US20020123104A1 - Human slit polypeptide and polynucleotides encoding same - Google Patents

Human slit polypeptide and polynucleotides encoding same Download PDF

Info

Publication number
US20020123104A1
US20020123104A1 US10/011,064 US1106401A US2002123104A1 US 20020123104 A1 US20020123104 A1 US 20020123104A1 US 1106401 A US1106401 A US 1106401A US 2002123104 A1 US2002123104 A1 US 2002123104A1
Authority
US
United States
Prior art keywords
leu
polypeptide
polynucleotide
ser
gly
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
US10/011,064
Inventor
Timothy Connolly
Bhanu Rajput
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.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US10/011,064 priority Critical patent/US20020123104A1/en
Publication of US20020123104A1 publication Critical patent/US20020123104A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy

Definitions

  • This invention relates to newly identified polynucleotides, polypeptides encoded by such polynucleotides, the use of such polynucleotides and polypeptides, as well as the production of such polynucleotides and polypeptides. More particularly, the polypeptides of the present invention are human Slit polypeptides. The invention also relates to identifying mesenchymal stem cells (MSCs) or other cells comprising such polypeptides or polynucleotides that encode the polypeptides.
  • MSCs mesenchymal stem cells
  • EGF epidermal growth factor
  • the Drosophila slit protein contains two types of repeated amino acid sequences: leucine rich repeats (“LRR”) and epidermal growth factor-like repeats (“EGF”). Its LRRs are arranged in four groups, each composed of four or five LRRs surrounded by conserved amino- and carboxy-flanking regions. The presence of both the LRRs and EGF-like repeats within a single protein make slit unusual in that such combination is not found in any other type of known protein. The absence of any potential transmembrane domains in a sequence having a typical signal sequence and two known extracellular-associated motifs suggests that the slit locus encodes a secreted extracellular protein.
  • LRR leucine rich repeats
  • EGF epidermal growth factor-like repeats
  • the LRR regions of the slit protein and such regions of related proteins participate in extracellular protein-protein interactions. Further, the EGF areas of the slit protein and such regions of related proteins participate in extracellular protein-protein reactions. Moreover, the slit protein is synthesized and secreted by midline glial cells can be come associated with axons. Among other functions, it influences the differentiation of midline cells from the neuroepithelium.
  • novel polypeptides and polynucleotides are of human origin and are found in human mesenchymal stem cells.
  • MSCs are the formative pluripotential blast cells found inter alia in bone marrow, blood, dermis and periosteum that are capable of differentiating into any of the specific types of mesenchymal or connective tissues (i.e. the tissues of the body that support the specialized elements; particularly adipose, osseous, cartilaginous, elastic, and fibrous connective tissues) depending upon various influences from bioactive factors, such as cytokines.
  • the polypeptide is designated as human Slit.
  • the human Slit polypeptide according to the present invention is of use in studing the culturing of MSCs and detection of their differentiation and development into multipotent cells.
  • nucleic acid molecules encoding such polypeptides, including mRNAs, cDNAs, genomic DNA as well as biologically active and diagnostically or therapeutically useful fragments, analogs and derivatives thereof.
  • nucleic acid probes comprising nucleic acid molecules of sufficient length to specifically hybridize to sequences of the present invention.
  • a process for producing such polypeptides by recombinant techniques which comprises culturing recombinant prokaryotic and/or eukaryotic host cells, containing a nucleic acid sequence of the present invention, under conditions promoting expression of said protein and subsequent recovery of said protein.
  • polypeptides or polynucleotides encoding such polypeptides for identifying human MSCs by utilizing the polynucleotides as probes or by expressing the polypeptides encoded thereby, using such polypeptides to produce an antibody specific for one of the polypeptides and then utilizing the antibody to identify the MSCs.
  • polypeptides and antibodies may be utilized to aid in the identification of MSCs from other species, as well as to investigate/identify MSC functions in humans or other species.
  • immunocyto-chemistry is utilized with an antibody specific for a polypeptide according to the invention as a means for monitoring the concentration of the polypeptide according to the invention in a culture solution.
  • the MSCs of the culture may thus be subjected to purification procedures to remove differentiated cells and help to maintain the MSCs in culture.
  • antibodies against such polypeptides and a method of employing such antibodies to detect diseases related to an overexpression or under expression of a polypeptide compressing a polypeptide with an amino acid sequence according to the present invention.
  • Such antibodies may be utilized to monitor the growth of MSCs in a culture or to detect the location of tumors in the body.
  • FIG. 1 shows a schematic of the transcript for a cDNA clone that encodes the mature slit polypeptide.
  • the various coding regions and repeats are identified by different types of cross-hatching as shown in the figure and identified by the legend below it.
  • FIG. 2 shows a cDNA sequence (SEQ ID. NO:1) that encodes the mature slit polypeptide (SEQ ID NO:2) of the present invention. Sequencing was performed using a 373 Automated DNA sequencer (Applied Biosystems, Inc.).
  • FIG. 3 is an illustration of amino acid sequence homology between the human slit polypeptide of the present invention (labelled as hSlit) and the Drosophila slit polypeptide (labelled as dSlit).
  • hSlit human slit polypeptide of the present invention
  • dSlit Drosophila slit polypeptide
  • FIG. 4 shows a photograph of a protein blot from expression of hSlit in human embryonic kidney cell line, BOSC 23. Untransfected BOSC cells do not express hSlit.
  • Lane 1 shows the results for untransfected BOSC cells; Lanes 2, 3 and 4, respectively show (2) BOSC cells transfected with pcDNA3.1/Myc-His/A vector, (3) pcDNA3.1/Myc-His/lacZ and (4) pcDNA3.1/Myc-His/hSlit cDNA.
  • isolated nucleic acids which encode the mature polypeptides comprises the deduced amino acid sequences of SEQ ID NO:2.
  • the mature forms of the slit polypeptide with and without an N-terminal methionine group (N-terminal methionine is the first amino acid of SEQ ID NO:2) are contemplated.
  • Polynucleotides encoding the polypeptide of the present invention have been isolated from a human MSC cDNA library.
  • the polynucleotide contains an open reading frame encoding the human slit polypeptide.
  • the protein exhibits a high degree of homology at the amino acid level to the Drosophila slit polypeptide with 40% identity (as shown in FIG. 3).
  • the human slit gene sequence according to SEQ ID NO:1 or an appropriate fragment may be utilized under stringent hybridization conditions to isolate from a cDNA library prepared from MSCs by procedures known in the art the cDNA encoding alleles of the mature slit polypeptide. Further, such full- or partial-length probes may be utilized to isolate genes (or cDNAs) encoding related polypeptides from non-human hosts under either stringent or highly-stringent hybridization conditions.
  • polypeptide having an amino acid sequence according to SEQ ID NO:2 or an immunogenic fragment may be utilized to produce antibodies specific for the polypeptide according to SEQ ID NO:2 and a fragment thereof.
  • Such antibodies are in turn useful to detect the presence of such polypeptides when they are expressed by a clone or a transformed host cell to indicate the presence of the respective polynucleotides encoding such polypeptides.
  • the polynucleotides of the present invention may be in the form of RNA or in the form of DNA, which DNA includes cDNA, genomic DNA, and synthetic DNA.
  • the DNA may be double-stranded or single-stranded, and if single stranded may be the coding strand or non-coding (anti-sense) strand.
  • the coding sequence which encodes the mature polypeptides may comprise an amino acid sequence identical to the coding sequence shown in FIG. 1 (SEQ ID NO:1) or may be a different coding sequence which coding sequence, as a result of the redundancy or degeneracy of the genetic code, encodes the same mature polypeptides comprising the polypeptide of SEQ. ID NO:2, the cDNA for which is shown in FIG. 1 (SEQ ID NO:1).
  • the polynucleotides which encode the mature polypeptides of the present invention comprise the polynucleotide sequence encoding the polypeptide of SEQ ID NO:2 and may include: only the coding sequence for the mature polypeptide; the coding sequence for the mature polypeptide and additional coding sequence such as a leader or secretory sequence or a proprotein sequence; the coding sequence for the mature polypeptide (and optionally additional coding sequence) and non-coding sequence, such as introns or non-coding sequence 5′ and/or 3′ of the coding sequence for the mature polypeptides.
  • polynucleotide encoding a polypeptide encompasses a polynucleotide which includes coding sequence for the polypeptide and may also include additional coding and/or non-coding sequence such as introns.
  • the present invention further relates to variants of the hereinabove described polynucleotides which encode fragments, analogs and derivatives of the mature polypeptide comprising amino acid sequence shown in SEQ ID NO:2.
  • the variant of the polynucleotides may be a naturally occurring allelic variant of the polynucleotides or a non-naturally occurring variant of the polynucleotides.
  • polynucleotides encoding the human slit polypeptide variants, analogs, derivatives and fragments, and variants, analogs and derivatives of the fragments which comprise the amino acid sequence of the polypeptide of SEQ ID NO:2 or of the deposit in which several, a few, 5 to 10, 1 to 5, 1 to 3, 2, 1 or no amino acid residues are substituted, deleted or added, in any combination.
  • silent substitutions, additions and deletions which do not alter the properties and activities of the human slit polypeptide.
  • conservative substitutions are also especially preferred in this regard.
  • Most highly preferred are mature polypeptides comprising the amino acid sequence set forth in SEQ ID NO:2 or of the deposit, without substitutions.
  • the present invention includes polynucleotides encoding the same mature polypeptides comprising the polypeptide as set forth in SEQ ID NO:2 as well as variants of such polynucleotides which variants encode a fragment, derivative or analog of the polypeptides set forth in SEQ ID NO:2.
  • polynucleotide variants include deletion variants, substitution variants and addition or insertion variants.
  • polynucleotide sequences comprising polynucleotide sequence variants of a starting polynucleotide sequence that are obtained by changing the starting polynucleotide sequence in at least one of the following ways (a) inserting at least one nucleotide into it, (b) deleting at least one nucleotide from it, (c) substituting at least one nucleotide for a nucleotide of it, or (d) a combination of at least two of (a), (b) and (c).
  • the starting polynucleotide sequence that is changed to obtain variant polynucleotide sequences is a member selected from (i) the coding portion of SEQ ID NO:1 and (ii) a redundant sequence encoding the same mature polypeptide as the coding portion of SEQ ID NO:1.
  • Each of the preferred variant polynucleotide sequences results from changing no more than a total of 10 percent of the coding sequence nucleotides of the starting polynucleotide sequence by such deletion, substitution, insertion or a combination thereof (i.e., not more than 10 nucleotides per 100 nucleotides).
  • variant polynucleotide sequences that result from changing no more than a total of 5 percent of the starting coding sequence nucleotides by deletion, insertion, substitution or a combination thereof. Even more preferred are variant polynucleotide sequences that result from changing no more than a total of 3 percent of the starting coding sequence nucleotides by deletion, insertion, substitution or a combination thereof. Such changes occur within the 5′ to 3′ portions of the coding sequence of the starting polynucleotide.
  • the polypeptides encoded by such variant polynucleotides may or may not retain the activity of the polypeptide encoded by the polynucleotide of SEQ ID NO:1.
  • polynucleotides may be employed as probes for the gene comprising the polynucleotide of SEQ ID NO:1, for the polynucleotide of SEQ ID NO:1 or for a redundant polynucleotide which encodes the same polypeptide that is encoded by the polynucleotide having a sequence according to SEQ ID NO:1.
  • the polynucleotides may have a coding sequence which is a naturally occurring allelic variant of the coding sequences comprising the coding portion of the polynucleotide sequence shown in FIG. 1 (of SEQ ID NO:1).
  • an allelic variant is an alternate form of a polynucleotide sequence which may have a substitution, deletion or addition of one or more nucleotides, which does not substantially alter the function of the encoded polypeptide.
  • the present invention also includes polynucleotides, wherein the coding sequence for the mature polypeptides may be fused in the same reading frame to a polynucleotide sequence which aids in expression and secretion of a polypeptide from a host cell, for example, a leader sequence which functions as a secretory sequence for controlling transport of a polypeptide from the cell.
  • the polypeptide having a leader sequence is a preprotein and may have the leader sequence cleaved by the host cell to form the mature form of the polypeptide.
  • the polynucleotides may also encode a proprotein which is the mature protein plus additional 5′ amino acid residues.
  • a mature protein having a prosequence is a proprotein and is an inactive form of the protein. Once the prosequence is cleaved an active mature protein remains.
  • the polynucleotides of the present invention may encode a mature protein, or for a protein having a prosequence or for a protein having both a prosequence and a presequence (leader sequence).
  • the polynucleotides of the present invention may also have the coding sequence fused in frame to a marker sequence which allows for purification of the polypeptides of the present invention.
  • the marker sequence may be a hexa-histidine tag supplied by a pQE-9 vector to provide for purification of the mature polypeptides fused to the marker in the case of a bacterial host, or, for example, the marker sequence may be a hemagglutinin (HA) tag when a mammalian host, e.g. COS-7 cells, is used.
  • the HA tag corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson, I., et al., Cell, 37:767 (1984)).
  • gene means the segment of DNA involved in producing a polypeptide chain; it includes regions preceding and following the coding region (leader and trailer) as well as intervening sequences (introns) between individual coding segments (exons).
  • Fragments of the full length gene of the present invention may be used as a hybridization probe for a cDNA library to isolate the full length cDNA and to isolate other cDNAs which have a high sequence similarity to the gene or similar biological activity.
  • Probes of this type preferably have at least 30 bases and may contain, for example, 50 or more bases.
  • the probe may also be used to identify a cDNA clone corresponding to a full length transcript and a genomic clone or clones that contain the complete gene including regulatory and promotor regions, exons, and introns.
  • An example of a screen comprises isolating the coding region of the gene by using the known DNA sequence to synthesize an oligonucleotide probe. Labeled oligonucleotides having a sequence complementary to that of the gene of the present invention are used to screen a library of human cDNA, genomic DNA or mRNA to determine which members of the library the probe hybridizes to.
  • the present invention further relates to polynucleotides which hybridize to the hereinabove-described sequences if there is at least 70%, preferably at least 90%, and more preferably at least 95% identity between the sequences.
  • the present invention particularly relates to polynucleotides which hybridize under stringent conditions to the hereinabove-described polynucleotides.
  • stringent conditions means hybridization will occur only if there is at least 95% and preferably at least 97% identity between the sequences.
  • polypeptides which hybridize to the hereinabove described polynucleotides in a preferred embodiment encode polypeptides which either retain substantially the same biological function or activity as the mature polypeptide comprising the amino acid sequence encoded by the cDNA of FIG. 1 (comprising SEQ ID NO:2).
  • the polynucleotide may have at least 20 bases, preferably at least 30 bases, and more preferably at least 50 bases which hybridize to a polynucleotide of the present invention and which has an identity thereto, as hereinabove described, and which may or may not retain activity.
  • such polynucleotides may be employed as probes for the gene comprising the polynucleotide of SEQ ID NO:1, for example, for recovery of the polynucleotide or as a diagnostic probe or as a PCR primer.
  • the present invention is directed to polynucleotides having at least a 70% identity, preferably at least 90% and more preferably at least a 95% identity to a polynucleotide which encodes the polypeptide of SEQ ID NO:2 and polynucleotides complementary thereto as well as portions thereof, which portions have at least 20, preferably at least 30 consecutive bases and may have at least 50 consecutive bases and to polypeptides encoded by such polynucleotides.
  • the present invention further relates to polypeptides which have the deduced amino acid sequence as set forth in SEQ ID NO:2, as well as fragments, analogs and derivatives of such polypeptides.
  • fragment when referring to the mature polypeptides comprising the polypeptide as set forth in SEQ ID NO:2, means polypeptides which retain essentially the same biological function or activity as such polypeptides.
  • an analog includes a proprotein which can be activated by cleavage of the proprotein portion to produce an active mature polypeptide.
  • particularly preferred embodiments of the invention in this regard are mature polypeptides comprising the amino acid sequence as set forth in SEQ ID NO:2, variants, analogs, derivatives and fragments thereof, and variants, analogs and derivatives of the fragments.
  • particularly preferred embodiments of the invention in this regard are polypeptides comprising the amino acid sequence of the human slit polypeptide encoded by the cDNA in the deposited clone, variants, analogs, derivatives and fragments thereof, and variants, analogs and derivatives of the fragments.
  • variants, analogs, derivatives and fragments, and variants, analogs and derivatives of the fragments comprising the amino acid sequence of the polypeptide as set forth in SEQ ID NO:2 or as encoded by the cDNA in the deposited clone, in which at least one amino acid residue per each 100 amino acids of the amino sequence is varied by at least one of (a) substituting an amino acid for it, (b) deleting at least one amino acid, (c) inserting at least one new amino acid, or (d) a combination of at least two of (a), (b) and (c).
  • variant polypeptides are obtained whose amino acid sequences are obtained by changing 5 to 10, 1 to 5, 1 to 3, or 1 to 2 amino acid residues per 100 amino acids in that at least one of (i) at least one new amino acid is substituted for an amino acid of SEQ ID NO:2 (or of a fragment of SEQ ID NO:2), (ii) at least one amino acid of SEQ ID NO:2 (or of a fragment of SEQ ID NO:2) is deleted, (ii) at least one new amino acid is inserted into SEQ ID NO:2 (or into a fragment of SEQ ID NO:2), or (iv) a combination of (i), (ii) or (iii).
  • polypeptides of the present invention may be recombinant polypeptides, natural polypeptides or synthetic polypeptides, preferably recombinant polypeptides.
  • the fragment, derivative or analog of the polypeptides comprising the amino acid sequence set forth in SEQ ID NO:2 may be (i) one in which one or more of the amino acid residues are substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code, or (ii) one in which one or more of the amino acid residues includes a substituent group, or (iii) one in which the mature polypeptide is fused with another compound, such as a compound to increase the half-life of the polypeptide (for example, polyethylene glycol), or (iv) one in which the additional amino acids are fused to the mature polypeptide, such as a leader or secretory sequence or a sequence which is employed for purification of the mature polypeptide or a proprotein sequence.
  • Such fragments, derivatives and analogs are deemed to be within the scope of those skilled in the art from the teachings herein.
  • polypeptides and polynucleotides of the present invention are preferably provided in an isolated form, and preferably are purified to homogeneity.
  • isolated means that the material is removed from its original environment (e.g., the natural environment if it is naturally occurring).
  • a naturally-occurring polynucleotide or polypeptide present in a living animal is not isolated, but the same polynucleotide or polypeptide, separated from some or all of the coexisting materials in the natural system, is isolated.
  • Such polynucleotides could be part of a vector and/or such polynucleotides or polypeptides could be part of a composition, and still be isolated in that such vector or composition is not part of its natural environment.
  • polypeptides of the present invention include polypeptides comprising the polypeptide of SEQ ID NO:2 (in particular the mature polypeptide) as well as polypeptides which have at least 70% similarity (preferably at least 70% identity) to the mature polypeptide comprising the amino acid sequence of SEQ ID NO:2, and which have at least 90% similarity (more preferably at least 90% identity) to the mature polypeptide comprising the amino acid sequence of SEQ ID NO:2 and still more preferably at least 95% similarity (still more preferably at least 95% identity) to the mature polypeptide comprising the amino acid sequence of SEQ ID NO:2 and also include portions of such polypeptides with such portion of the polypeptide generally containing at least 30 amino acids and more preferably at least 50 amino acids.
  • similarity between two polypeptides is determined by comparing the amino acid sequence and its conserved amino acid substitutes of one polypeptide to the sequence of a second polypeptide. For such a determination, two amino acid sequences are compared along a stretch of their sequences, any gap (or gaps) introduced in one sequence to improve the alignment and similarity to the other sequences is counted as spaces of dissimilarity equal to the number of amino acids corresponding to the gap which are present in the second sequence, and the total number of similar amino acids are divided by the total number of amino acids present in the comparison area which counts the spaces of gaps as part of the comparison area.
  • Fragments or portions of the polypeptides of the present invention may be employed for producing the corresponding full-length polypeptide by peptide synthesis; therefore, the fragments may be employed as intermediates for producing the full-length polypeptides. Fragments or portions of the polynucleotides of the present invention may be used to synthesize full-length polynucleotides of the present invention.
  • the present invention also relates to vectors which include polynucleotides of the present invention, host cells which are genetically engineered with vectors of the invention and the production of polypeptides of the invention by recombinant techniques.
  • Host cells are genetically engineered (transduced or transformed or transfected) with the vectors of this invention which may be, for example, a cloning vector or an expression vector.
  • the vector may be, for example, in the form of a plasmid, a viral particle, a phage, etc.
  • the engineered host cells can be cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transformants or amplifying the genes.
  • the culture conditions such as temperature, pH and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.
  • the polynucleotides of the present invention may be employed for producing polypeptides by recombinant techniques.
  • the polynucleotide may be included in any one of a variety of expression vectors for expressing a polypeptide.
  • Such vectors include chromosomal, nonchromosomal and synthetic DNA sequences, e.g., derivatives of SV40; bacterial plasmids; phage DNA; baculovirus; yeast plasmids; vectors derived from combinations of plasmids and phage DNA, viral DNA such as vaccinia, adenovirus, fowl pox virus, and pseudorabies.
  • any other vector may be used as long as it is replicable and viable in the host.
  • the appropriate DNA sequence may be inserted into the vector by a variety of procedures.
  • the DNA sequence is inserted into an appropriate restriction endonuclease site(s) by procedures known in the art. Such procedures and others are deemed to be within the scope of those skilled in the art.
  • the DNA sequence in the expression vector is operatively linked to an appropriate expression control sequence(s) (promoter) to direct mRNA synthesis.
  • promoters there may be mentioned: LTR or SV40 promoter, the E. coli. lac or trp, the phage lambda PL promoter and other promoters known to control expression of genes in prokaryotic or eukaryotic cells or their viruses.
  • the expression vector also contains a ribosome binding site for translation initiation and a transcription terminator.
  • the vector may also include appropriate sequences for amplifying expression.
  • the expression vectors preferably contain one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells such as dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, or such as tetracycline or ampicillin resistance in E. coli.
  • the vector containing the appropriate DNA sequence as hereinabove described, as well as an appropriate promoter or control sequence, may be employed to transform an appropriate host to permit the host to express the protein.
  • bacterial cells such as E. coli, Streptomyces, Salmonella typhimurium
  • fungal cells such as yeast
  • insect cells such as Drosophila S2 and Spodoptera Sf9
  • animal cells such as CHO, COS or Bowes melanoma
  • adenoviruses plant cells, etc.
  • the present invention also includes recombinant constructs comprising one or more of the sequences as broadly described above.
  • the constructs comprise a vector, such as a plasmid or viral vector, into which a sequence of the invention has been inserted, in a forward or reverse orientation.
  • the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the sequence.
  • a promoter operably linked to the sequence.
  • Bacterial pQE70, pQE60, pQE-9 (Qiagen), pBS, pD10, phagescript, psiX174, pBluescript SK, pBSKS, pNH8A, pNH16a, pNH18A, pNH46A (Stratagene); pTRC99a, pKK223-3, pKK233-3, pDR540, pRIT5 (Pharmacia).
  • Eukaryotic pWLNEO, pSV2CAT, pOG44, pXT1, pSG (Stratagene) pSVK3, pBPV, pMSG, pSVL (Pharmacia). However, any other plasmid or vector may be used as long as they are replicable and viable in the host.
  • Promoter regions can be selected from any desired gene using CAT (chloramphenicol transferase) vectors or other vectors with selectable markers.
  • Two appropriate vectors are pKK232-8 and pCM7.
  • Particular named bacterial promoters include lacl, lacZ, T3, T7, gpt, lambda P R , P L and trp.
  • Eukaryotic promoters include CMV immediate early, HSV thymidine kinase, early and late SV40, LTRs from retrovirus, and mouse metallothionein-I. Selection of the appropriate vector and promoter is well within the level of ordinary skill in the art.
  • the present invention relates to host cells containing the above-described constructs.
  • the host cell can be a higher eukaryotic cell, such as a mammalian cell, or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell.
  • Introduction of the construct into the host cell can be effected by calcium phosphate transfection, DEAE-Dextran mediated transfection, or electroporation (Davis, L., Dibner, M., Battey, I., Basic Methods in Molecular Biology, (1986)).
  • constructs in host cells can be used in a conventional manner to produce the gene products encoded by the recombinant sequences.
  • polypeptides of the invention can be synthetically produced by conventional peptide synthesizers.
  • Mature proteins can be expressed in mammalian cells, yeast, bacteria, or other cells under the control of appropriate promoters. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention. Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrook, et al., Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor, N.Y., (1989), the disclosure of which is hereby incorporated by reference.
  • Enhancers are cis-acting elements of DNA, usually about from 10 to 300 bp that act on a promoter to increase its transcription. Examples include the SV40 enhancer on the late side of the replication origin bp 100 to 270, a cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers.
  • recombinant expression vectors will include origins of replication and selectable markers permitting transformation of the host cell, e.g., the ampicillin resistance gene of E. coli and S. cerevisiae TRP1 gene, and a promoter derived from a highly-expressed gene to direct transcription of a downstream structural sequence.
  • promoters can be derived from operons encoding glycolytic enzymes such as 3-phosphoglycerate kinase (PGK), á-factor, acid phosphatase, or heat shock proteins, among others.
  • the heterologous structural sequence is assembled in appropriate phase with translation initiation and termination sequences, and preferably, a leader sequence capable of directing secretion of translated protein into the periplasmic space or extracellular medium.
  • the heterologous sequence can encode a fusion protein including an N-terminal identification peptide imparting desired characteristics, e.g., stabilization or simplified purification of expressed recombinant product.
  • Useful expression vectors for bacterial use are constructed by inserting a structural DNA sequence encoding a desired protein together with suitable translation initiation and termination signals in operable reading phase with a functional promoter.
  • the vector will comprise one or more phenotypic selectable markers and an origin of replication to ensure maintenance of the vector and to, if desirable, provide amplification within the host.
  • Suitable prokaryotic hosts for transformation include E. coli, Bacillus subtilis, Salmonella typhimurium and various species within the genera Pseudomonas, Streptomyces, and Staphylococcus, although others may also be employed as a matter of choice.
  • useful expression vectors for bacterial use can comprise a selectable marker and bacterial origin of replication derived from commercially available plasmids comprising genetic elements of the well known cloning vector pBR322 (ATCC 37017).
  • cloning vector pBR322 ATCC 37017
  • Such commercial vectors include, for example, pKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and pGEM1 (Promega Biotec, Madison, Wis., USA). These pBR322 “backbone” sections are combined with an appropriate promoter and the structural sequence to be expressed.
