cells (O207). Weakly detectable signals were detected in mesenteric lymph nodes K.10 K.O. (X203), total normal adult lung cells (O200) and total lung rag-1 cells (see Schwarz, et al. (1993) Immunodeficiency 4: 249-252; O205). Oíros did not show any signs of harm. The alpha signals suggest that the marker may be useful in distinguishing or characterizing dendritic cells and / or populations or subpopulations of macrophages. The distribution of SDCMP4 by PCR: positive signals in: dendritic cells flushed with GM-CSF and TNFa; monocytes acivated with PMA and ionomycin; granulocytes activated with PMA and lonomycin; and PBL (probably Langerhans cells); no detectable signals were found in: TF1 cell line, Jurkai, MRC5, JY, U937, CHA; activated T cells; or activated B cells. SDCMP4 was detected in DC (from CD34 + progenitors cultured 12 days in GM-CSF and FNTa), either not activated or activated with PMA and ionomycin. Signals were also detected in monocytes, granulocytes and PBL (both not activated or activated with PMA and ionomycin). Sequence databases show sequences of SDCMP4 in primary dendritic cells (frequent); bone marrow (one); eosinophils (a); abducted placenta (one) and T-cell lymphoma (two). The genes of SDCMP3 and SDCMP4 display considerable homology with the murine counterpart of human monocyte ASGPR (M-ASGPR). The homology is significant in the carbohydrate recognition domain which confers specificity to murine monocyte ASGPR for galactose and N-acetylgalactosamine (GalNAc). Sato, et al. (1992) L Biochem. 111: 331-336. In addition, murine monocyte ASGPR has a YENL incorporation signal in its kyphosic domain. A dendrogram of CRD sequences suggests a more accurate relationship of mouse and human SDCMP3 with SDCMP2 than with SDCMP4. These CRDs seem to be more closely related to each other than to the CRG of the hepatic ASGPR. The murine M-ASGPR functions as a receptor for endocytosis of galacfosylated glucoprofeins (Ozaki, et al (1992) J. Biol. Chem. 267: 9229-9235) and allows the recognition of malignant cells by tumoricidal macrophages (Kawakami, ef al. (1994) Jpn. J. Cancer Res. 85: 744-749). In this context, murine M-ASGPR was found expressed within meiastasis lung nodules of roots possessing ovarian metastatic ovarian cells OV2944-HM-1 (Imai, et al. (1995) Immunol., 86: 591-598). It should be noted that human M-ASGPR demonstrates a remarkable specificity for Tn antigen (Suzuki, et al (1996) Immunol 156: 128-135), which possesses a terminal GalNAc group linked to serine or ireine and it is associated with human carcinomas (Springer (1989) Mol.Immunol.26: 1-5; and 0rntoft, et al. (1990) Int. J. Cancer 45: 666-672). Based on sequence homology, it can be predicted that SDCMP also functions as an endocytic receptor for galactosylated glucoproleins. In addition, the incorporation of ligands by the mannose receptor, another endocytic lecithin of type C transmembrane, results in a highly efficient antigen presentation by DC through the MHC class II pathway. Celia, ef al. (1997) Current Opinion Immunol. 9: 10-16 By analogy, it is possible that SDCMP plays a similar role in the routing of ligands incorporated in an antigen presentation pathway. Thus, SDCMP4 could be a potential high efficiency target for loading antigens into DC to extend the presentation to cells in adjuvant therapy based on the immune response. This can be tackled by pulsing DC in vitro with either a galactosylated form of anigen or with mABs ani-SDCMP4 bound to anigen. The efficiency of in vitro presentation could be analyzed by the activation of antigen-specific T cells. This would focus on the presentation of tumor-associated antigens (TAA), due to the inherent therapeutic perspectives of such an approach. Of particular interest are TAAs associated with malignant melanoma. In addition, the specificity of M-ASGPR from human to Tn antigen makes this TAA carcinoma an option candidate to direct the SDCMP4. As shown recently, the exogenous antigen can be processed and presented in the MHC class I pathway. See Porgador and Gilboa (1995) J. Exp. Med. 182: 255-260; and Paglia, et al. (1996) J. Exp. Med. 183: 317-322. It is likely that specialized receivers perform this function in DC. These receptors in DC can be directed to help produce TAA specific cytotoxic T cells (CTL), with important therapeutic potential, since CTL seems to be involved in the induction of tumor rejection.
