US20040180825A1 - Semaphorin polypeptides - Google Patents

Semaphorin polypeptides Download PDF

Info

Publication number
US20040180825A1
US20040180825A1 US10/740,773 US74077303A US2004180825A1 US 20040180825 A1 US20040180825 A1 US 20040180825A1 US 74077303 A US74077303 A US 74077303A US 2004180825 A1 US2004180825 A1 US 2004180825A1
Authority
US
United States
Prior art keywords
semaphorin
polypeptide
dna
amino acid
seq
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/740,773
Inventor
Melanie Spriggs
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Immunex Corp
Original Assignee
Immunex Corp
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 Immunex Corp filed Critical Immunex Corp
Priority to US10/740,773 priority Critical patent/US20040180825A1/en
Publication of US20040180825A1 publication Critical patent/US20040180825A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • C07K14/4703Inhibitors; Suppressors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Definitions

  • the present invention relates to semaphorin polypeptides, nucleic acids encoding such semaphorin polypeptides, processes for producing recombinant semaphorin polypeptides, pharmaceutical compositions containing such polypeptides relates and processes for treating disorders associated with semaphorin activity.
  • the semaphorin gene family includes a large number of molecules that encode related transmembrane and secreted glycoproteins known to be neurologic regulators.
  • the semaphorins are generally well conserved in their extracellular domains which are typically about 500 amino acids in length. Semaphorin family proteins have been observed in neuronal and nonneuronal tissue and have been studied largely for their role in neuronal growth cone guidance.
  • the secreted semaphorins known as collapsin-1 and Drosophila semaphorin II are selectively involved in repulsive growth cone guidance during development. Flies having semaphorin II genes that are mutated so that their function is reduced exhibit abnormal behavior characteristics.
  • A39R open reading frame
  • Variola virus ORF also contains sequences that share homology with the vaccinia virus ORF A39R at the nucleotide level and the amino acid level.
  • AHV-sema has been found in the Alcelaphine Herpesvirus (AHV).
  • virus encoded semaphorins are produced by infected cells and are present in viruses that are lytic (poxviruses) and viruses that are not known to be neurotropic (AHV), it is unlikely that their primary function is to modify neurologic responses. It is more likely that the virus encoded semaphorins function to modify the immunologic response of the infected host and it is likely that mammalian homologues to virus encoded semaphorins function to modify the immunologic response.
  • viral semaphorins may function in the immune system as natural immunoregulators it would be beneficial to identify semaphorins that may be therapeutic agents for enhancing or diminishing the immune response.
  • the present invention pertains to novel semaphorins as isolated or homogeneous proteins.
  • the present invention provides semaphorin polypeptides, that are homologous to the viral semaphorins A39R and AHVSema.
  • DNAs encoding semaphorin polypeptides and expression vectors that include DNA encoding semaphorin polypeptides.
  • the present invention also includes host cells that have been transfected or transformed with expression vectors that include DNA encoding a semaphorin polypeptide, and processes for producing semaphorin polypeptides by culturing such host cells under conditions conducive to expressing semaphorin polypeptides.
  • the present invention further includes antibodies directed against semaphorin polypeptides.
  • processes for purifying or separating certain novel semaphorin polypeptides or cells that express novel semaphorins to which semaphorin receptor polypeptides bind include binding at least one semaphorin receptor to a solid phase matrix and contacting a mixture containing a semaphorin polypeptide to which the semaphorin receptor binds, or a mixture of cells expressing the semaphorin with the bound semaphorin receptor, and then separating the contacting surface and the solution.
  • the present invention additionally provides processes for treating mammals afflicted with a disease that is ameliorated by the interaction of semaphorins and their receptors.
  • processes involve activating immune cells that express receptors for the semaphorins of the present invention and include administering a therapeutically effective amount of semaphorin to a mammal afflicted with the disease.
  • the therapeutically effective amount is sufficient to activate immune system cells that express semaphorin receptors.
  • Such an activation results in the secretion of cytokines, the regulation of activation antigens or the migration of the cell to sites of immune activity.
  • the present invention provides novel semaphorin polypeptides, DNA encoding semaphorin polypeptides and recombinant expression vectors that include DNA encoding semaphorin polypeptides.
  • the present invention further provides methods for isolating semaphorin polypeptides and methods for producing recombinant semaphorin polypeptides by cultivating host cells transfected with the recombinant expression vectors under conditions appropriate for expressing semaphorin and recovering the expressed semaphorin polypeptide.
  • This invention additionally provides antibodies directed against semaphorin polypeptides.
  • Particular semaphorin embodiments of the present invention include polypeptides homologous to AHVsema.
  • the native semaphorin polypeptide described herein was discovered using data base search and comparison techniques that resulted in the identification of at least one EST having some homology to viral semaphorins. As described in Example 1, PCR techniques were used to identify and clone the full viral semaphorin homologue.
  • the human semaphorin of the present invention is found in placenta, testis, ovary, spleen, dendritic cells, and B cells.
  • Semaphorin polypeptides of the present invention bind to the semaphorin receptor, designated VESPR (described in copending application S/N 08/958,598, incorporated herein by reference).
  • VESPR semaphorin receptor
  • the semaphorins of the present invention are designated DCSema.
  • Example 1 describes identifying a native DCSema of the present invention.
  • the amino acid sequence of the identified native DCSema is disclosed in SEQ ID NO:2 and the DNA encoding the amino acid sequence is disclosed in SEQ ID NO:1.
  • the amino acid sequence presented in SEQ ID NO:2 is a secreted soluble polypeptide, but may additionally exist as a membrane bound protein.
  • the amino acid sequence of SEQ ID NO:2 has a predicted signal sequence that includes amino acids 1-44.
  • Also encompassed within the present invention are soluble DCSema polypeptides that lack the signal sequence. An example of such a polypeptide is amino acids 45-666 of SEQ ID NO:2.
  • the EST 151129 portion of SEQ ID NO:2 has a 28% identity to A39R and 44% identity to AHVsema, both of which bind to VESPR, a semaphorin receptor described in copending patent application S/N 08/958,598.
  • A39R and AHVsema share 29% identity.
  • AHVSema AHVSema encompass polypeptides having the amino acid sequence disclosed in SEQ ID NO:2, and proteins that are encoded by nucleic acids that contain the nucleic acid sequence of SEQ ID NO: 1.
  • semaphorin polypeptide refers to biologically active gene products of the DNA of SEQ ID NO: 1. Further encompassed by semaphorin polypeptides are soluble or truncated proteins that comprise primarily the binding portion of the protein, retain biological activity and are capable of being secreted. Specific examples of such soluble proteins are those comprising the sequence of amino acids 45-666 of SEQ ID NO:2.
  • biologically active as it refers to semaphorin polypeptides, means that the semaphorin polypeptide is capable of binding to at least one semaphorin receptor.
  • Assays suitable for determining DCSema binding are described infra and can include standard flow cytometry tests and slide binding tests.
  • Isolated means a DCSema polypeptide is free of association with other proteins or polypeptides, for example, as a purification product of recombinant host cell culture or as a purified extract.
  • a DCSema polypeptide variant as referred to herein means a polypeptide substantially homologous to native DCSema polypeptide, but which has an amino acid sequence different from that of the native polypeptides because of one or more deletions, insertions or substitutions.
  • the variant amino acid sequence preferably is at least 80% identical to a native DCSema amino acid sequence, most preferably at least 90% identical.
  • the percent identity may be determined, for example, by comparing sequence information using the GAP computer program, version 6.0 described by Devereux et al. ( Nucl. Acids Res. 12:387, 1984) and available from the University of Wisconsin Genetics Computer Group (UWGCG).
  • the preferred default parameters for the GAP program include: (1) a unary comparison matrix (containing a value of 1 for identities and 0 for non-identities) for nucleotides, and the weighted comparison matrix of Gribskov and Burgess, Nucl. Acids Res. 14:6745, 1986, as described by Schwartz and Dayhoff, eds., Atlas of Protein Sequence and Structure , National Biomedical Research Foundation, pp. 353-358, 1979; (2) a penalty of 3.0 for each gap and an additional 0.10 penalty for each symbol in each gap; and (3) no penalty for end caps.
  • Variants may comprise conservatively substituted sequences, meaning that a given amino acid residue is replaced by a residue having similar physiochemical characteristics.
  • conservative substitutions include substitution of one aliphatic residue for another, such as Ile, Val, Leu, or Ala for one another, or substitutions of one polar residue for another, such as between Lys and Arg; Glu and Asp; or Gin and Asn. Other such conservative substitutions, for example, substitutions of entire regions having similar hydrophobicity characteristics, are well known.
  • Naturally occurring variants or alleles are also encompassed by the invention. Examples of such variants are proteins that result from alternate mRNA splicing events or from proteolytic cleavage of the DCSema protein, wherein the binding property is retained.
  • Alternate splicing of mRNA may yield a truncated but biologically active polypeptide, such as a naturally occurring soluble form of the protein, for example.
  • Variations attributable to proteolysis include, for example, differences in the N-termini upon expression in different types of host cells, due to proteolytic removal of one or more terminal amino acids from the DCSema polypeptide (generally from 1-5 terminal amino acids).
  • Example 3 describes the construction of a novel viral Semaphorin/Fc fusion (DCSema/Fc) protein that may be utilized in studying the biological characteristics of DCSema and their receptor distribution.
  • DCSema/Fc viral Semaphorin/Fc fusion
  • Other antibody Fc regions may be substituted for the human IgG1 Fc region described in Example 3.
  • Suitable Fc regions are those that can bind with high affinity to protein A or protein G, and include the Fc region of human IgG1 or fragments of the human or murine IgG1 Fc region, e.g., fragments comprising at least the hinge region so that interchain disulfide bonds will form.
  • the viral DCSema/Fc fusion protein offers the advantage of being easily purified. In addition, disulfide bonds form between the Fc regions of two separate fusion protein chains, creating dimers.
  • the soluble DCSema polypeptides of the present invention may be isolated and identified by separating intact cells that express the desired protein from the culture medium in which the cells grow, and then assaying the medium (supernatant) for the presence of the desired protein. Separation can be accomplished using standard separation techniques including, including centrifugation. The presence of the DCSema polypeptide in the medium indicates that the protein was secreted from the cells in its expected soluble form. Because the DCSema polypeptides of the present invention are secreted soluble polypeptides they possess many advantages over membrane bound proteins. Purification of the proteins from recombinant host cells is feasible, since the soluble proteins are secreted from the cells. Further, soluble proteins are generally more suitable for intravenous administration.
  • DCSema proteins comprising less than the entire secreted polypeptide are included in the invention, e.g. soluble fragments such as amino acids 52-543 of SEQ ID NO:2 that include the “semaphorin domain,” which is part of an active binding site, and amino acids 45-644 of SEQ ID NO:2 that include the secreted protein without the signal peptide, are included in the invention.
  • DCSema polypeptides may additionally comprise one of the heterologous signal peptides described below that is functional within the host cells employed.
  • the protein may comprise the native signal peptide.
  • DCSema polypeptides can be expressed as a fusion protein comprising (from N— to C-terminus) the yeast ⁇ -factor signal peptide, a FLAG® peptide described below and in U.S. Pat. No. 5,011,912, and soluble DCSema polypeptide consisting of amino acids 45 to 666 of SEQ ID NO:2.
  • This recombinant fusion protein is expressed in and secreted from yeast cells.
  • the FLAG® peptide facilitates purification of the protein, and subsequently may be cleaved from the DCSema using bovine mucosal enterokinase.
  • Truncated DCSema polypeptides may be prepared by any of a number of conventional techniques.
  • a desired DNA sequence may be chemically synthesized using techniques known per se.
  • DNA fragments also may be produced by restriction endonuclease digestion of a full length cloned DNA sequence, and isolated by electrophoresis on agarose gels.
  • Linkers containing restriction endonuclease cleavage site(s) may be employed to insert the desired DNA fragment into an expression vector, or the fragment may be digested at cleavage sites naturally present therein.
  • the well known polymerase chain reaction procedure also may be employed to amplify a DNA sequence encoding a desired protein fragment.
  • known mutagenesis techniques may be employed to insert a stop codon at a desired point, e.g., immediately downstream of the codon for the last amino acid of the binding domain.
  • the invention provides isolated or homogeneous DCSema polypeptides, both recombinant and non-recombinant.
  • Variants and derivatives of native DCSema proteins that retain the desired biological activity may be obtained by mutations of nucleotide sequences coding for the native polypeptides. Alterations of the native amino acid sequence may be accomplished by any of a number of conventional methods. Mutations can be introduced at particular loci by synthesizing oligonucleotides containing a mutant sequence, flanked by restriction sites enabling ligation to fragments of the native sequence. Following ligation, the resulting reconstructed sequence encodes an analog having the desired amino acid insertion, substitution, or deletion.
  • oligonucleotide-directed site-specific mutagenesis procedures can be employed to provide an altered gene wherein predetermined codons can be altered by substitution, deletion or insertion.
  • Exemplary methods of making the alterations set forth above are disclosed by Walder et al. ( Gene 42:133, 1986); Bauer et al. ( Gene 37:73, 1985); Craik ( BioTechniques , January 1985, 12-19); Smith et al. ( Genetic Engineering: Principles and Methods . Plenum Press, 1981); Kunkel ( Proc. Natl. Acad. Sci. USA 82:488, 1985); Kunkel et al. ( Methods in Enzymol. 154:367, 1987); and U.S. Pat. Nos. 4,518,584 and 4,737,462 all of which are incorporated by reference.
  • Native DCSema polypeptides may be modified to create DCSema derivatives by forming covalent or aggregative conjugates with other chemical moieties, such as glycosyl groups, lipids, phosphate, acetyl groups and the like.
  • Covalent derivatives of the polypeptides may be prepared by linking the chemical moieties to functional groups on DCSema amino acid side chains or at the N-terminus or C-terminus of a DCSema polypeptide.
  • conjugates of DCSema polypeptides or their fragments with other proteins or polypeptides such as by synthesis in recombinant culture as N-terminal or C-terminal fusions.
  • the conjugate may comprise a signal or leader polypeptide sequence (e.g. the ⁇ -factor leader of Saccharomyces) at the N-terminus of a polypeptide of the present invention.
  • the signal or leader peptide co-translationally or post-translationally directs transfer of the conjugate from its site of synthesis to a site inside or outside of the cell membrane or cell wall.
  • DCSema polypeptide fusion proteins can comprise peptides added to facilitate purification and identification of semaphorin receptors that bind semaphorins of the present invention, including VESPR (see copending application S/N08/958,598.
  • peptides include, for example, poly-His or the antigenic identification peptides described in U.S. Pat. No. 5,011,912 and in Hopp et al., Bio/Technology 6:1204, 1988.
  • the invention further includes DCSema polypeptides with or without associated native-pattern glycosylation.
  • DCSema polypeptide expressed in yeast or mammalian expression systems e.g., COS-7 cells
  • Expression of DCSema polypeptides in bacterial expression systems, such as E. coli provides non-glycosylated molecules.
  • N-gycosylation sites in the DCSema extracellular domain can be modified to preclude glycosylation, allowing expression of a reduced carbohydrate analog in mammalian and yeast expression systems.
  • N-glycosylation sites in eukaryotic polypeptides are characterized by an amino acid triplet Asn-X-Y, wherein X is any amino acid except Pro and Y is Ser or Thr.
  • the native human DC Sema of the present invention comprises 4 such amino acid triplets, at amino acids 105-107, 157-159, 258-260 and 602-604 of SEQ ID NO:2.
  • Appropriate substitutions, additions or deletions to the nucleotide sequence encoding these triplets will result in prevention of attachment of carbohydrate residues at the Asn side chain.
  • Known procedures for inactivating N-glycosylation sites in proteins include those described in U.S. Pat. No. 5,071,972 and EP 276,846, hereby incorporated by reference.
  • sequences encoding Cys residues that are not essential for biological activity can be altered to cause the Cys residues to be deleted or replaced with other amino acids, preventing formation of incorrect intramolecular disulfide bridges upon renaturation.
  • Other equivalents are prepared by modification of adjacent dibasic amino acid residues to enhance expression in yeast systems in which KEX2 protease activity is present.
  • EP 212,914 discloses the use of site-specific mutagenesis to inactivate KEX2 protease processing sites in a protein.
  • KEX2 protease processing sites are inactivated by deleting, adding or substituting residues to alter Arg-Arg, Arg-Lys, and Lys-Arg pairs to eliminate the occurrence of these adjacent basic residues. Lys-Lys pairings are considerably less susceptible to KEX2 cleavage, and conversion of Arg-Lys or Lys-Arg to Lys-Lys represents a conservative and preferred approach to inactivating KEX2 sites.
  • the DC sema of SEQ ID NO:2 contains 7 KEX2 protease processing sites at amino acids 123-124, 135, 136, 192, 193, 204-205, 460461, 474-475 and 475-476, Nucleic acid sequences within the scope of the invention include isolated DNA and RNA sequences that hybridize to DCSema nucleotide sequences disclosed herein under conditions of moderate or high stringency, and that encode biologically active DCSema. Conditions of moderate stringency, as defined by Sambrook et al. Molecular Cloning: A Laboratory Manual, 2 ed. Vol. 1, pp.
  • a DNA sequence may vary from that shown in SEQ ID NO:1 and still encode a polypeptide having the amino acid sequence of SEQ ID NO:2.