  • the selected promoter is induced by appropriate means (e.g., temperature shift or chemical induction) and cells are cultured for an additional period.
  • Cells are typically harvested by centrifugation, disrupted by physical or chemical means, and the resulting crude extract retained for further purification.
  • Microbial cells employed in expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents, such methods are well know to those skilled in the art.
  • mammalian cell culture systems can also be employed to express recombinant protein.
  • mammalian expression systems include the COS-7 lines of monkey kidney fibroblasts, described by Gluzman, Cell, 23:175 (1981), and other cell lines capable of expressing a compatible vector, for example, the C127, 3T3, CHO, HeLa, 293 and BHK cell lines.
  • Mammalian expression vectors will comprise an origin of replication, a suitable promoter and enhancer, and also any necessary ribosome binding sites, polyadenylation site, splice donor and acceptor sites, transcriptional termination sequences, and 5′ flanking nontranscribed sequences. DNA sequences derived from the SV40 splice, and polyadenylation sites may be used to provide the required nontranscribed genetic elements.
  • polypeptides can be recovered and purified from recombinant cell cultures by methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Protein refolding steps can be used, as necessary, in completing configuration of the mature protein. Finally, high performance liquid chromatography (HPLC) can be employed for final purification steps.
  • HPLC high performance liquid chromatography
  • the polypeptides of the present invention may be a naturally purified product, or a product of chemical synthetic procedures, or produced by recombinant techniques from a prokaryotic or eukaryotic host (for example, by bacterial, yeast, higher plant, insect and mammalian cells in culture). Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosylated. Polypeptides of the invention may also include an initial methionine amino acid residue.
  • polynucleotides and polypeptides of the present invention may be employed as research reagents and materials for discovery of treatments and diagnostics for human disease.
  • the polynucleotides and polypeptides encoded by such polynucleotides may also be utilized for in vitro purposes related to scientific research, synthesis of DNA and manufacture of DNA vectors and for designing therapeutics and diagnostics for human disease.
  • the invention also provides a method for identifying human mesenchymal stem cells by contacting a mixture of mRNA from a cell sample with a polynucleotide unique to human slit and identifying any mRNA which has hybridized with the polynucleotide unique to human slit.
  • the polynucleotide unique to human slit is bound to a solid support.
  • the identification of slit cDNA enables the slit nucleic acid sequence to be utilized as a diagnostic reagent to identify human MSCs, such as by using gene expression array technology. Labeled (e.g.
  • polypeptides of the present invention and fragments and analogs and derivatives thereof may be identified by assays which detect MSC proliferation or other activity. Further, assays may be utilized which neutralize the production of the native slit in midline glia cells and subjecting such cells to a polypeptide sequence which is related to the native slit sequence but is different in order to verify the same functionality of polypeptides having both sequences.
  • This invention is also related to the use of the human slit polypeptide gene as part of a diagnostic assay for detecting diseases or susceptibility to diseases related to the presence of mutations in the human slit polypeptide nucleic acid sequences. Such diseases are related to under-expression or overexpression of the human slit polypeptides.
  • Nucleic acids for diagnosis may be obtained from a patient's cells, such as from blood, urine, saliva, tissue biopsy and autopsy material.
  • the genomic DNA may be used directly for detection or may be amplified enzymatically by using PCR (Saiki et al., Nature, 324:163-166 (1986)) prior to analysis.
  • RNA or cDNA may also be used for the same purpose.
  • PCR primers complementary to the nucleic acid encoding human slit polypeptide can be used to identify and analyze human slit polypeptide mutations.
  • deletions and insertions can be detected by a change in size of the amplified product in comparison to the normal genotype.
  • Point mutations can be identified by hybridizing amplified DNA to radiolabeled human slit polypeptide RNA or alternatively, radiolabeled human slit polypeptide antisense DNA sequences. Perfectly matched sequences can be distinguished from mismatched duplexes by RNase A digestion or by differences in melting temperatures.
  • DNA sequence differences may be achieved by detection of alteration in electrophoretic mobility of DNA fragments in gels with or without denaturing agents. Small sequence deletions and insertions can be visualized by high resolution gel electrophoresis. DNA fragments of different sequences may be distinguished on denaturing formamide gradient gels in which the mobilities of different DNA fragments are retarded in the gel at different positions according to their specific melting or partial melting temperatures (see, e.g., Myers et al., Science, 230:1242 (1985)).
  • Sequence changes at specific locations may also be revealed by nuclease protection assays, such as RNase and S1 protection or the chemical cleavage method (e.g., Cotton et al., PNAS, USA, 85:4397-4401 (1985)).
  • nuclease protection assays such as RNase and S1 protection or the chemical cleavage method (e.g., Cotton et al., PNAS, USA, 85:4397-4401 (1985)).
  • the detection of a specific DNA sequence may be achieved by methods such as hybridization, RNase protection, chemical cleavage, direct DNA sequencing or the use of restriction enzymes, (e.g., Restriction Fragment Length Polymorphisms (RFLP)) and Southern blotting of genomic DNA.
  • restriction enzymes e.g., Restriction Fragment Length Polymorphisms (RFLP)
  • mutations can also be detected by in situ analysis.
  • the present invention also relates to a diagnostic assay for detecting altered levels of the slit polypeptide in various tissues since an over-expression or under-expression of the proteins compared to normal control tissue samples may detect the presence of a disease or susceptibility to a disease, for example, reduced blood cell counts or malignancies such as cancers and tumors.
  • Assays used to detect levels of the slit polypeptide in a sample derived from a host are well-known to those of skill in the art and include radioimmunoassays, competitive-binding assays, Western Blot analysis, ELISA assays and “sandwich” assay.
  • An ELISA assay (Coligan, et al., Current Protocols in Immunology, 1(2), Chapter 6, (1991)) initially comprises preparing an antibody specific to the slit polypeptide antigen, preferably a monoclonal antibody.
  • a reporter antibody is prepared against the monoclonal antibody.
  • a detectable reagent such as radioactivity, fluorescence or, in this example, a horseradish peroxidase enzyme.
  • a sample is removed from a host and incubated on a solid support, e.g. a polystyrene dish, that binds the proteins in the sample. Any free protein binding sites on the dish are then covered by incubating with a non-specific protein like BSA.
  • the monoclonal antibody is incubated in the dish during which time the monoclonal antibodies attach to any slit polypeptide attached to the polystyrene dish. All unbound monoclonal antibody is washed out with buffer.
  • the reporter antibody linked to horseradish peroxidase is now placed in the dish resulting in binding of the reporter antibody to any monoclonal antibody bound to the slit polypeptide. Unattached reporter antibody is then washed out.
  • Peroxidase substrates are then added to the dish and the amount of color developed in a given time period is a measurement of the amount of the slit polypeptide present in a given volume of patient sample when compared against a standard curve.
  • a competition assay may be employed wherein antibodies specific to the slit polypeptide are attached to a solid support and labeled the slit polypeptide and a sample derived from the host are passed over the solid support and the amount of label detected, for example by liquid scintillation chromatography, can be correlated to a quantity of the slit polypeptide in the sample.
  • a “sandwich” assay is similar to an ELISA assay.
  • the slit polypeptide is passed over a solid support and binds to antibody attached to a solid support.
  • a second antibody is then bound to the slit polypeptide.
  • a third antibody which is labeled and specific to the second antibody is then passed over the solid support and binds to the second antibody and an amount can then be quantified.
  • This invention provides a method for identification of the receptors for the human slit polypeptides.
  • the gene encoding the receptor can be identified by numerous methods known to those of skill in the art, for example, ligand panning and FACS sorting (Coligan, et al., Current Protocols in Immun., 1(2), Chapter 5, (1991)).
  • expression cloning is employed wherein polyadenylated RNA is prepared from a cell responsive to the polypeptides, and a cDNA library created from this RNA is divided into pools and used to transfect COS cells or other cells that are not responsive to the polypeptides. Transfected cells which are grown on glass slides are exposed to the labeled polypeptides.
  • polypeptides can be labeled by a variety of means including iodination or inclusion of a recognition site for the slit polypeptides. Following fixation and incubation, the slides are subjected to autoradiographic analysis. Positive pools are identified and sub-pools are prepared and retransfected using an iterative sub-pooling and rescreening process, eventually yielding a single clones that encodes the putative receptor.
  • the labeled polypeptides can be photoaffinity linked with cell membrane or extract preparations that express the receptor molecule. Cross-linked material is resolved by PAGE analysis and exposed to X-ray film. The labeled complex containing the receptors of the polypeptides can be excised, resolved into peptide fragments, and subjected to protein microsequencing. The amino acid sequence obtained from microsequencing would be used to design a set of degenerate oligonucleotide probes to screen a cDNA library to identify the genes encoding the putative receptors.
  • This invention provides a method of screening compounds to identify agonists and antagonists to the human slit polypeptides of the present invention.
  • An agonist is a compound which has similar biological functions of the polypeptides, while antagonists block such functions.
  • Antagonists and agonists may be identified by the an MSC proliferation assay as is well known in the art.
  • Examples of potential the slit polypeptide antagonists include antibodies, or in some cases, oligonucleotides, which bind to the polypeptides.
  • Another example of a potential antagonist is a negative dominant mutant of the polypeptides. Negative dominant mutants are polypeptides which bind to the receptor of the wild-type polypeptide, but fail to retain biological activity.
  • Antisense constructs prepared using antisense technology are also potential antagonists.
  • Antisense technology can be used to control gene expression through triple-helix formation or antisense DNA or RNA, both of which methods are based on binding of a polynucleotide to DNA or RNA.
  • the 5′ coding portion of the polynucleotide sequence, which encodes the mature polypeptides of the present invention is used to design an antisense RNA oligonucleotide of from about 10 to 40 base pairs in length.
  • a DNA oligonucleotide is designed to be complementary to a region of the gene involved in transcription (triple- helix, see Lee et al., Nucl.
  • the antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into the polypeptides (antisense—Okano, J. Neurochem., 56:560 (1991); Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression, CRC Press, Boca Raton, Fla. (1988)).
  • the oligonucleotides described above can also be delivered to cells such that the antisense RNA or DNA may be expressed in vivo to inhibit production of the human slit polypeptide.
  • Another potential human slit antagonist is a peptide derivative of the polypeptides which are naturally or synthetically modified analogs of the polypeptides that have lost biological function yet still recognize and bind to the receptors of the polypeptides to thereby effectively block the receptors.
  • peptide derivatives include, but are not limited to, small peptides or peptide-like molecules.
  • the antagonists may be employed in a composition with a pharmaceutically acceptable carrier, e.g., as hereinafter described.
  • the human slit polypeptides and antagonists may be employed in combination with a suitable pharmaceutical carrier.
  • a suitable pharmaceutical carrier includes but is not limited to saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof.
  • a carrier includes but is not limited to saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof.
  • the formulation should suit the mode of administration.
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention.
  • a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention.
  • Associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • the polypeptides and agonists and antagonists may be employed in conjunction with other therapeutic compounds.
  • the pharmaceutical compositions may be administered in a convenient manner such as by the topical, intravenous, intraperitoneal, intramuscular, intratumor, subcutaneous, intranasal or intradermal routes.
  • the pharmaceutical compositions are administered in an amount which is effective for treating and/or prophylaxis of the specific indication.
  • the polypeptides will be administered in an amount of at least about 10 ⁇ g/kg body weight and in most cases they will be administered in an amount not in excess of about 8 mg/Kg body weight per day. In most cases, the dosage is from about 10 ⁇ g/kg to about 1 mg/kg body weight daily, taking into account the routes of administration, symptoms, etc.
  • the human slit polypeptides, and agonists or antagonists which are polypeptides, may be employed in accordance with the present invention by expression of such polypeptides in vivo, which is often referred to as “gene therapy.”
  • cells from a patient may be engineered with a polynucleotide (DNA or RNA) encoding a polypeptide ex vivo, with the engineered cells then being provided to a patient to be treated with the polypeptide.
  • a polynucleotide DNA or RNA
  • cells may be engineered by procedures known in the art by use of a retroviral particle containing RNA encoding a polypeptide of the present invention.
  • cells may be engineered in vivo for expression of a polypeptide in vivo by, for example, procedures known in the art.
  • a producer cell for producing a retroviral particle containing RNA encoding the polypeptide of the present invention may be administered to a patient for engineering cells in vivo and expression of the polypeptide in vivo.
  • the expression vehicle for engineering cells may be other than a retrovirus, for example, an adenovirus which may be used to engineer cells in vivo after combination with a suitable delivery vehicle.
  • Retroviruses from which the retroviral plasmid vectors hereinabove mentioned may be derived include, but are not limited to, Moloney Murine Leukemia Virus, spleen necrosis virus, retroviruses such as Rous Sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, gibbon ape leukemia virus, human immunodeficiency virus, adenovirus, Myeloproliferative Sarcoma Virus, and mammary tumor virus.
  • the retroviral plasmid vector is derived from Moloney Murine Leukemia Virus.
  • the vector includes one or more promoters.
  • Suitable promoters which may be employed include, but are not limited to, the retroviral LTR; the SV40 promoter; and the human cytomegalovirus (CMV) promoter described in Miller, et al., Biotechniques, Vol. 7, No. 9, 980-990 (1989), or any other promoter (e.g., cellular promoters such as eukaryotic cellular promoters including, but not limited to, the histone, pol 111, and â-actin promoters).
  • CMV cytomegalovirus
  • viral promoters which may be employed include, but are not limited to, adenovirus promoters, thymidine kinase (TK) promoters, and B19 parvovirus promoters. The selection of a suitable promoter will be apparent to those skilled in the art from the teachings contained herein.
  • the nucleic acid sequence encoding the polypeptide of the present invention is under the control of a suitable promoter.
  • suitable promoters which may be employed include, but are not limited to, adenoviral promoters, such as the adenoviral major late promoter; or heterologous promoters, such as the cytomegalovirus (CMV) promoter; the respiratory syncytial virus (RSV) promoter; inducible promoters, such as the MMT promoter, the metallothionein promoter; heat shock promoters; the albumin promoter; the ApoAl promoter; human globin promoters; viral thymidine kinase promoters, such as the Herpes Simplex thymidine kinase promoter; retroviral LTRs (including the modified retroviral LTRs hereinabove described); the â-actin promoter; and human growth hormone promoters.
  • the promoter also may be the native promoter which controls the gene encoding
  • the retroviral plasmid vector is employed to transduce packaging cell lines to form producer cell lines.
  • packaging cells which may be transfected include, but are not limited to, the PE501, PA317, é-2, é-AM, PA12, T19-14X, VT-19-17-H2, éCRE, éCRIP, GP+E-86, GP+envAm12, and DAN cell lines as described in Miller, Human Gene Therapy, Vol. 1, pgs. 5-14 (1990), which is incorporated herein by reference in its entirety.
  • the vector may transduce the packaging cells through any means known in the art. Such means include, but are not limited to, electroporation, the use of liposomes, and CaPO 4 precipitation.
  • the retroviral plasmid vector may be encapsulated into a liposome, or coupled to a lipid, and then administered to a host.
  • the producer cell line generates infectious retroviral vector particles which include the nucleic acid sequence(s) encoding the polypeptides.
  • retroviral vector particles then may be employed, to transduce eukaryotic cells, either in vitro or in vivo.
  • the transduced eukaryotic cells will express the nucleic acid sequence(s) encoding the polypeptide.
  • Eukaryotic cells which may be transduced include, but are not limited to, embryonic stem cells, embryonic carcinoma cells, as well as hematopoietic stem cells, hepatocytes, fibroblasts, myoblasts, keratinocytes, endothelial cells, and bronchial epithelial cells.
  • sequences of the present invention are also valuable for chromosome identification.
  • the sequence is specifically targeted to and can hybridize with a particular location on an individual human chromosome.
  • Few chromosome marking reagents based on actual sequence data (repeat polymorphisms) are presently available for marking chromosomal location.
  • the mapping of DNAs to chromosomes according to the present invention is an important first step in correlating those sequences with genes associated with disease.
  • sequences of the present invention are also valuable for chromosome identification.
  • the sequence is specifically targeted to and can hybridize with a particular location on an individual human chromosome.
  • Few chromosome marking reagents based on actual sequence data (repeat polymorphisms) are presently available for marking chromosomal location.
  • the mapping of DNAs to chromosomes according to the present invention is an important first step in correlating those sequences with genes associated with disease.
  • sequences can be mapped to chromosomes by preparing PCR primers (preferably 15-25 bp) from the cDNA. Computer analysis of the 3′ untranslated region is used to rapidly select primers that do not span more than one exon in the genomic DNA, thus complicating the amplification process. These primers are then used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene corresponding to the primer will yield an amplified fragment.
  • PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular DNA to a particular chromosome.
  • sublocalization can be achieved with panels of fragments from specific chromosomes or pools of large genomic clones in an analogous manner.
  • Other mapping strategies that can similarly be used to map to its chromosome include in situ hybridization, prescreening with labeled flow-sorted chromosomes and preselection by hybridization to construct chromosome specific-cDNA libraries.
  • Fluorescence in situ hybridization (FISH) of a cDNA clones to a metaphase chromosomal spread can be used to provide a precise chromosomal location in one step.
  • FISH Fluorescence in situ hybridization
  • a cDNA precisely localized to a chromosomal region associated with the disease could be one of between 50 and 500 potential causative genes. (This assumes 1 megabase mapping resolution and one gene per 20 kb).
  • polypeptides, their fragments or other derivatives, or analogs thereof, or cells expressing them can be used as an immunogen to produce antibodies thereto.
  • These antibodies can be, for example, polyclonal or monoclonal antibodies.
  • the present invention also includes chimeric, single chain, and humanized antibodies, as well as Fab fragments, or the product of an Fab expression library. Various procedures known in the art may be used for the production of such antibodies and fragments.
  • Antibodies generated against the polypeptides corresponding to a sequence of the present invention can be obtained by direct injection of the polypeptides into an animal or by administering the polypeptides to an animal, preferably a nonhuman. The antibody so obtained will then bind the polypeptides itself. In this manner, even a sequence encoding only a fragment of the polypeptides can be used to generate antibodies binding the whole native polypeptides. Such antibodies can then be used to isolate the polypeptide from tissue expressing that polypeptide.
  • Antibodies specific to the polypeptide of the present invention may be employed as a diagnostic to determine elevated or lowered levels of the polypeptide in a sample derived from a host by techniques known in the art. These elevated or lowered levels are indicative of certain disorders which are characterized by such levels of the protein of the present invention and members of its family.
  • any technique which provides antibodies produced by continuous cell line cultures can be used. Examples include the hybridoma technique (Kohler and Milstein, 1975, Nature, 256:495-497), the trioma technique, the human B-cell hybridoma technique (Kozbor et al., 1983, Immunology Today 4:72), and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole, et al., 1985, in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96).
  • Such antibodies to the polypeptides of the present invention may be utilized to detect the presence or the absence of the polypeptides of the present invention. Thus, they are useful in an assay to verify the successful insertion of the polynucleotides of the present invention (as part of a construct) into a host cell.
  • the protein encoded by the inserted polynucleotide according to the present invention when expressed by the transformed host cell, serves as a “marker” for the successful insertion of the polynucleotide that can be detected by an antibody for the marker.
  • Plasmids are designated by a lower case p preceded and/or followed by capital letters and/or numbers.
  • the starting plasmids herein are either commercially available, publicly available on an unrestricted basis, or can be constructed from available plasmids in accord with published procedures.
  • equivalent plasmids to those described are known in the art and will be apparent to the ordinarily skilled artisan.
  • “Digestion” of DNA refers to catalytic cleavage of the DNA with a restriction enzyme that acts only at certain sequences in the DNA.
  • the various restriction enzymes used herein are commercially available and their reaction conditions, cofactors and other requirements were used as would be known to the ordinarily skilled artisan.
  • For analytical purposes typically 1 ⁇ g of plasmid or DNA fragment is used with about 2 units of enzyme in about 20 ⁇ I of buffer solution.
  • For the purpose of isolating DNA fragments for plasmid construction typically 5 to 50 ⁇ g of DNA are digested with 20 to 250 units of enzyme in a larger volume. Appropriate buffers and substrate amounts for particular restriction enzymes are specified by the manufacturer. Incubation times of about 1 hour at 37° C. are ordinarily used, but may vary in accordance with the supplier's instructions. After digestion the reaction is electrophoresed directly on a polyacrylamide gel to isolate the desired fragment.
  • Oligonucleotides refers to either a single stranded polydeoxynucleotide or two complementary polydeoxynucleotide strands which may be chemically synthesized. Such synthetic oligonucleotides have no 5′ phosphate and thus will not ligate to another oligonucleotide without adding a phosphate with an ATP in the presence of a kinase. A synthetic oligonucleotide will ligate to a fragment that has not been dephosphorylated.
  • Ligase refers to the process of forming phosphodiester bonds between two double stranded nucleic acid fragments (Maniatis, T., et al., Id., p. 146). Unless otherwise provided, ligation may be accomplished using known buffers and conditions with 10 units to T4 DNA ligase (“ligase”) per 0.5 ⁇ g of approximately equimolar amounts of the DNA fragments to be ligated.
  • ligase T4 DNA ligase
  • Identity means, as utilized in the context of the present specification and claims, a homology comparison with respect to the degree of sameness between a first sequence and a second sequence (the first sequence may also be referred to as the “reference sequence”). Identity is expressed as the ratio N/D times 100 percent, where N is the number of identical aligned items (bases or amino acids) and D is the sum of the total number of items in the reference sequence and the total individual spaces (corresponding to items in the second sequence) introduced into the reference sequence as a result of its alignment with the second sequence.
  • the alignment by which the N/D ratio of identity is obtained is an alignment which gives essentially the largest possible percentage identity value, i.e., the largest N value (the largest number of aligned sequence items that are identical) and the smallest D value (the smallest number of individual gap spaces introduced into the reference sequence by the alignment). Ascertaining absolutely the highest possible identity value (or best alignment) is not required to report an “essentially largest identity value” since this means in the context of the present application that the percentage identity reported has a certainty deviation that limits any possible increases in the identity value due to an alternative alignment to less than one-half of a percentage point.
  • the sequence alignment utilized to obtain the N/D percentage identity may be performed by a manual method (hand and eye alignment) or by utilizing commercially available alignment software. The parameters of the alignment software may be adjusted until an identity value is obtained which has a certainty that limits any increase in the identity value to less than one-half of a percentage point with respect to the reported identity value.
  • “At Least X Percent Identity” means, as used in the context of the present specification or claims, a homology comparison with respect to the degree of sameness between a first sequence and a second sequence (the first sequence may also be referred to as the “reference sequence”) wherein the degree of sameness is equal to or exceeds the value “X” of the term.
  • the “identity” value (degree of sameness) of this term is expressed as the ratio N/D times 100 percent, where N is the number of identical aligned items (bases or amino acids) and D is the sum of the total number of items in the reference sequence and the total individual spaces (corresponding to items in the second sequence) introduced into the reference sequence as a result of its alignment with the second sequence.
  • the second sequence has “at least X percent identity” with respect to the reference sequence even though it may be possible to align the two sequence in a different manner such that the calculated value is less than X.
  • the sequence alignment utilized to obtain the N/D percentage identity may be performed by a manual method (hand and eye alignment) or by utilizing commercially available alignment software, provided that the “identity” value is calculated as hereinabove described.
  • the cDNA sequence coding for human slit is obtained from a cDNA library containing it (such as from MSCs or stem cells) and amplified by PCR using the oligonucleotide primers corresponding to the 5′ and 3′ end sequences of the processed slit nucleic acid sequence. Additional nucleotides corresponding to the slit gene are added to the 5′ and 3′ end sequences of the processed slit nucleic acid sequence.
  • Standard PCR amplification kits are available in the art and may be utilized for such amplification by following the PCR amplification instructions provided therewith.
  • Isolation of the full-length cDNA may be done utilizing methods standard in the art.
  • amplified cDNA may be utilized to produce the polypeptide which it encodes by utilizing methods standard in the art.
  • the cDNA sequence coding for human slit is obtained from a cDNA library and may be amplified as set forth in Example 1, above.
  • hslit cDNA fragment encompassing an EcoR1 site at the 5′-end and engineered to contain a Kpn1 site just before the termination codon was cloned into EcoR1, Kpn1 digested mammalian expression vector, pcDNA3,1/Myc-His/A (Invitrogen, Carlsbad, Calif.) such that the open reading frame of hSlit cDNA was in phase with the C-terminal myc epitope and the polyhistidine tag.
  • the pcDNA 3.1 vector was utilized in that it is designed for high level expression and purification of recombinant proteins in mammalian cells.
  • the human cytomegalovirus (CMV) promoter was utilized to provide high level expression in a wide range of mammalian cells.
  • the myc epitope and the his tag utilized allow tracking and purification of the expressed protein using commercially available (Invitrogen, Carlsbad, Calif.) anti-myc antibodies and metal-chelating resin, respectively.
  • BOSC 23 The human embryonic kidney cell line, BOSC 23, which does not express hSlit, and which can be transfected at a very high efficiency, was used for expression of hSlit.
  • BOSC 23 cells were transiently transfected with vector, or control plasmid, pcDNA3.1/Myc-His/lacZ, or pcDNA3.1/Myc-His/hSlit DNA using the standard calcium phosphate precipitation method. Forty-eight hours post-transfection, total cell lysates were prepared from the transfected and untransfected control cells and analyzed by Western blotting.
  • Protein content of the cell lysates was estimated using the BCA reagent (Pierce, Rockford, Ill.). Approximately 100 ⁇ g of protein from various samples was electrophoresed on a 7.5% SDS-PAGE gel, and electrophoretically transferred to Immobilon PVDF membrane (Milipore, Bedford, Mass.). The protein blot was probed with anti-myc antibodies using ECL detection reagents and protocol (Amersham, UK).
  • Lane 1 shows the results for untransfected BOSC cells
  • Lanes 2, 3 and 4 respectively, show (2) BOSC cells transfected with pcDNA3.1/Myc-His/A vector, (3) pcDNA3.1/Myc-His/lacZ and (4) pcDNA3.1/Myc-His/hSlit cDNA.