XIV. Isolation of a binding counterpart A DC prolein can be used as a specific binding reagent, taking advantage of its binding specificity, much as an antibody would be used. A binding reagent is labeled as described above, for example, with fluorescence or in another form, or immobilized to a frame for panning methods. The DC protein is used to select a cell line that has a binding. The staining techniques are used to delect or classify intracellular ligands or surface-expressed ligands, or the cells transformed by surface expression are selected by panning. The selection of intracellular expression is carried out through various staining or immunofluorescence procedures. See also McMahan, et al. (1991) EMBO J. 10: 2821-2832. For example, on day 0, pre-expose two-chamber permanox slides with 1 ml of fibrotectin per chamber, 10 ng / ml in PBS, for 30 minutes at room temperature. Rinse once with PBS. Then, plate COS cells at 2-3 x 105 cells per chamber in 1.5 ml of growth media. Incubate overnight at 37 ° C. On day 1 for each sample, prepare 0.5 ml of a 66 mg / ml solution of DEAE-dextran, 66 mM chloroquine, and 4 mg of DNA in serum-free DME. For each group, a positive conírol is prepared, for example, of human FLAG receptor-cDNA at a dilution of 1 and 1/200 and a negative simulation. Rinse the cells with serum-free DME. Add the DNA solution and incubate for 5 hours at 37 ° C. Remove the medium and add 0.5 and 10% DMSO in DME for 2.5 min. Stir and wash once with DME. Add 1.5 ml of growth medium and incubate overnight.
On day 2, change the medium. On days 3 or 4, the cells are fixed and stained. Rinse the cells twice with Hank's regulated saline solution (HBSS) and fix in 4% paraformaldehyde (PFA) / glucose for 5 min. Wash three times with HBSS. The slides can be stored at -80 ° C after all the liquid is removed. For each chamber, incubations of 0.5 ml are made in the following way. Add HBSS / saponin (0.1%) with 32 ml / ml of 1 M NaN for 20 min. Then, the cells are washed once with HBSS / saponin. Add proinin or protein / antibody complex to the cells and incubate for 30 minutes. Wash the cells twice with HBSS / saponin. If necessary, add the first antibody for 30 minutes. Add second antibody, for example, anti-mouse antibody as Vector at 1/200 dilution and incubate for 30 minutes. Prepare ELISA solution, for example, Vector Elite ABC radish peroxidase solution and preincubate for 30 minutes. Use, for example, one drop of solution A (avidin) and 1 drop of solution B (biotin) per 2.5 ml of HBSS / saponin. Wash the cells twice with HBSS / saponin. Add ABC HRP solution and incubate for 30 minutes. Wash the cells twice with HBSS, a second wash for 2 minutes, which closes the cells. Then, adding diaminobenzoic acid from the vector (DAB) lasts for 5 to 10 minutes. Use two drops of pH regulator plus 4 DAB gofas plus 2 drops of H2O2 in 5 ml of distilled water in glass. Carefully remove the chamber and rinse the slide in water. Air dry for a few minutes, then add a Crysal Mounf grog and a coverslip. Bake for 5 minutes at 85-90 ° C. Alternatively, other specific binding reagents to monocyte protein are used to purify or affinity select cells that express a receptor. See, for example, Sambrook, et al. o Ausubel, et al. Another strategy is to select a membrane-bound receiver by panning. The receptor cDNA is constructed as described above. The ligand can be immobilized and used to immobilize expression cells. Immobilization can be achieved by the use of appropriate antibodies which recognize, for example, a FLAG sequence of a monocyte proiein fusion construct, or by the use of antibodies produced with the first antibodies. The recursive cycles of selection and amplification lead to the enrichment of suitable clones and the eventual isolation of clones expressing the ligand. Phage display libraries can be selected by monocyte proteins. Appropriate labeling techniques, eg, anti-FLAG antibodies, will allow specific labeling of the appropriate clones. Many modifications and variations of this invention can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. The specific embodiments described herein are offered by way of example only, and the invention will be limited only to the terms of the appended claims, along with the full scope of equivalents to which the claims are entitled.