  • Such variant DNA sequences may result from silent mutations (e.g., occurring during PCR amplification), or may be the product of deliberate mutagenesis of a native sequence.
  • the invention provides equivalent isolated DNA sequences encoding biologically active human DCSema, selected from: (a) cDNA comprising the nucleotide sequence presented in SEQ ID NO: 1; (b) DNA capable of hybridization to a DNA of (a) under moderately stringent conditions and that encodes biologically active polypeptides and (c) DNA that is degenerate as a result of the genetic code to a DNA defined in (a), or (b) and that encodes biologically active human DCSema polypeptide. Polypeptides encoded by such DNA equivalent sequences are encompassed by the invention.
  • DNAs that are equivalent to the DNA sequence of SEQ ID NO:1 include those that hybridize under moderately and highly stringent conditions to the DNA sequence that encodes polypeptides comprising the sequence of SEQ ID NO:2.
  • proteins encoded by such DNA include, but are not limited to, polypeptide fragments and proteins that have inactivated N-glycosylation site(s), inactivated KEX2 protease processing site(s), or conservative amino acid substitution(s), as described above.
  • DCSema polypeptides encoded by DNA derived from other species, wherein the DNA will hybridize to the cDNA of SEQ ID NO:1, are also within the present invention.
  • DCSema polypeptide variants possessing the ability to bind semaphorin receptors may be identified by any suitable assay.
  • Biological activity of DCSema polypeptides of the present invention may be determined, for example, by competition for binding to the binding domain of semaphorin receptors, e.g. competitive binding assays, or for binding to a semaphorin receptor binding domain.
  • One type of a competitive binding assay for a DCSema polypeptide of the present invention uses a radiolabeled DCSema and intact semaphorin receptor-expressing cells. Instead of intact cells, one could substitute soluble semaphorin receptor:Fc fusion proteins bound to a solid phase through the interaction of a Protein A, Protein G or an antibody to the semaphorin receptor or Fc portions of the molecule, with the Fc region of the fusion protein.
  • Competitive binding assays can be performed following conventional methodology.
  • a soluble semaphorin receptor can be made to compete with an immobilized receptor for binding with a soluble semaphorin ligand.
  • a radiolabeled soluble semaphorin ligand can be antagonized by soluble VESPR in an assay for binding activity against a surface-bound semaphorin receptor.
  • Qualitative results can be obtained by competitive autoradiographic plate binding assays, or Scatchard plots may be utilized to generate quantitative results.
  • semaphorin binding proteins such as VESPR or anti-semaphorin antibodies
  • a solid phase such as a column chromatography matrix or a similar substrate suitable for identifying, separating or purifying cells that express semaphorin on their surface.
  • Binding of a semaphorin-binding protein to a solid phase contacting surface can be accomplished by any means, for example, by constructing a VESPR:Fc fusion protein and binding such to the solid phase through the interaction of Protein A or Protein G.
  • Various other means for fixing proteins to a solid phase are well known in the art and are suitable for use in the present invention.
  • magnetic microspheres can be coated with VESPR and held in the incubation vessel through a magnetic field. Suspensions of cell mixtures containing semaphorin-expressing cells are contacted with the solid phase that has VESPR polypeptides thereon. Cells having semaphorin on their surface bind to the fixed VESPR and unbound cells then are washed away.
  • This affinity-binding method is useful for purifying, screening or separating such semaphorin-expressing cells from solution.
  • Methods of releasing positively selected cells from the solid phase are known in the art and encompass, for example, the use of enzymes. Such enzymes are preferably non-toxic and non-injurious to the cells and are preferably directed to cleaving the cell-surface binding partner.
  • the enzyme preferably would cleave the semaphorin, thereby freeing the resulting cell suspension from the “foreign” semaphorin receptor material.
  • the purified cell population then may be used to repopulate mature (adult) tissues.
  • mixtures of cells suspected of containing semaphorin cells first can be incubated with a suitable biotinylated semaphorin receptor, e.g. VESPR. Incubation periods are typically at least one hour in duration to ensure sufficient binding to semaphorin
  • the resulting mixture then is passed through a column packed with avidin-coated beads, whereby the high affinity of biotin for avidin provides the binding of the cell to the beads.
  • avidin-coated beads is known in the art. See Berenson, et al. J. Cell. Biochem., 10D:239 (1986). Washing unbound material and releasing the bound cells is performed using conventional methods.
  • cells expressing DCSema polypeptides of the present invention can be separated using a semaphorin receptor, e.g. VESPR.
  • VESPR semaphorin receptor
  • other suitable semaphorin receptors, or an extracellular domain or a fragment thereof can be conjugated to a detectable moiety such as 125 I to detect semaphorin-expressing cells.
  • Radiolabeling with 125 I can be performed by any of several standard methodologies that yield a functional 125 I-molecule labeled to high specific activity or an iodinated or biotinylated antibody against the semaphorin receptor.
  • Another detectable moiety such as an enzyme that can catalyze a colorimetric or fluorometric reaction, biotin or avidin may be used.
  • Cells to be tested for DCSema polypeptide-expression can be contacted with a suitable labeled receptor, e.g. VESPR. After incubation, unbound labeled receptor is removed and binding is measured using the detectable moiety.
  • a suitable labeled receptor e.g. VESPR.
  • the binding characteristics of DCSema polypeptides may also be determined using a conjugated semaphorin receptor (for example, 125 I-semaphorin receptor:Fc) in competition assays similar to those described above.
  • a conjugated semaphorin receptor for example, 125 I-semaphorin receptor:Fc
  • intact cells expressing DCSema polypeptide of the present invention, bound to a solid substrate are used to measure the extent to which a sample containing a putative receptor variant competes for binding with a conjugated DCSema.
  • DCSema polypeptides of the present invention include the use of anti-DCSema antibodies, cell lines that proliferate in response to DCSema polypeptide, or recombinant cell lines that express DCSema and proliferate in the presence of a suitable semaphorin receptor.
  • the DCSema proteins disclosed herein also may be employed to measure the biological activity of any semaphorin receptor in terms of its binding affinity for its semaphorin ligand.
  • DCSema polypeptides of the present invention may be used in determining whether biological activity is retained after modification of a semaphorin receptor (e.g., chemical modification, truncation, mutation, etc.). The biological activity of a semaphorin receptor thus can be ascertained before it is used in a research study, or in the clinic, for example.
  • DCSema polypeptides disclosed herein find use as reagents in “quality assurance” studies, e.g., to monitor shelf life and stability of a receptor to which the DCSema binds under different conditions.
  • DCSema polypeptides of the present invention may be employed in a binding affinity study to measure the biological activity of a test semaphorin receptor that has been stored at different temperatures, or produced in different cell types. The binding affinity of the DCSema protein for the test receptor is compared to that of a standard or control semaphorin receptor to detect any adverse impact on biological activity of the test semaphorin receptor.
  • DCSema polypeptides described herein also find use as carriers for delivering agents attached thereto to cells expressing semaphorin receptors to which the semaphorin binds.
  • VESPR to which a DCSema of the present invention binds, is expressed in lung epithelial cells, stroma, intestinal epithelial cells and lymphoma cells.
  • DCSema polypeptides of the present invention can thus can be used to deliver diagnostic or therapeutic agents to these cells (or to other cell types found to express a suitable semaphorin receptor on cell surfaces) in in vitro or in vivo procedures.
  • Diagnostic and therapeutic agents that may be attached to a DCSema polypeptide of the present invention include, but are not limited to, drugs, toxins, radionuclides, chromophores, enzymes that catalyze a colorimetric or fluorometric reaction, and the like, with the particular agent being chosen according to the intended application.
  • drugs include those used in treating various forms of cancer, e.g., nitrogen mustards such as L-phenylalanine nitrogen mustard or cyclophosphamide, intercalating agents such as cis-diaminodichloroplatinum, antimetabolites such as 5-fluorouracil, vinca alkaloids such as vincristine, and antibiotics such as bleomycin, doxorubicin, daunorubicin, and derivatives thereof.
  • nitrogen mustards such as L-phenylalanine nitrogen mustard or cyclophosphamide
  • intercalating agents such as cis-diaminodichloroplatinum
  • antimetabolites such as 5-fluorouracil
  • vinca alkaloids such as vincristine
  • antibiotics such as bleomycin, doxorubicin, daunorubicin, and derivatives thereof.
  • Radionuclides suitable for diagnostic use include, but are not limited to, 123 I, 131 I, 99m Tc, 111 In, and 76 Br.
  • Radionuclides suitable for therapeutic use include, but are not limited to, 131 I, 211 At. 77 Br, 186 Re, 188 Re, 212 Pb, 212 Bi, 109 Pd, 64 Cu, and 67 Cu.
  • Such agents may be attached to the DCSema of the present invention by any suitable conventional procedure.
  • Semaphorin homologues of the present invention being proteins, include functional groups on amino acid side chains that can be reacted with functional groups on a desired agent to form covalent bonds, for example.
  • the protein or agent may be derivatized to generate or attach a desired reactive functional group.
  • the derivatization may involve attachment of one of the bifunctional coupling reagents available for attaching various molecules to proteins (Pierce Chemical Company, Rockford, Ill.). A number of techniques for radiolabeling proteins are known. Radionuclide metals may be attached to the receptor by using a suitable bifunctional chelating agent, for example.
  • Conjugates comprising molecules of the present invention and a suitable diagnostic or therapeutic agent (preferably covalently linked) are thus prepared. The conjugates are administered or otherwise employed in an amount appropriate for the particular application.
  • DCSema polypeptides of the present invention is as a research tool for studying the role that the DCSema, in conjunction with semaphorin receptors to which it binds, may play in immune regulation and viral infection.
  • the polypeptides of the present invention also may be employed in in vitro assays for detection of receptors to which it binds, e.g. VESPR, or the interactions thereof.
  • DCSema polypeptides of the present invention can be used as a research tool for studying the role that the ligand, in conjunction with its receptors, play in immune regulation.
  • VESPR a semaphorin•receptor
  • DCSema ligands including the DC Sema of the present invention, in the treatment of autoimmune disorders is expected to diminish unwanted symptoms associated with the autoimmune disorder by downregulating the antigen presenting capabilities of mature dendritic cells.
  • DCSema polypeptides of the invention can be formulated according to known methods used to prepare pharmaceutically useful compositions.
  • the molecules of the invention can be combined in admixture, either as the sole active material or with other known active materials, with pharmaceutically suitable diluents (e.g., Tris-HCl, acetate, phosphate), preservatives (e.g., Thimerosal, benzyl alcohol, parabens), emulsifiers, solubilizers, adjuvants and/or carriers.
  • diluents e.g., Tris-HCl, acetate, phosphate
  • preservatives e.g., Thimerosal, benzyl alcohol, parabens
  • emulsifiers e.g., solubilizers, adjuvants and/or carriers.
  • solubilizers solubilizers
  • adjuvants and/or carriers e.g., solubilizers, adj
  • compositions can contain DCSema polypeptide complexed with polyethylene glycol (PEG), metal ions, or incorporated into polymeric compounds such as polyacetic acid, polyglycolic acid, hydrogels, etc., or incorporated into liposomes, microemulsions, micelles, unilamellar or multilamellar vesicles, erythrocyte ghosts or spheroblasts.
  • PEG polyethylene glycol
  • metal ions or incorporated into polymeric compounds such as polyacetic acid, polyglycolic acid, hydrogels, etc.
  • liposomes such as polyacetic acid, polyglycolic acid, hydrogels, etc.
  • Such compositions will influence the physical state, solubility, stability, rate of in vivo release, and rate of in vivo clearance of DCSema.
  • DCSema polypeptides described herein can be conjugated to antibodies against tissue-specific receptors, ligands or antigens, or coupled to lig
  • DCSema polypeptides of the present invention can be administered topically, parenterally, or by inhalation.
  • parenteral includes subcutaneous injections, intravenous, intramuscular, intracisternal injection, or infusion techniques. These compositions will typically contain an effective amount of the polypeptide, alone or in combination with an effective amount of any other active material. Such dosages and desired drug concentrations contained in the compositions may vary depending upon many factors, including the intended use, patient's body weight and age, and route of administration. Preliminary doses can be determined according to animal tests, and the scaling of dosages for human administration can be performed according to art-accepted practices.
  • Polypeptides of the present invention may exist as oligomers, such as covalently-linked or non-covalently-linked dimers or trimers. Oligomers may be linked by disulfide bonds formed between cysteine residues on different DCSema molecules.
  • a dimer is created by fusing a DCSema to the Fc region of an antibody (e.g., IgG1) in a manner that does not interfere with binding of the DCSema to a semaphorin receptor-binding domain.
  • the Fc polypeptide preferably is fused to the C-terminus of a DCSema
  • a DCSema General preparation of fusion proteins comprising heterologous polypeptides fused to various portions of antibody-derived polypeptides (including the Fc domain) has been described, e.g., by Ashkenazi et al. ( PNAS USA 88:10535, 1991) and Byrn et al. ( Nature 344:677, 1990), hereby incorporated by reference.
  • a gene fusion encoding the DCSema/Fc fusion protein is inserted into an appropriate expression vector.
  • DCSema/Fc fusion proteins are allowed to assemble much like antibody molecules, whereupon interchain disulfide bonds form between Fc polypeptides, yielding divalent.
  • fusion proteins are made with both heavy and light chains of an antibody, it is possible to form a DCSema oligomer with as many as four semaphorin regions. Alternatively, one can link two DCSemas with a peptide linker.
  • Suitable host cells for expression of DCSema polypeptides of this invention include prokaryotes, yeast or higher eukaryotic cells.
  • Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, and mammalian cellular hosts are described, for example, in Pouwels et al. Cloning Vectors: A Laboratory Manual , Elsevier, N.Y., (1985). Cell-free translation systems could also be employed to produce the polypeptides of the present invention using RNAs derived from DNA constructs disclosed herein.
  • Prokaryotes include gram negative or gram positive organisms, for example, E. coli or Bacilli. Suitable prokaryotic host cells for transformation include, for example, E. coli, Bacillus subtilis, Salmonella typhimurium , and various other species within the genera Pseudomonas, Streptomyces, and Staphylococcus.
  • a semaphorin receptor polypeptide may include an N-terminal methionine residue to facilitate expression of the recombinant polypeptide in the prokaryotic host cell. The N-terminal Met may be cleaved from the expressed recombinant DCSema polypeptide.
  • DCSema polypeptides may be expressed in yeast host cells, preferably from the Saccharomyces genus (e.g., S. cerevisiae ). Other genera of yeast, such as Pichia, K. lactis or Kluyveromyces, may also be employed.
  • yeast vectors will often contain an origin of replication sequence from a 2 ⁇ yeast plasmid, an autonomously replicating sequence (ARS), a promoter region, sequences for polyadenylation, sequences for transcription termination, and a selectable marker gene.
  • Suitable promoter sequences for yeast vectors include, among others, promoters for metallothionein. 3-phosphoglycerate kinase (Hitzeman et al., J. Biol.
  • glycolytic enzymes Hess et al., J. Adv. Enzyme Reg. 7:149, 1968; and Holland et al., Biochem. 17:4900, 1978
  • enolase glyceraldehyde-3-phosphate dehydrogenase
  • hexokinase hexokinase
  • pyruvate decarboxylase phosphofructokinase
  • glucose-6-phosphate isomerase 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase.
  • yeast vectors and promoters for use in yeast expression are further described in Hitzeman, EPA-73,657 or in Fleer et. al., Gene, 107:285-195 (1991); and van den Berg et. al., Bio/technology, 8:135-139 (1990).
  • Another alternative is the glucose-repressible ADH2 promoter described by Russell et al. ( J. Biol. Chem. 258:2674, 1982) and Beier et al. ( Nature 300:724, 1982).
  • Shuttle vectors replicable in both yeast and E. coli may be constructed by inserting DNA sequences from pBR322 for selection and replication in E. coli (Amp r gene and origin of replication) into the above-described yeast vectors.
  • the yeast ⁇ -factor leader sequence may be employed to direct secretion of the VESPR or DCSema polypeptide.
  • the ⁇ -factor leader sequence is often inserted between the promoter sequence and the structural gene sequence. See, e.g., Kurjan et al., Cell 30:933, 1982: Bitter et al., Proc. Natl. Acad. Sci. USA 81:5330, 1984; U.S. Pat. No. 4,546,082; and EP 324,274.
  • Other leader sequences suitable for facilitating secretion of recombinant polypeptides from yeast hosts are known to those of skill in the art.
  • a leader sequence may be modified near its 3′ end to contain one or more restriction sites. This will facilitate fusion of the leader sequence to the structural gene.
  • Yeast transformation protocols are known to those of skill in the art.
  • One such protocol is described by Hinnen et al., Proc. Natl. Acad. Sci. USA 75:1929, 1978.
  • the Hinnen et al. protocol selects for Trp + transformants in a selective medium, wherein the selective medium consists of 0.67% yeast nitrogen base, 0.5% casamino acids, 2% glucose. 10 ⁇ g/ml adenine and 20 ⁇ g/ml uracil.
  • Yeast host cells transformed by vectors containing ADH2 promoter sequence may be grown for inducing expression in a “rich” medium.
  • a rich medium is one consisting of 1% yeast extract, 2% peptone, and 1% glucose supplemented with 80 ⁇ g/ml adenine and 80 ⁇ g/ml uracil. Depression of the ADH2 promoter occurs when glucose is exhausted from the medium.
  • Mammalian or insect host cell culture systems could also be employed to express recombinant polypeptides of the present invention.
  • Baculovirus systems for production of heterologous proteins in insect cells are reviewed by Luckow and Summers, Bio/Technology 6:47 (1988).
  • Established cell lines of mammalian origin also may be employed. Examples of suitable mammalian host cell lines include the COS-7 line of monkey kidney cells (ATCC CRL 1651) (Gluzman et al., Cell 23:175, 1981), L cells, C127 cells, 3T3 cells (ATCC CCL 163), Chinese hamster ovary (CHO) cells.