Abstract

This invention relates to newly identified polynucleotides, polypeptides encoded by such polynucleotides, the use of such polynucleotides and polypeptides, as well as the production of such polynucleotides and polypeptides. More particularly, the polypeptides of the present invention are human slit polypeptides. The invention also relates to identifying mesenchymal stem cells (MSCs) or other cells comprising such polypeptides or polynucleotides that encode the polypeptides.

Description

  • This invention relates to newly identified polynucleotides, polypeptides encoded by such polynucleotides, the use of such polynucleotides and polypeptides, as well as the production of such polynucleotides and polypeptides. More particularly, the polypeptides of the present invention are human Slit polypeptides. The invention also relates to identifying mesenchymal stem cells (MSCs) or other cells comprising such polypeptides or polynucleotides that encode the polypeptides. [0001]
  • Proteins containing epidermal growth factor (EGF)-like sequences have been shown to play an important role in many aspects of eukaryotic cell control, acting as signals for proliferation, growth inhibition, and differentiation. A common feature of these proteins is their involvement in extracellular events and ligand-receptor interactions. In characterizing genomic DNA identified by cross-hybridization to the sequence coding for the tandem EGF repeats of Notch in Drosophila, a related gene sequence from an unlinked locus that also has EGF repeats was discovered. Isolation and characterization of it, showed a corresponce to the slit locus. Further characterization of the related gene sequence established that null mutations to it would result in disruptions of the embryonic central nervous system (CNS) (Rothberg et al. 1988). Thus, it was shown to be involved in neurogenesis. [0002]
  • The Drosophila slit protein contains two types of repeated amino acid sequences: leucine rich repeats (“LRR”) and epidermal growth factor-like repeats (“EGF”). Its LRRs are arranged in four groups, each composed of four or five LRRs surrounded by conserved amino- and carboxy-flanking regions. The presence of both the LRRs and EGF-like repeats within a single protein make slit unusual in that such combination is not found in any other type of known protein. The absence of any potential transmembrane domains in a sequence having a typical signal sequence and two known extracellular-associated motifs suggests that the slit locus encodes a secreted extracellular protein. The LRR regions of the slit protein and such regions of related proteins participate in extracellular protein-protein interactions. Further, the EGF areas of the slit protein and such regions of related proteins participate in extracellular protein-protein reactions. Moreover, the slit protein is synthesized and secreted by midline glial cells can be come associated with axons. Among other functions, it influences the differentiation of midline cells from the neuroepithelium. [0003]
  • In accordance with one aspect of the present invention, there are provided novel polypeptides and polynucleotides, more particularly, the polypeptides of the present invention are of human origin and are found in human mesenchymal stem cells. MSCs are the formative pluripotential blast cells found inter alia in bone marrow, blood, dermis and periosteum that are capable of differentiating into any of the specific types of mesenchymal or connective tissues (i.e. the tissues of the body that support the specialized elements; particularly adipose, osseous, cartilaginous, elastic, and fibrous connective tissues) depending upon various influences from bioactive factors, such as cytokines. The polypeptide is designated as human Slit. The human Slit polypeptide according to the present invention, as well as biologically active and diagnostically or therapeutically useful fragments, analogs and derivatives thereof, is of use in studing the culturing of MSCs and detection of their differentiation and development into multipotent cells. [0004]
  • In accordance with another aspect of the present invention, there are provided isolated nucleic acid molecules encoding such polypeptides, including mRNAs, cDNAs, genomic DNA as well as biologically active and diagnostically or therapeutically useful fragments, analogs and derivatives thereof. [0005]
  • In accordance with another aspect of the present invention there are provided nucleic acid probes comprising nucleic acid molecules of sufficient length to specifically hybridize to sequences of the present invention. [0006]
  • In accordance with yet a further aspect of the present invention, there is provided a process for producing such polypeptides by recombinant techniques which comprises culturing recombinant prokaryotic and/or eukaryotic host cells, containing a nucleic acid sequence of the present invention, under conditions promoting expression of said protein and subsequent recovery of said protein. [0007]
  • In accordance with yet a further aspect of the present invention, there is provided a process for utilizing such polypeptides, or polynucleotides encoding such polypeptides for identifying human MSCs by utilizing the polynucleotides as probes or by expressing the polypeptides encoded thereby, using such polypeptides to produce an antibody specific for one of the polypeptides and then utilizing the antibody to identify the MSCs. Further such polynucleotides, polypeptides and antibodies may be utilized to aid in the identification of MSCs from other species, as well as to investigate/identify MSC functions in humans or other species. In a preferred aspect of the invention, immunocyto-chemistry is utilized with an antibody specific for a polypeptide according to the invention as a means for monitoring the concentration of the polypeptide according to the invention in a culture solution. The MSCs of the culture may thus be subjected to purification procedures to remove differentiated cells and help to maintain the MSCs in culture. [0008]
  • In accordance with yet a further aspect of the present invention, there are provided antibodies against such polypeptides and a method of employing such antibodies to detect diseases related to an overexpression or under expression of a polypeptide compressing a polypeptide with an amino acid sequence according to the present invention. Such antibodies (or active fragments) may be utilized to monitor the growth of MSCs in a culture or to detect the location of tumors in the body. [0009]
  • In accordance with another aspect of the present invention there is provided a method of diagnosing a disease or a susceptibility to a disease related to a mutation in the nucleic acid sequences and the proteins encoded by such nucleic acid sequences. [0010]
  • In accordance with yet a further aspect of the present invention, there is provided a process for utilizing such polypeptides, or polynucleotides encoding such polypeptides, for in vitro purposes related to scientific research, synthesis of DNA and manufacture of DNA vectors. [0011]
  • These and other aspects of the present invention should be apparent to those skilled in the art from the teachings herein. [0012]
  • The following drawings are illustrative of embodiments of the invention and are not meant to limit the scope of the invention as encompassed by the claims. [0013]
  • FIG. 1 shows a schematic of the transcript for a cDNA clone that encodes the mature slit polypeptide. The various coding regions and repeats are identified by different types of cross-hatching as shown in the figure and identified by the legend below it. [0014]
  • FIG. 2 shows a cDNA sequence (SEQ ID. NO:1) that encodes the mature slit polypeptide (SEQ ID NO:2) of the present invention. Sequencing was performed using a 373 Automated DNA sequencer (Applied Biosystems, Inc.). [0015]
  • FIG. 3 is an illustration of amino acid sequence homology between the human slit polypeptide of the present invention (labelled as hSlit) and the Drosophila slit polypeptide (labelled as dSlit). By aligning the two polypeptides in a manner that provides the essentially the largest number of aligned identical amino acids over the complete comparison area of the two sequences, and dividing the total number of identical amino acids by the total length of the comparison area (counting the individual spaces or gaps as part of the comparison area), a 40% identity between the two amino acid sequences was observed. Standard one-letter abbreviations for amino acids are used. [0016]
  • FIG. 4 shows a photograph of a protein blot from expression of hSlit in human embryonic kidney cell line, BOSC 23. Untransfected BOSC cells do not express hSlit. In FIG. 4, [0017] Lane 1 shows the results for untransfected BOSC cells; Lanes 2, 3 and 4, respectively show (2) BOSC cells transfected with pcDNA3.1/Myc-His/A vector, (3) pcDNA3.1/Myc-His/lacZ and (4) pcDNA3.1/Myc-His/hSlit cDNA.
  • In accordance with an aspect of the present invention, there are provided isolated nucleic acids (polynucleotides) which encode the mature polypeptides comprises the deduced amino acid sequences of SEQ ID NO:2. The mature forms of the slit polypeptide with and without an N-terminal methionine group (N-terminal methionine is the first amino acid of SEQ ID NO:2) are contemplated. [0018]
  • Polynucleotides encoding the polypeptide of the present invention have been isolated from a human MSC cDNA library. The polynucleotide contains an open reading frame encoding the human slit polypeptide. The protein exhibits a high degree of homology at the amino acid level to the Drosophila slit polypeptide with 40% identity (as shown in FIG. 3). [0019]
  • In accordance with a further aspect of the present invention the human slit gene sequence according to SEQ ID NO:1 or an appropriate fragment (full or partial length probes) may be utilized under stringent hybridization conditions to isolate from a cDNA library prepared from MSCs by procedures known in the art the cDNA encoding alleles of the mature slit polypeptide. Further, such full- or partial-length probes may be utilized to isolate genes (or cDNAs) encoding related polypeptides from non-human hosts under either stringent or highly-stringent hybridization conditions. Likewise the polypeptide having an amino acid sequence according to SEQ ID NO:2 or an immunogenic fragment may be utilized to produce antibodies specific for the polypeptide according to SEQ ID NO:2 and a fragment thereof. Such antibodies are in turn useful to detect the presence of such polypeptides when they are expressed by a clone or a transformed host cell to indicate the presence of the respective polynucleotides encoding such polypeptides. [0020]
  • The polynucleotides of the present invention may be in the form of RNA or in the form of DNA, which DNA includes cDNA, genomic DNA, and synthetic DNA. The DNA may be double-stranded or single-stranded, and if single stranded may be the coding strand or non-coding (anti-sense) strand. The coding sequence which encodes the mature polypeptides may comprise an amino acid sequence identical to the coding sequence shown in FIG. 1 (SEQ ID NO:1) or may be a different coding sequence which coding sequence, as a result of the redundancy or degeneracy of the genetic code, encodes the same mature polypeptides comprising the polypeptide of SEQ. ID NO:2, the cDNA for which is shown in FIG. 1 (SEQ ID NO:1). [0021]
  • The polynucleotides which encode the mature polypeptides of the present invention comprise the polynucleotide sequence encoding the polypeptide of SEQ ID NO:2 and may include: only the coding sequence for the mature polypeptide; the coding sequence for the mature polypeptide and additional coding sequence such as a leader or secretory sequence or a proprotein sequence; the coding sequence for the mature polypeptide (and optionally additional coding sequence) and non-coding sequence, such as introns or non-coding [0022] sequence 5′ and/or 3′ of the coding sequence for the mature polypeptides.
  • Thus, the term “polynucleotide encoding a polypeptide” encompasses a polynucleotide which includes coding sequence for the polypeptide and may also include additional coding and/or non-coding sequence such as introns. [0023]
  • The present invention further relates to variants of the hereinabove described polynucleotides which encode fragments, analogs and derivatives of the mature polypeptide comprising amino acid sequence shown in SEQ ID NO:2. The variant of the polynucleotides may be a naturally occurring allelic variant of the polynucleotides or a non-naturally occurring variant of the polynucleotides. [0024]
  • Further particularly preferred in this regard are polynucleotides encoding the human slit polypeptide variants, analogs, derivatives and fragments, and variants, analogs and derivatives of the fragments, which comprise the amino acid sequence of the polypeptide of SEQ ID NO:2 or of the deposit in which several, a few, 5 to 10, 1 to 5, 1 to 3, 2, 1 or no amino acid residues are substituted, deleted or added, in any combination. Especially preferred among these are silent substitutions, additions and deletions, which do not alter the properties and activities of the human slit polypeptide. Also especially preferred in this regard are conservative substitutions. Most highly preferred are mature polypeptides comprising the amino acid sequence set forth in SEQ ID NO:2 or of the deposit, without substitutions. [0025]
  • Thus, the present invention includes polynucleotides encoding the same mature polypeptides comprising the polypeptide as set forth in SEQ ID NO:2 as well as variants of such polynucleotides which variants encode a fragment, derivative or analog of the polypeptides set forth in SEQ ID NO:2. Such polynucleotide variants include deletion variants, substitution variants and addition or insertion variants. Preferred are polynucleotide sequences comprising polynucleotide sequence variants of a starting polynucleotide sequence that are obtained by changing the starting polynucleotide sequence in at least one of the following ways (a) inserting at least one nucleotide into it, (b) deleting at least one nucleotide from it, (c) substituting at least one nucleotide for a nucleotide of it, or (d) a combination of at least two of (a), (b) and (c). The starting polynucleotide sequence that is changed to obtain variant polynucleotide sequences is a member selected from (i) the coding portion of SEQ ID NO:1 and (ii) a redundant sequence encoding the same mature polypeptide as the coding portion of SEQ ID NO:1. Each of the preferred variant polynucleotide sequences results from changing no more than a total of 10 percent of the coding sequence nucleotides of the starting polynucleotide sequence by such deletion, substitution, insertion or a combination thereof (i.e., not more than 10 nucleotides per 100 nucleotides). More preferred are variant polynucleotide sequences that result from changing no more than a total of 5 percent of the starting coding sequence nucleotides by deletion, insertion, substitution or a combination thereof. Even more preferred are variant polynucleotide sequences that result from changing no more than a total of 3 percent of the starting coding sequence nucleotides by deletion, insertion, substitution or a combination thereof. Such changes occur within the 5′ to 3′ portions of the coding sequence of the starting polynucleotide. The polypeptides encoded by such variant polynucleotides may or may not retain the activity of the polypeptide encoded by the polynucleotide of SEQ ID NO:1. For example, such polynucleotides may be employed as probes for the gene comprising the polynucleotide of SEQ ID NO:1, for the polynucleotide of SEQ ID NO:1 or for a redundant polynucleotide which encodes the same polypeptide that is encoded by the polynucleotide having a sequence according to SEQ ID NO:1. However, preferred are the polynucleotides which encode variant polypeptides that retain substantially the same biological function or activity as the mature polypeptide comprising the amino acid sequence encoded by the cDNA of FIG. 1 (SEQ ID NO:2), or that of the amino acid sequence encoded by the polynucleotide of SEQ ID NO:1. [0026]
  • As hereinabove indicated, the polynucleotides may have a coding sequence which is a naturally occurring allelic variant of the coding sequences comprising the coding portion of the polynucleotide sequence shown in FIG. 1 (of SEQ ID NO:1). As known in the art, an allelic variant is an alternate form of a polynucleotide sequence which may have a substitution, deletion or addition of one or more nucleotides, which does not substantially alter the function of the encoded polypeptide. [0027]
  • The present invention also includes polynucleotides, wherein the coding sequence for the mature polypeptides may be fused in the same reading frame to a polynucleotide sequence which aids in expression and secretion of a polypeptide from a host cell, for example, a leader sequence which functions as a secretory sequence for controlling transport of a polypeptide from the cell. The polypeptide having a leader sequence is a preprotein and may have the leader sequence cleaved by the host cell to form the mature form of the polypeptide. The polynucleotides may also encode a proprotein which is the mature protein plus additional 5′ amino acid residues. A mature protein having a prosequence is a proprotein and is an inactive form of the protein. Once the prosequence is cleaved an active mature protein remains. [0028]
  • Thus, for example, the polynucleotides of the present invention may encode a mature protein, or for a protein having a prosequence or for a protein having both a prosequence and a presequence (leader sequence). [0029]
  • The polynucleotides of the present invention may also have the coding sequence fused in frame to a marker sequence which allows for purification of the polypeptides of the present invention. The marker sequence may be a hexa-histidine tag supplied by a pQE-9 vector to provide for purification of the mature polypeptides fused to the marker in the case of a bacterial host, or, for example, the marker sequence may be a hemagglutinin (HA) tag when a mammalian host, e.g. COS-7 cells, is used. The HA tag corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson, I., et al., Cell, 37:767 (1984)). [0030]
  • The term “gene” means the segment of DNA involved in producing a polypeptide chain; it includes regions preceding and following the coding region (leader and trailer) as well as intervening sequences (introns) between individual coding segments (exons). [0031]