LIST OF SEQUENCES (1. GENERAL INFORMATION: (i) APPLICANT: SCHERING CORPORATION (ii) TITLE OF THE INVENTION: ISOLATED MEMBRANE PROTEIN GENES OF MAMMALS, AND RELATED REAGENTS (iii) SEQUENCE NUMBER: 8 Uv) ADDRESS OF CORRESPONDENCE: (A) RECIPIENT: Schering Corportion (B) STREET: 2000 Galloping Hill Road (C) CITY: Kenilworth (D) STATE: New Jersey (E) COUNTRY: USA (F) C.P .: 07033-0530 (v) COMPUTER LEGIBLE FORM: (A) TYPE OF MEDIUM: Dis (B) COMPUTER: Apple Macintosh (C) OPERATING SYSTEM: acintosh 7.1 (D) SOFTWARE: icrosoft Word 6.0 (vi) CURRENT APPLICATION INFORMATION: (A) APPLICATION NUMBER: (B) SUBMISSION DATE: (C) CLASSIFICATION: (viii) EMPLOYEE / AGENT INFORMATION: (A) NAME: (B) REGISTRATION NUMBER: (C) REFERENCE NUMBER / CASE: (ix) TELECOMMUNICATION INFORMATION: (A) TELEPHONE: (908) 298 4000 (B) FAX: (908) 298 5388 (2) INFORMATION FOR SEQ ID NO: 1: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 850 base pairs (B) TYPE: nucleic acid (C) CHAIN TYPE: simple (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: cDNA (ix) FEATURE: (A) NAME / KEY: CDS (B) LOCATION: 108..593 (xi) SEQUENCE DESCRIPTION: SEQ ID NO: l: GTCCCTGAGC TCTAGCTTCT TTAAATGAAG CTGAGTCTCT GGGCAACATC TTTAGGGAGA GAGGTACAAA AGGTTCCTGG ACCTTCTCAA CACAGGGAGC CTGCATA ATG ATG CAA 116 Met Met Gln 1 GAG CAG CAÁ CCT CAÁ AGT ACÁ GAG AAA AGA GGC TGG TTG TCC CTG AGA 164 Glu Gln Gln Pro Gln Ser Thr Glu Lys Arg Gly Trp Leu Ser Leu Arg 5 10 15 CTC TGG TCT GTG GCT GGG ATT TCC ATT GCA CTC CTC AGT GCT TGC TTC 212 Leu Trp ger Val Ala Gly lie Ser lie Ala Leu Leu Ser Ala Cys Phe 20 25 30 35 ATT GTG AGC TGT GTA GTA ACT TAC CAT TTT ACA TAT GGT GAA ACT GGC 260 lie Val Ser Cys Val Val Thr Tyr His Phe Thr Tyr Gly Glu Thr Gly 40 45 50 AAA AGG CTG TCT GAA CTA CAC TCA TAT CAT TCA AGT CTT ACC TGC TTC 308 Lys Arg Leu Ser Glu Leu His Ser Tyr His Ser Ser Leu Thr Cys Phe 55 60 65 AGT GAA GGG ACA AAG GTG CCA GCC TGG GGA TGT TGC CCA GCT TCT TGG 356 Ser Glu Gly Thr Lys Val Pro Wing Trp Gly Cys Cys Pro Wing Ser Trp 70 75 80 AAG TCA TTT GGT TCC AGT TGC TAC TTC ATT TCC AGT GAA GAG AAG GTT 404 Lys Ser Phe Gly Ser Ser Cys Tyr Phe lie Ser Ser Glu Glu Lys Val 85 90 95 TGG TCT AAG AGT GAG CAG AAC TGT GTT GAG ATG GGA GCA CAT TTG GTT 452 Trp Ser Lys Ser Glu Gln Asn Cys Val Glu Met Gly Ala His Leu Val 100 105 110 115 GTG TTC AAC ACA GAA GCA GAG CAG AAT TTC ATT GTC CAG CAG CTG AAT 500 Val Phe Asn Thr Glu Wing Glu Gln Asn Phe lie Val Gln Gln Leu Asn 120 125 130 GAG TCA TTT TCT TAT TTT CTG GGG CTT TCA GAC CCA CAA GGT AAT AAT 548 Glu Ser Phe Ser Tyr Phe Leu Gly Leu Ser Asp Pro Gln Gly Asn Asn 135 140 145 AAT TGG CAÁ TGG ATT GAT AAG ACÁ CCT TAT GAG AAA AAT GTC AGG 593 Asn Trp Gln Trp lie Asp Lys Thr Pro Tyr Glu Lys Asn Val Arg 150 155 160 TGAGTGCAGT TCTGGGGCCT TGTTTACATA GAAAATCTAG GGAAATTTTG TTAGGAGTTA 653 CTAATAATGT TAATATTGGT AATTATGATA ACAGGATCTA ACAATTATTA AGCATTACTA 713 AGGATATGCA TTATCTCACT TAAACTTCAT GAAAACTTCT CTTTTTATGA ACTAATTTTA 773 CAGATAAAAA ATTAAATAAC TTGCCCCAAA TCAATAAACT AATAAGATGA GAAACTGGAT 833 GTCAACTCCA TGTCAAG 850 (2) INFORMATION FOR SEQ ID NO: 2: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 162 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 