  • Transcriptional and translational control sequences for mammalian host cell expression vectors may be excised from viral genomes. Commonly used promoter sequences and enhancer sequences are derived from Polyoma virus. Adenovirus 2, Simian Virus 40 (SV40), and human cytomegalovirus. DNA sequences derived from the SV40 viral genome, for example, SV40 origin, early and late promoter, enhancer, splice, and polyadenylation sites may be used to provide other genetic elements for expression of a structural gene sequence in a mammalian host cell.
  • SV40 Simian Virus 40
  • DNA sequences derived from the SV40 viral genome for example, SV40 origin, early and late promoter, enhancer, splice, and polyadenylation sites may be used to provide other genetic elements for expression of a structural gene sequence in a mammalian host cell.
  • Viral early and late promoters are particularly useful because both are easily obtained from a viral genome as a fragment which may also contain a viral origin of replication (Fiers et al., Nature 273:113, 1978). Smaller or larger SV40 fragments may also be used, provided the approximately 250 bp sequence extending from the Hind III site toward the Bgl I site located in the SV40 viral origin of replication site is included.
  • Exemplary expression vectors for use in mammalian host cells can be constructed as disclosed by Okayama and Berg (Mol. Cell. Biol. 3:280, 1983).
  • a useful system for stable high level expression of mammalian cDNAs in C127 murine mammary epithelial cells can be constructed substantially as described by Cosman et al. ( Mol. Immunol. 23:935, 1986).
  • a useful high expression vector, PMLSV N1/N4 described by Cosman et al., Nature 312:768, 1984 has been deposited as ATCC 39890. Additional useful mammalian expression vectors are described in EP-A-0367566, and in U.S. patent application Ser. No.
  • the vectors may be derived from retroviruses.
  • a heterologous signal sequence may be added, such as the signal sequence for IL-7 described in U.S. Pat. No. 4,965,195; the signal sequence for IL-2 receptor described in Cosman et al., Nature 312:768 (1984); the IL-4 signal peptide described in EP 367,566; the type I IL-1 receptor signal peptide described in U.S. Pat. No. 4,968,607; and the type II IL-1 receptor signal peptide described in EP 460,846.
  • DCSema polypeptides of the present invention as isolated, purified or homogeneous proteins may be produced by recombinant expression systems as described above or purified from naturally occurring cells.
  • the polypeptides can be purified to substantial homogeneity, as indicated by a single protein band upon analysis by SDS-polyacrylamide gel electrophoresis (SDS-PAGE).
  • One process for producing polypeptides of the present invention comprises culturing a host cell transformed with an expression vector comprising a DNA sequence that encodes the desired DCSema polypeptide under conditions sufficient to promote expression of the DCSema polypeptide.
  • the DCSema polypeptide is then recovered from culture medium or cell extracts, depending upon the expression system employed.
  • procedures for purifying a recombinant protein will vary according to such factors as the type of host cells employed and whether or not the recombinant protein is secreted into the culture medium.
  • the culture medium first may be concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit.
  • a purification matrix such as a gel filtration medium.
  • an anion exchange resin can be employed, for example, a matrix or substrate having pendant diethylaminoethyl (DEAE) groups.
  • the matrices can be acrylamide, agarose, dextran, cellulose or other types commonly employed in protein purification.
  • a cation exchange step can be employed.
  • Suitable cation exchangers include various insoluble matrices comprising sulfopropyl or carboxymethyl groups. Sulfopropyl groups are preferred.
  • one or more reversed-phase high performance liquid chromatography (RP-HPLC) steps employing hydrophobic RP-HPLC media, (e.g., silica gel having pendant methyl or other aliphatic groups) can be employed to further purify the polypeptide.
  • RP-HPLC reversed-phase high performance liquid chromatography
  • an affinity column comprising the receptor-binding domain of a DCSema of the present invention affinity-purify semaphorin receptor to which the DCSema binds.
  • a receptor can be removed from an affinity column using conventional techniques, e.g., in a high salt elution buffer and then dialyzed into a lower salt buffer for use or by changing pH or other components depending on the affinity matrix utilized.
  • Recombinant protein produced in bacterial culture can be isolated by initial disruption of the host cells, centrifugation, extraction from cell pellets if an insoluble polypeptide, or from the supernatant fluid if a soluble polypeptide, followed by one or more concentration, salting-out, ion exchange, affinity purification or size exclusion chromatography steps. Finally, RP-HPLC can be employed for final purification steps. Microbial cells can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents.
  • Transformed yeast host cells are preferably employed to express DCSemas of the present invention as a secreted polypeptide in order to simplify purification.
  • Secreted recombinant polypeptide from a yeast host cell fermentation can be purified by methods analogous to those disclosed by Urdal et al. ( J. Chromatog. 296:171, 1984). Urdal et al. describe two sequential, reversed-phase HPLC steps for purification of recombinant human IL-2 on a preparative HPLC column.
  • Useful fragments of the DCSema nucleic acids of the present invention include antisense or sense oligonucleotides comprising a single-stranded nucleic acid sequence (either RNA or DNA) capable of binding to target DCSema mRNA (sense) or DCSema DNA (antisense) sequences.
  • Antisense or sense oligonucleotides comprise a fragment of the coding region of DCSema cDNA. Such a fragment generally comprises at least about 14 nucleotides, preferably from about 14 to about 30 nucleotides.
  • binding of antisense or sense oligonucleotides to target nucleic acid sequences results, in the formation of duplexes that block transcription or translation of the target sequence by one of several means, including enhanced degradation of the duplexes, premature termination of transcription or translation, or by other means.
  • the antisense oligonucleotides thus may be used to block expression of proteins of the present invention.
  • Antisense or sense oligonucleotides further comprise oligonucleotides having modified sugar-phosphodiester backbones (or other sugar linkages, such as those described in WO91/06629) and wherein such sugar linkages are resistant to endogenous nucleases.
  • Such oligonucleotides with resistant sugar linkages are stable in vivo (i.e., capable of resisting enzymatic degradation) but retain sequence specificity to be able to bind to target nucleotide sequences.
  • Other examples of sense or antisense oligonucleotides include those oligonucleotides which are covalently linked to organic moieties, such as those described in WO 90/10448, and other moieties that increases affinity of the oligonucleotide for a target nucleic acid sequence, such as poly-(L-lysine).
  • intercalating agents such as ellipticine, and alkylating agents or metal complexes may be attached to sense or antisense oligonucleotides to modify binding specificities of the antisense or sense oligonucleotide for the target nucleotide sequence.
  • Antisense or sense oligonucleotides may be introduced into a cell containing the target nucleic acid sequence by any gene transfer method, including, for example, CaPO 4 -mediated DNA transfection, electroporation, or by using gene transfer vectors such as Epstein-Barr virus.
  • Antisense or sense oligonucleotides are preferably introduced into a cell containing the target nucleic acid sequence by insertion of the antisense or sense oligonucleotide into a suitable retroviral vector, then contacting the cell with the retrovirus vector containing the inserted sequence, either in vivo or ex vivo.
  • Suitable retroviral vectors include, but are not limited to, those derived from the murine retrovirus M-MuLV, N2 (a retrovirus derived from M-MuLV), or the double copy vectors designated DCT5A, DCT5B and DCT5C (see PCT Application US 90/02656).
  • Sense or antisense oligonucleotides also may be introduced into a cell containing the target nucleotide sequence by formation of a conjugate with a ligand binding molecule, as described in WO 91/04753.
  • Suitable ligand binding molecules include, but are not limited to, cell surface receptors, growth factors, other cytokines, or other ligands that bind to cell surface receptors.
  • conjugation of the ligand binding molecule does not substantially interfere with the ability of the ligand binding molecule to bind to its corresponding molecule or receptor, or block entry of the sense or antisense oligonucleotide or its conjugated version into the cell.
  • a sense or an antisense oligonucleotide may be introduced into a cell containing the target nucleic acid sequence by formation of an oligonucleotide-lipid complex, as described in WO 90/10448.
  • the sense or antisense oligonucleotide-lipid complex is preferably dissociated within the cell by an endogenous lipase.
  • EST 151129 is a partial sequence having neither an initiating ATG nor a termination codon. EST 151129 has sequence homology to A39R and AHVsema.
  • EST 151129 was utilized to isolate and identify the native human DCSema as follows:
  • a tissue source of EST 151129 was identified using phage library screening methods and PCR primers based upon this EST sequence. Oligonucleotide PCR primers having the following nucleotide sequences were synthesized: 5′-TGCTGGAACCTGGTGAATGG-3′ (SEQ ID NO:3) 5′-AGTGGAACAATGGCGTCTTC-3′ (SEQ ID NO:4)
  • PCR isolation and amplification methodologies were carried out using a panel of human tissue cDNA phase libraries as templates for the PCR reactions.
  • phage libraries Two of the phage libraries, human foreskin fibroblast and dermal fibroblast, were chosen for additional analysis.
  • the libraries were plated according to established procedures.
  • a radiolabeled probe was generated by incorporating 32 P-dCTP in the amplification of a PCR product using the EST 151129 as a template.
  • the PCR reaction mixture included 10 ng of EST151129 plasmid DNA, 50 pmoles of each PCR oligonucleotide primer identified below and specific for the 5′ end of EST 151129, 1 ⁇ Amplitaq buffer (Perkin-Elmer Cetus), 1.25 mM of dATP, dGTP, dTTP, 0.001 mM dCTP (Pharmacia,), 150 ⁇ Ci 32 P-dCTP, 0.5 ⁇ l Amplitaq taq polymerase (Perkin-Elmer Cetus,) in a 100 ⁇ l final reaction volume.
  • the PCR reaction cycles included one cycle at 94° C. for 5 min; twenty-five cycles at 94° C. for 1 min, 72° C. for 2 min and one cycle at 72° C. for 5 min using a Robocycler Gradient 40 (Stratagene, La Jolla. CA).
  • the primers used were as follows: 5′-TCCGCCCAGGGCCACCTAAGGAGCGGA-3′ (SEQ ID NO:7) 5′-TGTGCGGCTCAGTCTGGCCAAAGTCCA-3′ (SEQ ID NO:8)
  • EST 151129 sequence included nucleotides 115-1536, encoding amino acids 39-512 of the full length peptide.
  • the isolated cDNA provided nucleotides 1-114 and additional upstream non-coding nucleotides. These added nucleotides encode amino acids 1-38 of the full length human semaphorin, amino acid 1 being the initiating methionine.
  • Radiolabeled probes derived from SEQ ID NO:1 were used in Northern blot analyses of different human tissues to identify the tissue distribution of the human homologue of AHV Sema, designated DCSema.
  • the primers sequences were as follows: 5′ Primer: TCTACTACTT CTTCC (SEQ ID NO:5) 3′ Primer GGAATCCTAA TACGACTCAC TATAGGGAGG C GGGTTGGGA AGGC (SEQ ID NO:6)
  • the underlined portion of the downstream primer is a T7 site.
  • the riboprobe was generated using Ambion's MAXIscript SP6/T7 kit by combining 3 ⁇ L of RNAse free water, 2 ⁇ L 10 ⁇ transcription buffer, 1 ⁇ L each of 10 mM dATP/dCTP/dGTP, 5 ⁇ L 5′/3′ EST 151129 PCR Product, 5 ⁇ L Amersham [ ⁇ ] 32 P]UTP 10 mCi/mL, and 2 ⁇ L T7 RNA polymerase at room temperature. The combination was microfuged, spun briefly, and incubated at 37° C. for 30 minutes. Then 1 ⁇ L DNAse was added to the mixture and allowed to react for 15 minutes at 37° C. The reaction product was passed through two column volumes of G-25 (Boehringer). One microliter (1 ⁇ L) of the riboprobe was counted in a scintillation counter for 1 minute to determine cpm/ ⁇ L.
  • DC-Sema/Fc DNA encoding DC-Sema/Fc included a nucleotide sequence that encodes a murine IL-7 leader peptide, a FLAGTM octapeptide (described in U.S. Pat. No. 5,011,912), an Fc region of an immunoglobulin mutated to minimize binding to Fc receptor (described by Baum et al. Cir. Sh. 44:30, 1994), a flexible linker sequence and DNA encoding amino acids 45-666 of SEQ ID NO:2.
  • An expression vector containing the leader sequence, FLAG, mutated hu IgG Fc and flexible linker was prepared using conventional enzyme cutting and ligation techniques. The resulting vector was then restricted with Spe1 and Not1. The DC-Sema was inserted 5′ to 3′ after the flexible linker in a two-way ligation described below.
  • DC-Sema DNA To prepare the DC-Sema DNA, three primer pairs were designed and used to amplify three DNA fragments. Two of the fragments were amplified from a human foreskin fibroblast library phage clone containing the 3′ portion of the DCSema cDNA and one fragment was EST DNA (Research Genetics Imageclone ID #151129). The three amplified DNA fragments were combined in a PCR SOEing reaction to generate a DNA fragment encoding the entire peptide of DC-Sema (Horton et al., Biotechniques 8:528, 1990).
  • the upstream oligonucleotide primer introduced a Spe1 site upstream of amino acid 45 of the DC-sema peptide.
  • a downstream oligonucleotide primer introduced a Not1 site just downstream of the termination codon after amino acid 666.
  • the PCR fragment was then ligated into an expression vector (pDC409) containing the leader sequence.
  • Flag® sequence mutated human IgG Fc and a flexible linker region in a two-way ligation.
  • the resulting DNA construct was transfected into the monkey kidney cell lines CV-1/EBNA. After 7 days of culture in medium containing 0.5% low immunoglobulin bovine serum, a solution of 0.2% azide was added to the supernatant and the supernatant was filtered through a 0.22 ⁇ m filter.
  • DC-sema/polyHIS fusion protein DNA encoding DC-sema/polyHIS comprises sequences encoding the DC-sema gene from amino acid 1 to amino acid 666 (no stop codon), followed by a factor Xa cleavage site, a FLAG® octapeptide, a string of six histidine residues and a stop codon.
  • An expression vector containing the factor Xa cleavage site, flag, and polyHIS sequence was prepared using conventional enzyme cutting and fragment ligation techniques. The resulting vector was then restricted with Sal1 and SnaB 1.
  • the DCSema sequence was inserted 5′ to 3′ before the factor Xa cleavage site in a two-way ligation described below.
  • DCSema DNA To prepare the DCSema DNA, three primer pairs were designed and used to amplify three DNA fragments. Two of the fragments were from phage DNA and one fragment was EST DNA (Research Genetics Imageclone ID #151129). The three amplified DNA fragments were combined in a PCR SOEing reaction to generate a DNA fragment encoding the entire peptide of DCSema. This final DNA included a Sal1 site upstream of amino acid 1 of the DCSema peptide and a SnaB1 site downstream of amino acid 666.
  • the PCR product was then ligated into an expression vector pDC409 containing the factor Xa cleavage site, flag, and polyHIS sequence in a two-way ligation.
  • the resultant DNA construct (DCSema/polyHIS) was transiently transfected into the monkey cell line COS-1 (ATCC CRL-1650). Following a 7 day culture in medium containing 0.5% low immunoglobulin bovine serum, cell supernatants were harvested and a solution of 0.2% sodium azide was added to the supernatants.
  • the supernatants were filtered through a 0.22 ⁇ m filter, concentrated 10 fold with a prep scale concentrator (Millipore: Bedford, Mass.) and purified on a BioCad HPLC protein purification equipped with a Nickel NTA Superflow self pack resin column (Qiagen, Santa Clarita, Calif.). After the supernatant passed through the column, the column was washed with Buffer A (20 mM NaPO4, pH7.4; 300 mMNaCl; 50 mM Imidazole). Bound protein was then eluted from the column using a gradient elution techniques. Fractions containing protein were collected and analyzed on a 4-20% SDS-PAGE reducing gel. Peaks containing the fusion protein were pooled, concentrated 2 fold, and then dialyzed in PBS. The resulting DCSema/polyHis fusion protein was then filtered through a 0.22 ⁇ m sterile filter and recovered.
  • Buffer A (20 mM NaPO4, pH
  • the DC-Sema/Fc fusion protein prepared as described in Example 3 was used to screen cell lines for binding using quantitative binding studies according to standard flow cytometry methodologies. For each cell line screened, the procedure involved incubating approximately 100,000 of the cells blocked with 2% FCS (fetal calf serum), 5% normal goat serum and 5% rabbit serum in PBS for 1 hour. Then the blocked cells were incubated with 5 ⁇ g/mL of DC-Sema/Fc fusion protein in 2% FCS, 5% goat serum and 5% rabbit serum in PBS.
  • FCS fetal calf serum
  • rabbit serum fetal calf serum
  • This example illustrates a method for preparing antibodies to DC-Sema.
  • Purified DC-Sema/Fc is prepared as described in Example 3 above.
  • the purified protein is used to generate antibodies against DC-Sema as described in U.S. Pat. No. 4,411,993. Briefly, mice are immunized at 0, 2 and 6 weeks with 10 ⁇ g with DCSema/Fc.
  • the primary immunization is prepared with TITERMAX adjuvant, from Vaxcell, Inc., and subsequent immunizations are prepared with incomplete Freund's adjuvant (IFA).
  • IFA incomplete Freund's adjuvant
  • the mice are IV boosted with 3-4 ⁇ g DCSema/Fc in PBS.
  • splenocytes are harvested and fused with an Ag8.653 myeloma fusion partner using 50% aqueous PEG 1500 solution.
  • Hybridoma supernatants are screened for DC-Sema antibodies by dot blot assay against DCSema/FC and an irrelevant Fc protein.
  • Freshly isolated human monocytes were purified by first diluting 1:1 peripheral blood from healthy donors in low endotoxin PBS at pH 7.4 and room temperature. Then 35 mLs of the diluted blood was layered over 15 mLs of Isolymph (Gallard and Schlesinger Industries, Inc; Carle Place, N.Y.) and centrifuged at 2200 rpm for 25 minutes at room temperature. The plasma layers was reserved. The PBMC layer was harvested and washed three times to remove the Isolymph. The washed PBMC's were resuspended in X-Vivo 15 serum free media (BioWhittaker, Walkersville, Md.) and added to T175 flasks.
  • the flasks had been previously coated with 2% Gelatin (Sigma, St. Louis, Mo.) and pre-treated for 30 minutes with the reserved plasma layer.
  • the PBMC's were allowed to adhere for 90 minutes at 37° C., 5% CO 2 and then rinsed three times gently with 10 mL washes of low endotoxin PBS.
  • Adhered monocytes were harvested by incubating the cells in Enzyme Free Dissociation Buffer (Gibco, BRL) and washing the cells multiple times in PBS. Monocytes were centrifuged at 2500 rpm for 5 minutes, counted, and set up in 24 well dishes at 5 ⁇ 10 5 cells/well in 1 mL. The cultures were 95% pure.
  • Purified monocytes were cultured for 7-9 days in the presence of 20 ng/mL GM-CSF and 100 ng/mL IL-4 in order to allow cells to differentiate to a more dendritic cell-like phenotype. On day 7-9, cultures were treated with 1 ⁇ g/mL DCSema/Fc fusion protein (see Example 3) or a control Fc protein, and the next day cells and supernatants were harvested for analyses.
  • IL-6 and IL-8 were induced by DCSema protein. Heat inactivated DCSema and control proteins did not induce IL-6 or IL-8. Additionally, cytokine production was blocked by the inclusion of a mAb directed against DCSema.
  • monocytes were purified as described in Example 7 and DC-Sema/Fc fusion protein was prepared as described in Example 3. After incubating the DCSema/Fc fusion protein and purified, cultured monocytes for 20 hours, monocyte aggregation was observed.