  • Fragments of the full length gene of the present invention may be used as a hybridization probe for a cDNA library to isolate the full length cDNA and to isolate other cDNAs which have a high sequence similarity to the gene or similar biological activity. Probes of this type preferably have at least 30 bases and may contain, for example, 50 or more bases. The probe may also be used to identify a cDNA clone corresponding to a full length transcript and a genomic clone or clones that contain the complete gene including regulatory and promotor regions, exons, and introns. An example of a screen comprises isolating the coding region of the gene by using the known DNA sequence to synthesize an oligonucleotide probe. Labeled oligonucleotides having a sequence complementary to that of the gene of the present invention are used to screen a library of human cDNA, genomic DNA or mRNA to determine which members of the library the probe hybridizes to. [0032]
  • The present invention further relates to polynucleotides which hybridize to the hereinabove-described sequences if there is at least 70%, preferably at least 90%, and more preferably at least 95% identity between the sequences. The present invention particularly relates to polynucleotides which hybridize under stringent conditions to the hereinabove-described polynucleotides. As herein used, the term “stringent conditions” means hybridization will occur only if there is at least 95% and preferably at least 97% identity between the sequences. The polynucleotides which hybridize to the hereinabove described polynucleotides in a preferred embodiment encode polypeptides which either retain substantially the same biological function or activity as the mature polypeptide comprising the amino acid sequence encoded by the cDNA of FIG. 1 (comprising SEQ ID NO:2). [0033]
  • Alternatively, the polynucleotide may have at least 20 bases, preferably at least 30 bases, and more preferably at least 50 bases which hybridize to a polynucleotide of the present invention and which has an identity thereto, as hereinabove described, and which may or may not retain activity. For example, such polynucleotides may be employed as probes for the gene comprising the polynucleotide of SEQ ID NO:1, for example, for recovery of the polynucleotide or as a diagnostic probe or as a PCR primer. [0034]
  • Thus, the present invention is directed to polynucleotides having at least a 70% identity, preferably at least 90% and more preferably at least a 95% identity to a polynucleotide which encodes the polypeptide of SEQ ID NO:2 and polynucleotides complementary thereto as well as portions thereof, which portions have at least 20, preferably at least 30 consecutive bases and may have at least 50 consecutive bases and to polypeptides encoded by such polynucleotides. [0035]
  • The present invention further relates to polypeptides which have the deduced amino acid sequence as set forth in SEQ ID NO:2, as well as fragments, analogs and derivatives of such polypeptides. [0036]
  • The terms “fragment,” “derivative” and “analog” when referring to the mature polypeptides comprising the polypeptide as set forth in SEQ ID NO:2, means polypeptides which retain essentially the same biological function or activity as such polypeptides. Thus, an analog includes a proprotein which can be activated by cleavage of the proprotein portion to produce an active mature polypeptide. [0037]
  • Among the particularly preferred embodiments of the invention in this regard are mature polypeptides comprising the amino acid sequence as set forth in SEQ ID NO:2, variants, analogs, derivatives and fragments thereof, and variants, analogs and derivatives of the fragments. Alternatively, particularly preferred embodiments of the invention in this regard are polypeptides comprising the amino acid sequence of the human slit polypeptide encoded by the cDNA in the deposited clone, variants, analogs, derivatives and fragments thereof, and variants, analogs and derivatives of the fragments. [0038]
  • Further particularly preferred in this regard are variants, analogs, derivatives and fragments, and variants, analogs and derivatives of the fragments, comprising the amino acid sequence of the polypeptide as set forth in SEQ ID NO:2 or as encoded by the cDNA in the deposited clone, in which at least one amino acid residue per each 100 amino acids of the amino sequence is varied by at least one of (a) substituting an amino acid for it, (b) deleting at least one amino acid, (c) inserting at least one new amino acid, or (d) a combination of at least two of (a), (b) and (c). For example, variant polypeptides are obtained whose amino acid sequences are obtained by changing 5 to 10, 1 to 5, 1 to 3, or 1 to 2 amino acid residues per 100 amino acids in that at least one of (i) at least one new amino acid is substituted for an amino acid of SEQ ID NO:2 (or of a fragment of SEQ ID NO:2), (ii) at least one amino acid of SEQ ID NO:2 (or of a fragment of SEQ ID NO:2) is deleted, (ii) at least one new amino acid is inserted into SEQ ID NO:2 (or into a fragment of SEQ ID NO:2), or (iv) a combination of (i), (ii) or (iii). Especially preferred among these are silent substitutions, additions and deletions, which do not alter the properties and activities of such polypeptide as compared to those properties and activities of the human sift polypeptide. Also especially preferred in this regard are conservative substitutions. Most highly preferred are mature polypeptides comprising the amino acid sequence as set forth in SEQ ID NO:2, or of the deposited clone, without substitutions. [0039]
  • The polypeptides of the present invention may be recombinant polypeptides, natural polypeptides or synthetic polypeptides, preferably recombinant polypeptides. [0040]
  • The fragment, derivative or analog of the polypeptides comprising the amino acid sequence set forth in SEQ ID NO:2 may be (i) one in which one or more of the amino acid residues are substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code, or (ii) one in which one or more of the amino acid residues includes a substituent group, or (iii) one in which the mature polypeptide is fused with another compound, such as a compound to increase the half-life of the polypeptide (for example, polyethylene glycol), or (iv) one in which the additional amino acids are fused to the mature polypeptide, such as a leader or secretory sequence or a sequence which is employed for purification of the mature polypeptide or a proprotein sequence. Such fragments, derivatives and analogs are deemed to be within the scope of those skilled in the art from the teachings herein. [0041]
  • The polypeptides and polynucleotides of the present invention are preferably provided in an isolated form, and preferably are purified to homogeneity. [0042]
  • The term “isolated” means that the material is removed from its original environment (e.g., the natural environment if it is naturally occurring). For example, a naturally-occurring polynucleotide or polypeptide present in a living animal is not isolated, but the same polynucleotide or polypeptide, separated from some or all of the coexisting materials in the natural system, is isolated. Such polynucleotides could be part of a vector and/or such polynucleotides or polypeptides could be part of a composition, and still be isolated in that such vector or composition is not part of its natural environment. [0043]
  • The polypeptides of the present invention include polypeptides comprising the polypeptide of SEQ ID NO:2 (in particular the mature polypeptide) as well as polypeptides which have at least 70% similarity (preferably at least 70% identity) to the mature polypeptide comprising the amino acid sequence of SEQ ID NO:2, and which have at least 90% similarity (more preferably at least 90% identity) to the mature polypeptide comprising the amino acid sequence of SEQ ID NO:2 and still more preferably at least 95% similarity (still more preferably at least 95% identity) to the mature polypeptide comprising the amino acid sequence of SEQ ID NO:2 and also include portions of such polypeptides with such portion of the polypeptide generally containing at least 30 amino acids and more preferably at least 50 amino acids. [0044]
  • As known in the art “similarity” between two polypeptides is determined by comparing the amino acid sequence and its conserved amino acid substitutes of one polypeptide to the sequence of a second polypeptide. For such a determination, two amino acid sequences are compared along a stretch of their sequences, any gap (or gaps) introduced in one sequence to improve the alignment and similarity to the other sequences is counted as spaces of dissimilarity equal to the number of amino acids corresponding to the gap which are present in the second sequence, and the total number of similar amino acids are divided by the total number of amino acids present in the comparison area which counts the spaces of gaps as part of the comparison area. [0045]
  • Fragments or portions of the polypeptides of the present invention may be employed for producing the corresponding full-length polypeptide by peptide synthesis; therefore, the fragments may be employed as intermediates for producing the full-length polypeptides. Fragments or portions of the polynucleotides of the present invention may be used to synthesize full-length polynucleotides of the present invention. [0046]
  • The present invention also relates to vectors which include polynucleotides of the present invention, host cells which are genetically engineered with vectors of the invention and the production of polypeptides of the invention by recombinant techniques. [0047]
  • Host cells are genetically engineered (transduced or transformed or transfected) with the vectors of this invention which may be, for example, a cloning vector or an expression vector. The vector may be, for example, in the form of a plasmid, a viral particle, a phage, etc. The engineered host cells can be cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transformants or amplifying the genes. The culture conditions, such as temperature, pH and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan. [0048]
  • The polynucleotides of the present invention may be employed for producing polypeptides by recombinant techniques. Thus, for example, the polynucleotide may be included in any one of a variety of expression vectors for expressing a polypeptide. Such vectors include chromosomal, nonchromosomal and synthetic DNA sequences, e.g., derivatives of SV40; bacterial plasmids; phage DNA; baculovirus; yeast plasmids; vectors derived from combinations of plasmids and phage DNA, viral DNA such as vaccinia, adenovirus, fowl pox virus, and pseudorabies. However, any other vector may be used as long as it is replicable and viable in the host. [0049]
  • The appropriate DNA sequence may be inserted into the vector by a variety of procedures. In general, the DNA sequence is inserted into an appropriate restriction endonuclease site(s) by procedures known in the art. Such procedures and others are deemed to be within the scope of those skilled in the art. [0050]
  • The DNA sequence in the expression vector is operatively linked to an appropriate expression control sequence(s) (promoter) to direct mRNA synthesis. As representative examples of such promoters, there may be mentioned: LTR or SV40 promoter, the [0051] E. coli. lac or trp, the phage lambda PL promoter and other promoters known to control expression of genes in prokaryotic or eukaryotic cells or their viruses. The expression vector also contains a ribosome binding site for translation initiation and a transcription terminator. The vector may also include appropriate sequences for amplifying expression.
  • In addition, the expression vectors preferably contain one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells such as dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, or such as tetracycline or ampicillin resistance in [0052] E. coli.
  • The vector containing the appropriate DNA sequence as hereinabove described, as well as an appropriate promoter or control sequence, may be employed to transform an appropriate host to permit the host to express the protein. [0053]
  • As representative examples of appropriate hosts, there may be mentioned: bacterial cells, such as [0054] E. coli, Streptomyces, Salmonella typhimurium; fungal cells, such as yeast; insect cells such as Drosophila S2 and Spodoptera Sf9; animal cells such as CHO, COS or Bowes melanoma; adenoviruses; plant cells, etc. The selection of an appropriate host is deemed to be within the scope of those skilled in the art from the teachings herein.
  • More particularly, the present invention also includes recombinant constructs comprising one or more of the sequences as broadly described above. The constructs comprise a vector, such as a plasmid or viral vector, into which a sequence of the invention has been inserted, in a forward or reverse orientation. In a preferred aspect of this embodiment, the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the sequence. Large numbers of suitable vectors and promoters are known to those of skill in the art, and are commercially available. The following vectors are provided by way of example. Bacterial: pQE70, pQE60, pQE-9 (Qiagen), pBS, pD10, phagescript, psiX174, pBluescript SK, pBSKS, pNH8A, pNH16a, pNH18A, pNH46A (Stratagene); pTRC99a, pKK223-3, pKK233-3, pDR540, pRIT5 (Pharmacia). Eukaryotic: pWLNEO, pSV2CAT, pOG44, pXT1, pSG (Stratagene) pSVK3, pBPV, pMSG, pSVL (Pharmacia). However, any other plasmid or vector may be used as long as they are replicable and viable in the host. [0055]
  • Promoter regions can be selected from any desired gene using CAT (chloramphenicol transferase) vectors or other vectors with selectable markers. Two appropriate vectors are pKK232-8 and pCM7. Particular named bacterial promoters include lacl, lacZ, T3, T7, gpt, lambda P[0056] R, PL and trp. Eukaryotic promoters include CMV immediate early, HSV thymidine kinase, early and late SV40, LTRs from retrovirus, and mouse metallothionein-I. Selection of the appropriate vector and promoter is well within the level of ordinary skill in the art.
  • In a further embodiment, the present invention relates to host cells containing the above-described constructs. The host cell can be a higher eukaryotic cell, such as a mammalian cell, or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell. Introduction of the construct into the host cell can be effected by calcium phosphate transfection, DEAE-Dextran mediated transfection, or electroporation (Davis, L., Dibner, M., Battey, I., Basic Methods in Molecular Biology, (1986)). [0057]
  • The constructs in host cells can be used in a conventional manner to produce the gene products encoded by the recombinant sequences. Alternatively, the polypeptides of the invention can be synthetically produced by conventional peptide synthesizers. [0058]
  • Mature proteins can be expressed in mammalian cells, yeast, bacteria, or other cells under the control of appropriate promoters. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention. Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrook, et al., Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor, N.Y., (1989), the disclosure of which is hereby incorporated by reference. [0059]
  • Transcription of the DNA encoding the polypeptides of the present invention by higher eukaryotes is increased by inserting an enhancer sequence into the vector. Enhancers are cis-acting elements of DNA, usually about from 10 to 300 bp that act on a promoter to increase its transcription. Examples include the SV40 enhancer on the late side of the replication origin bp 100 to 270, a cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers. [0060]
  • Generally, recombinant expression vectors will include origins of replication and selectable markers permitting transformation of the host cell, e.g., the ampicillin resistance gene of [0061] E. coli and S. cerevisiae TRP1 gene, and a promoter derived from a highly-expressed gene to direct transcription of a downstream structural sequence. Such promoters can be derived from operons encoding glycolytic enzymes such as 3-phosphoglycerate kinase (PGK), á-factor, acid phosphatase, or heat shock proteins, among others. The heterologous structural sequence is assembled in appropriate phase with translation initiation and termination sequences, and preferably, a leader sequence capable of directing secretion of translated protein into the periplasmic space or extracellular medium. Optionally, the heterologous sequence can encode a fusion protein including an N-terminal identification peptide imparting desired characteristics, e.g., stabilization or simplified purification of expressed recombinant product.
  • Useful expression vectors for bacterial use are constructed by inserting a structural DNA sequence encoding a desired protein together with suitable translation initiation and termination signals in operable reading phase with a functional promoter. The vector will comprise one or more phenotypic selectable markers and an origin of replication to ensure maintenance of the vector and to, if desirable, provide amplification within the host. Suitable prokaryotic hosts for transformation include [0062] E. coli, Bacillus subtilis, Salmonella typhimurium and various species within the genera Pseudomonas, Streptomyces, and Staphylococcus, although others may also be employed as a matter of choice.
  • As a representative but nonlimiting example, useful expression vectors for bacterial use can comprise a selectable marker and bacterial origin of replication derived from commercially available plasmids comprising genetic elements of the well known cloning vector pBR322 (ATCC 37017). Such commercial vectors include, for example, pKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and pGEM1 (Promega Biotec, Madison, Wis., USA). These pBR322 “backbone” sections are combined with an appropriate promoter and the structural sequence to be expressed. [0063]
  • Following transformation of a suitable host strain and growth of the host strain to an appropriate cell density, the selected promoter is induced by appropriate means (e.g., temperature shift or chemical induction) and cells are cultured for an additional period. [0064]
  • Cells are typically harvested by centrifugation, disrupted by physical or chemical means, and the resulting crude extract retained for further purification. [0065]
  • Microbial cells employed in expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents, such methods are well know to those skilled in the art. [0066]
  • Various mammalian cell culture systems can also be employed to express recombinant protein. Examples of mammalian expression systems include the COS-7 lines of monkey kidney fibroblasts, described by Gluzman, Cell, 23:175 (1981), and other cell lines capable of expressing a compatible vector, for example, the C127, 3T3, CHO, HeLa, 293 and BHK cell lines. Mammalian expression vectors will comprise an origin of replication, a suitable promoter and enhancer, and also any necessary ribosome binding sites, polyadenylation site, splice donor and acceptor sites, transcriptional termination sequences, and 5′ flanking nontranscribed sequences. DNA sequences derived from the SV40 splice, and polyadenylation sites may be used to provide the required nontranscribed genetic elements. [0067]