2: Met Met Gln Glu Gln Gln Pro Gln Ser Thr Glu Lys Arg Gly Trp Leu 1 5 10 15 Ser Leu Arg Leu Trp Ser Val Ala Gly lie Ser lie Ala Leu Leu Ser 20 25 30 Ala Cys Phe lie Val Ser Cys Val Val Thr Tyr His Phe Thr Tyr Gly 35 40 45 Glu Thr Gly Lys Arg Leu Ser Glu Leu His Ser Tyr His Ser Ser Leu 50 55 60 Thr Cys Phe Ser Glu Gly Thr Lys Val Pro Wing Trp Gly Cys Cys Pro 65 70 75 80 Wing Ser Trp Lys Ser Phe Gly Ser Ser Cys Tyr Phe lie Ser Ser Glu 85 90 95 Glu Lys Val Trp Ser Lys Ser Glu Gln Asn Cys Val Glu Met Gly Ala 100 105 110 His Leu Val Val Phe Asn Thr Glu Ala Glu Gln Asn Phe lie Val Gln 115 120 125 Gln Leu Asn Glu Ser Phe Ser Tyr Phe Leu Gly Leu Ser Asp Pro Gln 130 135 140 Gly Asn Asn Asn Trp Gln Trp lie Asp Lys Thr Pro Tyr Glu Lys Asn 145 150 155 160 Val Arg (2) INFORMATION FOR SEQ ID NO: 3: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 630 base pairs (B) TYPE: nucleic acid (C) CHAIN TYPE: simple (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: cDNA (ix) FEATURE: (A) NAME / KEY: CDS (B) LOCATION: 1.627 (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 3: ATG GTG CAG GAA AGA CAA TCC CAA GGG AAG GGA GTC TGC TGG ACC CTG 48 Met Val Gln Glu Arg Gln Ser Gln Gly Lys Gly Val Cys Trp Thr Leu 1 5 10 15 AGA CTC TGG TCA GCT GCT GTG ATT TCC ATG TTA CTC TTG AGT ACC TGT 96 Arg Leu Trp Ser Ala Ala Val lie Met Leu Leu Leu Ser Thr Cys 20 25 30 TTC ATT GCG AGC TGT GTG GTG ACT TAC CA TTT ATT ATG GAC CAG CCC 144 Phe He Wing Ser Cys Val Val Thr Tyr Gln Phe He Met Asp Gln Pro 35 40 45 AGT AGA AGA CTA TAT GAA CTT CAC ACÁ TAC CAT TCC AGT CTC ACC TGC 192 Ser Arg Arg Leu Tyr Glu Leu His Thr Tyr His Ser Ser Leu Thr Cys 50 55 60 TTC AGT GAA GGG ACT ATG GTG TCA GAA AAA ATG TGG GGA TGC TGC CCA 240 Phe Ser Glu Gly Thr Met Val Ser Glu Lys Met Trp Gly Cys Cys Pro 65 70 75 80 AAT CAC TGG AAG TCA TTT GGC TCC AGC TGC TAC CTC ATT TCT ACC AAG 288 Asn His Trp Lys Ser Phe Gly Ser Ser Cys Tyr Leu Ser Ser Thr Lys 85 90 95 GAG AAC TTC TGG AGC ACC AGT GAG CAG AAC TGT GTT CAG ATG GGG GCT 336 Glu Asn Phe Trp Ser Thr Ser Glu Gln Asn Cys Val Gln Met Gly Ala 100 105 110 CAT CTG GTG GTG ATC AAT ACT GAA GCG GAG CAG AAT TTC ATC ACC CAG 384 His Leu Val Val He Asn Thr Glu Ala Glu Gln.Asn Phe He Thr Gln 115 120 125 CAG CTG AAT GAG TCA CTT TCT CT CT CTG GGT CTT TCG GAT CCA CAA Gln Leu Asn Glu Ser Leu Ser Tyr Phe Leu Gly Leu Ser Asp Pro Gln 130 135 140 GGT AAT GGC AAA TGG CAA TGG ATC GAT GAT ACT CCT TTC AGT CA AAT Gly Asn Gly Lys Trp Gln Trp He Asp Asp Thr Pro Phe Ser Gln Asn 145 150 155 160 GTC AGG TTC TGG CAC CCC CAT GAA CCC AAT CTT CCA GAA GAG CGG TGT Val Arg Phe Trp His Pro His Glu Pro Asn Leu Pro Glu Glu Arg Cys 165 170 175 GTT TCA ATA GTT TAC TGG AAT CCT TCG AAA TGG GGC TGG AAT GAT GTT 576 Val Ser He Val Tyr Trp Asn Pro Ser Lys Trp Gly Trp Asn Asp Val 180 185 190 TTC TGT GAT AGT AAA CAC AAT TCA ATA TGT GAA ATG AAG AAG ATT TAC 624 Phe Cys Asp Ser Lys His Asn Ser He Cys Glu Met Lys Lys He Tyr 195 200 205 CTA TGA 630 Leu (2) INFORMATION FOR SEQ ID NO: 4: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 209 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 4: Met Val Gln Glu Arg Gln Ser Gln Gly Lys Gly Val Cys Trp