Abstract

The invention is directed to semaphorin polypeptide as a purified and isolated protein, the DNA encoding the semaphorin polypeptide, host cells transfected with cDNAs encoding the polypeptide and methods for preparing the semaphorin polypeptide.

Description

    FIELD OF THE INVENTION
  • The present invention relates to semaphorin polypeptides, nucleic acids encoding such semaphorin polypeptides, processes for producing recombinant semaphorin polypeptides, pharmaceutical compositions containing such polypeptides relates and processes for treating disorders associated with semaphorin activity. [0001]
  • BACKGROUND OF THE INVENTION
  • The semaphorin gene family includes a large number of molecules that encode related transmembrane and secreted glycoproteins known to be neurologic regulators. The semaphorins are generally well conserved in their extracellular domains which are typically about 500 amino acids in length. Semaphorin family proteins have been observed in neuronal and nonneuronal tissue and have been studied largely for their role in neuronal growth cone guidance. For example, the secreted semaphorins known as collapsin-1 and Drosophila semaphorin II are selectively involved in repulsive growth cone guidance during development. Flies having semaphorin II genes that are mutated so that their function is reduced exhibit abnormal behavior characteristics. [0002]
  • Another semaphorin gene has been identified in several strains of poxvirus. This semaphorin is found in vaccinia virus (Copenhagen strain) and Ectromelia virus, and is encoded in an open reading frame (ORF) known as A39R. The A39R encoded protein has no transmembrane domain and no potential membrane linkage and is known to be a secreted protein. Variola virus ORF also contains sequences that share homology with the vaccinia virus ORF A39R at the nucleotide level and the amino acid level. Another viral semaphorin. AHV-sema, has been found in the Alcelaphine Herpesvirus (AHV). [0003]
  • Genes encoding mammalian (human, rat, and mouse) semaphorins have been identified, based upon their similarity to insect semaphorins. Functional studies of these semaphorins suggest that embryonic and adult neurons require a semaphorin to establish workable connections. Significantly, the fast response time of growth cone cultures to appropriate semaphorins suggests that semaphorin signaling involves a receptor-mediated signal transduction mechanism. Semaphorin ligands that are secreted into the extracellular milieu signal through receptor bearing cells in a located and systemic fashion. In order to further investigate the nature of cellular processes regulated by such local and systemic signaling, it would be beneficial to identify additional semaphorin receptors and ligands. Furthermore, because virus encoded semaphorins are produced by infected cells and are present in viruses that are lytic (poxviruses) and viruses that are not known to be neurotropic (AHV), it is unlikely that their primary function is to modify neurologic responses. It is more likely that the virus encoded semaphorins function to modify the immunologic response of the infected host and it is likely that mammalian homologues to virus encoded semaphorins function to modify the immunologic response. In view of the suggestion that viral semaphorins may function in the immune system as natural immunoregulators it would be beneficial to identify semaphorins that may be therapeutic agents for enhancing or diminishing the immune response. [0004]
  • SUMMARY OF THE INVENTION
  • The present invention pertains to novel semaphorins as isolated or homogeneous proteins. In particular, the present invention provides semaphorin polypeptides, that are homologous to the viral semaphorins A39R and AHVSema. Within the scope of the present invention are DNAs encoding semaphorin polypeptides, and expression vectors that include DNA encoding semaphorin polypeptides. The present invention also includes host cells that have been transfected or transformed with expression vectors that include DNA encoding a semaphorin polypeptide, and processes for producing semaphorin polypeptides by culturing such host cells under conditions conducive to expressing semaphorin polypeptides. The present invention further includes antibodies directed against semaphorin polypeptides. [0005]
  • Further within the scope of the present invention are processes for purifying or separating certain novel semaphorin polypeptides or cells that express novel semaphorins to which semaphorin receptor polypeptides bind. Such processes include binding at least one semaphorin receptor to a solid phase matrix and contacting a mixture containing a semaphorin polypeptide to which the semaphorin receptor binds, or a mixture of cells expressing the semaphorin with the bound semaphorin receptor, and then separating the contacting surface and the solution. [0006]
  • The present invention additionally provides processes for treating mammals afflicted with a disease that is ameliorated by the interaction of semaphorins and their receptors. Such processes involve activating immune cells that express receptors for the semaphorins of the present invention and include administering a therapeutically effective amount of semaphorin to a mammal afflicted with the disease. The therapeutically effective amount is sufficient to activate immune system cells that express semaphorin receptors. Such an activation results in the secretion of cytokines, the regulation of activation antigens or the migration of the cell to sites of immune activity. [0007]
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention provides novel semaphorin polypeptides, DNA encoding semaphorin polypeptides and recombinant expression vectors that include DNA encoding semaphorin polypeptides. The present invention further provides methods for isolating semaphorin polypeptides and methods for producing recombinant semaphorin polypeptides by cultivating host cells transfected with the recombinant expression vectors under conditions appropriate for expressing semaphorin and recovering the expressed semaphorin polypeptide. [0008]
  • This invention additionally provides antibodies directed against semaphorin polypeptides. [0009]
  • Particular semaphorin embodiments of the present invention include polypeptides homologous to AHVsema. The native semaphorin polypeptide described herein was discovered using data base search and comparison techniques that resulted in the identification of at least one EST having some homology to viral semaphorins. As described in Example 1, PCR techniques were used to identify and clone the full viral semaphorin homologue. The human semaphorin of the present invention is found in placenta, testis, ovary, spleen, dendritic cells, and B cells. Semaphorin polypeptides of the present invention bind to the semaphorin receptor, designated VESPR (described in copending application S/N 08/958,598, incorporated herein by reference). Evidence suggests that the interaction between the semaphorins of the present invention and their receptors are associated with the immune suppression of mature dendritic cells. For this reason, the semaphorins of the present invention are designated DCSema. [0010]
  • Example 1 describes identifying a native DCSema of the present invention. The amino acid sequence of the identified native DCSema is disclosed in SEQ ID NO:2 and the DNA encoding the amino acid sequence is disclosed in SEQ ID NO:1. The amino acid sequence presented in SEQ ID NO:2 is a secreted soluble polypeptide, but may additionally exist as a membrane bound protein. The amino acid sequence of SEQ ID NO:2 has a predicted signal sequence that includes amino acids 1-44. Also encompassed within the present invention are soluble DCSema polypeptides that lack the signal sequence. An example of such a polypeptide is amino acids 45-666 of SEQ ID NO:2. The EST 151129 portion of SEQ ID NO:2 has a 28% identity to A39R and 44% identity to AHVsema, both of which bind to VESPR, a semaphorin receptor described in copending patent application S/N 08/958,598. A39R and AHVsema share 29% identity. [0011]
  • The terms “semaphorin polypeptide”, “human semaphorin homologue”, “DCSema” and homologues of AHVSema encompass polypeptides having the amino acid sequence disclosed in SEQ ID NO:2, and proteins that are encoded by nucleic acids that contain the nucleic acid sequence of SEQ ID NO: 1. In addition, those polypeptides that have a high degree of similarity or a high degree of identity with the amino acid sequence of SEQ ID NO:2, which polypeptides are biologically active and bind at least one molecule or fragments of a molecule that is a semaphorin receptor. In addition, “semaphorin polypeptide”, “human semaphorin homologue”, “DCSema” refers to biologically active gene products of the DNA of SEQ ID NO: 1. Further encompassed by semaphorin polypeptides are soluble or truncated proteins that comprise primarily the binding portion of the protein, retain biological activity and are capable of being secreted. Specific examples of such soluble proteins are those comprising the sequence of amino acids 45-666 of SEQ ID NO:2. [0012]
  • The term “biologically active” as it refers to semaphorin polypeptides, means that the semaphorin polypeptide is capable of binding to at least one semaphorin receptor. Assays suitable for determining DCSema binding are described infra and can include standard flow cytometry tests and slide binding tests. [0013]
  • “Isolated” means a DCSema polypeptide is free of association with other proteins or polypeptides, for example, as a purification product of recombinant host cell culture or as a purified extract. [0014]
  • A DCSema polypeptide variant as referred to herein, means a polypeptide substantially homologous to native DCSema polypeptide, but which has an amino acid sequence different from that of the native polypeptides because of one or more deletions, insertions or substitutions. The variant amino acid sequence preferably is at least 80% identical to a native DCSema amino acid sequence, most preferably at least 90% identical. The percent identity may be determined, for example, by comparing sequence information using the GAP computer program, version 6.0 described by Devereux et al. ([0015] Nucl. Acids Res. 12:387, 1984) and available from the University of Wisconsin Genetics Computer Group (UWGCG). The preferred default parameters for the GAP program include: (1) a unary comparison matrix (containing a value of 1 for identities and 0 for non-identities) for nucleotides, and the weighted comparison matrix of Gribskov and Burgess, Nucl. Acids Res. 14:6745, 1986, as described by Schwartz and Dayhoff, eds., Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, pp. 353-358, 1979; (2) a penalty of 3.0 for each gap and an additional 0.10 penalty for each symbol in each gap; and (3) no penalty for end caps. Variants may comprise conservatively substituted sequences, meaning that a given amino acid residue is replaced by a residue having similar physiochemical characteristics. Examples of conservative substitutions include substitution of one aliphatic residue for another, such as Ile, Val, Leu, or Ala for one another, or substitutions of one polar residue for another, such as between Lys and Arg; Glu and Asp; or Gin and Asn. Other such conservative substitutions, for example, substitutions of entire regions having similar hydrophobicity characteristics, are well known. Naturally occurring variants or alleles are also encompassed by the invention. Examples of such variants are proteins that result from alternate mRNA splicing events or from proteolytic cleavage of the DCSema protein, wherein the binding property is retained. Alternate splicing of mRNA may yield a truncated but biologically active polypeptide, such as a naturally occurring soluble form of the protein, for example. Variations attributable to proteolysis include, for example, differences in the N-termini upon expression in different types of host cells, due to proteolytic removal of one or more terminal amino acids from the DCSema polypeptide (generally from 1-5 terminal amino acids).
  • Example 3 describes the construction of a novel viral Semaphorin/Fc fusion (DCSema/Fc) protein that may be utilized in studying the biological characteristics of DCSema and their receptor distribution. Other antibody Fc regions may be substituted for the human IgG1 Fc region described in Example 3. Suitable Fc regions are those that can bind with high affinity to protein A or protein G, and include the Fc region of human IgG1 or fragments of the human or murine IgG1 Fc region, e.g., fragments comprising at least the hinge region so that interchain disulfide bonds will form. The viral DCSema/Fc fusion protein offers the advantage of being easily purified. In addition, disulfide bonds form between the Fc regions of two separate fusion protein chains, creating dimers. [0016]
  • The soluble DCSema polypeptides of the present invention may be isolated and identified by separating intact cells that express the desired protein from the culture medium in which the cells grow, and then assaying the medium (supernatant) for the presence of the desired protein. Separation can be accomplished using standard separation techniques including, including centrifugation. The presence of the DCSema polypeptide in the medium indicates that the protein was secreted from the cells in its expected soluble form. Because the DCSema polypeptides of the present invention are secreted soluble polypeptides they possess many advantages over membrane bound proteins. Purification of the proteins from recombinant host cells is feasible, since the soluble proteins are secreted from the cells. Further, soluble proteins are generally more suitable for intravenous administration. [0017]
  • Truncated DCSema proteins comprising less than the entire secreted polypeptide are included in the invention, e.g. soluble fragments such as amino acids 52-543 of SEQ ID NO:2 that include the “semaphorin domain,” which is part of an active binding site, and amino acids 45-644 of SEQ ID NO:2 that include the secreted protein without the signal peptide, are included in the invention. When initially expressed within a host cell, DCSema polypeptides may additionally comprise one of the heterologous signal peptides described below that is functional within the host cells employed. Alternatively, the protein may comprise the native signal peptide. In one embodiment of the invention, DCSema polypeptides can be expressed as a fusion protein comprising (from N— to C-terminus) the yeast α-factor signal peptide, a FLAG® peptide described below and in U.S. Pat. No. 5,011,912, and soluble DCSema polypeptide consisting of amino acids 45 to 666 of SEQ ID NO:2. This recombinant fusion protein is expressed in and secreted from yeast cells. The FLAG® peptide facilitates purification of the protein, and subsequently may be cleaved from the DCSema using bovine mucosal enterokinase. [0018]
  • Truncated DCSema polypeptides may be prepared by any of a number of conventional techniques. A desired DNA sequence may be chemically synthesized using techniques known per se. DNA fragments also may be produced by restriction endonuclease digestion of a full length cloned DNA sequence, and isolated by electrophoresis on agarose gels. Linkers containing restriction endonuclease cleavage site(s) may be employed to insert the desired DNA fragment into an expression vector, or the fragment may be digested at cleavage sites naturally present therein. The well known polymerase chain reaction procedure also may be employed to amplify a DNA sequence encoding a desired protein fragment. As a further alternative, known mutagenesis techniques may be employed to insert a stop codon at a desired point, e.g., immediately downstream of the codon for the last amino acid of the binding domain. [0019]
  • As stated above, the invention provides isolated or homogeneous DCSema polypeptides, both recombinant and non-recombinant. Variants and derivatives of native DCSema proteins that retain the desired biological activity (e.g., the ability to bind to DCSema receptors) may be obtained by mutations of nucleotide sequences coding for the native polypeptides. Alterations of the native amino acid sequence may be accomplished by any of a number of conventional methods. Mutations can be introduced at particular loci by synthesizing oligonucleotides containing a mutant sequence, flanked by restriction sites enabling ligation to fragments of the native sequence. Following ligation, the resulting reconstructed sequence encodes an analog having the desired amino acid insertion, substitution, or deletion. [0020]
  • Alternatively, oligonucleotide-directed site-specific mutagenesis procedures can be employed to provide an altered gene wherein predetermined codons can be altered by substitution, deletion or insertion. Exemplary methods of making the alterations set forth above are disclosed by Walder et al. ([0021] Gene 42:133, 1986); Bauer et al. (Gene 37:73, 1985); Craik (BioTechniques, January 1985, 12-19); Smith et al. (Genetic Engineering: Principles and Methods. Plenum Press, 1981); Kunkel (Proc. Natl. Acad. Sci. USA 82:488, 1985); Kunkel et al. (Methods in Enzymol. 154:367, 1987); and U.S. Pat. Nos. 4,518,584 and 4,737,462 all of which are incorporated by reference.
  • Native DCSema polypeptides may be modified to create DCSema derivatives by forming covalent or aggregative conjugates with other chemical moieties, such as glycosyl groups, lipids, phosphate, acetyl groups and the like. Covalent derivatives of the polypeptides may be prepared by linking the chemical moieties to functional groups on DCSema amino acid side chains or at the N-terminus or C-terminus of a DCSema polypeptide. Other derivatives of the polypeptides within the scope of this invention include covalent or aggregative conjugates of DCSema polypeptides or their fragments with other proteins or polypeptides, such as by synthesis in recombinant culture as N-terminal or C-terminal fusions. For example, the conjugate may comprise a signal or leader polypeptide sequence (e.g. the α-factor leader of Saccharomyces) at the N-terminus of a polypeptide of the present invention. The signal or leader peptide co-translationally or post-translationally directs transfer of the conjugate from its site of synthesis to a site inside or outside of the cell membrane or cell wall. [0022]
  • DCSema polypeptide fusion proteins can comprise peptides added to facilitate purification and identification of semaphorin receptors that bind semaphorins of the present invention, including VESPR (see copending application S/N08/958,598. Such peptides include, for example, poly-His or the antigenic identification peptides described in U.S. Pat. No. 5,011,912 and in Hopp et al., [0023] Bio/Technology 6:1204, 1988.
  • The invention further includes DCSema polypeptides with or without associated native-pattern glycosylation. DCSema polypeptide expressed in yeast or mammalian expression systems (e.g., COS-7 cells) may be similar to or significantly different from a native DCSema polypeptide in molecular weight and glycosylation pattern, depending upon the choice of expression system. Expression of DCSema polypeptides in bacterial expression systems, such as [0024] E. coli, provides non-glycosylated molecules.
  • Equivalent DNA constructs that encode various additions or substitutions of amino acid residues or sequences, or deletions of terminal or internal residues or sequences not needed for biological activity or binding are encompassed by the invention. For example, N-gycosylation sites in the DCSema extracellular domain can be modified to preclude glycosylation, allowing expression of a reduced carbohydrate analog in mammalian and yeast expression systems. N-glycosylation sites in eukaryotic polypeptides are characterized by an amino acid triplet Asn-X-Y, wherein X is any amino acid except Pro and Y is Ser or Thr. The native human DC Sema of the present invention comprises 4 such amino acid triplets, at amino acids 105-107, 157-159, 258-260 and 602-604 of SEQ ID NO:2. Appropriate substitutions, additions or deletions to the nucleotide sequence encoding these triplets will result in prevention of attachment of carbohydrate residues at the Asn side chain. Alteration of a single nucleotide, chosen so that Asn is replaced by a different amino acid, for example, is sufficient to inactivate an N-glycosylation site. Known procedures for inactivating N-glycosylation sites in proteins include those described in U.S. Pat. No. 5,071,972 and EP 276,846, hereby incorporated by reference. [0025]