  • The polypeptides can be recovered and purified from recombinant cell cultures by methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Protein refolding steps can be used, as necessary, in completing configuration of the mature protein. Finally, high performance liquid chromatography (HPLC) can be employed for final purification steps. [0068]
  • The polypeptides of the present invention may be a naturally purified product, or a product of chemical synthetic procedures, or produced by recombinant techniques from a prokaryotic or eukaryotic host (for example, by bacterial, yeast, higher plant, insect and mammalian cells in culture). Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosylated. Polypeptides of the invention may also include an initial methionine amino acid residue. [0069]
  • The polynucleotides and polypeptides of the present invention may be employed as research reagents and materials for discovery of treatments and diagnostics for human disease. For example, the polynucleotides and polypeptides encoded by such polynucleotides may also be utilized for in vitro purposes related to scientific research, synthesis of DNA and manufacture of DNA vectors and for designing therapeutics and diagnostics for human disease. [0070]
  • The invention also provides a method for identifying human mesenchymal stem cells by contacting a mixture of mRNA from a cell sample with a polynucleotide unique to human slit and identifying any mRNA which has hybridized with the polynucleotide unique to human slit. In a preferred embodiment the polynucleotide unique to human slit is bound to a solid support. Thus, for example, the identification of slit cDNA enables the slit nucleic acid sequence to be utilized as a diagnostic reagent to identify human MSCs, such as by using gene expression array technology. Labeled (e.g. fluorescent or radiolabeled) mixtures of total cellular mRNA hybridize to cognate elements of slit on a chip based array and allow for the accurate detection of genes specific to MSCs. This technology is described, for example, in Schena, [0071] Bioessays, 18(5):427-431 (May 1996) and O'Donnell-Maloney & Little, Genet Anal., 13(6):151-157 (Dec. 1996).
  • The polypeptides of the present invention and fragments and analogs and derivatives thereof may be identified by assays which detect MSC proliferation or other activity. Further, assays may be utilized which neutralize the production of the native slit in midline glia cells and subjecting such cells to a polypeptide sequence which is related to the native slit sequence but is different in order to verify the same functionality of polypeptides having both sequences. [0072]
  • This invention is also related to the use of the human slit polypeptide gene as part of a diagnostic assay for detecting diseases or susceptibility to diseases related to the presence of mutations in the human slit polypeptide nucleic acid sequences. Such diseases are related to under-expression or overexpression of the human slit polypeptides. [0073]
  • Individuals carrying mutations in the human slit polypeptide gene may be detected at the DNA level by a variety of techniques. Nucleic acids for diagnosis may be obtained from a patient's cells, such as from blood, urine, saliva, tissue biopsy and autopsy material. The genomic DNA may be used directly for detection or may be amplified enzymatically by using PCR (Saiki et al., Nature, 324:163-166 (1986)) prior to analysis. RNA or cDNA may also be used for the same purpose. As an example, PCR primers complementary to the nucleic acid encoding human slit polypeptide can be used to identify and analyze human slit polypeptide mutations. For example, deletions and insertions can be detected by a change in size of the amplified product in comparison to the normal genotype. Point mutations can be identified by hybridizing amplified DNA to radiolabeled human slit polypeptide RNA or alternatively, radiolabeled human slit polypeptide antisense DNA sequences. Perfectly matched sequences can be distinguished from mismatched duplexes by RNase A digestion or by differences in melting temperatures. [0074]
  • Genetic testing based on DNA sequence differences may be achieved by detection of alteration in electrophoretic mobility of DNA fragments in gels with or without denaturing agents. Small sequence deletions and insertions can be visualized by high resolution gel electrophoresis. DNA fragments of different sequences may be distinguished on denaturing formamide gradient gels in which the mobilities of different DNA fragments are retarded in the gel at different positions according to their specific melting or partial melting temperatures (see, e.g., Myers et al., Science, 230:1242 (1985)). [0075]
  • Sequence changes at specific locations may also be revealed by nuclease protection assays, such as RNase and S1 protection or the chemical cleavage method (e.g., Cotton et al., PNAS, USA, 85:4397-4401 (1985)). [0076]
  • Thus, the detection of a specific DNA sequence may be achieved by methods such as hybridization, RNase protection, chemical cleavage, direct DNA sequencing or the use of restriction enzymes, (e.g., Restriction Fragment Length Polymorphisms (RFLP)) and Southern blotting of genomic DNA. [0077]
  • In addition to more conventional gel-electrophoresis and DNA sequencing, mutations can also be detected by in situ analysis. [0078]
  • The present invention also relates to a diagnostic assay for detecting altered levels of the slit polypeptide in various tissues since an over-expression or under-expression of the proteins compared to normal control tissue samples may detect the presence of a disease or susceptibility to a disease, for example, reduced blood cell counts or malignancies such as cancers and tumors. Assays used to detect levels of the slit polypeptide in a sample derived from a host are well-known to those of skill in the art and include radioimmunoassays, competitive-binding assays, Western Blot analysis, ELISA assays and “sandwich” assay. An ELISA assay (Coligan, et al., Current Protocols in Immunology, 1(2), Chapter 6, (1991)) initially comprises preparing an antibody specific to the slit polypeptide antigen, preferably a monoclonal antibody. In addition a reporter antibody is prepared against the monoclonal antibody. To the reporter antibody is attached a detectable reagent such as radioactivity, fluorescence or, in this example, a horseradish peroxidase enzyme. A sample is removed from a host and incubated on a solid support, e.g. a polystyrene dish, that binds the proteins in the sample. Any free protein binding sites on the dish are then covered by incubating with a non-specific protein like BSA. Next, the monoclonal antibody is incubated in the dish during which time the monoclonal antibodies attach to any slit polypeptide attached to the polystyrene dish. All unbound monoclonal antibody is washed out with buffer. The reporter antibody linked to horseradish peroxidase is now placed in the dish resulting in binding of the reporter antibody to any monoclonal antibody bound to the slit polypeptide. Unattached reporter antibody is then washed out. Peroxidase substrates are then added to the dish and the amount of color developed in a given time period is a measurement of the amount of the slit polypeptide present in a given volume of patient sample when compared against a standard curve. [0079]
  • A competition assay may be employed wherein antibodies specific to the slit polypeptide are attached to a solid support and labeled the slit polypeptide and a sample derived from the host are passed over the solid support and the amount of label detected, for example by liquid scintillation chromatography, can be correlated to a quantity of the slit polypeptide in the sample. [0080]
  • A “sandwich” assay is similar to an ELISA assay. In a “sandwich” assay the slit polypeptide is passed over a solid support and binds to antibody attached to a solid support. A second antibody is then bound to the slit polypeptide. A third antibody which is labeled and specific to the second antibody is then passed over the solid support and binds to the second antibody and an amount can then be quantified. [0081]
  • This invention provides a method for identification of the receptors for the human slit polypeptides. The gene encoding the receptor can be identified by numerous methods known to those of skill in the art, for example, ligand panning and FACS sorting (Coligan, et al., Current Protocols in Immun., 1(2), [0082] Chapter 5, (1991)). Preferably, expression cloning is employed wherein polyadenylated RNA is prepared from a cell responsive to the polypeptides, and a cDNA library created from this RNA is divided into pools and used to transfect COS cells or other cells that are not responsive to the polypeptides. Transfected cells which are grown on glass slides are exposed to the labeled polypeptides. The polypeptides can be labeled by a variety of means including iodination or inclusion of a recognition site for the slit polypeptides. Following fixation and incubation, the slides are subjected to autoradiographic analysis. Positive pools are identified and sub-pools are prepared and retransfected using an iterative sub-pooling and rescreening process, eventually yielding a single clones that encodes the putative receptor.
  • As an alternative approach for receptor identification, the labeled polypeptides can be photoaffinity linked with cell membrane or extract preparations that express the receptor molecule. Cross-linked material is resolved by PAGE analysis and exposed to X-ray film. The labeled complex containing the receptors of the polypeptides can be excised, resolved into peptide fragments, and subjected to protein microsequencing. The amino acid sequence obtained from microsequencing would be used to design a set of degenerate oligonucleotide probes to screen a cDNA library to identify the genes encoding the putative receptors. [0083]
  • This invention provides a method of screening compounds to identify agonists and antagonists to the human slit polypeptides of the present invention. An agonist is a compound which has similar biological functions of the polypeptides, while antagonists block such functions. Antagonists and agonists may be identified by the an MSC proliferation assay as is well known in the art. [0084]
  • Examples of potential the slit polypeptide antagonists include antibodies, or in some cases, oligonucleotides, which bind to the polypeptides. Another example of a potential antagonist is a negative dominant mutant of the polypeptides. Negative dominant mutants are polypeptides which bind to the receptor of the wild-type polypeptide, but fail to retain biological activity. [0085]
  • Antisense constructs prepared using antisense technology are also potential antagonists. Antisense technology can be used to control gene expression through triple-helix formation or antisense DNA or RNA, both of which methods are based on binding of a polynucleotide to DNA or RNA. For example, the 5′ coding portion of the polynucleotide sequence, which encodes the mature polypeptides of the present invention, is used to design an antisense RNA oligonucleotide of from about 10 to 40 base pairs in length. A DNA oligonucleotide is designed to be complementary to a region of the gene involved in transcription (triple- helix, see Lee et al., Nucl. Acids Res., 6:3073 (1979); Cooney et al., Science, 241:456 (1988); and Dervan et al., Science, 251: 1360 (1991)), thereby preventing transcription and the production of the human slit polypeptide. The antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into the polypeptides (antisense—Okano, J. Neurochem., 56:560 (1991); Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression, CRC Press, Boca Raton, Fla. (1988)). The oligonucleotides described above can also be delivered to cells such that the antisense RNA or DNA may be expressed in vivo to inhibit production of the human slit polypeptide. [0086]
  • Another potential human slit antagonist is a peptide derivative of the polypeptides which are naturally or synthetically modified analogs of the polypeptides that have lost biological function yet still recognize and bind to the receptors of the polypeptides to thereby effectively block the receptors. Examples of peptide derivatives include, but are not limited to, small peptides or peptide-like molecules. [0087]
  • The antagonists may be employed in a composition with a pharmaceutically acceptable carrier, e.g., as hereinafter described. [0088]
  • The human slit polypeptides and antagonists may be employed in combination with a suitable pharmaceutical carrier. Such compositions comprise a therapeutically effective amount of the polypeptide, and a pharmaceutically acceptable carrier or excipient. Such a carrier includes but is not limited to saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof. The formulation should suit the mode of administration. [0089]
  • The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention. Associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration. In addition, the polypeptides and agonists and antagonists may be employed in conjunction with other therapeutic compounds. [0090]
  • The pharmaceutical compositions may be administered in a convenient manner such as by the topical, intravenous, intraperitoneal, intramuscular, intratumor, subcutaneous, intranasal or intradermal routes. The pharmaceutical compositions are administered in an amount which is effective for treating and/or prophylaxis of the specific indication. In general, the polypeptides will be administered in an amount of at least about 10 μg/kg body weight and in most cases they will be administered in an amount not in excess of about 8 mg/Kg body weight per day. In most cases, the dosage is from about 10 μg/kg to about 1 mg/kg body weight daily, taking into account the routes of administration, symptoms, etc. [0091]
  • The human slit polypeptides, and agonists or antagonists which are polypeptides, may be employed in accordance with the present invention by expression of such polypeptides in vivo, which is often referred to as “gene therapy.”[0092]
  • Thus, for example, cells from a patient may be engineered with a polynucleotide (DNA or RNA) encoding a polypeptide ex vivo, with the engineered cells then being provided to a patient to be treated with the polypeptide. Such methods are well-known in the art. For example, cells may be engineered by procedures known in the art by use of a retroviral particle containing RNA encoding a polypeptide of the present invention. [0093]
  • Similarly, cells may be engineered in vivo for expression of a polypeptide in vivo by, for example, procedures known in the art. As known in the art, a producer cell for producing a retroviral particle containing RNA encoding the polypeptide of the present invention may be administered to a patient for engineering cells in vivo and expression of the polypeptide in vivo. These and other methods for administering a polypeptide of the present invention by such method should be apparent to those skilled in the art from the teachings of the present invention. For example, the expression vehicle for engineering cells may be other than a retrovirus, for example, an adenovirus which may be used to engineer cells in vivo after combination with a suitable delivery vehicle. [0094]
  • Retroviruses from which the retroviral plasmid vectors hereinabove mentioned may be derived include, but are not limited to, Moloney Murine Leukemia Virus, spleen necrosis virus, retroviruses such as Rous Sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, gibbon ape leukemia virus, human immunodeficiency virus, adenovirus, Myeloproliferative Sarcoma Virus, and mammary tumor virus. In one embodiment, the retroviral plasmid vector is derived from Moloney Murine Leukemia Virus. [0095]
  • The vector includes one or more promoters. Suitable promoters which may be employed include, but are not limited to, the retroviral LTR; the SV40 promoter; and the human cytomegalovirus (CMV) promoter described in Miller, et al., [0096] Biotechniques, Vol. 7, No. 9, 980-990 (1989), or any other promoter (e.g., cellular promoters such as eukaryotic cellular promoters including, but not limited to, the histone, pol 111, and â-actin promoters). Other viral promoters which may be employed include, but are not limited to, adenovirus promoters, thymidine kinase (TK) promoters, and B19 parvovirus promoters. The selection of a suitable promoter will be apparent to those skilled in the art from the teachings contained herein.
  • The nucleic acid sequence encoding the polypeptide of the present invention is under the control of a suitable promoter. Suitable promoters which may be employed include, but are not limited to, adenoviral promoters, such as the adenoviral major late promoter; or heterologous promoters, such as the cytomegalovirus (CMV) promoter; the respiratory syncytial virus (RSV) promoter; inducible promoters, such as the MMT promoter, the metallothionein promoter; heat shock promoters; the albumin promoter; the ApoAl promoter; human globin promoters; viral thymidine kinase promoters, such as the Herpes Simplex thymidine kinase promoter; retroviral LTRs (including the modified retroviral LTRs hereinabove described); the â-actin promoter; and human growth hormone promoters. The promoter also may be the native promoter which controls the gene encoding the polypeptide. [0097]
  • The retroviral plasmid vector is employed to transduce packaging cell lines to form producer cell lines. Examples of packaging cells which may be transfected include, but are not limited to, the PE501, PA317, é-2, é-AM, PA12, T19-14X, VT-19-17-H2, éCRE, éCRIP, GP+E-86, GP+envAm12, and DAN cell lines as described in Miller, [0098] Human Gene Therapy, Vol. 1, pgs. 5-14 (1990), which is incorporated herein by reference in its entirety. The vector may transduce the packaging cells through any means known in the art. Such means include, but are not limited to, electroporation, the use of liposomes, and CaPO4 precipitation. In one alternative, the retroviral plasmid vector may be encapsulated into a liposome, or coupled to a lipid, and then administered to a host.
  • The producer cell line generates infectious retroviral vector particles which include the nucleic acid sequence(s) encoding the polypeptides. Such retroviral vector particles then may be employed, to transduce eukaryotic cells, either in vitro or in vivo. The transduced eukaryotic cells will express the nucleic acid sequence(s) encoding the polypeptide. Eukaryotic cells which may be transduced include, but are not limited to, embryonic stem cells, embryonic carcinoma cells, as well as hematopoietic stem cells, hepatocytes, fibroblasts, myoblasts, keratinocytes, endothelial cells, and bronchial epithelial cells. The sequences of the present invention are also valuable for chromosome identification. The sequence is specifically targeted to and can hybridize with a particular location on an individual human chromosome. Moreover, there is a current need for identifying particular sites on the chromosome. Few chromosome marking reagents based on actual sequence data (repeat polymorphisms) are presently available for marking chromosomal location. The mapping of DNAs to chromosomes according to the present invention is an important first step in correlating those sequences with genes associated with disease. [0099]
  • The sequences of the present invention are also valuable for chromosome identification. The sequence is specifically targeted to and can hybridize with a particular location on an individual human chromosome. Moreover, there is a current need for identifying particular sites on the chromosome. Few chromosome marking reagents based on actual sequence data (repeat polymorphisms) are presently available for marking chromosomal location. The mapping of DNAs to chromosomes according to the present invention is an important first step in correlating those sequences with genes associated with disease. [0100]
  • Briefly, sequences can be mapped to chromosomes by preparing PCR primers (preferably 15-25 bp) from the cDNA. Computer analysis of the 3′ untranslated region is used to rapidly select primers that do not span more than one exon in the genomic DNA, thus complicating the amplification process. These primers are then used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene corresponding to the primer will yield an amplified fragment. [0101]
  • PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular DNA to a particular chromosome. Using the present invention with the same oligonucleotide primers, sublocalization can be achieved with panels of fragments from specific chromosomes or pools of large genomic clones in an analogous manner. Other mapping strategies that can similarly be used to map to its chromosome include in situ hybridization, prescreening with labeled flow-sorted chromosomes and preselection by hybridization to construct chromosome specific-cDNA libraries. [0102]
  • Fluorescence in situ hybridization (FISH) of a cDNA clones to a metaphase chromosomal spread can be used to provide a precise chromosomal location in one step. This technique can be used with cDNA as short as 50 or 60 bases. For a review of this technique, see Verma et al., Human Chromosomes: a Manual of Basic Techniques, Pergamon Press, New York (1988). [0103]
  • Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be correlated with genetic map data. Such data are found, for example, in V. McKusick, Mendelian Inheritance in Man (available on line through Johns Hopkins University Welch Medical Library). The relationship between genes and diseases that have been mapped to the same chromosomal region are then identified through linkage analysis (coinheritance of physically adjacent genes). [0104]
  • Next, it is necessary to determine the differences in the cDNA or genomic sequence between affected and unaffected individuals. If a mutation is observed in some or all of the affected individuals but not in any normal individuals, then the mutation is likely to be the causative agent of the disease. [0105]
  • With current resolution of physical mapping and genetic mapping techniques, a cDNA precisely localized to a chromosomal region associated with the disease could be one of between 50 and 500 potential causative genes. (This assumes 1 megabase mapping resolution and one gene per 20 kb). [0106]
  • The polypeptides, their fragments or other derivatives, or analogs thereof, or cells expressing them can be used as an immunogen to produce antibodies thereto. These antibodies can be, for example, polyclonal or monoclonal antibodies. The present invention also includes chimeric, single chain, and humanized antibodies, as well as Fab fragments, or the product of an Fab expression library. Various procedures known in the art may be used for the production of such antibodies and fragments. [0107]
  • Antibodies generated against the polypeptides corresponding to a sequence of the present invention can be obtained by direct injection of the polypeptides into an animal or by administering the polypeptides to an animal, preferably a nonhuman. The antibody so obtained will then bind the polypeptides itself. In this manner, even a sequence encoding only a fragment of the polypeptides can be used to generate antibodies binding the whole native polypeptides. Such antibodies can then be used to isolate the polypeptide from tissue expressing that polypeptide. [0108]
  • Antibodies specific to the polypeptide of the present invention may be employed as a diagnostic to determine elevated or lowered levels of the polypeptide in a sample derived from a host by techniques known in the art. These elevated or lowered levels are indicative of certain disorders which are characterized by such levels of the protein of the present invention and members of its family. [0109]
  • For preparation of monoclonal antibodies, any technique which provides antibodies produced by continuous cell line cultures can be used. Examples include the hybridoma technique (Kohler and Milstein, 1975, Nature, 256:495-497), the trioma technique, the human B-cell hybridoma technique (Kozbor et al., 1983, Immunology Today 4:72), and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole, et al., 1985, in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96). [0110]
  • Techniques described for the production of single chain antibodies (U.S. Pat. No. 4,946,778) can be adapted to produce single chain antibodies to immunogenic polypeptide products of this invention. Also, transgenic mice may be used to express humanized antibodies to immunogenic polypeptide products of this invention. [0111]
  • Such antibodies to the polypeptides of the present invention may be utilized to detect the presence or the absence of the polypeptides of the present invention. Thus, they are useful in an assay to verify the successful insertion of the polynucleotides of the present invention (as part of a construct) into a host cell. Thus, the protein encoded by the inserted polynucleotide according to the present invention, when expressed by the transformed host cell, serves as a “marker” for the successful insertion of the polynucleotide that can be detected by an antibody for the marker. [0112]
  • “Plasmids” are designated by a lower case p preceded and/or followed by capital letters and/or numbers. The starting plasmids herein are either commercially available, publicly available on an unrestricted basis, or can be constructed from available plasmids in accord with published procedures. In addition, equivalent plasmids to those described are known in the art and will be apparent to the ordinarily skilled artisan. [0113]
  • “Digestion” of DNA refers to catalytic cleavage of the DNA with a restriction enzyme that acts only at certain sequences in the DNA. The various restriction enzymes used herein are commercially available and their reaction conditions, cofactors and other requirements were used as would be known to the ordinarily skilled artisan. For analytical purposes, typically 1 ìg of plasmid or DNA fragment is used with about 2 units of enzyme in about 20 ìI of buffer solution. For the purpose of isolating DNA fragments for plasmid construction, typically 5 to 50 ìg of DNA are digested with 20 to 250 units of enzyme in a larger volume. Appropriate buffers and substrate amounts for particular restriction enzymes are specified by the manufacturer. Incubation times of about 1 hour at 37° C. are ordinarily used, but may vary in accordance with the supplier's instructions. After digestion the reaction is electrophoresed directly on a polyacrylamide gel to isolate the desired fragment. [0114]
  • Size separation of the cleaved fragments is performed using 8 percent polyacrylamide gel described by Goeddel, D. et al., Nucleic Acids Res., 8:4057 (1980). [0115]
  • “Oligonucleotides” refers to either a single stranded polydeoxynucleotide or two complementary polydeoxynucleotide strands which may be chemically synthesized. Such synthetic oligonucleotides have no 5′ phosphate and thus will not ligate to another oligonucleotide without adding a phosphate with an ATP in the presence of a kinase. A synthetic oligonucleotide will ligate to a fragment that has not been dephosphorylated. [0116]
  • “Ligation” refers to the process of forming phosphodiester bonds between two double stranded nucleic acid fragments (Maniatis, T., et al., Id., p. 146). Unless otherwise provided, ligation may be accomplished using known buffers and conditions with 10 units to T4 DNA ligase (“ligase”) per 0.5 ìg of approximately equimolar amounts of the DNA fragments to be ligated. [0117]
  • “Identity” means, as utilized in the context of the present specification and claims, a homology comparison with respect to the degree of sameness between a first sequence and a second sequence (the first sequence may also be referred to as the “reference sequence”). Identity is expressed as the ratio N/D times 100 percent, where N is the number of identical aligned items (bases or amino acids) and D is the sum of the total number of items in the reference sequence and the total individual spaces (corresponding to items in the second sequence) introduced into the reference sequence as a result of its alignment with the second sequence. Further, the alignment by which the N/D ratio of identity is obtained is an alignment which gives essentially the largest possible percentage identity value, i.e., the largest N value (the largest number of aligned sequence items that are identical) and the smallest D value (the smallest number of individual gap spaces introduced into the reference sequence by the alignment). Ascertaining absolutely the highest possible identity value (or best alignment) is not required to report an “essentially largest identity value” since this means in the context of the present application that the percentage identity reported has a certainty deviation that limits any possible increases in the identity value due to an alternative alignment to less than one-half of a percentage point. The sequence alignment utilized to obtain the N/D percentage identity may be performed by a manual method (hand and eye alignment) or by utilizing commercially available alignment software. The parameters of the alignment software may be adjusted until an identity value is obtained which has a certainty that limits any increase in the identity value to less than one-half of a percentage point with respect to the reported identity value. [0118]
  • “At Least X Percent Identity” means, as used in the context of the present specification or claims, a homology comparison with respect to the degree of sameness between a first sequence and a second sequence (the first sequence may also be referred to as the “reference sequence”) wherein the degree of sameness is equal to or exceeds the value “X” of the term. The “identity” value (degree of sameness) of this term is expressed as the ratio N/D times 100 percent, where N is the number of identical aligned items (bases or amino acids) and D is the sum of the total number of items in the reference sequence and the total individual spaces (corresponding to items in the second sequence) introduced into the reference sequence as a result of its alignment with the second sequence. If any alignment exists for the second sequence and the reference sequence which results in a sameness value (NID×100%) that is equal to or greater than the value of “X” in the phrase “at least X percent identity” then the second sequence has “at least X percent identity” with respect to the reference sequence even though it may be possible to align the two sequence in a different manner such that the calculated value is less than X. The sequence alignment utilized to obtain the N/D percentage identity may be performed by a manual method (hand and eye alignment) or by utilizing commercially available alignment software, provided that the “identity” value is calculated as hereinabove described. [0119]
  • Unless otherwise stated, transformation was performed as described in the method of Graham, F. and Van der Eb, A., Virology, 52:456-457 (1973). [0120]
  • The present invention will be further described with reference to the following examples; however, it is to be understood that the present invention is not limited to such examples. All parts or amounts, unless otherwise specified, are by weight.[0121]
  • In order to facilitate understanding of the invention the following examples providing certain frequently occurring methods and/or terms will be described. [0122]
  • EXAMPLE 1 PCR Amplification of Human Slit
  • The cDNA sequence coding for human slit is obtained from a cDNA library containing it (such as from MSCs or stem cells) and amplified by PCR using the oligonucleotide primers corresponding to the 5′ and 3′ end sequences of the processed slit nucleic acid sequence. Additional nucleotides corresponding to the slit gene are added to the 5′ and 3′ end sequences of the processed slit nucleic acid sequence. [0123]
  • For example, the following PCR primers may be utilized for amplification of the cDNA: [0124]
    5′primer = TCCTCGGGCTCCACGCGTCTT (SEQ ID NO:3), and
    3′primer = GGTACATATACGCAGATGGTG (SEQ ID NO:4).
  • Standard PCR amplification kits are available in the art and may be utilized for such amplification by following the PCR amplification instructions provided therewith. [0125]
  • Isolation of the full-length cDNA may be done utilizing methods standard in the art. [0126]
  • Furthermore, the amplified cDNA may be utilized to produce the polypeptide which it encodes by utilizing methods standard in the art. [0127]
  • EXAMPLE 2 Expression and Purification of Human Slit
  • The cDNA sequence coding for human slit is obtained from a cDNA library and may be amplified as set forth in Example 1, above. [0128]
  • A. Construction of Expression Plasmid [0129]
  • The full-length hslit cDNA fragment encompassing an EcoR1 site at the 5′-end and engineered to contain a Kpn1 site just before the termination codon was cloned into EcoR1, Kpn1 digested mammalian expression vector, pcDNA3,1/Myc-His/A (Invitrogen, Carlsbad, Calif.) such that the open reading frame of hSlit cDNA was in phase with the C-terminal myc epitope and the polyhistidine tag. [0130]
  • The pcDNA 3.1 vector was utilized in that it is designed for high level expression and purification of recombinant proteins in mammalian cells. The human cytomegalovirus (CMV) promoter was utilized to provide high level expression in a wide range of mammalian cells. The myc epitope and the his tag utilized allow tracking and purification of the expressed protein using commercially available (Invitrogen, Carlsbad, Calif.) anti-myc antibodies and metal-chelating resin, respectively. [0131]
  • B. Transfection of BOSC 23 Cells [0132]
  • The human embryonic kidney cell line, BOSC 23, which does not express hSlit, and which can be transfected at a very high efficiency, was used for expression of hSlit. BOSC 23 cells were transiently transfected with vector, or control plasmid, pcDNA3.1/Myc-His/lacZ, or pcDNA3.1/Myc-His/hSlit DNA using the standard calcium phosphate precipitation method. Forty-eight hours post-transfection, total cell lysates were prepared from the transfected and untransfected control cells and analyzed by Western blotting. [0133]
  • C. Western Analysis [0134]
  • Protein content of the cell lysates was estimated using the BCA reagent (Pierce, Rockford, Ill.). Approximately 100 ìg of protein from various samples was electrophoresed on a 7.5% SDS-PAGE gel, and electrophoretically transferred to Immobilon PVDF membrane (Milipore, Bedford, Mass.). The protein blot was probed with anti-myc antibodies using ECL detection reagents and protocol (Amersham, UK). [0135]
  • Such procedures are standard in the art. Briefly, the blot was incubated with 5% milk to block non-specific binding sites, followed by incubation with a 1:5000 dilution of anti-myc mouse monoclonal antibodies, and finally incubation with a 1:3000 dilution of anti-mouse Ig linked to horse radish peroxidase. The antibody binding was detected using ECL detection reagents and exposure to X-ray film. [0136]
  • D. Western Analysis Results [0137]
  • The results are shown in the FIG. 4. In FIG. 4, [0138] Lane 1 shows the results for untransfected BOSC cells; Lanes 2, 3 and 4, respectively, show (2) BOSC cells transfected with pcDNA3.1/Myc-His/A vector, (3) pcDNA3.1/Myc-His/lacZ and (4) pcDNA3.1/Myc-His/hSlit cDNA.
  • Numerous modifications and variations of the present invention are possible in light of the above teachings and, therefore, within the scope of the appended claims. Further, the invention may be readily adapted and practiced otherwise than as particularly described. [0139]
  • 1 5 1 5176 DNA Artificial Sequence Description of Artificial SequencecDNA coding for the human slit polypeptide of SEQ ID NO2 1 gccggccccg ccgatggagc tgctgttgct gccgccgccg cctcccggag cgccccgctc 60 cgcccgcgcc ccgtgcgcct gagcaccgag ctcgccctcc tccgccgcta actccgccgc 120 ccgctcccca ggccgcccgc gctccccgcg cgcctcctcg ggctccacgc gtcttgcccc 180 gcagaggcag cctcctccag gagcggggcc ctgcacacca tggcccccgg gtgggcaggg 240 gtcggcgccg ccgtgcgcgc ccgcctggcg ctggccttgg cgctggcgag cgtcctgagt 300 gggcctccag ccgtcgcctg ccccaccaag tgtacctgct ccgctgccag cgtggactgc 360 cacgggctgg gcctccgcgc ggttcctcgg ggcatccccc gcaacgctga gcgccttgac 420 ctggacagaa ataatatcac caggatcacc aagatggact tcgctgggct caagaacctc 480 cgagtcttgc atctggaaga caaccaggtc agcgtcatcg agagaggcgc cttccaggac 540 ctgaagcagc tagagcgact gcgcctgaac aagaataagc tgcaagtcct tccagaattg 600 cttttccaga gcacgccgaa gctcaccaga ctagatttga gtgaaaacca gatccagggg 660 atcccgagga aggcgttccg cggcatcacc gatgtgaaga acctgcaact ggacaacaac 720 cacatcagct gcattgaaga tggagccttc cgagcgctgc gcgatttgga gatccttacc 780 ctcaacaaca acaacatcag tcgcatcctg gtcaccagct tcaaccacat gccgaagatc 840 cgaactctgc gcctccactc caaccacctg tactgcgact gccacctggc ctggctctcg 900 gattggctgc gacagcgacg gacagttggc cagttcacac tctgcatggc tcctgtgcat 960 ttgaggggct tcaacgtggc ggatgtgcag aagaaggagt acgtgtgccc agccccccac 1020 tcggagcccc catcctgcaa tgccaactcc atctcctgcc cttcgccctg cacgtgcagc 1080 aataacatcg tggactgtcg aggaaagggc ttgatggaga ttcctgccaa cttgccggag 1140 ggcatcgtcg aaatacgcct agaacagaac tccatcaaag ccatccctgc aggagccttc 1200 acccagtaca agaaactgaa gcgaatagac atcagcaaga atcagatatc ggatattgct 1260 ccagatgcct tccagggcct gaaatcactc acatcgctgg tcctgtatgg gaacaagatc 1320 accgagattg ccaagggact gtttgatggg ctggtgtccc tacagctgct cctcctcaat 1380 gccaacaaga tcaactgcct gcgggtgaac acgtttcagg acctgcagaa cctcaacttg 1440 ctctccctgt atgacaacaa gctgcagacc atcagcaagg ggctcttcgc ccctctgcag 1500 tccatccaga cactccactt agcccaaaac ccatttgtgt gcgactgcca cttgaagtgg 1560 ctggccgact acctccagga caaccccatc gagacaagcg gggcccgctg cagcagcccg 1620 cgccgactcg ccaacaagcg catcagccag atcaagagca agaagttccg ctgctcaggc 1680 tccgaggatt accgcagcag gttcagcagc gagtgcttca tggacctcgt gtgccccgag 1740 aagtgtcgct gtgagggcac gattgtggac tgctccaacc agaagctggt ccgcatccca 1800 agccacctcc ctgaatatgt caccgacctg cgactgaatg acaatgaggt atctgttctg 1860 gaggccactg gcatcttcaa gaagttgccc aacctgcgga aaataaatct gagtaacaat 1920 aagatcaagg aggtgcgaga gggagctttc gatggagcag ccagcgtgca ggagctgatg 1980 ctgacaggga accagctgga gaccgtgcac gggcgcgtgt tccgtggcct cagtggcctc 2040 aaaaccttga tgctgaggag taacttgatc agctgtgtga gtaatgacac ctttgccggc 2100 ctgagttcgg tgagactgct gtccctctat gacaatcgga tcaccaccat cacccctggg 2160 gccttcacca cgcttgtctc ccctgtccac cataaacctc ctgtccaacc ccttcaactg 2220 caactgccac tggcctggct cggcaagtgg ttgaggaaga ggcggatcgt cagtgggaac 2280 cctaggtgcc agaagccatt tttcctcaag gagatcccca tccaggatgt ggccatccag 2340 gacttcacct gtgatggcaa cgaggagagt agttgccagc tgagcccgcg ctgcccggag 2400 cagtgcacct gtatggagac agtggtgcga tgcagcaaca aggggctccg cgccctcccc 2460 agaggcatgc ccaaggatgt gaccgagctg tacctggaag gaaaccacct aacagccgtg 2520 cccagagagc tgtccgccct ccgacacctg acgcttattg acctgagcaa caacagcatc 2580 agcatgctga ccaattacac cttcagtaac atgtctcacc tctccactct gatcctgagc 2640 tacaaccggc tgaggtgcat ccccgtccac gccttcaacg ggctgcggtc cctgcgagtg 2700 ctaaccctcc atggcaatga catttccagc gttcctgaag gctccttcaa cgacctcaca 2760 tctctttccc atctggcgct gggaaccaac ccactccact gtgactgcag tcttcggtgg 2820 ctgtcggagt gggtgaaggc ggggtacaag gagcctggca tcgcccgctg cagtagccct 2880 gagcccatgg ctgacaggct cctgctcacc accccaaccc accgcttcca gtgcaaaggg 2940 ccagtggaca tcaacattgt ggccaaatgc aatgcctgcc tctccagccc gtgcaagaat 3000 aacgggacat gcacccagga ccctgtggag ctgtaccgct gtgcctgccc ctacagctac 3060 aagggcaagg actgcactgt gcccatcaac acctgcatcc agaacccctg tcagcatgga 3120 ggcacctgcc acctgagtga cagccacaag gatgggttca gctgctcctg ccctctgggc 3180 tttgaggggc agcggtgtga gatcaaccca gatgactgtg aggacaacga ctgcgaaaac 3240 aatgccacct gcgtggacgg gatcaacaac tacgtgtgta tctgtccgcc taactacaca 3300 ggtgagctat gcgacgaggt gattgaccac tgtgtgcctg agctgaacct ctgtcagcat 3360 gaggccaagt gcatccccct ggacaaagga ttcagctgcg agtgtgtccc tggctacagc 3420 gggaagctct gtgagacaga caatgatgac tgtgtggccc acaagtgccg ccacggggcc 3480 cagtgcgtgg acacaatcaa tggctacaca tgcacctgcc cccagggctt cagtggaccc 3540 ttctgtgaac accccccacc catggtccta ctgcagacca gcccatgcga ccagtacgag 3600 tgccagaacg gggcccagtg catcgtggtg cagcaggagc ccacctgccg ctgcccacca 3660 ggcttcgccg gccccagatg cgagaagctc atcactgtca acttcgtggg caaagactcc 3720 tacgtggaac tggcctccgc caaggtccga ccccaggcca acatctccct gcaggtggcc 3780 actgacaagg acaacggcat ccttctctac aaaggagaca atgaccccct ggcactggag 3840 ctgtaccagg gccacgtgcg gctggtctat gacagcgtga gttcccctcc aaccacagtg 3900 tacagtgtgg agacagtgaa tgatgggcag tttcacagtg tggaggtggt gacgctaaac 3960 cagaccctga acttagtagt ggacaaagga actccaaaga gcttggggaa gttccagaag 4020 cagccagcag tgggcatcaa cagccccctc taccttggag gcatccccac ctccaccggc 4080 ctctccgcct tgcgccaggg cacggaccgg cctctaggcg gcttccacgg atgcatccat 4140 gaggtgcgca tcaacaacga gctgcaggac ttcaaggccc tcccaccaca gtccctgggg 4200 gtgtcaccag gctgcaagtc ctgcaccgtg tgcaagcacg gcctgtgccg ctccgtggag 4260 aaggacagcg tggtgtgcga gtgccgccca ggctggaccg gcccactctg cgatcaggag 4320 gcccgggacc cctgcctcgg ccacagatgc caccatggaa aatgtgtggc aactgggacc 4380 tcatacatgt gcaagtgtgc cgagggctat ggaggggact tgtgtgacaa caagaatgac 4440 tctgccaatg cctgctcagc cttcaagtgt caccatgggc agtgccacat ctcagaccaa 4500 ggggagccct actgcctgtg ccagcccggc tttagcggcg agcactgcca acaagagaat 4560 ccgtgcctgg gaccagtagt ccgagaggtg atccgccgcc agaaaggtta tgcatcatgt 4620 gccacagcct ccaaggtgcc catcatggaa tgtcgtgggg gctgtgggcc ccagtgctgc 4680 cagcccaccc gcagcaagcg gcggaaatac gtcttccagt gcacggacgg ctcctcgttt 4740 gtagaagagg tggagagaca gttagagtgc ggctgcctcg cgtgttccta agcccctgcc 4800 cgcctgcctg ccacctctcg gactccagct tgatggagtt gggacagcca tgtgggaccc 4860 ccgggtgatt cagcatgaag gaaatgaagc tggagaggaa ggtaaagaag aagagaatat 4920 taagtatatt gtaaaataaa caaaaaatag aacttatttt tattatggaa agtgactatt 4980 ttcatctttt attatataaa tatattacac catctgcgta tatgtaccat atagtgagtt 5040 atttttacca agttttgtgt tgtgtatttg ttgtgttttt aaaaatagct gtttaaaaat 5100 ttaagaaaaa aatagactaa taaaaatgct ttaaaacaaa aggataagaa taaagaatga 5160 tagcctgtct gaggaa 5176 2 1523 PRT Homo sapiens 2 Met Ala Pro Gly Trp Ala Gly Val Gly Ala Ala Val Arg Ala Arg Leu 1 5 10 15 Ala Leu Ala Leu Ala Leu Ala Ser Val Leu Ser Gly Pro Pro Ala Val 20 25 30 Ala Cys Pro Thr Lys Cys Thr Cys Ser Ala Ala Ser Val Asp Cys His 35 40 45 Gly Leu Gly Leu Arg Ala Val Pro Arg Gly Ile Pro Arg Asn Ala Glu 50 55 60 Arg Leu Asp Leu Asp Arg Asn Asn Ile Thr Arg Ile Thr Lys Met Asp 65 70 75 80 Phe Ala Gly Leu Lys Asn Leu Arg Val Leu His Leu Glu Asp Asn Gln 85 90 95 Val Ser Val Ile Glu Arg Gly Ala Phe Gln Asp Leu Lys Gln Leu Glu 100 105 110 Arg Leu Arg Leu Asn Lys Asn Lys Leu Gln Val Leu Pro Glu Leu Leu 115 120 125 Phe Gln Ser Thr Pro Lys Leu Thr Arg Leu Asp Leu Ser Glu Asn Gln 130 135 140 Ile Gln Gly Ile Pro Arg Lys Ala Phe Arg Gly Ile Thr Asp Val Lys 145 150 155 160 Asn Leu Gln Leu Asp Asn Asn His Ile Ser Cys Ile Glu Asp Gly Ala 165 170 175 Phe Arg Ala Leu Arg Asp Leu Glu Ile Leu Thr Leu Asn Asn Asn Asn 180 185 190 Ile Ser Arg Ile Leu Val Thr Ser Phe Asn His Met Pro Lys Ile Arg 195 200 205 Thr Leu Arg Leu His Ser Asn His Leu Tyr Cys Asp Cys His Leu Ala 210 215 220 Trp Leu Ser Asp Trp Leu Arg Gln Arg Arg Thr Val Gly Gln Phe Thr 225 230 235 240 Leu Cys Met Ala Pro Val His Leu Arg Gly Phe Asn Val Ala Asp Val 245 250 255 Gln Lys Lys Glu Tyr Val Cys Pro Ala Pro His Ser Glu Pro Pro Ser 260 265 270 Cys Asn Ala Asn Ser Ile Ser Cys Pro Ser Pro Cys Thr Cys Ser Asn 275 280 285 Asn Ile Val Asp Cys Arg Gly Lys Gly Leu Met Glu Ile Pro Ala Asn 290 295 300 Leu Pro Glu Gly Ile Val Glu Ile Arg Leu Glu Gln Asn Ser Ile Lys 305 310 315 320 Ala Ile Pro Ala Gly Ala Phe Thr Gln Tyr Lys Lys Leu Lys Arg Ile 325 330 335 Asp Ile Ser Lys Asn Gln Ile Ser Asp Ile Ala Pro Asp Ala Phe Gln 340 345 350 Gly Leu Lys Ser Leu Thr Ser Leu Val Leu Tyr Gly Asn Lys Ile Thr 355 360 365 Glu Ile Ala Lys Gly Leu Phe Asp Gly Leu Val Ser Leu Gln Leu Leu 370 375 380 Leu Leu Asn Ala Asn Lys Ile Asn Cys Leu Arg Val Asn Thr Phe Gln 385 390 395 400 Asp Leu Gln Asn Leu Asn Leu Leu Ser Leu Tyr Asp Asn Lys Leu Gln 405 410 415 Thr Ile Ser Lys Gly Leu Phe Ala Pro Leu Gln Ser Ile Gln Thr Leu 420 425 430 His Leu Ala Gln Asn Pro Phe Val Cys Asp Cys His Leu Lys Trp Leu 435 440 445 Ala Asp Tyr Leu Gln Asp Asn Pro Ile Glu Thr Ser Gly Ala Arg Cys 450 455 460 Ser Ser Pro Arg Arg Leu Ala Asn Lys Arg Ile Ser Gln Ile Lys Ser 465 470 475 480 Lys Lys Phe Arg Cys Ser Gly Ser Glu Asp Tyr Arg Ser Arg Phe Ser 485 490 495 Ser Glu Cys Phe Met Asp Leu Val Cys Pro Glu Lys Cys Arg Cys Glu 500 505 510 Gly Thr Ile Val Asp Cys Ser Asn Gln Lys Leu Val Arg Ile Pro Ser 515 520 525 His Leu Pro Glu Tyr Val Thr Asp Leu Arg Leu Asn Asp Asn Glu Val 530 535 540 Ser Val Leu Glu Ala Thr Gly Ile Phe Lys Lys Leu Pro Asn Leu Arg 545 550 555 560 Lys Ile Asn Leu Ser Asn Asn Lys Ile Lys Glu Val Arg Glu Gly Ala 565 570 575 Phe Asp Gly Ala Ala Ser Val Gln Glu Leu Met Leu Thr Gly Asn Gln 580 585 590 Leu Glu Thr Val His Gly Arg Val Phe Arg Gly Leu Ser Gly Leu Lys 595 600 605 Thr Leu Met Leu Arg Ser Asn Leu Ile Ser Cys Val Ser Asn Asp Thr 610 615 620 Phe Ala Gly Leu Ser Ser Val Arg Leu Leu Ser Leu Tyr Asp Asn Arg 625 630 635 640 Ile Thr Thr Ile Thr Pro Gly Ala Phe Thr Thr Leu Val Ser Pro Val 645 650 655 His His Lys Pro Pro Val Gln Pro Leu Gln Leu Gln Leu Pro Leu Ala 660 665 670 Trp Leu Gly Lys Trp Leu Arg Lys Arg Arg Ile Val Ser Gly Asn Pro 675 680 685 Arg Cys Gln Lys Pro Phe Phe Leu Lys Glu Ile Pro Ile Gln Asp Val 690 695 700 Ala Ile Gln Asp Phe Thr Cys Asp Gly Asn Glu Glu Ser Ser Cys Gln 705 710 715 720 Leu Ser Pro Arg Cys Pro Glu Gln Cys Thr Cys Met Glu Thr Val Val 725 730 735 Arg Cys Ser Asn Lys Gly Leu Arg Ala Leu Pro Arg Gly Met Pro Lys 740 745 750 Asp Val Thr Glu Leu Tyr Leu Glu Gly Asn His Leu Thr Ala Val Pro 755 760 765 Arg Glu Leu Ser Ala Leu Arg His Leu Thr Leu Ile Asp Leu Ser Asn 770 775 780 Asn Ser Ile Ser Met Leu Thr Asn Tyr Thr Phe Ser Asn Met Ser His 785 790 795 800 Leu Ser Thr Leu Ile Leu Ser Tyr Asn Arg Leu Arg Cys Ile Pro Val 805 810 815 His Ala Phe Asn Gly Leu Arg Ser Leu Arg Val Leu Thr Leu His Gly 820 825 830 Asn Asp Ile Ser Ser Val Pro Glu Gly Ser Phe Asn Asp Leu Thr Ser 835 840 845 Leu Ser His Leu Ala Leu Gly Thr Asn Pro Leu His Cys Asp Cys Ser 850 855 860 Leu Arg Trp Leu Ser Glu Trp Val Lys Ala Gly Tyr Lys Glu Pro Gly 865 870 875 880 Ile Ala Arg Cys Ser Ser Pro Glu Pro Met Ala Asp Arg Leu Leu Leu 885 890 895 Thr Thr Pro Thr His Arg Phe Gln Cys Lys Gly Pro Val Asp Ile Asn 900 905 910 Ile Val Ala Lys Cys Asn Ala Cys Leu Ser Ser Pro Cys Lys Asn Asn 915 920 925 Gly Thr Cys Thr Gln Asp Pro Val Glu Leu Tyr Arg Cys Ala Cys Pro 930 935 940 Tyr Ser Tyr Lys Gly Lys Asp Cys Thr Val Pro Ile Asn Thr Cys Ile 945 950 955 960 Gln Asn Pro Cys Gln His Gly Gly Thr Cys His Leu Ser Asp Ser His 965 970 975 Lys Asp Gly Phe Ser Cys Ser Cys Pro Leu Gly Phe Glu Gly Gln Arg 980 985 990 Cys Glu Ile Asn Pro Asp Asp Cys Glu Asp Asn Asp Cys Glu Asn Asn 995 1000 1005 Ala Thr Cys Val Asp Gly Ile Asn Asn Tyr Val Cys Ile Cys Pro Pro 1010 1015 1020 Asn Tyr Thr Gly Glu Leu Cys Asp Glu Val Ile Asp His Cys Val Pro 1025 1030 1035 1040 Glu Leu Asn Leu Cys Gln His Glu Ala Lys Cys Ile Pro Leu Asp Lys 1045 1050 1055 Gly Phe Ser Cys Glu Cys Val Pro Gly Tyr Ser Gly Lys Leu Cys Glu 1060 1065 1070 Thr Asp Asn Asp Asp Cys Val Ala His Lys Cys Arg His Gly Ala Gln 1075 1080 1085 Cys Val Asp Thr Ile Asn Gly Tyr Thr Cys Thr Cys Pro Gln Gly Phe 1090 1095 1100 Ser Gly Pro Phe Cys Glu His Pro Pro Pro Met Val Leu Leu Gln Thr 1105 1110 1115 1120 Ser Pro Cys Asp Gln Tyr Glu Cys Gln Asn Gly Ala Gln Cys Ile Val 1125 1130 1135 Val Gln Gln Glu Pro Thr Cys Arg Cys Pro Pro Gly Phe Ala Gly Pro 1140 1145 1150 Arg Cys Glu Lys Leu Ile Thr Val Asn Phe Val Gly Lys Asp Ser Tyr 1155 1160 1165 Val Glu Leu Ala Ser Ala Lys Val Arg Pro Gln Ala Asn Ile Ser Leu 1170 1175 1180 Gln Val Ala Thr Asp Lys Asp Asn Gly Ile Leu Leu Tyr Lys Gly Asp 1185 1190 1195 1200 Asn Asp Pro Leu Ala Leu Glu Leu Tyr Gln Gly His Val Arg Leu Val 1205 1210 1215 Tyr Asp Ser Val Ser Ser Pro Pro Thr Thr Val Tyr Ser Val Glu Thr 1220 1225 1230 Val Asn Asp Gly Gln Phe His Ser Val Glu Val Val Thr Leu Asn Gln 1235 1240 1245 Thr Leu Asn Leu Val Val Asp Lys Gly Thr Pro Lys Ser Leu Gly Lys 1250 1255 1260 Phe Gln Lys Gln Pro Ala Val Gly Ile Asn Ser Pro Leu Tyr Leu Gly 1265 1270 1275 1280 Gly Ile Pro Thr Ser Thr Gly Leu Ser Ala Leu Arg Gln Gly Thr Asp 1285 1290 1295 Arg Pro Leu Gly Gly Phe His Gly Cys Ile His Glu Val Arg Ile Asn 1300 1305 1310 Asn Glu Leu Gln Asp Phe Lys Ala Leu Pro Pro Gln Ser Leu Gly Val 1315 1320 1325 Ser Pro Gly Cys Lys Ser Cys Thr Val Cys Lys His Gly Leu Cys Arg 1330 1335 1340 Ser Val Glu Lys Asp Ser Val Val Cys Glu Cys Arg Pro Gly Trp Thr 1345 1350 1355 1360 Gly Pro Leu Cys Asp Gln Glu Ala Arg Asp Pro Cys Leu Gly His Arg 1365 1370 1375 Cys His His Gly Lys Cys Val Ala Thr Gly Thr Ser Tyr Met Cys Lys 1380 1385 1390 Cys Ala Glu Gly Tyr Gly Gly Asp Leu Cys Asp Asn Lys Asn Asp Ser 1395 1400 1405 Ala Asn Ala Cys Ser Ala Phe Lys Cys His His Gly Gln Cys His Ile 1410 1415 1420 Ser Asp Gln Gly Glu Pro Tyr Cys Leu Cys Gln Pro Gly Phe Ser Gly 1425 1430 1435 1440 Glu His Cys Gln Gln Glu Asn Pro Cys Leu Gly Gln Val Val Arg Glu 1445 1450 1455 Val Ile Arg Arg Gln Lys Gly Tyr Ala Ser Cys Ala Thr Ala Ser Lys 1460 1465 1470 Val Pro Ile Met Glu Cys Arg Gly Gly Cys Gly Pro Gln Cys Cys Gln 1475 1480 1485 Pro Thr Arg Ser Lys Arg Arg Lys Tyr Val Phe Gln Cys Thr Asp Gly 1490 1495 1500 Ser Ser Phe Val Glu Glu Val Glu Arg His Leu Glu Cys Gly Cys Leu 1505 1510 1515 1520 Ala Cys Ser 3 21 DNA Artificial Sequence Description of Artificial SequencePCR primer for amplification of cDNA of SEQ ID NO1 3 tcctcgggct ccacgcgtct t 21 4 21 DNA Artificial Sequence Description of Artificial SequencePCR primer for amplification of cDNA of SEQ ID NO1 4 ggtacatata cgcagatggt g 21 5 1480 PRT Drosophila melanogaster 5 Met Ala Ala Pro Ser Arg Thr Thr Leu Met Pro Pro Pro Phe Arg Leu 1 5 10 15 Gln Leu Arg Leu Leu Ile Leu Pro Ile Leu Leu Leu Leu Arg His Asp 20 25 30 Ala Val His Ala Glu Pro Tyr Ser Gly Gly Phe Gly Ser Ser Ala Val 35 40 45 Ser Ser Gly Gly Leu Gly Ser Val Gly Ile His Ile Pro Gly Gly Gly 50 55 60 Val Gly Val Ile Thr Glu Ala Arg Cys Pro Arg Val Cys Ser Cys Thr 65 70 75 80 Gly Leu Asn Val Asp Cys Ser His Arg Gly Leu Thr Ser Val Pro Arg 85 90 95 Lys Ile Ser Ala Asp Val Glu Arg Leu Glu Leu Gln Gly Asn Asn Leu 100 105 110 Thr Val Ile Tyr Glu Thr Asp Phe Gln Arg Leu Thr Lys Leu Arg Met 115 120 125 Leu Gln Leu Thr Asp Asn Gln Ile His Thr Ile Glu Arg Asn Ser Phe 130 135 140 Gln Asp Leu Val Ser Leu Glu Arg Leu Asp Ile Ser Asn Asn Val Ile 145 150 155 160 Thr Thr Val Gly Arg Arg Val Phe Lys Gly Ala Gln Ser Leu Arg Ser 165 170 175 Leu Gln Leu Asp Asn Asn Gln Ile Thr Cys Leu Asp Glu His Ala Phe 180 185 190 Lys Gly Leu Val Glu Leu Glu Ile Leu Thr Leu Asn Asn Asn Asn Leu 195 200 205 Thr Ser Leu Pro His Asn Ile Phe Gly Gly Leu Gly Arg Leu Arg Ala 210 215 220 Leu Arg Leu Ser Asp Asn Pro Phe Ala Cys Asp Cys His Leu Ser Trp 225 230 235 240 Leu Ser Arg Phe Leu Arg Ser Ala Thr Arg Leu Ala Pro Tyr Thr Arg 245 250 255 Cys Gln Ser Pro Ser Gln Leu Lys Gly Gln Asn Val Ala Asp Leu His 260 265 270 Asp Gln Glu Phe Lys Cys Ser Gly Leu Thr Glu His Ala Pro Met Glu 275 280 285 Cys Gly Ala Glu Asn Ser Cys Pro His Pro Cys Arg Cys Ala Asp Gly 290 295 300 Ile Val Asp Cys Arg Glu Lys Ser Leu Thr Ser Val Pro Val Thr Leu 305 310 315 320 Pro Asp Asp Thr Thr Asp Val Leu Leu Glu Gln Asn Phe Ile Thr Glu 325 330 335 Leu Pro Pro Lys Ser Phe Ser Ser Phe Arg Arg Leu Arg Arg Ile Asp 340 345 350 Leu Ser Asn Asn Asn Ile Ser Arg Ile Ala His Asp Ala Leu Ser Gly 355 360 365 Leu Lys Gln Leu Thr Thr Leu Val Leu Tyr Gly Asn Lys Ile Lys Asp 370 375 380 Leu Pro Ser Gly Val Phe Lys Gly Leu Gly Ser Leu Arg Leu Leu Leu 385 390 395 400 Leu Asn Ala Asn Glu Ile Ser Cys Ile Arg Lys Asp Ala Phe Arg Asp 405 410 415 Leu His Ser Leu Ser Leu Leu Ser Leu Tyr Asp Asn Asn Ile Gln Ser 420 425 430 Leu Ala Asn Gly Thr Phe Asp Ala Met Lys Ser Met Lys Thr Val His 435 440 445 Leu Ala Lys Asn Pro Phe Ile Cys Asp Cys Asn Leu Arg Trp Leu Ala 450 455 460 Asp Tyr Leu His Lys Asn Pro Ile Glu Thr Ser Gly Ala Arg Cys Glu 465 470 475 480 Ser Pro Lys Arg Met His Arg Arg Arg Ile Glu Ser Leu Arg Glu Glu 485 490 495 Lys Phe Lys Cys Ser Trp Gly Glu Leu Arg Met Lys Leu Ser Gly Glu 500 505 510 Cys Arg Met Asp Ser Asp Cys Pro Ala Met Cys His Cys Glu Gly Thr 515 520 525 Thr Val Asp Cys Thr Gly Arg Arg Leu Lys Glu Ile Pro Arg Asp Ile 530 535 540 Pro Leu His Thr Thr Glu Leu Leu Leu Asn Asp Asn Glu Leu Gly Arg 545 550 555 560 Ile Ser Ser Asp Gly Leu Phe Gly Arg Leu Pro His Leu Val Lys Leu 565 570 575 Glu Leu Lys Arg Asn Gln Leu Thr Gly Ile Glu Pro Asn Ala Phe Glu 580 585 590 Gly Ala Ser His Ile Gln Glu Leu Gln Leu Gly Glu Asn Lys Ile Lys 595 600 605 Glu Ile Ser Asn Lys Met Phe Leu Gly Leu His Gln Leu Lys Thr Leu 610 615 620 Asn Leu Tyr Asp Asn Gln Ile Ser Cys Val Met Pro Gly Ser Phe Glu 625 630 635 640 His Leu Asn Ser Leu Thr Ser Leu Asn Leu Ala Ser Asn Pro Phe Asn 645 650 655 Cys Asn Cys His Leu Ala Trp Phe Ala Glu Cys Val Arg Lys Lys Ser 660 665 670 Leu Asn Gly Gly Ala Ala Arg Cys Gly Ala Pro Ser Lys Val Arg Asp 675 680 685 Val Gln Ile Lys Asp Leu Pro His Ser Glu Phe Lys Cys Ser Ser Glu 690 695 700 Asn Ser Glu Gly Cys Leu Gly Asp Gly Tyr Cys Pro Pro Ser Cys Thr 705 710 715 720 Cys Thr Gly Thr Val Val Ala Cys Ser Arg Asn Gln Leu Lys Glu Ile 725 730 735 Pro Arg Gly Ile Pro Ala Glu Thr Ser Glu Leu Tyr Leu Glu Ser Asn 740 745 750 Glu Ile Glu Gln Ile His Tyr Glu Arg Ile Arg His Leu Arg Ser Leu 755 760 765 Thr Arg Leu Asp Leu Ser Asn Asn Gln Ile Thr Ile Leu Ser Asn Tyr 770 775 780 Thr Phe Ala Asn Leu Thr Lys Leu Ser Thr Leu Ile Ile Ser Tyr Asn 785 790 795 800 Lys Leu Gln Cys Leu Gln Arg His Ala Leu Ser Gly Leu Asn Asn Leu 805 810 815 Arg Val Val Ser Leu His Gly Asn Arg Ile Ser Met Leu Pro Glu Gly 820 825 830 Ser Phe Glu Asp Leu Lys Ser Leu Thr His Ile Ala Leu Gly Ser Asn 835 840 845 Pro Leu Tyr Cys Asp Cys Gly Leu Lys Trp Phe Ser Asp Trp Ile Lys 850 855 860 Leu Asp Tyr Val Glu Pro Gly Ile Ala Arg Cys Ala Glu Pro Glu Gln 865 870 875 880 Met Lys Asp Lys Leu Ile Leu Ser Thr Pro Ser Ser Ser Phe Val Cys 885 890 895 Arg Gly Arg Val Arg Asn Asp Ile Leu Ala Lys Cys Asn Ala Cys Phe 900 905 910 Glu Gln Pro Cys Gln Asn Gln Ala Gln Cys Val Ala Leu Pro Gln Arg 915 920 925 Glu Tyr Gln Cys Leu Cys Gln Pro Gly Tyr His Gly Lys His Cys Glu 930 935 940 Phe Met Ile Asp Ala Cys Tyr Gly Asn Pro Cys Arg Asn Asn Ala Thr 945 950 955 960 Cys Thr Val Leu Glu Glu Gly Arg Phe Ser Cys Gln Cys Ala Pro Gly 965 970 975 Tyr Thr Gly Ala Arg Cys Glu Thr Asn Ile Asp Asp Cys Leu Gly Glu 980 985 990 Ile Lys Cys Gln Asn Asn Ala Thr Cys Ile Asp Gly Val Glu Ser Tyr 995 1000 1005 Lys Cys Glu Cys Gln Pro Gly Phe Ser Gly Glu Phe Cys Asp Thr Lys 1010 1015 1020 Ile Gln Phe Cys Ser Pro Glu Phe Asn Pro Cys Ala Asn Cys Ala Lys 1025 1030 1035 1040 Cys Met Asp His Phe Thr His Tyr Ser Cys Asp Cys Gln Ala Gly Phe 1045 1050 1055 His Gly Thr Asn Cys Thr Asp Asn Ile Asp Asp Cys Gln Asn His Met 1060 1065 1070 Cys Gln Asn Gly Gly Thr Cys Val Asp Gly Ile Asn Asp Tyr Gln Cys 1075 1080 1085 Arg Cys Pro Asp Asp Tyr Thr Gly Lys Tyr Cys Glu Gly His Asn Met 1090 1095 1100 Ile Ser Met Met Tyr Pro Gln Thr Ser Pro Cys Gln Asn His Glu Cys 1105 1110 1115 1120 Lys His Gly Val Cys Phe Gln Pro Asn Ala Gln Gly Ser Asp Tyr Leu 1125 1130 1135 Cys Arg Cys His Pro Gly Tyr Thr Gly Lys Trp Cys Glu Tyr Leu Thr 1140 1145 1150 Ser Ile Ser Phe Val His Asn Asn Ser Phe Val Glu Leu Glu Pro Leu 1155 1160 1165 Arg Thr Arg Pro Glu Ala Asn Val Thr Ile Val Phe Ser Ser Ala Glu 1170 1175 1180 Gln Asn Gly Ile Leu Met Tyr Asp Gly Gln Asp Ala His Leu Ala Val 1185 1190 1195 1200 Glu Leu Phe Asn Gly Arg Ile Arg Val Ser Tyr Asp Val Gly Asn His 1205 1210 1215 Pro Val Ser Thr Asn Tyr Ser Phe Glu Met Val Ala Asp Gly Lys Tyr 1220 1225 1230 His Ala Val Glu Leu Leu Ala Ile Lys Lys Asn Phe Thr Leu Arg Val 1235 1240 1245 Asp Arg Gly Leu Ala Arg Ser Ile Ile Asn Glu Gly Ser Asn Asp Tyr 1250 1255 1260 Leu Lys Leu Thr Thr Pro Met Phe Leu Gly Gly Leu Pro Val Asp Pro 1265 1270 1275 1280 Ala Gln Gln Ala Tyr Lys Asn Trp Gln Ile Arg Asn Leu Thr Ser Phe 1285 1290 1295 Lys Gly Cys Met Lys Glu Val Trp Ile Asn His Lys Leu Val Asp Phe 1300 1305 1310 Gly Asn Ala Gln Arg Gln Gln Lys Ile Thr Pro Gly Cys Ala Leu Leu 1315 1320 1325 Glu Gly Glu Gln Gln Glu Glu Glu Asp Asp Glu Gln Asp Phe Met Asp 1330 1335 1340 Glu Thr Pro His Ile Lys Glu Glu Pro Val Asp Pro Cys Leu Glu Asn 1345 1350 1355 1360 Lys Cys Arg Arg Gly Ser Arg Cys Val Pro Asn Ser Asn Ala Arg Asp 1365 1370 1375 Gly Tyr Gln Cys Lys Cys Lys His Gly Gln Arg Gly Arg Tyr Cys Asp 1380 1385 1390 Gln Gly Glu Gly Ser Thr Glu Pro Pro Thr Val Thr Ala Ala Ser Thr 1395 1400 1405 Cys Arg Lys Glu Gln Val Arg Glu Tyr Tyr Thr Glu Asn Asp Cys Arg 1410 1415 1420 Ser Arg Gln Pro Leu Lys Tyr Ala Lys Cys Val Gly Gly Cys Gly Asn 1425 1430 1435 1440 Gln Cys Cys Ala Ala Lys Ile Val Arg Arg Arg Lys Val Arg Met Val 1445 1450 1455 Cys Ser Asn Asn Arg Lys Tyr Ile Lys Asn Leu Asp Ile Val Arg Lys 1460 1465 1470 Cys Gly Cys Thr Lys Lys Cys Tyr 1475 1480

Claims (20)

What is claimed is:
1. An isolated polynucleotide comprising a polynucleotide sequence which is a member selected from the group consisting of:
(a) a polynucleotide encoding amino acid 2 to 1523 of SEQ ID NO:2;
(b) a variant polynucleotide sequence of (a), wherein said variant polynucleotide sequence varies from the polynucleotide sequence of (a) by a member selected from (i) nucleotide substitution, (ii) nucleotide deletion, (iii) nucleotide insertion, and (iv) a combination of (i), (ii) or (iii), and said variant polynucleotide will hybridize to the complement of a polynucleotide of (a),
(c) the full complement of (a); and
(d) the full complement of (b).
2. An isolated polynucleotide comprising a polynucleotide having at least 95% identity to a member selected from the group consisting of:
(a) a polynucleotide encoding amino acid 2 to 1523 of SEQ ID NO:2; and
(b) the full complement of (a).
3. The isolated polynucleotide of claim 1 wherein said member is (a) or (b).
4. The isolated polynucleotide of claim 1 comprising a polynucleotide encoding a polypeptide comprising amino acids 2 to 1523 of SEQ ID NO:2.
5. The isolated polynucleotide of claim 1, wherein said member is (a) or (b) and the polynucleotide is DNA.
6. A recombinant vector comprising the polynucleotide of claim 3, wherein said polynucleotide is DNA.
7. A recombinant host cell comprising the polynucleotide of claim 3, wherein said polynucleotide is DNA.
8. A method for producing a polypeptide comprising expressing from the recombinant cell of claim 7 the polypeptide encoded by said polynucleotide.
9. A process for producing a mature slit polypeptide comprising:
expressing from a recombinant cell containing the polynucleotide of claim 4 the polypeptide encoded by said polynucleotide.
10. The isolated polynucleotide of claim 1 comprising the nucleotides of the sequence of SEQ ID NO:1.
11. An isolated polypeptide comprising:
a mature polypeptide having an amino acid sequence encoded by a polynucleotide which is at least 95% identical to the polynucleotide of claim 4.
12. The isolated polypeptide of claim 11, comprising amino acids 1 to 1523 of sequence of SEQ ID NO:2.
13. An antibody against the polypeptide of claim 11.
14. An antagonist against the polypeptide of claim 11.
15. A method for the treatment of a patient having need of a human slit polypeptide comprising: administering to the patient a therapeutically effective amount of the polypeptide of claim 11.
16. The method of claim 15 wherein said therapeutically effective amount of the polypeptide is administered by providing to the patient DNA encoding said polypeptide and expressing said polypeptide in vivo.
17. A method for the treatment of a patient having need to inhibit the activity of a human slit polypeptide comprising: administering to the patient a therapeutically effective amount of the antagonist of claim 14.
18. A method for the treatment of a patient having need of a human slit polypeptide comprising: administering to the patient a therapeutically effective amount of the agonist of claim 15.
19. A process for diagnosing a disease or a susceptibility to a disease related to expression of the polypeptide of claim 11 comprising:
determining a mutation in the nucleic acid sequence encoding said polypeptide.
20. A diagnostic process comprising:
analyzing for the presence of the polypeptide of claim 11 in a sample derived from a host.
US10/011,064 1997-10-31 2001-12-03 Human slit polypeptide and polynucleotides encoding same Abandoned US20020123104A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/011,064 US20020123104A1 (en) 1997-10-31 2001-12-03 Human slit polypeptide and polynucleotides encoding same

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US6394697P 1997-10-31 1997-10-31
US9642098P 1998-08-13 1998-08-13
US09/182,024 US6342370B1 (en) 1997-10-31 1998-10-29 Human slit polypeptide and polynucleotides encoding same
US10/011,064 US20020123104A1 (en) 1997-10-31 2001-12-03 Human slit polypeptide and polynucleotides encoding same

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/182,024 Division US6342370B1 (en) 1997-10-31 1998-10-29 Human slit polypeptide and polynucleotides encoding same

Publications (1)

Publication Number Publication Date
US20020123104A1 true US20020123104A1 (en) 2002-09-05

Family

ID=26743971

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/182,024 Expired - Lifetime US6342370B1 (en) 1997-10-31 1998-10-29 Human slit polypeptide and polynucleotides encoding same
US10/011,064 Abandoned US20020123104A1 (en) 1997-10-31 2001-12-03 Human slit polypeptide and polynucleotides encoding same

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/182,024 Expired - Lifetime US6342370B1 (en) 1997-10-31 1998-10-29 Human slit polypeptide and polynucleotides encoding same

Country Status (3)

Country Link
US (2) US6342370B1 (en)
AU (1) AU1203999A (en)
WO (1) WO1999023219A1 (en)

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1030918A2 (en) * 1997-11-14 2000-08-30 The Regents of the University of California Slit polypeptide and method of modulation of robo with ligands
JP2002539218A (en) * 1999-03-18 2002-11-19 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Composition for promoting nerve regeneration
WO2001046418A1 (en) * 1999-12-21 2001-06-28 Zymogenetics, Inc. Human slit polypeptide zslit3
ES2263651T3 (en) * 2000-08-30 2006-12-16 Avi Biopharma, Inc. METHOD FOR ANALIDIS OF OLIGONUCLEOTID ANALOGS.
WO2004090112A2 (en) * 2003-04-01 2004-10-21 United States Of America Department Of Veteran's Affairs Stem-cell, precursor cell, or target cell-based treatment of multi-organ failure and renal dysfunction
CA2512667A1 (en) * 2005-01-07 2006-07-07 Takahiro Ochiya Human hepatocyte-like cells and uses thereof
BRPI0520280A2 (en) * 2005-05-10 2009-04-28 Us Of America Dept Of Veteran therapy of renal dysfunction and multiple organ failure with mesenchymal stem cells and medium conditioned by exposure to mesenchymal stem cells
US20110002972A1 (en) * 2007-09-14 2011-01-06 Scil Technology Gmbh Neuroendocrine factors for treatment of degenerative diseases
US9301975B2 (en) 2009-05-01 2016-04-05 Biocardia, Inc. Method of preparing autologous cells and method of use for therapy
EP2625263B1 (en) 2010-10-08 2020-03-11 Terumo BCT, Inc. Configurable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
JP6633522B2 (en) 2013-11-16 2020-01-22 テルモ ビーシーティー、インコーポレーテッド Cell growth in bioreactors
EP3613841B1 (en) 2014-03-25 2022-04-20 Terumo BCT, Inc. Passive replacement of media
JP6830059B2 (en) 2014-09-26 2021-02-17 テルモ ビーシーティー、インコーポレーテッド Scheduled cell feeding
WO2017004592A1 (en) 2015-07-02 2017-01-05 Terumo Bct, Inc. Cell growth with mechanical stimuli
US11104874B2 (en) 2016-06-07 2021-08-31 Terumo Bct, Inc. Coating a bioreactor
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
EP3601521A2 (en) 2017-03-31 2020-02-05 Terumo BCT, Inc. Cell expansion
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992010518A1 (en) 1990-12-07 1992-06-25 Yale University Purified slit protein and sequence elements thereof
JPH1087699A (en) 1996-07-16 1998-04-07 Asahi Chem Ind Co Ltd Slit-like polypeptide
JPH1118777A (en) * 1997-07-09 1999-01-26 Asahi Chem Ind Co Ltd New slit-like polypeptide
JPH1175846A (en) * 1997-09-02 1999-03-23 Asahi Chem Ind Co Ltd Slit-formed polypeptide
EP1030918A2 (en) * 1997-11-14 2000-08-30 The Regents of the University of California Slit polypeptide and method of modulation of robo with ligands

Also Published As

Publication number Publication date
US6342370B1 (en) 2002-01-29
AU1203999A (en) 1999-05-24
WO1999023219A1 (en) 1999-05-14

Similar Documents

Publication Publication Date Title
EP0750628B1 (en) Fibroblast growth factor-10
US6342370B1 (en) Human slit polypeptide and polynucleotides encoding same
US7482326B2 (en) Endothelial-monocyte activating polypeptide III
US20070154908A1 (en) Connective Tissue Growth Factor-2
US20090197278A9 (en) Antibodies to hADA2
EP1323824A2 (en) Use of human keratinocyte growth factor-2 for the preparation of pharmaceutical compositions
US7282351B2 (en) Prostatic growth factor
WO1996018730A1 (en) Prostatic growth factor
US6358702B1 (en) Polynucleotides encoding human Hox C10
US7071312B2 (en) Epidermal differentiation factor
US5654172A (en) Gabaa receptor epsilon subunit
US6573360B1 (en) Human ABH
AU710568B2 (en) Human vascular IBP-like growth factor
US6537539B2 (en) Immune cell cytokine
US20030022312A1 (en) Human hepatoma-derived growth factor-2
US20020119487A1 (en) Human stem cell antigen 2
US20050066266A9 (en) Human CCN-like growth factor
EP1230359A1 (en) Chordin-like homologs
US20030157103A1 (en) Human amine transporter antibodies
EP1284292A2 (en) Human B-cell Translocation Gene-3
EP1496065A2 (en) Breast specific genes and proteins
WO1997025338A1 (en) Immune cell cytokine

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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