Thr Leu 1 5 10 15 Arg Leu Trp Be Ala Ala Val Be He Met Met Leu Leu Leu Be Thr Cys 20 25 30 Phe He Wing Ser Cys Val Val Thr Tyr Gln Phe He Met Asp Gln Pro 35 40 45 Being Arg Arg Leu Tyr Glu Leu His Thr Tyr His Being Being Leu Thr Cys 50 55 60 Phe Ser Glu Gly Thr Met Val Being Glu Lys Met Trp Gly Cys Cys Pro 65 70 75 80 Asn His Trp Lys Ser Phe Gly Ser Ser Cys Tyr Leu He Ser Thr Lys 85 90 95 Glu Asn Phe Trp Ser Thr Ser Glu Gln Asn Cys Val Gln Met Gly Ala 100 105 110 His Leu Val Val He Asn Thr Glu Wing Glu Gln Asn Phe He Thr Gln 115 120 125 Gln Leu Asn Glu Ser Leu Ser Tyr Phe Leu Gly Leu Ser Asp Pro Gln 130 135 140 Gly Asn Gly Lys Trp Gln Trp He Asp Asp Thr Pro Phe Ser Gln Asn 145 150 155 160 Val Arg * "Phe Trp His Pro His Glu Pro Asn Leu Pro Glu Glu Arg Cys 165 170 175 Val Ser He Val Tyr Trp Asn Pro Ser Lys Trp Gly Trp Asn Asp Val 180 185 190 Phe Cys Asp Ser Lys His Asn Ser He Cys Glu Met Lys Lys He Tyr 195 200 205 Leu (2) INFORMATION FOR SEQ ID NO: 5: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 1018 base pairs (B) TYPE: nucleic acid (C) TYPE OF CHAIN: simple (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: cDNA (ix) FEATURE: (A) NAME / KEY: CDS (B) LOCATION: 160..900 (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 5: ATCTGGTTGA ACTACTTAAG CTTAATTTGT TAAACTCCGG TAAGTACCTA GCCCACATGA 60 TTTGACTCAG AGATTCTCTT TTGTCCACAG ACAGTCATCT CAGGAGCAGA AAGAAAAGAG 120 CTCCCAAATG CTATATCTAT TCAGGGGCTC TCAAGAACA ATG GAA TAT CAT CCT 174 Met Glu Tyr His Pro 1 5 GAT TTA GAA AAT TTG GAT GAA GAT TAT ACT CAA TTA CAC TTC GAC 222 Asp Leu Glu Asn Leu Asp Glu Asp Gly Tyr Thr Gln Leu His Phe Asp 10 15 20 TCT CAA AGC AAT ACC AGG ATA GCT GTT GTT TCA GAG AAA GGA TCG TGT 270 Ser Gln Ser Asn Thr Arg He Wing Val Val Ser Glu Lys Gly Ser Cys 25 30 35 GCT GCA TCT CCT CCT TGG CGC CTC ATT GCT GTA ATT TTG GGA ATC CTA 318 Wing Wing Pro Pro Pro Trp Arg Leu He Wing Val He Leu Gly He Leu 40 45 50 TGC TTG GTA ATA CTG GTG ATA GCT GTG GTC CTG GGT ACC ATG GCT ATT 366 Cys Leu Val He Leu Val He Wing Val Val Leu Gly Thr Met Wing He 55 60 65 TGG AGA TCC AAT TCA GGA AGC AAC ACÁ TTG GAG AAT GGC TAC TTT CTA 414 Trp Arg Ser Asn Ser Gly Ser Asn Thr Leu Glu Asn Gly Tyr Phe Leu 70 75 80 85 TCA AGA AAT AAA GAG AAC CAC AGT CAÁ CCC ACÁ CAÁ TCA TCT TTA GAA Ser Arg Asn Lys Glu Asn His Ser Gln Pro Thr Gln Ser Ser Leu Glu 90 95 100 GAC AGT GTG ACT CCT ACC AAA GCT GTC AAA ACC ACA GGG GTT CTT TCC 510 Asp Ser Val Thr Pro Thr Lys Wing Val Lys Thr Thr Gly Val Leu Ser 105 110 115 AGC CCT TGT CCT CCT AAT TGG ATT ATA TAT GAG AAG AGC TGT TAT CTA 558 Pro Pro Cys Pro Pro Asn Trp He He Tyr Glu Lys Ser Cys Tyr Leu 120 125 130 TTC AGC ATG TCA CTA AAT TCC TGG GAT GGA AGT AAA AGA CAA TGC TGG 606 Phe Ser Met Ser Leu Asn Ser Trp Asp Gly Ser Lys Arg Gln Cys Trp 135 140 145 CAA CTG GGC TCT AAT CTC CTA AAG ATA GAC AGC TCA AAT GAA TTG GGA 654 Gln Leu Gly Ser Asn Leu Leu Lys He Asp Ser Ser Asn Glu Leu Gly 150 155 160 165 TTT ATA GTA AAA CAA GTG TCT TCC CA CA CCT GAT AAT TCA TTT TGG ATA 702 Phe He Val Lys Gln Val Ser Ser Gln Pro Asp Asn Ser Phe Trp He 170 175 180 GGC CTT TCT CGG CCC CAG ACT GAG GTA CCA TGG CTC TGG GAG GAT GGA 750 Gly Leu Ser Arg Pro Gln