  • In another example, sequences encoding Cys residues that are not essential for biological activity can be altered to cause the Cys residues to be deleted or replaced with other amino acids, preventing formation of incorrect intramolecular disulfide bridges upon renaturation. Other equivalents are prepared by modification of adjacent dibasic amino acid residues to enhance expression in yeast systems in which KEX2 protease activity is present. EP 212,914 discloses the use of site-specific mutagenesis to inactivate KEX2 protease processing sites in a protein. KEX2 protease processing sites are inactivated by deleting, adding or substituting residues to alter Arg-Arg, Arg-Lys, and Lys-Arg pairs to eliminate the occurrence of these adjacent basic residues. Lys-Lys pairings are considerably less susceptible to KEX2 cleavage, and conversion of Arg-Lys or Lys-Arg to Lys-Lys represents a conservative and preferred approach to inactivating KEX2 sites. The DC sema of SEQ ID NO:2 contains 7 KEX2 protease processing sites at amino acids 123-124, 135, 136, 192, 193, 204-205, 460461, 474-475 and 475-476, Nucleic acid sequences within the scope of the invention include isolated DNA and RNA sequences that hybridize to DCSema nucleotide sequences disclosed herein under conditions of moderate or high stringency, and that encode biologically active DCSema. Conditions of moderate stringency, as defined by Sambrook et al. [0026] Molecular Cloning: A Laboratory Manual, 2 ed. Vol. 1, pp. 101-104, Cold Spring Harbor Laboratory Press, (1989), include use of a prewashing solution of 5×SSC, 0.5% SDS, 1.0 mM EDTA (pH 8.0) and hybridization conditions of about 55° C., 5×SSC, overnight. Conditions of severe stringency include higher temperatures of hybridization and washing. The skilled artisan will recognize that the temperature and wash solution salt concentration may be adjusted as necessary according to factors such as the length of the nucleic acid molecule and the relative amount of A, T[U, C and G nucleotides.
  • Due to the known degeneracy of the genetic code wherein more than one codon can encode the same amino acid, a DNA sequence may vary from that shown in SEQ ID NO:1 and still encode a polypeptide having the amino acid sequence of SEQ ID NO:2. Such variant DNA sequences may result from silent mutations (e.g., occurring during PCR amplification), or may be the product of deliberate mutagenesis of a native sequence. [0027]
  • The invention provides equivalent isolated DNA sequences encoding biologically active human DCSema, selected from: (a) cDNA comprising the nucleotide sequence presented in SEQ ID NO: 1; (b) DNA capable of hybridization to a DNA of (a) under moderately stringent conditions and that encodes biologically active polypeptides and (c) DNA that is degenerate as a result of the genetic code to a DNA defined in (a), or (b) and that encodes biologically active human DCSema polypeptide. Polypeptides encoded by such DNA equivalent sequences are encompassed by the invention. [0028]
  • DNAs that are equivalent to the DNA sequence of SEQ ID NO:1 include those that hybridize under moderately and highly stringent conditions to the DNA sequence that encodes polypeptides comprising the sequence of SEQ ID NO:2. Examples of proteins encoded by such DNA, include, but are not limited to, polypeptide fragments and proteins that have inactivated N-glycosylation site(s), inactivated KEX2 protease processing site(s), or conservative amino acid substitution(s), as described above. DCSema polypeptides encoded by DNA derived from other species, wherein the DNA will hybridize to the cDNA of SEQ ID NO:1, are also within the present invention. [0029]
  • DCSema polypeptide variants possessing the ability to bind semaphorin receptors may be identified by any suitable assay. Biological activity of DCSema polypeptides of the present invention may be determined, for example, by competition for binding to the binding domain of semaphorin receptors, e.g. competitive binding assays, or for binding to a semaphorin receptor binding domain. [0030]
  • One type of a competitive binding assay for a DCSema polypeptide of the present invention uses a radiolabeled DCSema and intact semaphorin receptor-expressing cells. Instead of intact cells, one could substitute soluble semaphorin receptor:Fc fusion proteins bound to a solid phase through the interaction of a Protein A, Protein G or an antibody to the semaphorin receptor or Fc portions of the molecule, with the Fc region of the fusion protein. Competitive binding assays can be performed following conventional methodology. In one embodiment, a soluble semaphorin receptor can be made to compete with an immobilized receptor for binding with a soluble semaphorin ligand. For example, a radiolabeled soluble semaphorin ligand can be antagonized by soluble VESPR in an assay for binding activity against a surface-bound semaphorin receptor. Qualitative results can be obtained by competitive autoradiographic plate binding assays, or Scatchard plots may be utilized to generate quantitative results. [0031]
  • Alternatively, semaphorin binding proteins, such as VESPR or anti-semaphorin antibodies, can be bound to a solid phase such as a column chromatography matrix or a similar substrate suitable for identifying, separating or purifying cells that express semaphorin on their surface. Binding of a semaphorin-binding protein to a solid phase contacting surface can be accomplished by any means, for example, by constructing a VESPR:Fc fusion protein and binding such to the solid phase through the interaction of Protein A or Protein G. Various other means for fixing proteins to a solid phase are well known in the art and are suitable for use in the present invention. For example, magnetic microspheres can be coated with VESPR and held in the incubation vessel through a magnetic field. Suspensions of cell mixtures containing semaphorin-expressing cells are contacted with the solid phase that has VESPR polypeptides thereon. Cells having semaphorin on their surface bind to the fixed VESPR and unbound cells then are washed away. This affinity-binding method is useful for purifying, screening or separating such semaphorin-expressing cells from solution. Methods of releasing positively selected cells from the solid phase are known in the art and encompass, for example, the use of enzymes. Such enzymes are preferably non-toxic and non-injurious to the cells and are preferably directed to cleaving the cell-surface binding partner. In the case of semaphorin-VESPR interactions, the enzyme preferably would cleave the semaphorin, thereby freeing the resulting cell suspension from the “foreign” semaphorin receptor material. The purified cell population then may be used to repopulate mature (adult) tissues. [0032]
  • Alternatively, mixtures of cells suspected of containing semaphorin cells first can be incubated with a suitable biotinylated semaphorin receptor, e.g. VESPR. Incubation periods are typically at least one hour in duration to ensure sufficient binding to semaphorin The resulting mixture then is passed through a column packed with avidin-coated beads, whereby the high affinity of biotin for avidin provides the binding of the cell to the beads. Use of avidin-coated beads is known in the art. See Berenson, et al. [0033] J. Cell. Biochem., 10D:239 (1986). Washing unbound material and releasing the bound cells is performed using conventional methods.
  • As described above, cells expressing DCSema polypeptides of the present invention can be separated using a semaphorin receptor, e.g. VESPR. In an alternative method, VESPR, other suitable semaphorin receptors, or an extracellular domain or a fragment thereof can be conjugated to a detectable moiety such as [0034] 125I to detect semaphorin-expressing cells. Radiolabeling with 125I can be performed by any of several standard methodologies that yield a functional 125I-molecule labeled to high specific activity or an iodinated or biotinylated antibody against the semaphorin receptor. Another detectable moiety such as an enzyme that can catalyze a colorimetric or fluorometric reaction, biotin or avidin may be used. Cells to be tested for DCSema polypeptide-expression can be contacted with a suitable labeled receptor, e.g. VESPR. After incubation, unbound labeled receptor is removed and binding is measured using the detectable moiety.
  • The binding characteristics of DCSema polypeptides may also be determined using a conjugated semaphorin receptor (for example, [0035] 125I-semaphorin receptor:Fc) in competition assays similar to those described above. In this case, however, intact cells expressing DCSema polypeptide of the present invention, bound to a solid substrate, are used to measure the extent to which a sample containing a putative receptor variant competes for binding with a conjugated DCSema.
  • Other means of assaying for DCSema polypeptides of the present invention include the use of anti-DCSema antibodies, cell lines that proliferate in response to DCSema polypeptide, or recombinant cell lines that express DCSema and proliferate in the presence of a suitable semaphorin receptor. [0036]
  • The DCSema proteins disclosed herein also may be employed to measure the biological activity of any semaphorin receptor in terms of its binding affinity for its semaphorin ligand. As one example, DCSema polypeptides of the present invention may be used in determining whether biological activity is retained after modification of a semaphorin receptor (e.g., chemical modification, truncation, mutation, etc.). The biological activity of a semaphorin receptor thus can be ascertained before it is used in a research study, or in the clinic, for example. [0037]
  • DCSema polypeptides disclosed herein find use as reagents in “quality assurance” studies, e.g., to monitor shelf life and stability of a receptor to which the DCSema binds under different conditions. To illustrate. DCSema polypeptides of the present invention may be employed in a binding affinity study to measure the biological activity of a test semaphorin receptor that has been stored at different temperatures, or produced in different cell types. The binding affinity of the DCSema protein for the test receptor is compared to that of a standard or control semaphorin receptor to detect any adverse impact on biological activity of the test semaphorin receptor. [0038]
  • DCSema polypeptides described herein also find use as carriers for delivering agents attached thereto to cells expressing semaphorin receptors to which the semaphorin binds. As described in copending application S/N 958,598, VESPR, to which a DCSema of the present invention binds, is expressed in lung epithelial cells, stroma, intestinal epithelial cells and lymphoma cells. DCSema polypeptides of the present invention can thus can be used to deliver diagnostic or therapeutic agents to these cells (or to other cell types found to express a suitable semaphorin receptor on cell surfaces) in in vitro or in vivo procedures. [0039]
  • Diagnostic and therapeutic agents that may be attached to a DCSema polypeptide of the present invention include, but are not limited to, drugs, toxins, radionuclides, chromophores, enzymes that catalyze a colorimetric or fluorometric reaction, and the like, with the particular agent being chosen according to the intended application. Examples of drugs include those used in treating various forms of cancer, e.g., nitrogen mustards such as L-phenylalanine nitrogen mustard or cyclophosphamide, intercalating agents such as cis-diaminodichloroplatinum, antimetabolites such as 5-fluorouracil, vinca alkaloids such as vincristine, and antibiotics such as bleomycin, doxorubicin, daunorubicin, and derivatives thereof. Among the toxins are ricin, abrin, diptheria toxin, [0040] Pseudomonas aeruginosa exotoxin A, ribosomal inactivating proteins, mycotoxins such as trichothecenes, and derivatives and fragments (e.g., single chains) thereof. Radionuclides suitable for diagnostic use include, but are not limited to, 123I, 131I, 99m Tc, 111In, and 76Br. Radionuclides suitable for therapeutic use include, but are not limited to, 131I, 211At. 77Br, 186Re, 188Re, 212Pb, 212Bi, 109Pd, 64Cu, and 67Cu.
  • Such agents may be attached to the DCSema of the present invention by any suitable conventional procedure. Semaphorin homologues of the present invention, being proteins, include functional groups on amino acid side chains that can be reacted with functional groups on a desired agent to form covalent bonds, for example. Alternatively, the protein or agent may be derivatized to generate or attach a desired reactive functional group. The derivatization may involve attachment of one of the bifunctional coupling reagents available for attaching various molecules to proteins (Pierce Chemical Company, Rockford, Ill.). A number of techniques for radiolabeling proteins are known. Radionuclide metals may be attached to the receptor by using a suitable bifunctional chelating agent, for example. Conjugates comprising molecules of the present invention and a suitable diagnostic or therapeutic agent (preferably covalently linked) are thus prepared. The conjugates are administered or otherwise employed in an amount appropriate for the particular application. [0041]
  • Another use of the DCSema polypeptides of the present invention is as a research tool for studying the role that the DCSema, in conjunction with semaphorin receptors to which it binds, may play in immune regulation and viral infection. The polypeptides of the present invention also may be employed in in vitro assays for detection of receptors to which it binds, e.g. VESPR, or the interactions thereof. Similarly, DCSema polypeptides of the present invention can be used as a research tool for studying the role that the ligand, in conjunction with its receptors, play in immune regulation. [0042]
  • Furthermore, it is known that administration of IL-12 to tumor bearing animals results in tumor regression and the establishment of a tumor-specific immune response. Thus, using a DCSema ligand to bind with VESPR in order to enhance or promote IL-12 can induce a curative immune response against aggressive micrometastasizing tumors. [0043]
  • VESPR, a semaphorin•receptor, binds with a binding partner to down-regulate expression of MHC Class II molecules and CD86, a co-stimulatory molecule, on dendritic cells, cultured with GM-CSF and IL-4 (see copending application S/N 60/085,497). By analogy, this suggests that the interaction between DCSema of the present invention and its receptors are associated with the immune suppression of mature dendritic cells. Thus, the use of DCSema ligands, including the DC Sema of the present invention, in the treatment of autoimmune disorders is expected to diminish unwanted symptoms associated with the autoimmune disorder by downregulating the antigen presenting capabilities of mature dendritic cells. [0044]
  • DCSema polypeptides of the invention can be formulated according to known methods used to prepare pharmaceutically useful compositions. The molecules of the invention can be combined in admixture, either as the sole active material or with other known active materials, with pharmaceutically suitable diluents (e.g., Tris-HCl, acetate, phosphate), preservatives (e.g., Thimerosal, benzyl alcohol, parabens), emulsifiers, solubilizers, adjuvants and/or carriers. Suitable carriers and their formulations are described in Remington's Pharmaceutical Sciences, 16th ed. 1980, Mack Publishing Co. In addition, such compositions can contain DCSema polypeptide complexed with polyethylene glycol (PEG), metal ions, or incorporated into polymeric compounds such as polyacetic acid, polyglycolic acid, hydrogels, etc., or incorporated into liposomes, microemulsions, micelles, unilamellar or multilamellar vesicles, erythrocyte ghosts or spheroblasts. Such compositions will influence the physical state, solubility, stability, rate of in vivo release, and rate of in vivo clearance of DCSema. DCSema polypeptides described herein can be conjugated to antibodies against tissue-specific receptors, ligands or antigens, or coupled to ligands of tissue-specific receptors. [0045]
  • DCSema polypeptides of the present invention can be administered topically, parenterally, or by inhalation. The term “parenteral” includes subcutaneous injections, intravenous, intramuscular, intracisternal injection, or infusion techniques. These compositions will typically contain an effective amount of the polypeptide, alone or in combination with an effective amount of any other active material. Such dosages and desired drug concentrations contained in the compositions may vary depending upon many factors, including the intended use, patient's body weight and age, and route of administration. Preliminary doses can be determined according to animal tests, and the scaling of dosages for human administration can be performed according to art-accepted practices. [0046]
  • Polypeptides of the present invention may exist as oligomers, such as covalently-linked or non-covalently-linked dimers or trimers. Oligomers may be linked by disulfide bonds formed between cysteine residues on different DCSema molecules. In one embodiment of the invention, a dimer is created by fusing a DCSema to the Fc region of an antibody (e.g., IgG1) in a manner that does not interfere with binding of the DCSema to a semaphorin receptor-binding domain. The Fc polypeptide preferably is fused to the C-terminus of a DCSema General preparation of fusion proteins comprising heterologous polypeptides fused to various portions of antibody-derived polypeptides (including the Fc domain) has been described, e.g., by Ashkenazi et al. ([0047] PNAS USA 88:10535, 1991) and Byrn et al. (Nature 344:677, 1990), hereby incorporated by reference. A gene fusion encoding the DCSema/Fc fusion protein is inserted into an appropriate expression vector. DCSema/Fc fusion proteins are allowed to assemble much like antibody molecules, whereupon interchain disulfide bonds form between Fc polypeptides, yielding divalent. If fusion proteins are made with both heavy and light chains of an antibody, it is possible to form a DCSema oligomer with as many as four semaphorin regions. Alternatively, one can link two DCSemas with a peptide linker.
  • Suitable host cells for expression of DCSema polypeptides of this invention include prokaryotes, yeast or higher eukaryotic cells. Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, and mammalian cellular hosts are described, for example, in Pouwels et al. [0048] Cloning Vectors: A Laboratory Manual, Elsevier, N.Y., (1985). Cell-free translation systems could also be employed to produce the polypeptides of the present invention using RNAs derived from DNA constructs disclosed herein.
  • Prokaryotes include gram negative or gram positive organisms, for example, [0049] E. coli or Bacilli. Suitable prokaryotic host cells for transformation include, for example, E. coli, Bacillus subtilis, Salmonella typhimurium, and various other species within the genera Pseudomonas, Streptomyces, and Staphylococcus. In a prokaryotic host cell, such as E. coli, a semaphorin receptor polypeptide may include an N-terminal methionine residue to facilitate expression of the recombinant polypeptide in the prokaryotic host cell. The N-terminal Met may be cleaved from the expressed recombinant DCSema polypeptide.