Thr Glu Val Pro Trp Leu Trp Glu Asp Gly 185 190 195 TCA ACT TTC TCT TCT AAC TTA TTT CAG ATC AGA ACC ACA GCT ACC CAA 798 Ser Thr Phe Ser Ser Asn Leu Phe Gln He Arg Thr Thr Ala Thr Gln 200 205 210 GAA AAC CCA TCT CCA AAT TGT GTA TGG ATT CAC GTG TCA GTC ATT TAT 846 Glu Asn Pro Ser Pro Asn Cys Val Trp He His Val Ser Val He Tyr 215 220 225 GAC CAÁ CTG TGT AGT GTG CCC TCA TAT AGT ATT TGT GAG AAG AAG TTT 894 Asp Gln Leu Cys Ser Val Pro Ser Tyr Ser He Cys Glu Lys Lys Phe 230 235 240 245 TCA ATG TAAGGGGAAG GGTGGAGAAG GAGAGAGAAA TATGTGAGGT AGTTAAGGAG 950 Ser Met GACAGAAAAC AGAACAGAAA AGAGTAACAG CTGAGGGTCA AGATAAATGC AGAAAATGTT 1010 TAGAGAGC 1018 (2) INFORMATION FOR SEQ ID NO: 6: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 247 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 6: Met Glu Tyr His Pro Asp Leu Glu Asn Leu Asp Glu Asp Gly Tyr Thr 1 5 10 15 Gln Leu His Phe Asp Ser Gln Ser Asn Thr Arg He Wing Val Val Ser 20 25 30 Glu Lys Gly Ser Cys Wing Wing Pro Pro Pro Trp Arg Leu He Wing Val 35 40 45 He Leu Gly He Leu Cys Leu Val He Leu Val He Wing Val Val Leu 50 55 60 Gly Thr Met Wing He Trp Arg Ser Asn Ser Gly Ser Asn Thr Leu Glu 65 70 75 80 Asn Gly Tyr Phe Leu Ser Arg Asn Lys Glu Asn His Ser Gln Pro Thr 85 90 95 Gln Ser Ser Leu Glu Asp Ser Val Thr Pro Thr Lys Wing Val Lys Thr 100 105 110 Thr Gly Val Leu Ser Ser Pro Cys Pro Pro Asn Trp He He Tyr Glu 115 120 125 Lys Ser Cys Tyr Leu Phe Ser Met Ser Leu Asn Ser Trp Asp Gly Ser 130 135 140 Lys Arg Gln Cys Trp Gln Leu Gly Ser Asn Leu Leu Lys He Asp Ser 145 150 155 160 Being Asn Glu Leu Gly Phe He Val Lys Gln Val Being Ser Gln Pro Asp 165 170 175 Asn Ser Phe Trp He Gly Leu Ser Arg Pro Gln Thr Glu Val Pro Trp 180 185 190 Leu Trp Glu Asp Gly Be Thr Phe Be Ser Asn Leu Phe Gln He Arg 195 200 205 Thr Thr Ala Thr Gln Glu Asn Pro Ser Pro Asn Cys Val Trp He His 210 215 220 Val Ser Val He Tyr Asp Gln Leu Cys Ser Val Pro Ser Tyr Ser He 225 230 235 240 Cys Glu Lys Lys Phe Ser Met 245 (2) INFORMATION FOR? EQ ID NO: 7: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 880 base pairs (B) TYPE: nucleic acid (C) CHAIN TYPE: simple (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: cDNA (ix) FEATURE: (A) NAME / KEY: CDS (B) LOCATION: 160..762 (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 7: ATCTGGTTGA ACTACTTAAG CTTAATTTGT TAAACTCCGG TAAGTACCTA GCCCACATGA TTTGACTCAG AGATTCTCTT TTGTCCACAG ACAGTCATCT CAGGAGCCGA AAGAAAAGAG 120 CTCCCAAATG CTATATCTAT TCAGGGGCTC TCAAGAACA ATG GAA TAT CAT CCT 174 Met Glu Tyr His Pro 1 5 GAT TTA GAA AAT TTG GAT GAA GAT GGA TAT ACT CAA TTA CAC TTC GAC 222 Asp Leu Glu Asn Leu Asp Glu Asp Gly Tyr Thr Gln Leu His Phe Asp 10 15 20 TCT CAA AGC AAT ACC ATG ATA GCT GTT GTT TCA GAG AAA GGA TCG TGT 270 be Gln Ser Asn Thr Met He Wing Val Val Ser Glu Lys Gly Ser Cys 25 30 35 GCT GCA TCT CCT CCT TGG CGC CTC ATT GCT ATT TTG GGA ATC CTA 318 Wing Wing Pro Pro Trp Arg Leu He Wing Val He Leu Gly He Leu 40 45 50 TGC TTG GTA ATA CTG GTG ATA GCT GTG GTC CTG GGT ACC ATG GGG GTT Cys Leu Val He Leu Val He Wing Val Val Leu Gly Thr Met Gly Val 55 60 65 CTT TCC AGC CCT TGT CCT CCT AAT TGG ATT ATA TAT GAG AAG AGC TGT Leu Ser Pro Pro Cys Pro Pro Asn Trp He He Tyr Glu Lys Ser Cys 70 75 80 85 TAT CTA TTC AGC ATG TCA CTA AAT TCC TGG GAT GGA AGT AAA AGA CAA Tyr Leu Phe Ser Met Ser Leu Asn Ser Trp Asp Gly Ser Lys Arg Gln 90 95 100 TGC TGG CAÁ CTG GGC TCT AAT CTC CTA AAG ATA GAC AGC TCA AAT GAA 510 Cys Trp Gln Leu Gly Ser Asn Leu Leu Lys He Asp Ser Ser Asn Glu 105 110 115 TTG GGA TTT ATA GTA AAA CAA GTG TCT TCC CA CA CCT GAT AAT TCA TTT 558 Leu Gly Phe He Val Lys Gln Val Ser Ser Gln Pro Asp Asn Ser Phe 120 125 130 TGG ATA GGC CTT TCT CGG CCC CAG ACT GAG GTA CCA TGG CTC TGG GAG 606 Trp He Gly Leu Ser Arg Pro Gln Thr Glu Val Pro Trp Leu Trp Glu 135 140 145 GAT GGA TCA ACA TTC TCT TCT AAC TTA TTT CAG ATC AGA ACC ACCT GCT 654 Asp Gly Ser Thr Phe Ser Ser Asn Leu Phe Gln He Arg Thr Thr Ala 150 155 160 165 ACC CAA GAA AAC CCA TCT CCA AAT TGT GTA TGG ATT CAC GTG TCA GTC 702 Tnr Gln GlU Asn Pro Ser Pro Asn cy? Val TrP Ile His Val Ser Val 170 175 180 ATT TAT GAC CAG CTG TGT AGT GTG CCC TCA TAT AGT ATT TGG GAG AAG 750 He Tyr Asp Gln Leu Cys Ser Val Pro Ser Tyr Ser He Cys Glu Lys 185 190 195 AAG TTT TCA ATG TAAGGGGAAG GGTGGAGAAG GAGAGAGAAA TATGTGAGGT 802 Lys Phe Ser Met 200 AGTTAAGGAG GACAGAAAAC AGAACAGAAA AGAGTAACAG CTGAGGGTCA AGATAAATGC 862 AGAAAATGTT TAGAGAGC (2) INFORMATION FOR SEQ ID NO: 8: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 201 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 8: Met Glu Tyr His Pro Asp Leu Glu Asn Leu Asp Glu Asp Gly Tyr Thr 1 5 10 15 Gln Leu His Phe Asp Ser Gln Ser Asn Thr Met He Wing Val Val Ser 20 25 30 Glu Lys Gly Ser Cys Wing Wing Pro Pro Pro Trp Arg Leu He Wing Val 35 40 45 He Leu Gly He Leu Cys Leu Val He Leu Val He Wing Val Val Leu 50 55 60 Gly Thr Met Gly Val Leu Ser Pro Cys Pro Pro Asn Trp He He 65 70 75 80 Tyr Glu Lys Ser Cys Tyr Leu Phe Ser Met Ser Leu Asn Ser Trp Asp 85 90 95 Gly Ser Lys Arg Gln Cys Trp Gln Leu Gly Ser Asn Leu Leu Lys He 100 105 110 Asp Being Ser Asn Glu Leu Gly Phe He Val Lys Gln Val Being Ser Gln 115 120 125 Pro Asp Asn Ser Phe Trp He Gly Leu Ser Arg Pro Gln Thr Glu Val 130 135 140 Pro Trp Leu Trp Glu Asp Gly Ser Thr Phe Ser Ser Asn Leu Phe Gln 145 150 155 160 He Arg Thr Thr Wing Thr Gln Glu Asn Pro Ser Pro Asn Cys Val Trp 165 170 175 He His Val Val Val Tyr Asp Gln Leu Cys Ser Val Pro Ser Tyr 180 185 190 Ser He Cys Glu Lys Lys Phe Ser Met 195 200

Claims (20)

1 6 NOVELTY OF THE INVENTION CLAIMS
1. - A binding compound comprising an antibody binding site that specifically binds to: a) a primate or rodent SDCMP3 protein; or b) a primate SDCMP4 protein.
2. The binding compound according to claim 1, further characterized in that: a) said antibody binding site is: 1) specifically immunoreactive with a protein of SEQ ID NO: 2 or 4; 2) specifically immunoreactive with a protein of SEQ ID NO: 6 or 8; 3) produced confers a purified or recombinant human SDCMP3 protein of SEQ ID NO: 2 or rodent SDCMP3 protein of SEQ ID NO: 4; 4) produced against a purified or recombinantly human SDCMP4 protein produced from SEQ ID NO: 6 or 8; 5) in a monoclonal antibody, Fab, or F (ab) 2; or b) said binding compound is: 1) detectably labeled; 2) sterile; or 3) in a regulated composition of pH.