  • DCSema polypeptides may be expressed in yeast host cells, preferably from the Saccharomyces genus (e.g., [0050] S. cerevisiae). Other genera of yeast, such as Pichia, K. lactis or Kluyveromyces, may also be employed. Yeast vectors will often contain an origin of replication sequence from a 2μ yeast plasmid, an autonomously replicating sequence (ARS), a promoter region, sequences for polyadenylation, sequences for transcription termination, and a selectable marker gene. Suitable promoter sequences for yeast vectors include, among others, promoters for metallothionein. 3-phosphoglycerate kinase (Hitzeman et al., J. Biol. Chem. 255:2073, 1980) or other glycolytic enzymes (Hess et al., J. Adv. Enzyme Reg. 7:149, 1968; and Holland et al., Biochem. 17:4900, 1978), such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase. 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase. Other suitable vectors and promoters for use in yeast expression are further described in Hitzeman, EPA-73,657 or in Fleer et. al., Gene, 107:285-195 (1991); and van den Berg et. al., Bio/technology, 8:135-139 (1990). Another alternative is the glucose-repressible ADH2 promoter described by Russell et al. (J. Biol. Chem. 258:2674, 1982) and Beier et al. (Nature 300:724, 1982). Shuttle vectors replicable in both yeast and E. coli may be constructed by inserting DNA sequences from pBR322 for selection and replication in E. coli (Ampr gene and origin of replication) into the above-described yeast vectors.
  • The yeast α-factor leader sequence may be employed to direct secretion of the VESPR or DCSema polypeptide. The α-factor leader sequence is often inserted between the promoter sequence and the structural gene sequence. See, e.g., Kurjan et al., [0051] Cell 30:933, 1982: Bitter et al., Proc. Natl. Acad. Sci. USA 81:5330, 1984; U.S. Pat. No. 4,546,082; and EP 324,274. Other leader sequences suitable for facilitating secretion of recombinant polypeptides from yeast hosts are known to those of skill in the art. A leader sequence may be modified near its 3′ end to contain one or more restriction sites. This will facilitate fusion of the leader sequence to the structural gene.
  • Yeast transformation protocols are known to those of skill in the art. One such protocol is described by Hinnen et al., [0052] Proc. Natl. Acad. Sci. USA 75:1929, 1978. The Hinnen et al. protocol selects for Trp+ transformants in a selective medium, wherein the selective medium consists of 0.67% yeast nitrogen base, 0.5% casamino acids, 2% glucose. 10 μg/ml adenine and 20 μg/ml uracil.
  • Yeast host cells transformed by vectors containing ADH2 promoter sequence may be grown for inducing expression in a “rich” medium. An example of a rich medium is one consisting of 1% yeast extract, 2% peptone, and 1% glucose supplemented with 80 μg/ml adenine and 80 μg/ml uracil. Depression of the ADH2 promoter occurs when glucose is exhausted from the medium. [0053]
  • Mammalian or insect host cell culture systems could also be employed to express recombinant polypeptides of the present invention. Baculovirus systems for production of heterologous proteins in insect cells are reviewed by Luckow and Summers, [0054] Bio/Technology 6:47 (1988). Established cell lines of mammalian origin also may be employed. Examples of suitable mammalian host cell lines include the COS-7 line of monkey kidney cells (ATCC CRL 1651) (Gluzman et al., Cell 23:175, 1981), L cells, C127 cells, 3T3 cells (ATCC CCL 163), Chinese hamster ovary (CHO) cells. HeLa cells, and BHK (ATCC CRL 10) cell lines, and the CV-1/EBNA-1 cell line derived from the African green monkey kidney cell line CVI (ATCC CCL 70) as described by McMahan et al. (EMBO J. 10: 2821, 1991).
  • Transcriptional and translational control sequences for mammalian host cell expression vectors may be excised from viral genomes. Commonly used promoter sequences and enhancer sequences are derived from Polyoma virus. Adenovirus 2, Simian Virus 40 (SV40), and human cytomegalovirus. DNA sequences derived from the SV40 viral genome, for example, SV40 origin, early and late promoter, enhancer, splice, and polyadenylation sites may be used to provide other genetic elements for expression of a structural gene sequence in a mammalian host cell. Viral early and late promoters are particularly useful because both are easily obtained from a viral genome as a fragment which may also contain a viral origin of replication (Fiers et al., [0055] Nature 273:113, 1978). Smaller or larger SV40 fragments may also be used, provided the approximately 250 bp sequence extending from the Hind III site toward the Bgl I site located in the SV40 viral origin of replication site is included.
  • Exemplary expression vectors for use in mammalian host cells can be constructed as disclosed by Okayama and Berg (Mol. Cell. Biol. 3:280, 1983). A useful system for stable high level expression of mammalian cDNAs in C127 murine mammary epithelial cells can be constructed substantially as described by Cosman et al. ([0056] Mol. Immunol. 23:935, 1986). A useful high expression vector, PMLSV N1/N4, described by Cosman et al., Nature 312:768, 1984 has been deposited as ATCC 39890. Additional useful mammalian expression vectors are described in EP-A-0367566, and in U.S. patent application Ser. No. 07/701,415, filed May 16, 1991, incorporated by reference herein. The vectors may be derived from retroviruses. In place of the native signal sequence, and in addition to an initiator methionine, a heterologous signal sequence may be added, such as the signal sequence for IL-7 described in U.S. Pat. No. 4,965,195; the signal sequence for IL-2 receptor described in Cosman et al., Nature 312:768 (1984); the IL-4 signal peptide described in EP 367,566; the type I IL-1 receptor signal peptide described in U.S. Pat. No. 4,968,607; and the type II IL-1 receptor signal peptide described in EP 460,846.
  • DCSema polypeptides of the present invention, as isolated, purified or homogeneous proteins may be produced by recombinant expression systems as described above or purified from naturally occurring cells. The polypeptides can be purified to substantial homogeneity, as indicated by a single protein band upon analysis by SDS-polyacrylamide gel electrophoresis (SDS-PAGE). [0057]
  • One process for producing polypeptides of the present invention comprises culturing a host cell transformed with an expression vector comprising a DNA sequence that encodes the desired DCSema polypeptide under conditions sufficient to promote expression of the DCSema polypeptide. The DCSema polypeptide is then recovered from culture medium or cell extracts, depending upon the expression system employed. As is known to the skilled artisan, procedures for purifying a recombinant protein will vary according to such factors as the type of host cells employed and whether or not the recombinant protein is secreted into the culture medium. [0058]
  • For example, when expression systems that secrete the recombinant protein are employed, the culture medium first may be concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit. Following the concentration step, the concentrate can be applied to a purification matrix such as a gel filtration medium. Alternatively, an anion exchange resin can be employed, for example, a matrix or substrate having pendant diethylaminoethyl (DEAE) groups. The matrices can be acrylamide, agarose, dextran, cellulose or other types commonly employed in protein purification. Alternatively, a cation exchange step can be employed. Suitable cation exchangers include various insoluble matrices comprising sulfopropyl or carboxymethyl groups. Sulfopropyl groups are preferred. Finally, one or more reversed-phase high performance liquid chromatography (RP-HPLC) steps employing hydrophobic RP-HPLC media, (e.g., silica gel having pendant methyl or other aliphatic groups) can be employed to further purify the polypeptide. Some or all of the foregoing purification steps, in various combinations, are well known and can be employed to provide a substantially homogeneous recombinant protein. [0059]
  • It is possible to utilize an affinity column comprising the receptor-binding domain of a DCSema of the present invention affinity-purify semaphorin receptor to which the DCSema binds. Such a receptor can be removed from an affinity column using conventional techniques, e.g., in a high salt elution buffer and then dialyzed into a lower salt buffer for use or by changing pH or other components depending on the affinity matrix utilized. [0060]
  • Recombinant protein produced in bacterial culture can be isolated by initial disruption of the host cells, centrifugation, extraction from cell pellets if an insoluble polypeptide, or from the supernatant fluid if a soluble polypeptide, followed by one or more concentration, salting-out, ion exchange, affinity purification or size exclusion chromatography steps. Finally, RP-HPLC can be employed for final purification steps. Microbial cells can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents. [0061]
  • Transformed yeast host cells are preferably employed to express DCSemas of the present invention as a secreted polypeptide in order to simplify purification. Secreted recombinant polypeptide from a yeast host cell fermentation can be purified by methods analogous to those disclosed by Urdal et al. ([0062] J. Chromatog. 296:171, 1984). Urdal et al. describe two sequential, reversed-phase HPLC steps for purification of recombinant human IL-2 on a preparative HPLC column.
  • Useful fragments of the DCSema nucleic acids of the present invention include antisense or sense oligonucleotides comprising a single-stranded nucleic acid sequence (either RNA or DNA) capable of binding to target DCSema mRNA (sense) or DCSema DNA (antisense) sequences. Antisense or sense oligonucleotides, according to the present invention, comprise a fragment of the coding region of DCSema cDNA. Such a fragment generally comprises at least about 14 nucleotides, preferably from about 14 to about 30 nucleotides. The ability to derive an antisense or a sense oligonucleotide, based upon a cDNA sequence encoding a given protein is described in, for example, Stein and Cohen ([0063] Cancer Res. 48:2659, 1988) and van der Krol et al. (BioTechniques 6:958, 1988).
  • Binding of antisense or sense oligonucleotides to target nucleic acid sequences results, in the formation of duplexes that block transcription or translation of the target sequence by one of several means, including enhanced degradation of the duplexes, premature termination of transcription or translation, or by other means. The antisense oligonucleotides thus may be used to block expression of proteins of the present invention. Antisense or sense oligonucleotides further comprise oligonucleotides having modified sugar-phosphodiester backbones (or other sugar linkages, such as those described in WO91/06629) and wherein such sugar linkages are resistant to endogenous nucleases. Such oligonucleotides with resistant sugar linkages are stable in vivo (i.e., capable of resisting enzymatic degradation) but retain sequence specificity to be able to bind to target nucleotide sequences. Other examples of sense or antisense oligonucleotides include those oligonucleotides which are covalently linked to organic moieties, such as those described in WO 90/10448, and other moieties that increases affinity of the oligonucleotide for a target nucleic acid sequence, such as poly-(L-lysine). Further still, intercalating agents, such as ellipticine, and alkylating agents or metal complexes may be attached to sense or antisense oligonucleotides to modify binding specificities of the antisense or sense oligonucleotide for the target nucleotide sequence. [0064]
  • Antisense or sense oligonucleotides may be introduced into a cell containing the target nucleic acid sequence by any gene transfer method, including, for example, CaPO[0065] 4-mediated DNA transfection, electroporation, or by using gene transfer vectors such as Epstein-Barr virus. Antisense or sense oligonucleotides are preferably introduced into a cell containing the target nucleic acid sequence by insertion of the antisense or sense oligonucleotide into a suitable retroviral vector, then contacting the cell with the retrovirus vector containing the inserted sequence, either in vivo or ex vivo. Suitable retroviral vectors include, but are not limited to, those derived from the murine retrovirus M-MuLV, N2 (a retrovirus derived from M-MuLV), or the double copy vectors designated DCT5A, DCT5B and DCT5C (see PCT Application US 90/02656).
  • Sense or antisense oligonucleotides also may be introduced into a cell containing the target nucleotide sequence by formation of a conjugate with a ligand binding molecule, as described in WO 91/04753. Suitable ligand binding molecules include, but are not limited to, cell surface receptors, growth factors, other cytokines, or other ligands that bind to cell surface receptors. Preferably, conjugation of the ligand binding molecule does not substantially interfere with the ability of the ligand binding molecule to bind to its corresponding molecule or receptor, or block entry of the sense or antisense oligonucleotide or its conjugated version into the cell. [0066]
  • Alternatively, a sense or an antisense oligonucleotide may be introduced into a cell containing the target nucleic acid sequence by formation of an oligonucleotide-lipid complex, as described in WO 90/10448. The sense or antisense oligonucleotide-lipid complex is preferably dissociated within the cell by an endogenous lipase. [0067]
  • In addition to the above, the following examples are provided to illustrate particular embodiments and not to limit the scope of the invention. [0068]
  • EXAMPLE 1 Isolating a Human Semaphorin Homologue Designated DCSema
  • Using sequence alignment techniques and comparative sequence analysis of ESTs, a human homologue of AHV Sema semaphorin was identified as follows. The nucleotide sequence of viral A39R was used in a Unigene comparative sequence search. This search resulted in the identification of EST #151129 (accession # [H029021], deposited June 20, 1995. EST 151129 is a partial sequence having neither an initiating ATG nor a termination codon. EST 151129 has sequence homology to A39R and AHVsema. [0069]
  • EST 151129 was utilized to isolate and identify the native human DCSema as follows: [0070]
  • A tissue source of EST 151129 was identified using phage library screening methods and PCR primers based upon this EST sequence. Oligonucleotide PCR primers having the following nucleotide sequences were synthesized: [0071]
    5′-TGCTGGAACCTGGTGAATGG-3′ (SEQ ID NO:3)
    5′-AGTGGAACAATGGCGTCTTC-3′ (SEQ ID NO:4)
  • PCR isolation and amplification methodologies were carried out using a panel of human tissue cDNA phase libraries as templates for the PCR reactions. [0072]
  • Two of the phage libraries, human foreskin fibroblast and dermal fibroblast, were chosen for additional analysis. The libraries were plated according to established procedures. A radiolabeled probe was generated by incorporating [0073] 32P-dCTP in the amplification of a PCR product using the EST 151129 as a template. The PCR reaction mixture included 10 ng of EST151129 plasmid DNA, 50 pmoles of each PCR oligonucleotide primer identified below and specific for the 5′ end of EST 151129, 1× Amplitaq buffer (Perkin-Elmer Cetus), 1.25 mM of dATP, dGTP, dTTP, 0.001 mM dCTP (Pharmacia,), 150 μCi 32P-dCTP, 0.5 μl Amplitaq taq polymerase (Perkin-Elmer Cetus,) in a 100 μl final reaction volume. The PCR reaction cycles included one cycle at 94° C. for 5 min; twenty-five cycles at 94° C. for 1 min, 72° C. for 2 min and one cycle at 72° C. for 5 min using a Robocycler Gradient 40 (Stratagene, La Jolla. CA).
  • The primers used were as follows: [0074]
    5′-TCCGCCCAGGGCCACCTAAGGAGCGGA-3′ (SEQ ID NO:7)
    5′-TGTGCGGCTCAGTCTGGCCAAAGTCCA-3′ (SEQ ID NO:8)
  • Approximately 5×10[0075] 5 cpm/mL of purified probe was used to hybridize with the human dermal fibroblast library on nylon membrane filters in the same manner described in Example 5 for probing human foreskin fibroblast and human dermal fibroblast libraries.
  • A cDNA that overlapped with the 5′ end of EST 151129 was isolated. Using sequence alignments it was determined that EST 151129 sequence included nucleotides 115-1536, encoding amino acids 39-512 of the full length peptide. The isolated cDNA provided nucleotides 1-114 and additional upstream non-coding nucleotides. These added nucleotides encode amino acids 1-38 of the full length human semaphorin, amino acid 1 being the initiating methionine. [0076]
  • In order to isolate cDNA overlapping the 3′ end of EST 151129, another radiolabeled PCR probe was generated as described above but using PCR oligonucleotide primers specific for the 3′ end of EST 151129: [0077]
    5′-AGCCAGGTGCCCCTGGACCT-3′ (SEQ ID NO:9)
    5′-CTCGAGGCCAAGAATTCGGC-3′ (SEQ ID NO:10)
  • Approximately 5×10[0078] 5 cpm/mL of purified probe was used to hybridize with the human foreskin fibroblast library on nylon membrane filters following the hybridization and washing protocol described above. Three independent cDNA clones were isolated which overlapped with the 3′ end of EST151129 and extended the 3′ end of EST 151129 from nucleotide 1537 to 2001. The isolated clones contained additional downstream non-coding nucleotides. Nucleotides 1537-2001 translate into amino acids 513-666 of the full length peptide plus a stop codon. The full cDNA sequence of the human DCSema homologue is provided in SEQ ID NO: 1. The encoded amino acid sequence is shown in SEQ ID NO:2.
  • EXAMPLE 2 Northern Blot Analysis for Tissue Expressing the Human Homologue
  • Radiolabeled probes derived from SEQ ID NO:1 were used in Northern blot analyses of different human tissues to identify the tissue distribution of the human homologue of AHV Sema, designated DCSema. [0079]
  • Human poly A+ multiple tissue northern blots were purchased form Clontech Laboratories, Palo Alto, Calif. (Cat. #s 7760-1,7759-1, 7756-1, 757-1). The northern blot filters were prehybridized, probed, and washed according to manufacturer's instructions. The probe was an antisense riboprobe specific for EST 151129, an EST discovered using the viral AHV Sema sequence in sequence alignment analyses and comparative analysis methods. The template for the riboprobe was generated using PCR techniques and oligonucleotide primers that were designed to span nucleotides 613 to 978 of EST 151129(HO2902/H). The primers sequences were as follows: [0080]
    5′ Primer: TCTACTACTT CTTCC (SEQ ID NO:5)
    3′ Primer GGAATCCTAA TACGACTCAC TATAGGGAGG CGGGTTGGGA AGGC (SEQ ID NO:6)
  • The underlined portion of the downstream primer is a T7 site. [0081]
  • The riboprobe was generated using Ambion's MAXIscript SP6/T7 kit by combining 3 μL of RNAse free water, 2 μL 10× transcription buffer, 1 μL each of 10 mM dATP/dCTP/dGTP, 5 μL 5′/3′ EST 151129 PCR Product, 5 μL Amersham [α][0082] 32P]UTP 10 mCi/mL, and 2 μL T7 RNA polymerase at room temperature. The combination was microfuged, spun briefly, and incubated at 37° C. for 30 minutes. Then 1 μL DNAse was added to the mixture and allowed to react for 15 minutes at 37° C. The reaction product was passed through two column volumes of G-25 (Boehringer). One microliter (1 μL) of the riboprobe was counted in a scintillation counter for 1 minute to determine cpm/μL.
  • After probing the northern blots, they were washed once for 30 minutes with 2×SSC, 0.05% SDS at 63° C. and three times for 30 minutes with 0.1×SSC, 0.1% SDS and then exposed to x-ray film. The developed film indicated that the human DCSema homologue is found in placenta, brain, spinal cord, testes and spleen. Weak hybridization signals were observed in skeletal muscle, lymph node, ovary, and bone marrow. [0083]
  • EXAMPLE 3 Preparing a DC-Sema/Fc Fusion Protein