3. A method that binds the binding compound according to claim 2, which comprises bringing said binding compound into conjoint with a biological sample comprising an antigen to form a complex of binding compound: antigen.
4. The method according to claim 3, further characterized in that said biological sample is human, and in that said binding compound is an antibody.
5. A defection device comprising said binding compound according to claim 1 and: a) instructional material for the use of said binding compound for said detection; or b) a compartment that provides segregation of said binding compound.
6. A purely or isolated sub-epidermal polypeptide, which binds specifically to a binding compound according to claim 1.
7. The polypeptide according to claim 6, further characterized in that: a) it comprises at least one fragment of at least 14 amino acid residues of a primate or rodent SDCMP3 protein; b) comprises at least one fragment of at least 14 amino acid residues of a primate SDCMP4 protein; c) is a soluble polypeptide; d) is marked in a reliable manner; e) it is in a sterile solution; f) is in a regulated pH solution; g) binds to a sialic acid residue; h) is produced recombinantly; or ¡) has a sequence of natural polypeptides.
8. An isolated nucleic acid encoding a polypeptide of claim 7.
9. The nucleic acid according to claim 8, further characterized in that: a) it comprises at least 30 nucleotides of the coding portion of SEQ ID NO : 1 or 3; or b) comprises at least 30 nucleolides of the coding portion of SEQ ID NO: 5 or 7.
10. A vector comprising the nucleic acid of claim 8.
11. A cell transfected with said nucleic acid of claim 8.
12. The cell according to claim 11, further characterized in that said nucleic acid consists of the protein coding for portions of SEQ ID NO: 1, 3 or 5. 13.- A method that uses at least one chain of said nucleic acid of claim 8 for forming a nucleic acid duplex, said method comprises a step of contacting said strand with a sample comprising a complementary strand capable of specific hybridization. 14. The method according to claim 13, further characterized in that: a) it allows the detection of said duplex; or b) allows the histological location of said duplex. 15. A method for using the binding compound of claim 1, comprising a step of contacting said binding compound with a sample to form a complex of binding compound: amphigen. 16. The method according to claim 15, further characterized in that: a) said sample is a biological sample, which includes a body fluid; b) said antigen is in a cell; or c) said antigen is subsequently purified. 17. A method for using said polypeptide of claim 6, comprising contacting said polypeptide with a sample to form a binding complex: polypeptide. 18. The method according to claim 17, further characterized in that said polypeptide is subsequently purified ..}. 19. The use of: a) a joining compound of claim 1; b) a proinin SDCMP3 or SDCMP4; or c) a nucleic acid encoding SDCMP3 or SDCMP4, for the manufacture of a composition for modulating the physiology or function of dendritic cells. 20. The use as claimed in claim 19, wherein said composition is combined with an antigen, which includes a cell surface, MHC class I antigen or MHC class II antigen.
MXPA/A/2000/009063A 1998-03-17 2000-09-14 Isolated mammalian membrane protein genes and related reagents MXPA00009063A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US09/040,111 1998-03-17

Publications (1)

Publication Number Publication Date
MXPA00009063A true MXPA00009063A (en) 2001-07-09

Family

ID=

Similar Documents

Publication Publication Date Title
US7691373B2 (en) Isolated mammalian membrane proteins; related agents
US6562333B1 (en) Purified mammalian CTLA-8 antigens and related reagents
JPH11502420A (en) Mammalian chemokines CCF8 and chemokine receptor CCKR3
JP4156034B2 (en) Isolated mammalian monocytic cell genes; related reagents
WO1995006738A1 (en) B70(b7-2):ctla-4 bonding protein
CA2323083A1 (en) Isolated mammalian membrane protein genes and related reagents
US5811284A (en) Nucleic acids encoding kp43 protein and antigenic fragments thereof
US5965401A (en) Purified mammalian NK antigens and related reagents
US20030162955A1 (en) Isolated mammalian membrane protein genes; related reagents
US6361939B1 (en) Isolated mammalian dendritic cell genes; related reagents
MXPA00009063A (en) Isolated mammalian membrane protein genes and related reagents
WO1998023747A2 (en) Isolated mammalian dendritic cell genes; related reagents
WO1997020046A1 (en) Dnam, an nk antigen and adhesion molecule of the immunoglobulin superfamily
CA2715850A1 (en) Isolated dendritic cell membrane protein genes
MXPA00000356A (en) Isolated dendritic cell membrane protein genes
CZ473699A3 (en) Nearly pure or recombinant polypeptide DCMP1 and DCMP2, fusion protein, binding compound, nucleic acid encoding DCMP1 or DCMP2, expression vector, host cell for the expression vector and recombinant preparation process of the polypeptide