  • This example describes preparing a DC-Sema/Fc DNA construct and subsequently expressing a DC-Sema/immunoglobulin Fc fusion protein referred to as DC-Sema/Fc. DNA encoding DC-Sema/Fc included a nucleotide sequence that encodes a murine IL-7 leader peptide, a FLAG™ octapeptide (described in U.S. Pat. No. 5,011,912), an Fc region of an immunoglobulin mutated to minimize binding to Fc receptor (described by Baum et al. [0084] Cir. Sh. 44:30, 1994), a flexible linker sequence and DNA encoding amino acids 45-666 of SEQ ID NO:2. An expression vector containing the leader sequence, FLAG, mutated hu IgG Fc and flexible linker was prepared using conventional enzyme cutting and ligation techniques. The resulting vector was then restricted with Spe1 and Not1. The DC-Sema was inserted 5′ to 3′ after the flexible linker in a two-way ligation described below.
  • To prepare the DC-Sema DNA, three primer pairs were designed and used to amplify three DNA fragments. Two of the fragments were amplified from a human foreskin fibroblast library phage clone containing the 3′ portion of the DCSema cDNA and one fragment was EST DNA (Research Genetics Imageclone ID #151129). The three amplified DNA fragments were combined in a PCR SOEing reaction to generate a DNA fragment encoding the entire peptide of DC-Sema (Horton et al., Biotechniques 8:528, 1990). In the final fragment, the upstream oligonucleotide primer introduced a Spe1 site upstream of amino acid 45 of the DC-sema peptide. A downstream oligonucleotide primer introduced a Not1 site just downstream of the termination codon after amino acid 666. [0085]
  • The PCR fragment was then ligated into an expression vector (pDC409) containing the leader sequence. Flag® sequence, mutated human IgG Fc and a flexible linker region in a two-way ligation. The resulting DNA construct was transfected into the monkey kidney cell lines CV-1/EBNA. After 7 days of culture in medium containing 0.5% low immunoglobulin bovine serum, a solution of 0.2% azide was added to the supernatant and the supernatant was filtered through a 0.22 μm filter. Then approximately 1 L of culture supernatant was passed through a BioCad Protein A HPLC protein purification system using a 4.6×100 mm Protein A column (POROS 20A from PerSeptive Biosystems) at 10 mL/min. The Protein A column binds the Fc Portion of the fusion protein in the supernatant, immobilizing the fusion protein and allowing other components of the supernatant to pass through the column. The column was washed with 30 mL of PBS solution and bound fusion protein was eluted from the HPLC column with citric acid adjusted to pH 3.0. Eluted purified fusion protein was neutralized as it eluted using 1M HEPES solution at pH 7.4. [0086]
  • EXAMPLE 4 Preparing DC-Sema/polyHIS Fusion Protein
  • This example describes preparing a DC-Sema/polyHIS DNA construct and subsequently expressing a DCSema/poly histidine tagged protein referred to as DC-sema/polyHIS fusion protein. DNA encoding DC-sema/polyHIS comprises sequences encoding the DC-sema gene from amino acid 1 to amino acid 666 (no stop codon), followed by a factor Xa cleavage site, a FLAG® octapeptide, a string of six histidine residues and a stop codon. An expression vector containing the factor Xa cleavage site, flag, and polyHIS sequence was prepared using conventional enzyme cutting and fragment ligation techniques. The resulting vector was then restricted with Sal1 and SnaB 1. The DCSema sequence was inserted 5′ to 3′ before the factor Xa cleavage site in a two-way ligation described below. [0087]
  • To prepare the DCSema DNA, three primer pairs were designed and used to amplify three DNA fragments. Two of the fragments were from phage DNA and one fragment was EST DNA (Research Genetics Imageclone ID #151129). The three amplified DNA fragments were combined in a PCR SOEing reaction to generate a DNA fragment encoding the entire peptide of DCSema. This final DNA included a Sal1 site upstream of amino acid 1 of the DCSema peptide and a SnaB1 site downstream of amino acid 666. [0088]
  • The PCR product was then ligated into an expression vector pDC409 containing the factor Xa cleavage site, flag, and polyHIS sequence in a two-way ligation. The resultant DNA construct (DCSema/polyHIS) was transiently transfected into the monkey cell line COS-1 (ATCC CRL-1650). Following a 7 day culture in medium containing 0.5% low immunoglobulin bovine serum, cell supernatants were harvested and a solution of 0.2% sodium azide was added to the supernatants. The supernatants were filtered through a 0.22 μm filter, concentrated 10 fold with a prep scale concentrator (Millipore: Bedford, Mass.) and purified on a BioCad HPLC protein purification equipped with a Nickel NTA Superflow self pack resin column (Qiagen, Santa Clarita, Calif.). After the supernatant passed through the column, the column was washed with Buffer A (20 mM NaPO4, pH7.4; 300 mMNaCl; 50 mM Imidazole). Bound protein was then eluted from the column using a gradient elution techniques. Fractions containing protein were collected and analyzed on a 4-20% SDS-PAGE reducing gel. Peaks containing the fusion protein were pooled, concentrated 2 fold, and then dialyzed in PBS. The resulting DCSema/polyHis fusion protein was then filtered through a 0.22 μm sterile filter and recovered. [0089]
  • EXAMPLE 5 Screening Cell Lines for Binding to DCSema
  • The DC-Sema/Fc fusion protein prepared as described in Example 3 was used to screen cell lines for binding using quantitative binding studies according to standard flow cytometry methodologies. For each cell line screened, the procedure involved incubating approximately 100,000 of the cells blocked with 2% FCS (fetal calf serum), 5% normal goat serum and 5% rabbit serum in PBS for 1 hour. Then the blocked cells were incubated with 5 μg/mL of DC-Sema/Fc fusion protein in 2% FCS, 5% goat serum and 5% rabbit serum in PBS. Following the incubation the sample was washed 2 times with FACS buffer (2% FCS in PBS) and then treated with mouse anti human Fc/biotin (purchased from Jackson Research) and SAPE (streptavidin-phycoerythrin purchased from Molecular Probes). This treatment causes the antihuman Fc/biotin to bind to any bound DC-Sema/Fc and the SAPE to bind to the anti human Fc/biotin resulting in a fluorescent identifying label on DC-Sema/Fc which is bound to cells. The cells were analyzed for any bound protein using fluorescent detection flow cytometry. Table I details the results of the flow cytometry studies. +indicates that binding was detected between the cell surface and DC-Sema. −indicates that no binding was detected between the cell surface and A39R. [0090]
    TABLE I
    Cell Line DC-Sema Binding Result
    CB23 (Human Cord Blood B Cell Line) +
    MP-1 (Human B Cell Lymphoma) +
    PB B (Human Peripheral Blood B Cells) +
    U937 (Human Monocyte-Type Cell) +
    W126 (Human Lung Epithelium) +
    RAJI (Burkitt's Lymphoma) +
    Primary Human Monocytes +
    THP-1 (Human Promonocytes) +
  • EXAMPLE 6 Monoclonal Antibodies to DC-Sema
  • This example illustrates a method for preparing antibodies to DC-Sema. Purified DC-Sema/Fc is prepared as described in Example 3 above. The purified protein is used to generate antibodies against DC-Sema as described in U.S. Pat. No. 4,411,993. Briefly, mice are immunized at 0, 2 and 6 weeks with 10 μg with DCSema/Fc. The primary immunization is prepared with TITERMAX adjuvant, from Vaxcell, Inc., and subsequent immunizations are prepared with incomplete Freund's adjuvant (IFA). At 11 weeks, the mice are IV boosted with 3-4 μg DCSema/Fc in PBS. Three days after the IV boost, splenocytes are harvested and fused with an Ag8.653 myeloma fusion partner using 50% aqueous PEG 1500 solution. Hybridoma supernatants are screened for DC-Sema antibodies by dot blot assay against DCSema/FC and an irrelevant Fc protein. [0091]
  • EXAMPLE 7 Cytokine Induction from Freshly Isolated Human Monocytes
  • Freshly isolated human monocytes were purified by first diluting 1:1 peripheral blood from healthy donors in low endotoxin PBS at pH 7.4 and room temperature. Then 35 mLs of the diluted blood was layered over 15 mLs of Isolymph (Gallard and Schlesinger Industries, Inc; Carle Place, N.Y.) and centrifuged at 2200 rpm for 25 minutes at room temperature. The plasma layers was reserved. The PBMC layer was harvested and washed three times to remove the Isolymph. The washed PBMC's were resuspended in X-Vivo 15 serum free media (BioWhittaker, Walkersville, Md.) and added to T175 flasks. The flasks had been previously coated with 2% Gelatin (Sigma, St. Louis, Mo.) and pre-treated for 30 minutes with the reserved plasma layer. The PBMC's were allowed to adhere for 90 minutes at 37° C., 5% CO[0092] 2 and then rinsed three times gently with 10 mL washes of low endotoxin PBS. Adhered monocytes were harvested by incubating the cells in Enzyme Free Dissociation Buffer (Gibco, BRL) and washing the cells multiple times in PBS. Monocytes were centrifuged at 2500 rpm for 5 minutes, counted, and set up in 24 well dishes at 5×105 cells/well in 1 mL. The cultures were 95% pure.
  • Purified monocytes were cultured for 7-9 days in the presence of 20 ng/mL GM-CSF and 100 ng/mL IL-4 in order to allow cells to differentiate to a more dendritic cell-like phenotype. On day 7-9, cultures were treated with 1 μg/mL DCSema/Fc fusion protein (see Example 3) or a control Fc protein, and the next day cells and supernatants were harvested for analyses. [0093]
  • The monocyte supernatants were examined for the presence of proinflammatory cytokines. In all donors tested, IL-6 and IL-8 was induced by DCSema protein. Heat inactivated DCSema and control proteins did not induce IL-6 or IL-8. Additionally, cytokine production was blocked by the inclusion of a mAb directed against DCSema. [0094]
  • The results of this experiment demonstrate that DCSema, or homologues of this protein, by interact with its receptor, can induce cytokine production by freshly isolated monocytes. [0095]
  • EXAMPLE 8 Monocyte Aggregation Studies
  • In order to examine human monocyte response to the interaction of a DCSema to its receptor on monocytes, monocytes were purified as described in Example 7 and DC-Sema/Fc fusion protein was prepared as described in Example 3. After incubating the DCSema/Fc fusion protein and purified, cultured monocytes for 20 hours, monocyte aggregation was observed. [0096]
  • This work confirms that the receptor for the DCSema of the present invention is expressed on monocytes and that the interaction between DCSema and it receptor results in monocyte aggregation. Similar to B cells, monocyte aggregation is indicative of their activation. [0097]
  • 1 10 1 2001 DNA Homo sapiens CDS (1)..(2001) 1 atg acg cct cct ccg ccc gga cgt gcc gcc ccc agc gca ccg cgc gcc 48 Met Thr Pro Pro Pro Pro Gly Arg Ala Ala Pro Ser Ala Pro Arg Ala 1 5 10 15 cgc gtc cct ggc ccg ccg gct cgg ttg ggg ctt ccg ctg cgg ctg cgg 96 Arg Val Pro Gly Pro Pro Ala Arg Leu Gly Leu Pro Leu Arg Leu Arg 20 25 30 ctg ctg ctg ctg ctt tgg gcg gcc gcc gcc tcc gcc cag ggc cac cta 144 Leu Leu Leu Leu Leu Trp Ala Ala Ala Ala Ser Ala Gln Gly His Leu 35 40 45 agg agc gga ccc cgc atc ttc gcc gtc tgg aaa ggc cat gta ggg cag 192 Arg Ser Gly Pro Arg Ile Phe Ala Val Trp Lys Gly His Val Gly Gln 50 55 60 gac cgg gtg gac ttt ggc cag act gag ccg cac acg gtg ctt ttc cac 240 Asp Arg Val Asp Phe Gly Gln Thr Glu Pro His Thr Val Leu Phe His 65 70 75 80 gag cca ggc agc tcc tct gtg tgg gtg gga gga cgt ggc aag gtc tac 288 Glu Pro Gly Ser Ser Ser Val Trp Val Gly Gly Arg Gly Lys Val Tyr 85 90 95 ctc ttt gac ttc ccc gag ggc aag aac gca tct gtg cgc acg gtg aat 336 Leu Phe Asp Phe Pro Glu Gly Lys Asn Ala Ser Val Arg Thr Val Asn 100 105 110 atc ggc tcc aca aag ggg tcc tgt ctg gat aag cgg gac tgc gag aac 384 Ile Gly Ser Thr Lys Gly Ser Cys Leu Asp Lys Arg Asp Cys Glu Asn 115 120 125 tac atc act ctc ctg gag agg cgg agt gag ggg ctg ctg gcc tgt ggc 432 Tyr Ile Thr Leu Leu Glu Arg Arg Ser Glu Gly Leu Leu Ala Cys Gly 130 135 140 acc aac gcc cgg cac ccc agc tgc tgg aac ctg gtg aat ggc act gtg 480 Thr Asn Ala Arg His Pro Ser Cys Trp Asn Leu Val Asn Gly Thr Val 145 150 155 160 gtg cca ctt ggc gag atg aga ggc tac gcc ccc ttc agc ccg gac gag 528 Val Pro Leu Gly Glu Met Arg Gly Tyr Ala Pro Phe Ser Pro Asp Glu 165 170 175 aac tcc ctg gtt ctg ttt gaa ggg gac gag gtg tat tcc acc atc cgg 576 Asn Ser Leu Val Leu Phe Glu Gly Asp Glu Val Tyr Ser Thr Ile Arg 180 185 190 aag cag gaa tac aat ggg aag atc cct cgg ttc cgc cgc atc cgg ggc 624 Lys Gln Glu Tyr Asn Gly Lys Ile Pro Arg Phe Arg Arg Ile Arg Gly 195 200 205 gag agt gag ctg tac acc agt gat act gtc atg cag aac cca cag ttc 672 Glu Ser Glu Leu Tyr Thr Ser Asp Thr Val Met Gln Asn Pro Gln Phe 210 215 220 atc aaa gcc acc atc gtg cac caa gac cag gct tac gat gac aag atc 720 Ile Lys Ala Thr Ile Val His Gln Asp Gln Ala Tyr Asp Asp Lys Ile 225 230 235 240 tac tac ttc ttc cga gag gac aat cct gac aag aat cct gag gct cct 768 Tyr Tyr Phe Phe Arg Glu Asp Asn Pro Asp Lys Asn Pro Glu Ala Pro 245 250 255 ctc aat gtg tcc cgt gtg gcc cag ttg tgc agg ggg gac cag ggt ggg 816 Leu Asn Val Ser Arg Val Ala Gln Leu Cys Arg Gly Asp Gln Gly Gly 260 265 270 gaa agt tca ctg tca gtc tcc aag tgg aac act ttt ctg aaa gcc atg 864 Glu Ser Ser Leu Ser Val Ser Lys Trp Asn Thr Phe Leu Lys Ala Met 275 280 285 ctg gta tgc agt gat gct gcc acc aac aag aac ttc aac agg ctg caa 912 Leu Val Cys Ser Asp Ala Ala Thr Asn Lys Asn Phe Asn Arg Leu Gln 290 295 300 gac gtc ttc ctg ctc cct gac ccc agc ggc cag tgg agg gac acc agg 960 Asp Val Phe Leu Leu Pro Asp Pro Ser Gly Gln Trp Arg Asp Thr Arg 305 310 315 320 gtc tat ggt gtt ttc tcc aac ccc tgg aac tac tca gcc gtc tgt gtg 1008 Val Tyr Gly Val Phe Ser Asn Pro Trp Asn Tyr Ser Ala Val Cys Val 325 330 335 tat tcc ctc ggt gac att gac aag gtc ttc cgt acc tcc tca ctc aag 1056 Tyr Ser Leu Gly Asp Ile Asp Lys Val Phe Arg Thr Ser Ser Leu Lys 340 345 350 ggc tac cac tca agc ctt ccc aac ccg cgg cct ggc aag tgc ctc cca 1104 Gly Tyr His Ser Ser Leu Pro Asn Pro Arg Pro Gly Lys Cys Leu Pro 355 360 365 gac cag cag ccg ata ccc aca gag acc ttc cag gtg gct gac cgt cac 1152 Asp Gln Gln Pro Ile Pro Thr Glu Thr Phe Gln Val Ala Asp Arg His 370 375 380 cca gag gtg gcg cag agg gtg gag ccc atg ggg cct ctg aag acg cca 1200 Pro Glu Val Ala Gln Arg Val Glu Pro Met Gly Pro Leu Lys Thr Pro 385 390 395 400 ttg ttc cac tct aaa tac cac tac cag aaa gtg gcc gtt cac cgc atg 1248 Leu Phe His Ser Lys Tyr His Tyr Gln Lys Val Ala Val His Arg Met 405 410 415 caa gcc agc cac ggg gag acc ttt cat gtg ctt tac cta act aca gac 1296 Gln Ala Ser His Gly Glu Thr Phe His Val Leu Tyr Leu Thr Thr Asp 420 425 430 agg ggc act atc cac aag gtg gtg gaa ccg ggg gag cag gag cac agc 1344 Arg Gly Thr Ile His Lys Val Val Glu Pro Gly Glu Gln Glu His Ser 435 440 445 ttc gcc ttc aac atc atg gag atc cag ccc ttc cgc cgc gcg gct gcc 1392 Phe Ala Phe Asn Ile Met Glu Ile Gln Pro Phe Arg Arg Ala Ala Ala 450 455 460 atc cag acc atg tcg ctg gat gct gag cgg agg aag ctg tat gtg agc 1440 Ile Gln Thr Met Ser Leu Asp Ala Glu Arg Arg Lys Leu Tyr Val Ser 465 470 475 480 tcc cag tgg gag gtg agc cag gtg ccc ctg gac ctg tgt gag gtc tat 1488 Ser Gln Trp Glu Val Ser Gln Val Pro Leu Asp Leu Cys Glu Val Tyr 485 490 495 ggc ggg ggc tgc cac ggt tgc ctc atg tcc cga gac ccc tac tgc ggc 1536 Gly Gly Gly Cys His Gly Cys Leu Met Ser Arg Asp Pro Tyr Cys Gly 500 505 510 tgg gac cag ggc cgc tgc atc tcc atc tac agc tcc gaa cgg tca gtg 1584 Trp Asp Gln Gly Arg Cys Ile Ser Ile Tyr Ser Ser Glu Arg Ser Val 515 520 525 ctg caa tcc att aat cca gcc gag cca cac aag gag tgt ccc aac ccc 1632 Leu Gln Ser Ile Asn Pro Ala Glu Pro His Lys Glu Cys Pro Asn Pro 530 535 540 aaa cca gac aag gcc cca ctg cag aag gtt tcc ctg gcc cca aac tct 1680 Lys Pro Asp Lys Ala Pro Leu Gln Lys Val Ser Leu Ala Pro Asn Ser 545 550 555 560 cgc tac tac ctg agc tgc ccc atg gaa tcc cgc cac gcc acc tac tca 1728 Arg Tyr Tyr Leu Ser Cys Pro Met Glu Ser Arg His Ala Thr Tyr Ser 565 570 575 tgg cgc cac aag gag aac gtg gag cag agc tgc gaa cct ggt cac cag 1776 Trp Arg His Lys Glu Asn Val Glu Gln Ser Cys Glu Pro Gly His Gln 580 585 590 agc ccc aac tgc atc ctg ttc atc gag aac ctc acg gcg cag cag tac 1824 Ser Pro Asn Cys Ile Leu Phe Ile Glu Asn Leu Thr Ala Gln Gln Tyr 595 600 605 ggc cac tac ttc tgc gag gcc cag gag ggc tcc tac ttc cgc gag gct 1872 Gly His Tyr Phe Cys Glu Ala Gln Glu Gly Ser Tyr Phe Arg Glu Ala 610 615 620 cag cac tgg cag ctg ctg ccc gag gac ggc atc atg gcc gag cac ctg 1920 Gln His Trp Gln Leu Leu Pro Glu Asp Gly Ile Met Ala Glu His Leu 625 630 635 640 ctg ggt cat gcc tgt gcc ctg gct gcc tcc ctc tgg ctg ggg gtg ctg 1968 Leu Gly His Ala Cys Ala Leu Ala Ala Ser Leu Trp Leu Gly Val Leu 645 650 655 ccc aca ctc act ctt ggc ttg ctg gtc cac tag 2001 Pro Thr Leu Thr Leu Gly Leu Leu Val His 660 665 2 666 PRT Homo sapiens 2 Met Thr Pro Pro Pro Pro Gly Arg Ala Ala Pro Ser Ala Pro Arg Ala 1 5 10 15 Arg Val Pro Gly Pro Pro Ala Arg Leu Gly Leu Pro Leu Arg Leu Arg 20 25 30 Leu Leu Leu Leu Leu Trp Ala Ala Ala Ala Ser Ala Gln Gly His Leu 35 40 45 Arg Ser Gly Pro Arg Ile Phe Ala Val Trp Lys Gly His Val Gly Gln 50 55 60 Asp Arg Val Asp Phe Gly Gln Thr Glu Pro His Thr Val Leu Phe His 65 70 75 80 Glu Pro Gly Ser Ser Ser Val Trp Val Gly Gly Arg Gly Lys Val Tyr 85 90 95 Leu Phe Asp Phe Pro Glu Gly Lys Asn Ala Ser Val Arg Thr Val Asn 100 105 110 Ile Gly Ser Thr Lys Gly Ser Cys Leu Asp Lys Arg Asp Cys Glu Asn 115 120 125 Tyr Ile Thr Leu Leu Glu Arg Arg Ser Glu Gly Leu Leu Ala Cys Gly 130 135 140 Thr Asn Ala Arg His Pro Ser Cys Trp Asn Leu Val Asn Gly Thr Val 145 150 155 160 Val Pro Leu Gly Glu Met Arg Gly Tyr Ala Pro Phe Ser Pro Asp Glu 165 170 175 Asn Ser Leu Val Leu Phe Glu Gly Asp Glu Val Tyr Ser Thr Ile Arg 180 185 190 Lys Gln Glu Tyr Asn Gly Lys Ile Pro Arg Phe Arg Arg Ile Arg Gly 195 200 205 Glu Ser Glu Leu Tyr Thr Ser Asp Thr Val Met Gln Asn Pro Gln Phe 210 215 220 Ile Lys Ala Thr Ile Val His Gln Asp Gln Ala Tyr Asp Asp Lys Ile 225 230 235 240 Tyr Tyr Phe Phe Arg Glu Asp Asn Pro Asp Lys Asn Pro Glu Ala Pro 245 250 255 Leu Asn Val Ser Arg Val Ala Gln Leu Cys Arg Gly Asp Gln Gly Gly 260 265 270 Glu Ser Ser Leu Ser Val Ser Lys Trp Asn Thr Phe Leu Lys Ala Met 275 280 285 Leu Val Cys Ser Asp Ala Ala Thr Asn Lys Asn Phe Asn Arg Leu Gln 290 295 300 Asp Val Phe Leu Leu Pro Asp Pro Ser Gly Gln Trp Arg Asp Thr Arg 305 310 315 320 Val Tyr Gly Val Phe Ser Asn Pro Trp Asn Tyr Ser Ala Val Cys Val 325 330 335 Tyr Ser Leu Gly Asp Ile Asp Lys Val Phe Arg Thr Ser Ser Leu Lys 340 345 350 Gly Tyr His Ser Ser Leu Pro Asn Pro Arg Pro Gly Lys Cys Leu Pro 355 360 365 Asp Gln Gln Pro Ile Pro Thr Glu Thr Phe Gln Val Ala Asp Arg His 370 375 380 Pro Glu Val Ala Gln Arg Val Glu Pro Met Gly Pro Leu Lys Thr Pro 385 390 395 400 Leu Phe His Ser Lys Tyr His Tyr Gln Lys Val Ala Val His Arg Met 405 410 415 Gln Ala Ser His Gly Glu Thr Phe His Val Leu Tyr Leu Thr Thr Asp 420 425 430 Arg Gly Thr Ile His Lys Val Val Glu Pro Gly Glu Gln Glu His Ser 435 440 445 Phe Ala Phe Asn Ile Met Glu Ile Gln Pro Phe Arg Arg Ala Ala Ala 450 455 460 Ile Gln Thr Met Ser Leu Asp Ala Glu Arg Arg Lys Leu Tyr Val Ser 465 470 475 480 Ser Gln Trp Glu Val Ser Gln Val Pro Leu Asp Leu Cys Glu Val Tyr 485 490 495 Gly Gly Gly Cys His Gly Cys Leu Met Ser Arg Asp Pro Tyr Cys Gly 500 505 510 Trp Asp Gln Gly Arg Cys Ile Ser Ile Tyr Ser Ser Glu Arg Ser Val 515 520 525 Leu Gln Ser Ile Asn Pro Ala Glu Pro His Lys Glu Cys Pro Asn Pro 530 535 540 Lys Pro Asp Lys Ala Pro Leu Gln Lys Val Ser Leu Ala Pro Asn Ser 545 550 555 560 Arg Tyr Tyr Leu Ser Cys Pro Met Glu Ser Arg His Ala Thr Tyr Ser 565 570 575 Trp Arg His Lys Glu Asn Val Glu Gln Ser Cys Glu Pro Gly His Gln 580 585 590 Ser Pro Asn Cys Ile Leu Phe Ile Glu Asn Leu Thr Ala Gln Gln Tyr 595 600 605 Gly His Tyr Phe Cys Glu Ala Gln Glu Gly Ser Tyr Phe Arg Glu Ala 610 615 620 Gln His Trp Gln Leu Leu Pro Glu Asp Gly Ile Met Ala Glu His Leu 625 630 635 640 Leu Gly His Ala Cys Ala Leu Ala Ala Ser Leu Trp Leu Gly Val Leu 645 650 655 Pro Thr Leu Thr Leu Gly Leu Leu Val His 660 665 3 20 DNA Artificial Sequence PRIMER 3 tgctggaacc tggtgaatgg 20 4 20 DNA Artificial Sequence PRIMER 4 agtggaacaa tggcgtcttc 20 5 15 DNA Artificial Sequence PRIMER 5 tctactactt cttcc 15 6 44 DNA Artificial Sequence PRIMER 6 ggaatcctaa tacgactcac tatagggagg cgggttggga aggc 44 7 27 DNA Artificial Sequence PRIMER 7 tccgcccagg gccacctaag gagcgga 27 8 27 DNA Artificial Sequence PRIMER 8 tgtgcggctc agtctggcca aagtcca 27 9 20 DNA Artificial Sequence PRIMER 9 agccaggtgc ccctggacct 20 10 20 DNA Artificial Sequence PRIMER 10 ctcgaggcca agaattcggc 20

Claims (23)

What is claimed is:
1. A semaphorin polypeptide comprising an amino acid sequence that is at least 80% identical to the amino acid sequence of SEQ ID NO:2, the semaphorin polypeptide capable of binding at least one semaphorin receptor.
2. An isolated semaphorin polypeptide encoded by DNA sequences selected from the group consisting of:
(a) nucleotides x1—2001 of SEQ ID NO:1, wherein x1 is nucleotide 1 or 135; and
(b) DNA sequences that hybridize under moderately stringent conditions to the DNA of (a); and which DNA sequences encode a polypeptide that binds semaphorin receptors.
3. A semaphorin polypeptide comprising an amino acid sequence that is at least 80% identical to an amino acid sequence selected from the group consisting of:
(a) amino acids x1 to 666 of SEQ ID NO: 2, wherein x1 is amino acid 1 or 45. a fragment of the sequence of (a), wherein the fragment is capable of binding a semaphorin receptor.
4. A semaphorin polypeptide comprising an amino acid sequence selected from the group consisting of:
(a) amino acids x1 to 666 of SEQ ID NO: 2, wherein x1 is amino acid 1 or 45, and
(b) a fragment of the sequence of (a), wherein the fragment is capable of binding a semaphorin receptor.
5. An isolated DNA encoding a semaphorin polypeptide, the DNA selected from the group consisting of:
(a) DNA capable of hybridizing under moderately stringent conditions to a nucleotide sequence consisting essentially of SEQ ID NO: 1; and
(b) DNA capable of hybridizing under moderately stringent conditions to DNA complementary to the sequence of (a).
6. An isolated DNA encoding a semaphorin polypeptide, the DNA selected from the group consisting of:
(a) nucleotides x1—2001 of SEQ ID NO:1, wherein x1 is nucleotide 1 or 135; and
(b) DNA sequences that hybridize under moderately stringent conditions to the cDNA of (a); and which DNA sequences encode a polypeptide that binds semaphorin receptor; and
(c) DNA sequences that, due to the degeneracy of the genetic code, encode semaphorin polypeptides having the amino acid sequence of the polypeptides encoded by the DNA sequences of (a) or (b).
7. An isolated DNA encoding an semaphorin polypeptide wherein the polypeptide comprises an amino acid sequence that is at least 80% identical to the amino acid sequence of SEDQ ID NO:2.
8. The isolated DNA of claim 7 wherein the semaphorin polypeptide comprises the amino acid sequence of SEQ ID NO:2.
9. An isolated DNA encoding a semaphorin polypeptide, wherein the polypeptide comprises an amino acid sequence that is at least 80% identical to a sequence selected from the group consisting of:
(a) amino acids x1—666 of SEQ ID NO:2, wherein x1 is amino acid 1 or 45; and
(b) a fragment of the sequence of (a), wherein the polypeptide binds a semaphorin receptor.
10. A DNA of claim 9 wherein the, semaphorin polypeptide comprises an amino acid sequence selected from the group consisting of:
(a) amino acids x1—666 of SEQ ID NO:2, wherein x1 is amino acid 1 or 45; and
(b) a fragment of (a).
11. A DNA that is at least 80% identical to DNA that encodes an amino acid sequence selected from the group consisting:
(a) amino acids x1—666 of SEQ ID NO:2, wherein x1 is amino acid 1 or 45; and
(b) a fragment of (a).
12. A fusion protein comprising amino acids x1—666 where x1 is amino acid 1 or 45 of SEQ ID NO:2
13. A recombinant expression vector comprising DNA of claim 6.
14. A process for preparing a semaphorin polypeptide, the process comprising culturing a host cell transformed with an expression vector of claim 14 under conditions that promote expression of the polypeptide, and recovering the polypeptide.
15. A composition comprising a suitable diluent carrier and a polypeptide of claim 3.
16. An antibody that is immunoreactive with a polypeptide of claim 2.
17. A process for treating an inflammatory disease in a mammal afflicted with the disease, the process comprising administering an amount of semaphorin polypeptide.
18. A method of separating cells having semaphorin polypeptide on the surface thereof from a mixture of cells in suspension, comprising contacting the cells in the mixture with a contacting surface having a semaphorin, and separating the contacting surface and the suspension.
19. An isolated DNA selected from the group consisting of:
(a) DNA of SEQ ID NO:5;
(b) DNA sequences that hybridize under moderately stringent conditions to the cDNA of (a); and which DNA sequences encode a polypeptide the binds semaphorin receptor;
(c) DNA sequences that, due to the degeneracy of the genetic code, encode semaphorin polypeptides having the amino acid sequence of the polypeptides encoded by the DNA sequences of (a) or (b).
20. An isolated DNA encoding a soluble semaphorin polypeptide, wherein the soluble polypeptide comprises an amino acid sequence that is at least 90% identical to a sequence selected from the group consisting of:
(a) amino acids x1 to 666 of SEQ ID NO:6, wherein x1 is amino acid 1 or 45;
(b) a fragment of the sequence of (a), wherein the soluble polypeptide binds a semaphorin receptor.
21. A DNA of claim 29 wherein the soluble polypeptide comprises an amino acid sequence selected from the group consisting of:
(a) amino acids x1 to 666 of SEQ ID NO:6, wherein x1 is amino acid 1 or 45; and
(b) a fragment of (a).
22. A recombinant expression vector comprising DNA of claim 21.
23. A process for preparing a semaphorin polypeptide, the process comprising culturing a host cell transformed with an expression vector of claim 31 under conditions that promote expression of the polypeptide, and recovering the polypeptide.
US10/740,773 1998-05-14 2003-12-19 Semaphorin polypeptides Abandoned US20040180825A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/740,773 US20040180825A1 (en) 1998-05-14 2003-12-19 Semaphorin polypeptides

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US8549798P 1998-05-14 1998-05-14
PCT/US1999/009831 WO1999058676A2 (en) 1998-05-14 1999-05-05 Semaphorin polypeptides
US09/689,012 US6670135B1 (en) 1998-05-14 2000-10-12 Semaphorin polypeptides
US10/740,773 US20040180825A1 (en) 1998-05-14 2003-12-19 Semaphorin polypeptides

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/689,012 Division US6670135B1 (en) 1998-05-14 2000-10-12 Semaphorin polypeptides

Publications (1)

Publication Number Publication Date
US20040180825A1 true US20040180825A1 (en) 2004-09-16

Family

ID=22192003

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/689,012 Expired - Fee Related US6670135B1 (en) 1998-05-14 2000-10-12 Semaphorin polypeptides
US10/740,773 Abandoned US20040180825A1 (en) 1998-05-14 2003-12-19 Semaphorin polypeptides

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/689,012 Expired - Fee Related US6670135B1 (en) 1998-05-14 2000-10-12 Semaphorin polypeptides

Country Status (7)

Country Link
US (2) US6670135B1 (en)
EP (1) EP1076697B1 (en)
AT (1) ATE408687T1 (en)
AU (1) AU3787499A (en)
DE (1) DE69939587D1 (en)
ES (1) ES2315011T3 (en)
WO (1) WO1999058676A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080279771A1 (en) * 2005-10-29 2008-11-13 Ge Healthcare Limited Novel Imaging Agents for Cancer

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0892047A3 (en) 1997-07-09 2000-03-08 Hoechst Marion Roussel Deutschland GmbH Human and murine semaphorin L
AU2669501A (en) * 1999-11-30 2001-06-12 Smithkline Beecham Plc New use

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6225285B1 (en) * 1998-03-11 2001-05-01 Exelixis Pharmaceuticals, Inc. Semaphorin K1

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0892047A3 (en) * 1997-07-09 2000-03-08 Hoechst Marion Roussel Deutschland GmbH Human and murine semaphorin L
WO1999021997A1 (en) * 1997-10-28 1999-05-06 Immunex Corporation Viral encoded semaphorin protein receptor dna and polypeptides
EP0933425A1 (en) 1998-01-30 1999-08-04 Smithkline Beecham Plc Semaphorin family polypeptides and polynucleotides
WO1999045114A2 (en) * 1998-03-03 1999-09-10 Zymogenetics, Inc. Human semaphorin zsmf-7

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6225285B1 (en) * 1998-03-11 2001-05-01 Exelixis Pharmaceuticals, Inc. Semaphorin K1

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080279771A1 (en) * 2005-10-29 2008-11-13 Ge Healthcare Limited Novel Imaging Agents for Cancer

Also Published As

Publication number Publication date
EP1076697A2 (en) 2001-02-21
WO1999058676A2 (en) 1999-11-18
AU3787499A (en) 1999-11-29
ATE408687T1 (en) 2008-10-15
US6670135B1 (en) 2003-12-30
WO1999058676A3 (en) 2000-01-20
ES2315011T3 (en) 2009-03-16
EP1076697B1 (en) 2008-09-17
DE69939587D1 (en) 2008-10-30

Similar Documents

Publication Publication Date Title
US7939640B2 (en) Antibodies that bind B7L-1
US7659385B2 (en) Polynucleotides encoding molecules designated LDCAM
NZ502884A (en) DNA and peptide sequences encoding the cytokine LERK-6 that bind to the hek or elk receptor
NZ295022A (en) Lerk-6 polypeptide derivatives, proteins, dna and host cells thereof
US6670135B1 (en) Semaphorin polypeptides
EP1027436A1 (en) Viral encoded semaphorin protein receptor dna and polypeptides
US7214497B2 (en) Viral encoded semaphorin protein receptor DNA and polypeptides
US6174689B1 (en) Viral encoded semaphorin protein receptor DNA and polypeptides
AU748168B2 (en) Viral encoded semaphorin protein receptor DNA and polypeptides
US6562949B1 (en) Antibodies to viral encoded semaphorin protein receptor polypeptides
JP3621883B2 (en) Semaphorin protein receptor DNA and polypeptide encoded by virus
US20130084603A1 (en) B7l-1 polynucleotides

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION