US20010041350A1 - Human DNA ligase III - Google Patents

Human DNA ligase III Download PDF

Info

Publication number
US20010041350A1
US20010041350A1 US09/879,228 US87922801A US2001041350A1 US 20010041350 A1 US20010041350 A1 US 20010041350A1 US 87922801 A US87922801 A US 87922801A US 2001041350 A1 US2001041350 A1 US 2001041350A1
Authority
US
United States
Prior art keywords
antibody
amino acid
acid sequence
lys
dna
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
US09/879,228
Inventor
Ying-Fei Wei
Guo-Liang Yu
William Haseltine
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.)
Human Genome Sciences Inc
Original Assignee
Human Genome Sciences Inc
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
Priority claimed from PCT/US1995/003939 external-priority patent/WO1996030524A1/en
Application filed by Human Genome Sciences Inc filed Critical Human Genome Sciences Inc
Priority to US09/879,228 priority Critical patent/US20010041350A1/en
Publication of US20010041350A1 publication Critical patent/US20010041350A1/en
Priority to US10/464,768 priority patent/US20030211582A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/93Ligases (6)
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • C12N2799/026Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from a baculovirus

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.
  • the polypeptide of the present invention has been putatively identified as Human DNA Ligase m.
  • the invention also relates to inhibiting the action of such polypeptides.
  • DNA strand breaks and gaps are generated transiently during replication, repair and recombination. In mammalian cell nuclei, rejoining of such strand breaks depends on several different DNA polymerases and DNA ligase enzymes.
  • Reactivated AMP residue of the DNA ligase-adenylate intermediate is transferred to the 5′ phosphate terminus of a single strand break in double stranded DNA to generate a covalent DNA-AMP complex with a 5′-5′ phosphoanhydride bond.
  • This reaction intermediate has also been isolated for microbial and mammalian DNA ligase enzymes, but is more short lived than the adenylylated enzyme.
  • unadenylylated DNA ligase enzymes required for the generation of a phosphodiester bond catalyze displacement of the AMP residue through attack by the adjacent 3′-hydroxyl group on the adenylylated site.
  • DNA Ligase I The occurrence of three different DNA ligase enzymes, DNA Ligase I, II and III, was established previously by biochemical and immunological characterization of purified enzymes (Tomkinson, A. E. et al., J. Biol. Chem., 266:21728-21735 (1991) and Roberts, E., et al., J. Biol. Chem., 269:3789-3792 (1994)).
  • the inter-relationship between these proteins was unclear as a cDNA clone has only been available for DNA Ligase I, the major enzyme of this type in proliferating cells (Barnes, D. E., et al., PNAS USA, 87:6679-6683 (1990)).
  • DNA Ligase I appears to be the joining of Okazaki fragments during lagging-strand DNA replication (Waga, S., et al., J. Biol. Chem. 269:10923-10934 (1994); Li, C., et al., Nucl. Acids Res., 22:632-638 (1994); and Prigent, C., et al., Mol. Cell. Biol., 14:310-317 (1994)).
  • a full-length human cDNA encoding DNA Ligase I has been obtained by functional complementation of a S. cereviasiae cdc9 temperature-sensitive DNA ligase mutant (Barker, D. G., Eur. J. Biochem., 162:659-67 (1987)).
  • the full-length cDNA encodes a 102-kDa protein of 919 amino acid residues.
  • the active site lysine residue is located at position 568. It also effectively seals single-strand breaks in DNA and joins restriction enzyme DNA fragments with staggered ends. The enzyme is also able to catalyze blunt-end joining of DNA.
  • DNA Ligase I can join oligo (dT) molecules hydrogen-bonded to poly (dA), but the enzyme differs from T4 DNA Ligase II and III in being unable to ligate oligo (dT) with a poly (rA) complementary strand.
  • DNA Ligase III is more firmly associated with the cell nuclei. This enzyme is a labile protein, which is rapidly inactivated at 42° C. DNA Ligase III resembles other eukaryotic DNA Ligase enzymes in requiring ATP as cofactor, but the enzyme differs from DNA Ligase I in having a higher association for ATP. DNA Ligase III catalyzes the formation of phosphodiester bonds with an oligo (dT).poly (rA) substrate, but not with an oligo (rA).poly (dT) substrate, so it differs completely from DNA Ligase I in this regard (Arrand, J. E. et al., J. Biol. Chem., 261:9079-82 (1986)).
  • DNA Ligase III repairs single strand breaks in DNA efficiently, but it is unable to perform either blunt-end joining or AMP-dependent relaxation of super-coiled DNA (Elder, R. H. et a., Eur. J. Biochem., 203:53-58 (1992)).
  • mutant rodent cells deficient in XRCC1 protein exhibit reduced DNA Ligase III activity, defective strand break repair, an anomalously high level of sister chromatid exchanges, are hyper-sensitive to simple alkylating agents and ionizing radiation, and have an altered mutation spectrum after exposure to ethyl methanesulfonate (Caldecott, K. W., et al., Mol. Cell. Biol., 14:68-76 (1994); Ljungquist, S., et al., Mutat.
  • a purified mammalian protein fraction active in repair and recombination processes in vitro was shown to contain a ligase with the properties of Human DNA Ligase III, but no detectable amounts of Human DNA Ligase I (Jessberger, R., et al., J. Biol. Chem., 268:15070-15079 (1993)).
  • the role of the distinct enzyme, DNA Ligase II remains unclear, although an observed increase in DNA Ligase II activity during meiotic prophase suggests a role in meiotic recombination (Higashitani, A., et al., Cell Struct. Funct., 15:67-72 (1990)).
  • the polynucleotide of the present invention and polypeptide encoded thereby have been putatively identified as human DNA Ligase III as a result of size, amino acid sequence homology to DNA Ligase II and ability to bind XRCC1 protein.
  • the gene sequence of DNA Ligase III was not known.
  • novel mature polypeptides which are human DNA Ligase III, as well as biologically active and diagnostically or therapeutically useful fragments, analogs and derivatives thereof.
  • nucleic acid molecules encoding human DNA Ligase III, including mRNAs, DNAs, cDNAs, genomic DNAs as well as analogs and biologically active and diagnostically or therapeutically useful fragments thereof.
  • a process for producing such polypeptides by recombinant techniques comprising culturing recombinant prokaryotic and/or eukaryotic host cells, containing a human DNA Ligase III nucleic acid sequence, under conditions promoting expression of said protein and subsequent recovery of said protein.
  • a method of treating conditions which are related to insufficient human DNA Ligase III activity via gene therapy comprising inserting the DNA Ligase III gene into a patient's cells either in vivo or ex vivo.
  • the gene is expressed in transduced cells and as a result, the protein encoded by the gene may be used therapeutically, for example, to prevent disorders associated with defects in DNA, for example, abnormal cellular proliferation, for example cancers, leukemia and tumors, to treat severe immunosuppression, stunted growth and lymphoma, as well as cellular hypersensitivity to DNA-damaging agents.
  • nucleic acid probes comprising nucleic acid molecules of sufficient length to specifically hybridize to human DNA Ligase III sequences which may be used diagnostically to detect a mutation in the gene encoding DNA Ligase III.
  • antagonists to such polypeptides which may be manufactured intracellularly or administered through gene therapy for inhibiting the action of such polypeptides, for example, to target and destroy undesired cells, e.g., cancer cells.
  • diagnostic assays for detecting mutations in the polynucleotide sequences of the present invention for detecting diseases related to a lack of Human DNA Ligase III activity are provided.
  • FIGS. 1 A- 1 J show the cDNA sequence (SEQ ID NO:1) and the corresponding deduced amino sequence (SEQ ID NO:2) of the DNA Ligase III polypeptide.
  • the standard one letter abbreviation for amino acids is used.
  • the vertical arrow indicates the active site lysine.
  • an isolated nucleic acid (polynucleotide) (SEQ ID NO:1) which encodes for the mature polypeptide having the deduced amino acid sequence of FIGS. 1 A- 1 J (SEQ ID NO:2) or for the mature polypeptide encoded by the cDNA of the clone deposited as ATCC Deposit No. 97052 on Feb. 6, 1995 with the American Tissue Culture Collection, 10801 University Boulevard., Manassas, Va. 20110-2209, USA. The strain is being maintained under the terms of the Budapest Treaty and will be made available to a patent office signatory to the Budapest Treaty.
  • a polynucleotide encoding a polypeptide of the present invention may be obtained from testis, prostate, heart and thymus.
  • the polynucleotide of this invention was discovered in a cDNA library derived from human testis. It is structurally related to the DNA ligase family. It contains an open reading frame encoding a protein of 922 amino acid residues. The protein exhibits the highest degree of homology to vaccine virus DNA ligase with 56% identity and 73% similarity over the entire protein.
  • E-KYDG-R SEQ ID NO:11
  • the region flanking the conserved lysine residue is an active site motif that is essential for the formation of an enzyme-adenylate reaction intermediate (Tomkinson, A. E., et al., PNAS USA, 88:400-404 (1991)).
  • the conserved lysine residue is indicated by a vertical arrow and the active site motif is underlined in FIGS. 1 A- 1 J. Further a putative zinc finger motif shown at residues 18 to 55 in FIGS. 1 A- 1 J is underlined by a broken line.
  • the 100 kDa in vitro translation product of the DNA ligase III cDNA interacts with human XRCC1 protein which is a characteristic of DNA Ligase III (Caldecott, K.
  • Histidine-tagged recombinant XRCC1 protein was incubated with [ 35 S] methionine-labeled in vitro translation product of the cDNA to allow formation of XRCC1-protein complexes, after which NTA-agarose beads were added to affinity-bind XRCC1-His. The agarose beads were washed to remove non-specifically associated polypeptides prior to elution of XRCC1-His with 200 mM imidazole. XRCC1-his binds the product of the cDNA.
  • the longest open reading frame of the cDNA encoding DNA ligase III extends from 73 bp to 3099 bp within the cDNA clone and would encode a polypeptide of 1009 amino acids, approximately 150 kDa molecular mass.
  • the next downstream ATG at 334 bp occurs in a typical translation start consensus and defines an open reading frame of 2766 bp (922 amino acids).
  • the protein produced in this case would be approximately 103 kDa, consistent with both the observed molecular mass of the in vitro translation product and the apparent molecular mass of authentic DNA Ligase III purified from HeLa cells by standard chromatographic procedures. This indicates that this cDNA represents a full length cDNA clone.
  • a 5′-truncated cDNA clone lacking the first 78 bp (and the first ATG codon) produced an in vitro translation product of identical electrophoretic mobility to that encoded by the fill length clone, in support of assignment of the ATG at 334 bp as the translation initiation codon.
  • the DNA Ligase III amino acid sequence shows extensive amino acid homology to Human DNA Ligase I.
  • the DNA Ligase III sequence is identical at 8 of 12 residues flanking the active site lysine of DNA Ligase I, and both contain the minimum active site consensus for all ATP-dependent DNA ligases, -K-DG-R- (SEQ ID NO:10), with lys421 (DNA Ligase III) being the putative active lysine.
  • lys421 DNA Ligase III
  • human DNA Ligase I and III differ by 9 amino acids in the size of the region between the two motifs (active lysine and minimum active site motifs).
  • the 3′ flanking motif is located 37 amino acids from the C-terminus of DNA Ligase I, whereas the DNA Ligase III sequence extends a further 195 residues.
  • the C-terminus of the DNA Ligase III shows weak homology to several proteins, including approximately 20% identity to a 144 amino acid sequence within the C-terminal quarter of both human and murine XRCC1.
  • DNA Ligase I and III show very limited sequence homology beyond about 30 residues upstream of their active sites, and DNA Ligase I has an extended hydrophilic N-terminal region with no homology to DNA Ligase.
  • N-terminal 112 amino acids of the DNA Ligase III cDNA show approximately 30% identity to residues 3 to 107, and also residues 108 to 217, of human poly (ADP ribose) polymerase (PARP). These same two regions contain two evolutionarily conserved zinc finger motifs within the DNA-binding domain of PARP.
  • PARP human poly (ADP ribose) polymerase
  • sequence 411 -(K) CPNGMFSEIKYDGERVQVH (K)- 431 (SEQ ID NO:9) in the predicted amino acid sequence of the DNA ligase III cDNA, with Lys 421 the putative active lysine, is identical to the active site tryptic peptide identified in the purified bovine DNA Ligase II protein and different from that of DNA Ligase I (Tomkinson, A. E., et al., PNAS USA, 88:400-404 (1991)).
  • the polynucleotide 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 polypeptide may be identical to the coding sequence shown in FIGS. 1 A- 1 J (SEQ ID NO:1) or that of the deposited clone 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 polypeptide as the DNA of FIGS. 1 A- 1 J (SEQ ID NO:1) or the deposited cDNA.
  • the polynucleotide which encodes for the mature polypeptide of FIGS. 1 A- 1 J (SEQ ID NO:2) or for the mature polypeptide encoded by the deposited cDNA may include: only the coding sequence for the mature polypeptide; 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 polypeptide.
  • polynucleotide encoding a polypeptide encompasses a polynucleotide which includes only coding sequence for the polypeptide as well as a polynucleotide which includes additional coding and/or non-coding sequence.
  • the present invention further relates to variants of the hereinabove described polynucleotides which encode for fragments, analogs and derivatives of the polypeptide having the deduced amino acid sequence of FIGS. 1 A- 1 J (SEQ ID NO:2) or the polypeptide encoded by the cDNA of the deposited clone.
  • the variant of the polynucleotide may be a naturally occurring allelic variant of the polynucleotide or a non-naturally occurring variant of the polynucleotide.
  • the present invention includes polynucleotides encoding the same mature polypeptide as shown in FIGS. 1 A- 1 J (SEQ ID NO:2) or the same mature polypeptide encoded by the cDNA of the deposited clone as well as variants of such polynucleotides which variants encode for a fragment, derivative or analog of the polypeptide of FIGS. 1 A- 1 J (SEQ ID NO:2) or the polypeptide encoded by the cDNA of the deposited clone.
  • nucleotide variants include deletion variants, substitution variants and addition or insertion variants.
  • the polynucleotide may have a coding sequence which is a naturally occurring allelic variant of the coding sequence shown in FIGS. 1 A- 1 J (SEQ ID NO:1) or of the coding sequence of the deposited clone.
  • 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 polynucleotides of the present invention may also have the coding sequence fused in frame to a marker sequence which allows for purification of the polypeptide 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 polypeptide 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 promoter 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 encoded by the cDNAs of FIGS. 1 A- 1 J (SEQ ID NO:1) or the deposited cDNA(s).
  • the polynucleotide may have at least 20 bases, preferably 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 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 as well as fragments thereof, which fragments have at least 30 bases and preferably at least 50 bases and to polypeptides encoded by such polynucleotides.
  • the present invention further relates to a DNA Ligase III polypeptide which has the deduced amino acid sequence of FIGS. 1 A- 1 J (SEQ ID NO:2) or which has the amino acid sequence encoded by the deposited cDNA, as well as fragments, analogs and derivatives of such polypeptide.
  • fragment when referring to the polypeptide of FIGS. 1 A- 1 J (SEQ ID NO:2) or that encoded by the deposited cDNA, means a polypeptide which retains essentially the same biological function or activity as such polypeptide.
  • the polypeptide of the present invention may be a recombinant polypeptide, a natural polypeptide or a synthetic polypeptide, preferably a recombinant polypeptide.
  • the fragment, derivative or analog of the polypeptide of FIGS. 1 A- 1 J (SEQ ID NO:2) or that encoded by the deposited cDNA 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, which is employed for purification of the mature polypeptide.
  • 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.
  • 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).
  • 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 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 polypeptide of SEQ ID NO:2 and more preferably at least 90% similarity (more preferably at least 90% identity) to the polypeptide of SEQ ID NO:2 and still more preferably at least 95% similarity (still more preferably at least 95% identity) to the polypeptide 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.
  • 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 DNA Ligase III 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.
  • 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 (Pharrnacia).
  • Eukaryotic pWLNEO, pSV2CAT, pOG44, pXT1, pSG (Stratagene) pSVK3, pBPV, pMSG, pSVL (Pharrnacia).
  • 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 lacI, lacZ, T3, T7, gpt, lambda P P , 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 product encoded by the recombinant sequence.
  • the 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 including 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), ⁇ -factor, acid phosphatase, or heat shock proteins, among others.
  • PGK 3-phosphoglycerate kinase
  • the heterologous structural sequence is assembled in appropriate phase with translation, initiation and termination sequences.
  • 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 GEMI (Promega Biotec, Madison, Wis., USA).
  • 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 known 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 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.
  • the DNA Ligase II polypeptide 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.
  • DNA Ligase III polypeptides and agonists and antagonists which are polypeptides, described below, 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 III, and ⁇ -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 ApoAI 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 ⁇ -actin promoter; and human growth hormone promoters.
  • the promoter also may be the native promoter which controls the gene encoding the
  • 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, ⁇ -2, ⁇ -AM, PA12, T19-14X, VT-19-17-H2, ⁇ CRE, ⁇ CRIP, 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. 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 DNA Ligase III polypeptide may be used to repair single-strand breaks in DNA which result from DNA-damaging agents, e.g., UV radiation.
  • DNA-damaging agents e.g., UV radiation.
  • Several human syndromes result from autosomal recessive inheritance for the DNA ligase gene. These syndromes cause severe immunodeficiency and greatly increases the susceptibility of abnormal cellular differentiation due to the disrepair of DNA while at the cellular level they are characterized by chromosome instability and hypersensitivity to DNA-damaging agents. These syndromes include Fanconi's anemia and Blackfan-diamond anemia.
  • the polypeptide of the present invention may also be employed to treat severe immunosuppression which is the result of a defect in the DNA Ligase III gene.
  • DNA Ligase III may also be employed to treat stunted growth and lymphoma which result from defective rejoining of DNA.
  • the most common neoplastic chromosomal abnormality in this region is a translocation between chromosomes 15 and 17 seen in acute myeloid leukemia subtype m3 which involves the disruption of the retinoic acid receptor a gene (Chomienne, H., et al., Nature, 347:558-561 (1990)).
  • DNA Ligase III gene and gene product may be employed to treat these neoplasias.
  • Fragments of the full length Ligase III gene may be used as a hybridization probe for a cDNA library to isolate other genes which have a high sequence similarity to the DNA Ligase III gene or have similar biological activity.
  • Probes of this type have at least 20 bases. Preferably, however, the probes 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 DNA Ligase III gene including regulatory and promoter regions, exons, and introns.
  • An example of a screen comprises isolating the coding region of the DNA Ligase III 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 polypeptide and/or polynucleotide of the present invention may also be employed in relation to scientific research, synthesis of DNA and for the manufacture of DNA vectors.
  • the polypeptide and/or polynucleotide of the present invention may be sold into the research market.
  • DNA Ligase III may be used for ligation of DNA sequences in vitro in a manner similar to other DNA ligase enzymes of the art.
  • This invention also provides a method of screening compounds to identify those which enhance or inhibit the DNA joining reaction catalyzed by human DNA Ligase III.
  • An example of such a method comprises combining ATP, DNA Ligase III and DNA having single-strand breaks with the compound under conditions where the DNA Ligase would normally cleave ATP to AMP and the AMP is transferred to the 5′ phosphate terminus of a single strand break in double-stranded DNA to generate a covalent DNA-AMP complex with the single strand break being subsequently repaired.
  • the DNA having the single-strand breaks may be supplied in the above example by mutant cells which are deficient in proteins that are responsible for strand break repair, for example, mutant rodent cells deficient in XRCC1 and the cdc9 S. cerevisiae DNA ligase mutant. The ability of the compound to enhance or block the catalysis of this reaction could then be measured to determine if the compound is an effective agonist or antagonist.
  • Human DNA Ligase III is produced and functions intracellularly, therefore, any antagonist must be intra-cellular.
  • Potential antagonists to human DNA Ligase III include antibodies which are produced intracellularly.
  • an antibody identified as antagonizing DNA Ligase III may be produced intracellularly as a single chain antibody by procedures known in the art, such as transforming the appropriate cells with DNA encoding the single chain antibody to prevent the function of human DNA Ligase III.
  • Another potential antagonist is an antisense construct prepared using antisense technology.
  • 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 for 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 DNA Ligase III (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 DNA Ligase III.
  • Yet another potential antagonist includes a mutated form, or mutein, of DNA Ligase III which recognizes DNA but does not repair single-strand breaks and, therefore, acts to prevent human DNA Ligase III from functioning.
  • the antagonists may be employed to target undesired cells, e.g., cancer cells and leukemic cells, since the prevention of DNA Ligase III prevents repair of single-strand breaks in DNA and will eventually result in death of the cell.
  • compositions comprise a therapeutically effective amount of the molecule and a pharmaceutically acceptable carrier or excipient.
  • 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 pharmaceutical compositions of the present invention may be employed in conjunction with other therapeutic compounds.
  • the pharmaceutical compositions may be administered in a convenient manner such as by the oral, topical, intravenous, intraperitoneal, intramuscular, 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, they are 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.
  • This invention also provides the use of the human DNA Ligase III gene as a diagnostic. For example, some diseases result from inherited defective genes. These genes can be detected by comparing the sequence of the defective gene with that of a normal one. That is, a mutant gene would be associated with hypersensitivity to DNA-damaging agents and an elevated susceptibility to abnormal cell growth, for example, tumors, leukemia and cancer.
  • Nucleic acids used 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. Deletions or 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 DNA Ligase III RNA or alternatively, radiolabeled DNA Ligase III 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 protection 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 protection 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, and Southern blotting of genomic DNA. Also, mutations may be detected by in situ analysis.
  • some diseases are a result of, or are characterized by, changes in gene expression which can be detected by changes in the mRNA.
  • the DNA Ligase III gene can be used as a reference to identify individuals expressing a decreased level of DNA Ligase III protein, e.g., by Northern blotting.
  • 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 clone to a metaphase chromosomal spread can be used to provide a precise chromosomal location in one step.
  • FISH Fluorescence in situ hybridization
  • This technique can be used with cDNA as short as 50 or 60 bases. For example, 2,000 bp is good, 4,000 is better, and more than 4,000 is probably not necessary to get good results a reasonable percentage of the time.
  • Verna et al. Human Chromosomes: a Manual of Basic Techniques, Pergamon Press, New York (1988).
  • 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. hi 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.
  • 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).
  • 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 enzymes 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 ⁇ l 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 of T4 DNA ligase (“ligase”) per 0.5 ⁇ g of approximately equimolar amounts of the DNA fragments to be ligated.
  • ligase T4 DNA ligase
  • the DNA sequence encoding DNA Ligase III is initially amplified using PCR oligonucleotide primers corresponding to the 5′ and 3 ⁇ 0 end sequences of the processed DNA Ligase III gene.
  • the 5′ oligonucleotide primer has the sequence 5′ CGCGGATCCATGGCTGAGCAACGGTTCTG 3′ (SEQ ID NO:3) contains a Bam HI restriction enzyme site (underlined) followed by 20 nucleotides of DNA Ligase III coding sequence starting from the presumed terminal amino acid of the processed protein codon.
  • the 3′ sequence 5′ GCGTCTAGACTAGCAGGGAGCTACCAG 3′ contains complementary sequences to a XbaI site (underlined) and is followed by 18 nucleotides of DNA Ligase III at C-terminal of DNA Ligase III.
  • the restriction enzyme sites correspond to the restriction enzyme sites on the bacterial expression vector pQE-9 (Qiagen, Inc. Chatsworth, Calif.).
  • pQE-9 encodes antibiotic resistance (Amp r ), a bacterial origin of replication (ori), an IPTG-regulatable promoter operator (P/O), a ribosome binding site (RBS), a 6-His tag and restriction enzyme sites.
  • pQE-9 is then digested with Bam HI and Pst I.
  • the amplified sequences are ligated into pQE-9 and inserted in frame with the sequence encoding for the histidine tag and the RBS.
  • the ligation mixture is then used to transform E. coli strain M15/rep 4 (Qiagen, Inc.) under the trademark M15/rep 4 by the procedure described in Sambrook, J. et al., Molecular Cloning: A Laboratory Manual, Cold Spring Laboratory Press, (1989).
  • M15/rep4 contains multiple copies of the plasmid pREP4, which expresses the lacI repressor and also confers kanamycin resistance (Kan r ).
  • Transformants are identified by their ability to grow on LB plates and ampicillin/kanamycin resistant colonies are selected. Plasmid DNA is isolated and confirmed by restriction analysis. Clones containing the desired constructs are grown overnight (O/N) in liquid culture in LB media supplemented with both Amp (100 ug/ml) and Kan (25 ug/ml). The O/N culture is used to inoculate a large culture at a ratio of 1:100 to 1:250. The cells are grown to an optical density 600 (O.D. 600 ) of between 0.4 and 0.6. IPTG (“Isopropyl-B-D-thiogalacto pyranoside”) is then added to a final concentration of 1 mM.
  • O.D. 600 optical density 600
  • IPTG induces by inactivating the lacI repressor, clearing the P/O leading to increased gene expression.
  • Cells are grown an extra 3 to 4 hours. Cells are then harvested by centrifugation. The cell pellet is solubilized in the chaotropic agent 6 Molar Guanidine HCl. After clarification, solubilized protein extract is purified from this solution by chromatography on a Nickel-Chelate column under conditions that allow for tight binding by proteins containing the 6-His tag (Hochuli, E. et al., J.
  • the 5′ primer has the sequence 5′ CGCGAATCCATGGCTGAGCAACGGTTCTG 3′ (SEQ ID NO:5) and contains a BamHI restriction enzyme site (in bold) followed first by 20 nucleotides of N-terminal sequence (the initiation codon for translation “ATG” is underlined).
  • the 3′ primer has the sequence 5′ GCGTCTAGACTAGCAGGGAGCTACCAG 3′ (SEQ ID NO:6) and contains the cleavage site for the restriction endonuclease XbaI (in bold) and 18 nucleotides complementary to the C-terminal sequence of the DNA Ligase III gene.
  • the amplified sequences were isolated from a 1% agarose gel using a commercially available kit (“Geneclean,” BIO 101 Inc., La Jolla, Calif.). The fragment was then digested with the endonucleases BamHI and XbaI and then purified again on a 1% agarose gel. This fragment is designated F2.
  • the vector pA2 (modification of pVL941 vector, discussed below) is used for the expression of the DNA Ligase II protein using the baculovirus expression system (for review see: Summers, M. D. and Smith, G. E. 1987, A manual of methods for baculovirus vectors and insect cell culture procedures, Texas Agricultural Experimental Station Bulletin No. 1555).
  • This expression vector contains the strong polyhedrin promoter of the Autographa califormica nuclear polyhidrosis virus (AcMNPV) followed by the recognition sites for the restriction endonucleases BamHI and XbaI.
  • the polyadenylation site of the simian virus (SV)40 is used for efficient polyadenylation.
  • the beta-galactosidase gene from E. coli is inserted in the same orientation as the polyhedrin promoter followed by the polyadenylation signal of the polyhedrin gene.
  • the polyhedrin sequences are flanked at both sides by viral sequences for the cell-mediated homologous recombination of co-transfected wild-type viral DNA.
  • Many other baculovirus vectors could be used in place of pRG1 such as pAc373, pVL941 and pAcIM1 (Luckow, V. A. and Summers, M. D., Virology, 170:31-39).
  • the plasmid is digested with the restriction enzymes BamHI and XbaI and then dephosphorylated using calf intestinal phosphatase by procedures known in the art.
  • the DNA is then isolated from a 1% agarose gel using the commercially available kit (“Geneclean” BIO 101 Inc., La Jolla, Calif.). This vector DNA is designated V2.
  • Fragment F2 and the dephosphorylated plasmid V2 were ligated with T4 DNA ligase.
  • E. coli HB101 cells are then transformed and bacteria identified that contained the plasmid (pBac DNA Ligase III) with the DNA Ligase III gene using the enzymes BamHI and XbaI. The sequence of the cloned fragment is confirmed by DNA sequencing.
  • the plate is then incubated for 5 hours at 27° C. After 5 hours the transfection solution is removed from the plate and 1 ml of Grace's insect medium supplemented with 10% fetal calf serum is added. The plate is put back into an incubator and cultivation continued at 27° C. for four days.
  • plaque assay After four days the supematant is collected and a plaque assay performed similar as described by Summers and Smith (supra). As a modification an agarose gel with “Blue Gal” (Life Technologies Inc., Gaithersburg) is used which allows an easy isolation of blue stained plaques. (A detailed description of a “plaque assay” can also be found in the user's guide for insect cell culture and baculovirology distributed by Life Technologies Inc., Gaithersburg, page 9-10).
  • the viruses are added to the cells and blue stained plaques are picked with the tip of an Eppendorf pipette.
  • the agar containing the recombinant viruses is then resuspended in an Eppendorf tube containing 200 ⁇ l of Grace's medium.
  • the agar is removed by a brief centrifugation and the supernatant containing the recombinant baculovirus is used to infect Sf9 cells seeded in 35 mm dishes.
  • the supernatants of these culture dishes are harvested and then stored at 4° C.
  • Sf9 cells are grown in Grace's medium supplemented with 10% heat-inactivated FBS.
  • the cells are infected with the recombinant baculovirus V-DNA Ligase III at a multiplicity of infection (MOI) of 2.
  • MOI multiplicity of infection
  • the medium is removed and replaced with SF900 III medium minus methionine and cysteine (Life Technologies Inc., Gaithersburg).
  • 5 ⁇ Ci of 35 S-methionine and 5 ⁇ Ci 35 S cysteine (Amersham) are added.
  • the cells are further incubated for 16 hours before they are harvested by centrifugation and the labeled proteins visualized by SDS-PAGE and autoradiography.
  • Plasmid, DNA Ligase III HA is derived from a vector pcDNAI/Amp (Invitrogen) containing: 1) SV40 origin of replication, 2) ampicillin resistance gene, 3) E. coli replication origin, 4) CMV promoter followed by a polylinker region, a SV40 intron and polyadenylation site.
  • a DNA fragment encoding the entire DNA Ligase III precursor and a HA tag fused in frame to its 3′ end was cloned into the polylinker region of the vector, therefore, the recombinant protein expression is directed under the CMV promoter.
  • the HA tag correspond to an epitope derived from the influenza hemagglutinin protein as previously described (I.
  • HA tag to the target protein allows detection of the recombinant protein with an antibody that recognizes the HA epitope.
  • the DNA sequence encoding DNA Ligase III is constructed by PCR using two primers: the 5′ primer 5′ CGC ⁇ overscore (GAATCC) ⁇ ATGGCTGAGCAACGGTTCTG 3′ (SEQ ID NO:7) contains an BamHI site (underlined) followed by 20 nucleotides of DNA Ligase III coding sequence starting from the initiation codon; the 3′ sequence 5′ GCG ⁇ overscore (TCTAGA) ⁇ TCAAGCGTAGTCTGGGACGTCGTATGGGTAGCAGGGAGCTACCA GTC 3′ (SEQ ID NO:8) contains complementary sequences to an Xbal site (underlined), translation stop codon, HA tag and the last 17 nucleotides of the DNA Ligase III coding sequence (not including the stop codon).
  • the PCR product contains an BamHI site, DNA Ligase III coding sequence followed by HA tag fused in frame, a translation termination stop codon next to the HA tag, and an XbaI site.
  • the PCR amplified DNA fragment and the vector, pcDNAI/Amp are digested with BamHI and XbaI restriction enzyme and ligated.
  • the ligation mixture is transformed into E. coli strain SURE (Stratagene Cloning Systems, La Jolla, Calif.) the transformed culture is plated on ampicillin media plates and resistant colonies are selected. Plasmid DNA is isolated from transformants and examined by restriction analysis for the presence of the correct fragment.
  • COS cells are transfected with the expression vector by DEAE-DEXTRAN method (J. Sambrook, E. Fritsch, T. Maniatis, Molecular Cloning: A Laboratory Manual, Cold Spring Laboratory Press, (1989)).
  • the expression of the DNA Ligase III HA protein is detected by radiolabeling and immunoprecipitation method (E. Harlow, D. Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, (1988)). Cells are labeled for 8 hours with 35 S-cysteine two days post transfection.
  • Culture media are then collected and cells are lysed with detergent (RIPA buffer (150 mM NaCl, 0.1% SDS, 1% NP-40, 0.5% DOC, 50 mM Tris, pH 7.5) (Wilson, I. et al., Id. 37:767 (1984)). Both cell lysate and culture media are precipitated with a HA specific monoclonal antibody. Proteins precipitated are analyzed on 15% SDS-PAGE gels.
  • RIPA buffer 150 mM NaCl, 0.1% SDS, 1% NP-40, 0.5% DOC, 50 mM Tris, pH 7.5
  • Northern blot analysis may be performed to examine the levels of expression of DNA Ligase III in human tissues.
  • Total cellular RNA samples are isolated with RNAzolTM B system (Biotecx Laboratories, Inc. Houston, Tex.) About 15 ⁇ g of total RNA isolated from each human tissue specified is separated on 1% agarose gel and blotted onto a nylon filter (Sambrook, Fritsch, and Maniatis, Molecular Cloning, Cold Spring Harbor Press, (1989)).
  • the labeling reaction is done according to the Stratagene Prime-It kit with 50 ng DNA fragment.
  • the labeled DNA is purified with a Select-G-50 column (5 Prime-3 Prime, Inc. Boulder, Colo.).
  • the filter containing the particular RNA blot is then hybridized with radioactive labeled full length DNA Ligase III gene at 1,000,000 cpm/ml in 0.5 M NaPO 4 , pH 7.4 and 7% SDS overnight at 65° C. After wash twice at room temperature and twice at 60° C. with 0.5 ⁇ SSC, 0.1% SDS, the filter is then exposed at ⁇ 70° C. overnight with an intensifying screen.
  • the message RNA for DNA Ligase II is abundant in the testis, prostate, heart, and thymus.
  • DNA ligase III was subcloned as a Sal I/Not I restriction fragment into the multiple cloning sire of pSPORT (Life Technologies), with the 5′ end proximal to the T7 promoter; the DNA ligase III plasmid constructs (1 ⁇ g) was linearized with either Not I or Xho I (New England Biolabs), downstream of the cDNA insert, then transcribed and capped at 36° C. for 30 minutes with T7 polymerase and the mCAP RNA capping kit (Stratagene). The reactions were terminated by incubation with 10 units RNase-free DNase at 37° C. for 5 minutes.
  • the in vitro transcription products were resuspended in 20 ⁇ l 10 mM Tris-HCl/1 mM EDTA, pH 8.0 (TE).
  • the transcript (0 to 5 ⁇ l, made up to a final volume of 5 ⁇ l with water) was translated in 20 ⁇ l rabbit reticulocyte lysate (Amersham) at 30° C. for 90 minutes.
  • reaction was supplemented with 20 ⁇ Ci [ 35 S]methionine (3000 Ci mmol ⁇ 1 , Amersham).
  • Translations were terminated by incubation with 5 ⁇ l of 400 ml ⁇ 1 RNase A/50 mM EDTA at 37° C. for 15 minutes (30 ⁇ l final volume).
  • Samples (5 ⁇ l) of translations carried out in the presence of [ 35 S]methionine were analyzed by electrophoresis in SDS-7.5% polyacrylamide gels and autoradiography.
  • Non-radiolabeled translation products were assayed for ability to form protein-adenylate complexes after removal of ATP by chromatography through spun 1 ml columns of Sephadex G50 (Pharmacia) equilibrated with TE.
  • Fibroblasts are obtained from a subject by skin biopsy. The resulting tissue is placed in tissue-culture medium and separated into small pieces. Small chunks of the tissue are placed on a wet surface of a tissue culture flask, approximately ten pieces are placed in each flask. The flask is turned upside down, closed tight and left at room temperature over night. After 24 hours at room temperature, the flask is inverted and the chunks of tissue remain fixed to the bottom of the flask and fresh media (e.g., Ham's F12 media, with 10% FBS, penicillin and streptomycin, is added. This is then incubated at 37° C. for approximately one week. At this time, fresh media is added and subsequently changed every several days. After an additional two weeks in culture, a monolayer of fibroblasts emerge. The monolayer is trypsinized and scaled into larger flasks.
  • fresh media e.g., Ham's F12 media, with 10% FBS, penicillin and streptomycin
  • Moloney murine leukemia virus is digested and treated with calf intestinal phosphatase.
  • the linear vector is fractionated on agarose gel and purified, using glass beads.
  • the DNA Ligase III cDNA (see FIGS. 1 A- 1 J) is isolated and the ends of this fragment are treated with DNA polymerase in order to fill in the recessed ends and create blunt ends.
  • PE501 packaging cells are grown in tissue culture to confluent density in Dulbecco's Modified Eagles Medium (DMEM) with 10% calf serum (CS), penicillin and streptomycin.
  • DMEM Dulbecco's Modified Eagles Medium
  • CS calf serum
  • penicillin and streptomycin The Moloney murine leukemia virus vector containing the gene is then added to the media and the packaging cells are transduced with the vector.
  • the packaging cells now produce infectious viral particles containing the DNA Ligase III gene.
  • Fresh media is added to the transduced producer cells, and subsequently the media is harvested from a 10 cm plate of confluent producer cells.
  • the spent media containing the infectious viral particles, is filtered through a millipore filter to remove detached producer cells and this media is then used to infect fibroblast cells.
  • Media is removed from a sub-confluent plate of fibroblasts and quickly replaced with the media from the producer cells.
  • the engineered fibroblasts are then injected into the into a host, for example, a rat, either alone or after having been grown to confluence on cytodex 3 microcarrier beads.
  • the fibroblasts now produce the protein product and the biological actions of DNA Ligase III are conveyed to the host.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

A human DNA Ligase III polypeptide and DNA (RNA) encoding such polypeptide and a procedure for producing such polypeptide by recombinant techniques is disclosed. Also disclosed are methods for utilizing such polypeptide via gene therapy for the treatment of disorders associated with a defect in DNA Ligase III. Antagonists against such polypeptides and their use as a therapeutic to destroy unwanted cells are also disclosed. Diagnostic assays to detect mutant DNA Ligase III genes are also disclosed.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application is a divisional of and claims priority under 35 U.S.C. § 120 to U.S. application Ser. No. 09/054,775, filed Apr. 3, 1998, which is a divisional of and claims priority under 35 U.S.C. § 120 to U.S. application Ser. No. 08/464,402, filed Jun. 5, 1995 (now U.S. Pat. No. 5,858,705, issued Jan. 12, 1999), which is a continuation-in-part of and claims priority under 35 U.S.C. § 120 to PCT/US95/03939, filed Mar. 31, 1995.[0001]
  • FIELD OF THE INVENTION
  • 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. The polypeptide of the present invention has been putatively identified as Human DNA Ligase m. The invention also relates to inhibiting the action of such polypeptides. [0002]
  • BACKGROUND OF THE INVENTION
  • DNA strand breaks and gaps are generated transiently during replication, repair and recombination. In mammalian cell nuclei, rejoining of such strand breaks depends on several different DNA polymerases and DNA ligase enzymes. [0003]
  • The mechanism for joining of DNA strand interruptions by DNA ligase enzymes has been widely described. The reaction is initiated by the formation of a covalent enzyme-adenylate complex. Mammalian and viral DNA ligase enzymes employ ATP as cofactor, whereas bacterial DNA ligase enzymes use NAD to generate the adenylyl group. The ATP is cleaved to AMP and pyrophosphate with the adenylyl residue linked by a phosphoramidate bond to the ε-amino group of a specific lysine residue at the active site of the protein (Gumport, R. I., et al., [0004] PNAS, 68:2559-63 (1971)). Reactivated AMP residue of the DNA ligase-adenylate intermediate is transferred to the 5′ phosphate terminus of a single strand break in double stranded DNA to generate a covalent DNA-AMP complex with a 5′-5′ phosphoanhydride bond. This reaction intermediate has also been isolated for microbial and mammalian DNA ligase enzymes, but is more short lived than the adenylylated enzyme. In the final step of DNA ligation, unadenylylated DNA ligase enzymes required for the generation of a phosphodiester bond catalyze displacement of the AMP residue through attack by the adjacent 3′-hydroxyl group on the adenylylated site.
  • The occurrence of three different DNA ligase enzymes, DNA Ligase I, II and III, was established previously by biochemical and immunological characterization of purified enzymes (Tomkinson, A. E. et al., J. Biol. Chem., 266:21728-21735 (1991) and Roberts, E., et al., J. Biol. Chem., 269:3789-3792 (1994)). However, the inter-relationship between these proteins was unclear as a cDNA clone has only been available for DNA Ligase I, the major enzyme of this type in proliferating cells (Barnes, D. E., et al., PNAS USA, 87:6679-6683 (1990)). The main function of DNA Ligase I appears to be the joining of Okazaki fragments during lagging-strand DNA replication (Waga, S., et al., J. Biol. Chem. 269:10923-10934 (1994); Li, C., et al., Nucl. Acids Res., 22:632-638 (1994); and Prigent, C., et al., Mol. Cell. Biol., 14:310-317 (1994)). [0005]
  • A full-length human cDNA encoding DNA Ligase I has been obtained by functional complementation of a [0006] S. cereviasiae cdc9 temperature-sensitive DNA ligase mutant (Barker, D. G., Eur. J. Biochem., 162:659-67 (1987)). The full-length cDNA encodes a 102-kDa protein of 919 amino acid residues. There is no marked sequence homology to other known proteins except for microbial DNA ligase enzymes. The active site lysine residue is located at position 568. It also effectively seals single-strand breaks in DNA and joins restriction enzyme DNA fragments with staggered ends. The enzyme is also able to catalyze blunt-end joining of DNA. DNA Ligase I can join oligo (dT) molecules hydrogen-bonded to poly (dA), but the enzyme differs from T4 DNA Ligase II and III in being unable to ligate oligo (dT) with a poly (rA) complementary strand.
  • Human DNA Ligase III is more firmly associated with the cell nuclei. This enzyme is a labile protein, which is rapidly inactivated at 42° C. DNA Ligase III resembles other eukaryotic DNA Ligase enzymes in requiring ATP as cofactor, but the enzyme differs from DNA Ligase I in having a higher association for ATP. DNA Ligase III catalyzes the formation of phosphodiester bonds with an oligo (dT).poly (rA) substrate, but not with an oligo (rA).poly (dT) substrate, so it differs completely from DNA Ligase I in this regard (Arrand, J. E. et al., [0007] J. Biol. Chem., 261:9079-82 (1986)).
  • DNA Ligase III repairs single strand breaks in DNA efficiently, but it is unable to perform either blunt-end joining or AMP-dependent relaxation of super-coiled DNA (Elder, R. H. et a., [0008] Eur. J. Biochem., 203:53-58 (1992)).
  • Clues as to the physiological role of DNA Ligase III have come from its physical interaction in a high salt-resistant complex with another nuclear protein, the XRCC1 gene product (Caldecott, K. W., et al., [0009] Mol. Cell. Biol., 14:68-76 (1994) and Ljungquist, S., et al., Mutat. Res., 314:177-186 (1994)). The XRCC1 gene encodes a 70 kDa protein, that by itself does not appear to join DNA strand breaks (Caldecott, K. W., et al., Mol. Cell. Biol., 14:68-76 (1994); Ljungquist, S., et al., Mutat. Res., 314:177-186 (1994) and Thompson, L. H., et al., Mol. Cell. Biol., 10:6160-6171 (1990)). However, mutant rodent cells deficient in XRCC1 protein exhibit reduced DNA Ligase III activity, defective strand break repair, an anomalously high level of sister chromatid exchanges, are hyper-sensitive to simple alkylating agents and ionizing radiation, and have an altered mutation spectrum after exposure to ethyl methanesulfonate (Caldecott, K. W., et al., Mol. Cell. Biol., 14:68-76 (1994); Ljungquist, S., et al., Mutat. Res., 314:177-186 (1994); Thompson, L. H., et al., Mol. Cell. Biol., 10:6160-6171 (1990); and Op het Veld, C. W., et al., Cancer Res., 54:3001-3006 (1994)). These data indicate that XRCC1 mutant cells are defective in base excision-repair, and strongly suggest that both DNA Ligase III and XRCC1 are active in this process (Dianov, G., and Lindahl, T., Curr. Biol., 4:1069-1076 (1994)).
  • A purified mammalian protein fraction active in repair and recombination processes in vitro was shown to contain a ligase with the properties of Human DNA Ligase III, but no detectable amounts of Human DNA Ligase I (Jessberger, R., et al., [0010] J. Biol. Chem., 268:15070-15079 (1993)). The role of the distinct enzyme, DNA Ligase II, remains unclear, although an observed increase in DNA Ligase II activity during meiotic prophase suggests a role in meiotic recombination (Higashitani, A., et al., Cell Struct. Funct., 15:67-72 (1990)). Comparison of 32P-adenylylated DNA Ligase II and III by partial or complete proteolytic cleavage patterns indicated that these two enzymes share extensive amino acid sequence similarity or identity flanking their active sites, but that they are quite different from DNA Ligase I (Roberts, E., et al., J. Biol. Chem., 269:3789-3792 (1994)). Neither DNA Ligase I, II nor III is exclusively a mitochondrial enzyme.
  • BRIEF SUMMARY OF THE INVENTION
  • The polynucleotide of the present invention and polypeptide encoded thereby have been putatively identified as human DNA Ligase III as a result of size, amino acid sequence homology to DNA Ligase II and ability to bind XRCC1 protein. Heretofore, the gene sequence of DNA Ligase III was not known. [0011]
  • In accordance with one aspect of the present invention, there are provided novel mature polypeptides which are human DNA Ligase III, as well as biologically active and diagnostically or therapeutically useful fragments, analogs and derivatives thereof. [0012]
  • In accordance with another aspect of the present invention, there are provided isolated nucleic acid molecules encoding human DNA Ligase III, including mRNAs, DNAs, cDNAs, genomic DNAs as well as analogs and biologically active and diagnostically or therapeutically useful fragments thereof. [0013]
  • In accordance with yet a further aspect of the present invention, there is provided a process for producing such polypeptides by recombinant techniques comprising culturing recombinant prokaryotic and/or eukaryotic host cells, containing a human DNA Ligase III nucleic acid sequence, under conditions promoting expression of said protein and subsequent recovery of said protein. [0014]
  • 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. [0015]
  • In accordance with another aspect of the present invention there is provided a method of treating conditions which are related to insufficient human DNA Ligase III activity via gene therapy comprising inserting the DNA Ligase III gene into a patient's cells either in vivo or ex vivo. The gene is expressed in transduced cells and as a result, the protein encoded by the gene may be used therapeutically, for example, to prevent disorders associated with defects in DNA, for example, abnormal cellular proliferation, for example cancers, leukemia and tumors, to treat severe immunosuppression, stunted growth and lymphoma, as well as cellular hypersensitivity to DNA-damaging agents. [0016]
  • In accordance with yet a further aspect of the present invention, there is also provided nucleic acid probes comprising nucleic acid molecules of sufficient length to specifically hybridize to human DNA Ligase III sequences which may be used diagnostically to detect a mutation in the gene encoding DNA Ligase III. [0017]
  • In accordance with yet another aspect of the present invention, there are provided antagonists to such polypeptides, which may be manufactured intracellularly or administered through gene therapy for inhibiting the action of such polypeptides, for example, to target and destroy undesired cells, e.g., cancer cells. [0018]
  • In accordance with still another aspect of the present invention, there are provided diagnostic assays for detecting mutations in the polynucleotide sequences of the present invention for detecting diseases related to a lack of Human DNA Ligase III activity. [0019]
  • These and other aspects of the present invention should be apparent to those skilled in the art from the teachings herein.[0020]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • 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. [0021]
  • FIGS. [0022] 1A-1J show the cDNA sequence (SEQ ID NO:1) and the corresponding deduced amino sequence (SEQ ID NO:2) of the DNA Ligase III polypeptide. The standard one letter abbreviation for amino acids is used. The vertical arrow indicates the active site lysine.
  • DETAILED DESCRIPTION
  • In accordance with an aspect of the present invention, there is provided an isolated nucleic acid (polynucleotide) (SEQ ID NO:1) which encodes for the mature polypeptide having the deduced amino acid sequence of FIGS. [0023] 1A-1J (SEQ ID NO:2) or for the mature polypeptide encoded by the cDNA of the clone deposited as ATCC Deposit No. 97052 on Feb. 6, 1995 with the American Tissue Culture Collection, 10801 University Blvd., Manassas, Va. 20110-2209, USA. The strain is being maintained under the terms of the Budapest Treaty and will be made available to a patent office signatory to the Budapest Treaty.
  • A polynucleotide encoding a polypeptide of the present invention may be obtained from testis, prostate, heart and thymus. The polynucleotide of this invention was discovered in a cDNA library derived from human testis. It is structurally related to the DNA ligase family. It contains an open reading frame encoding a protein of 922 amino acid residues. The protein exhibits the highest degree of homology to vaccine virus DNA ligase with 56% identity and 73% similarity over the entire protein. It is also important that there is a conserved active lysine residue at [0024] position 421 which is bordered on either side by a hydrophobic amino acid residue, and the sequence E-KYDG-R (SEQ ID NO:11) is also conserved and is common to enzymes from different sources such as mammalian cells, yeasts, vaccinia virus and bacteriophage T7.
  • The region flanking the conserved lysine residue is an active site motif that is essential for the formation of an enzyme-adenylate reaction intermediate (Tomkinson, A. E., et al., [0025] PNAS USA, 88:400-404 (1991)). The conserved lysine residue is indicated by a vertical arrow and the active site motif is underlined in FIGS. 1A-1J. Further a putative zinc finger motif shown at residues 18 to 55 in FIGS. 1A-1J is underlined by a broken line. The 100 kDa in vitro translation product of the DNA ligase III cDNA interacts with human XRCC1 protein which is a characteristic of DNA Ligase III (Caldecott, K. W., et al., Mol. Cell. Biol., 14:68-76 (1994)). Histidine-tagged recombinant XRCC1 protein was incubated with [35S] methionine-labeled in vitro translation product of the cDNA to allow formation of XRCC1-protein complexes, after which NTA-agarose beads were added to affinity-bind XRCC1-His. The agarose beads were washed to remove non-specifically associated polypeptides prior to elution of XRCC1-His with 200 mM imidazole. XRCC1-his binds the product of the cDNA. Recovery of radiolabeled polypeptides is dependent on addition of XRCC1-His. Approximately 50% of the full length 100 kDa translation product, and as much as 90% of some of the truncated polypeptides, were recovered with XRCC1-His. These results indicate that the cDNA clone encodes a 100 kDa polypeptide.
  • The longest open reading frame of the cDNA encoding DNA ligase III extends from 73 bp to 3099 bp within the cDNA clone and would encode a polypeptide of 1009 amino acids, approximately 150 kDa molecular mass. The next downstream ATG at 334 bp occurs in a typical translation start consensus and defines an open reading frame of 2766 bp (922 amino acids). The protein produced in this case would be approximately 103 kDa, consistent with both the observed molecular mass of the in vitro translation product and the apparent molecular mass of authentic DNA Ligase III purified from HeLa cells by standard chromatographic procedures. This indicates that this cDNA represents a full length cDNA clone. Furthermore, a 5′-truncated cDNA clone lacking the first 78 bp (and the first ATG codon) produced an in vitro translation product of identical electrophoretic mobility to that encoded by the fill length clone, in support of assignment of the ATG at 334 bp as the translation initiation codon. [0026]
  • The DNA Ligase III amino acid sequence shows extensive amino acid homology to Human DNA Ligase I. The DNA Ligase III sequence is identical at 8 of 12 residues flanking the active site lysine of DNA Ligase I, and both contain the minimum active site consensus for all ATP-dependent DNA ligases, -K-DG-R- (SEQ ID NO:10), with lys421 (DNA Ligase III) being the putative active lysine. Although their amino acid sequences are not co-linear at optimum alignment, human DNA Ligase I and III differ by 9 amino acids in the size of the region between the two motifs (active lysine and minimum active site motifs). [0027]
  • The 3′ flanking motif is located 37 amino acids from the C-terminus of DNA Ligase I, whereas the DNA Ligase III sequence extends a further 195 residues. The C-terminus of the DNA Ligase III shows weak homology to several proteins, including approximately 20% identity to a 144 amino acid sequence within the C-terminal quarter of both human and murine XRCC1. [0028]
  • In their N-terminal regions, DNA Ligase I and III show very limited sequence homology beyond about 30 residues upstream of their active sites, and DNA Ligase I has an extended hydrophilic N-terminal region with no homology to DNA Ligase. [0029]
  • The N-terminal 112 amino acids of the DNA Ligase III cDNA show approximately 30% identity to residues 3 to 107, and also residues 108 to 217, of human poly (ADP ribose) polymerase (PARP). These same two regions contain two evolutionarily conserved zinc finger motifs within the DNA-binding domain of PARP. [0030]
  • The highly conserved motif flanking the 3′ boundary of the region of homology between DNA Ligase I and III is unique to ATP-dependent DNA ligases and is not found in the RNA capping enzymes. Similarly to vaccinia virus DNA Ligase, Human DNA Ligase III does not contain the region 2 motif which is present in the capping enzymes, and Human DNA Ligase I (Shuman, S., et al. PNAS USA, in press (1994)). [0031]
  • There is near identity of peptides within the predicted amino acid sequence of the DNA Ligase III cDNA with sequenced tryptic peptides from the 70 kDa bovine DNA Ligase II protein (Wang, Y-C. J., et al., [0032] J. Biol. Chem., 269:31923-31928 (1994)). These tryptic peptides span the region between the active site and the conserved DNA Ligase-specific motif, and are also highly homologous to the corresponding region of the vaccinia virus DNA ligase. The sequence 411-(K) CPNGMFSEIKYDGERVQVH (K)-431 (SEQ ID NO:9) in the predicted amino acid sequence of the DNA ligase III cDNA, with Lys421 the putative active lysine, is identical to the active site tryptic peptide identified in the purified bovine DNA Ligase II protein and different from that of DNA Ligase I (Tomkinson, A. E., et al., PNAS USA, 88:400-404 (1991)).
  • The polynucleotide 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 polypeptide may be identical to the coding sequence shown in FIGS. [0033] 1A-1J (SEQ ID NO:1) or that of the deposited clone 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 polypeptide as the DNA of FIGS. 1A-1J (SEQ ID NO:1) or the deposited cDNA.
  • The polynucleotide which encodes for the mature polypeptide of FIGS. [0034] 1A-1J (SEQ ID NO:2) or for the mature polypeptide encoded by the deposited cDNA may include: only the coding sequence for the mature polypeptide; 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 polypeptide.
  • Thus, the term “polynucleotide encoding a polypeptide” encompasses a polynucleotide which includes only coding sequence for the polypeptide as well as a polynucleotide which includes additional coding and/or non-coding sequence. [0035]
  • The present invention further relates to variants of the hereinabove described polynucleotides which encode for fragments, analogs and derivatives of the polypeptide having the deduced amino acid sequence of FIGS. [0036] 1A-1J (SEQ ID NO:2) or the polypeptide encoded by the cDNA of the deposited clone. The variant of the polynucleotide may be a naturally occurring allelic variant of the polynucleotide or a non-naturally occurring variant of the polynucleotide.
  • Thus, the present invention includes polynucleotides encoding the same mature polypeptide as shown in FIGS. [0037] 1A-1J (SEQ ID NO:2) or the same mature polypeptide encoded by the cDNA of the deposited clone as well as variants of such polynucleotides which variants encode for a fragment, derivative or analog of the polypeptide of FIGS. 1A-1J (SEQ ID NO:2) or the polypeptide encoded by the cDNA of the deposited clone. Such nucleotide variants include deletion variants, substitution variants and addition or insertion variants.
  • As hereinabove indicated, the polynucleotide may have a coding sequence which is a naturally occurring allelic variant of the coding sequence shown in FIGS. [0038] 1A-1J (SEQ ID NO:1) or of the coding sequence of the deposited clone. 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.
  • 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 polypeptide 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 polypeptide 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., [0039] Cell, 37:767 (1984)).
  • 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). [0040]
  • 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 promoter 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. [0041]
  • 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 encoded by the cDNAs of FIGS. [0042] 1A-1J (SEQ ID NO:1) or the deposited cDNA(s).
  • Alternatively, the polynucleotide may have at least 20 bases, preferably 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 polynucleotide of SEQ ID NO:1, for example, for recovery of the polynucleotide or as a diagnostic probe or as a PCR primer. [0043]
  • 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 as well as fragments thereof, which fragments have at least 30 bases and preferably at least 50 bases and to polypeptides encoded by such polynucleotides. [0044]
  • The deposit(s) referred to herein will be maintained under the terms of the Budapest Treaty on the International Recognition of the Deposit of Micro-organisms for purposes of Patent Procedure. These deposits are provided merely as convenience to those of skill in the art and are not an admission that a deposit is required under 35 U.S.C. §112. The sequence of the polynucleotides contained in the deposited materials, as well as the amino acid sequence of the polypeptides encoded thereby, are incorporated herein by reference and are controlling in the event of any conflict with any description of sequences herein. A license may be required to make, use or sell the deposited materials, and no such license is hereby granted. [0045]
  • The present invention further relates to a DNA Ligase III polypeptide which has the deduced amino acid sequence of FIGS. [0046] 1A-1J (SEQ ID NO:2) or which has the amino acid sequence encoded by the deposited cDNA, as well as fragments, analogs and derivatives of such polypeptide.
  • The terms “fragment,” “derivative” and “analog” when referring to the polypeptide of FIGS. [0047] 1A-1J (SEQ ID NO:2) or that encoded by the deposited cDNA, means a polypeptide which retains essentially the same biological function or activity as such polypeptide.
  • The polypeptide of the present invention may be a recombinant polypeptide, a natural polypeptide or a synthetic polypeptide, preferably a recombinant polypeptide. [0048]
  • The fragment, derivative or analog of the polypeptide of FIGS. [0049] 1A-1J (SEQ ID NO:2) or that encoded by the deposited cDNA 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, which is employed for purification of the mature polypeptide. Such fragments, derivatives and analogs are deemed to be within the scope of those skilled in the art from the teachings herein.
  • The polypeptides and polynucleotides of the present invention are preferably provided in an isolated form, and preferably are purified to homogeneity. [0050]
  • 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). [0051]
  • 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. [0052]
  • The polypeptides of the present invention include 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 polypeptide of SEQ ID NO:2 and more preferably at least 90% similarity (more preferably at least 90% identity) to the polypeptide of SEQ ID NO:2 and still more preferably at least 95% similarity (still more preferably at least 95% identity) to the polypeptide 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. [0053]
  • 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. [0054]
  • 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. [0055]
  • 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. [0056]
  • 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 DNA Ligase III 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. [0057]
  • 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. [0058]
  • 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. [0059]
  • 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 [0060] 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 [0061] 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. [0062]
  • As representative examples of appropriate hosts, there may be mentioned: bacterial cells, such as [0063] 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 (Pharrnacia). Eukaryotic: pWLNEO, pSV2CAT, pOG44, pXT1, pSG (Stratagene) pSVK3, pBPV, pMSG, pSVL (Pharrnacia). However, any other plasmid or vector may be used as long as they are replicable and viable in the host. [0064]
  • 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 lacI, lacZ, T3, T7, gpt, lambda P[0065] P, 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)). [0066]
  • The constructs in host cells can be used in a conventional manner to produce the gene product encoded by the recombinant sequence. Alternatively, the polypeptides of the invention can be synthetically produced by conventional peptide synthesizers. [0067]
  • 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. [0068]
  • 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 including 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. [0069]
  • 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 [0070] 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), α-factor, acid phosphatase, or heat shock proteins, among others. The heterologous structural sequence is assembled in appropriate phase with translation, initiation and termination sequences. 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 [0071] 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 GEMI (Promega Biotec, Madison, Wis., USA). These pBR322 “backbone” sections are combined with an appropriate promoter and the structural sequence to be expressed. [0072]
  • 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. [0073]
  • Cells are typically harvested by centrifugation, disrupted by physical or chemical means, and the resulting crude extract retained for further purification. [0074]
  • 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 known to those skilled in the art. [0075]
  • 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 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. [0076]
  • The DNA Ligase II polypeptide 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. [0077]
  • 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. [0078]
  • The DNA Ligase III polypeptides and agonists and antagonists which are polypeptides, described below, may be employed in accordance with the present invention by expression of such polypeptides in vivo, which is often referred to as “gene therapy.”[0079]
  • 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. [0080]
  • 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. [0081]
  • 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. [0082]
  • 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., [0083] 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 III, and β-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 ApoAI 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 β-actin promoter; and human growth hormone promoters. The promoter also may be the native promoter which controls the gene encoding the polypeptide. [0084]
  • 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, φ-2, φ-AM, PA12, T19-14X, VT-19-17-H2, φCRE, φCRIP, GP+E-86, GP+envAm12, and DAN cell lines as described in Miller, [0085] 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. hi 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. [0086]
  • Once the DNA Ligase III polypeptide is being expressed intracellularly via gene therapy, it may be used to repair single-strand breaks in DNA which result from DNA-damaging agents, e.g., UV radiation. Several human syndromes result from autosomal recessive inheritance for the DNA ligase gene. These syndromes cause severe immunodeficiency and greatly increases the susceptibility of abnormal cellular differentiation due to the disrepair of DNA while at the cellular level they are characterized by chromosome instability and hypersensitivity to DNA-damaging agents. These syndromes include Fanconi's anemia and Blackfan-diamond anemia. [0087]
  • The polypeptide of the present invention may also be employed to treat severe immunosuppression which is the result of a defect in the DNA Ligase III gene. DNA Ligase III may also be employed to treat stunted growth and lymphoma which result from defective rejoining of DNA. [0088]
  • Chromosome abnormalities in the 17q11-12 region, to which the DNA Ligase III gene has been mapped, are associated with several diseases including several neoplasias. The most common neoplastic chromosomal abnormality in this region is a translocation between chromosomes 15 and 17 seen in acute myeloid leukemia subtype m3 which involves the disruption of the retinoic acid receptor a gene (Chomienne, H., et al., [0089] Nature, 347:558-561 (1990)). However, chromosomal abnormalities in this region are frequently reported in both acute myeloid and lymphoblastic leukemias and are seen sporadically in several other cancers (Mitelman, F., Catalog of Chromosome Aberrations in Cancer (Fourth Edition), Wiley Liss, New York (1991)). Accordingly, the DNA Ligase III gene and gene product may be employed to treat these neoplasias.
  • Fragments of the full length Ligase III gene may be used as a hybridization probe for a cDNA library to isolate other genes which have a high sequence similarity to the DNA Ligase III gene or have similar biological activity. Probes of this type have at least 20 bases. Preferably, however, the probes 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 DNA Ligase III gene including regulatory and promoter regions, exons, and introns. [0090]
  • An example of a screen comprises isolating the coding region of the DNA Ligase III 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. [0091]
  • The polypeptide and/or polynucleotide of the present invention may also be employed in relation to scientific research, synthesis of DNA and for the manufacture of DNA vectors. The polypeptide and/or polynucleotide of the present invention may be sold into the research market. Thus, for example DNA Ligase III may be used for ligation of DNA sequences in vitro in a manner similar to other DNA ligase enzymes of the art. [0092]
  • This invention also provides a method of screening compounds to identify those which enhance or inhibit the DNA joining reaction catalyzed by human DNA Ligase III. An example of such a method comprises combining ATP, DNA Ligase III and DNA having single-strand breaks with the compound under conditions where the DNA Ligase would normally cleave ATP to AMP and the AMP is transferred to the 5′ phosphate terminus of a single strand break in double-stranded DNA to generate a covalent DNA-AMP complex with the single strand break being subsequently repaired. The DNA having the single-strand breaks may be supplied in the above example by mutant cells which are deficient in proteins that are responsible for strand break repair, for example, mutant rodent cells deficient in XRCC1 and the cdc9 [0093] S. cerevisiae DNA ligase mutant. The ability of the compound to enhance or block the catalysis of this reaction could then be measured to determine if the compound is an effective agonist or antagonist.
  • Human DNA Ligase III is produced and functions intracellularly, therefore, any antagonist must be intra-cellular. Potential antagonists to human DNA Ligase III include antibodies which are produced intracellularly. For example, an antibody identified as antagonizing DNA Ligase III may be produced intracellularly as a single chain antibody by procedures known in the art, such as transforming the appropriate cells with DNA encoding the single chain antibody to prevent the function of human DNA Ligase III. [0094]
  • Another potential antagonist is an antisense construct prepared using antisense technology. 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 for 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 DNA Ligase III. The antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into the DNA Ligase III (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 DNA Ligase III. [0095]
  • Yet another potential antagonist includes a mutated form, or mutein, of DNA Ligase III which recognizes DNA but does not repair single-strand breaks and, therefore, acts to prevent human DNA Ligase III from functioning. [0096]
  • The antagonists may be employed to target undesired cells, e.g., cancer cells and leukemic cells, since the prevention of DNA Ligase III prevents repair of single-strand breaks in DNA and will eventually result in death of the cell. [0097]
  • The small molecule agonists and antagonists of the present invention may be employed in combination with a suitable pharmaceutical carrier. Such compositions comprise a therapeutically effective amount of the molecule 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. [0098]
  • 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 pharmaceutical compositions of the present invention may be employed in conjunction with other therapeutic compounds. [0099]
  • The pharmaceutical compositions may be administered in a convenient manner such as by the oral, topical, intravenous, intraperitoneal, intramuscular, 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, they are 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. [0100]
  • This invention also provides the use of the human DNA Ligase III gene as a diagnostic. For example, some diseases result from inherited defective genes. These genes can be detected by comparing the sequence of the defective gene with that of a normal one. That is, a mutant gene would be associated with hypersensitivity to DNA-damaging agents and an elevated susceptibility to abnormal cell growth, for example, tumors, leukemia and cancer. [0101]
  • Individuals carrying mutations in the human DNA Ligase III gene may be detected at the DNA level by a variety of techniques. Nucleic acids used 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., [0102] Nature, 324:163-166 (1986)) prior to analysis. RNA or cDNA may also be used for the same purpose. Deletions or 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 DNA Ligase III RNA or alternatively, radiolabeled DNA Ligase III antisense DNA sequences. Perfectly matched sequences can be distinguished from mismatched duplexes by RNase A digestion or by differences in melting temperatures.
  • 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., [0103] Science, 230:1242 (1985)).
  • Sequence changes at specific locations may also be revealed by nuclease protection assays, such as RNase protection and S1 protection or the chemical cleavage method (e.g., Cotton et al., [0104] PNAS, USA, 85:4397-4401 (1985)).
  • 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, and Southern blotting of genomic DNA. Also, mutations may be detected by in situ analysis. [0105]
  • In addition, some diseases are a result of, or are characterized by, changes in gene expression which can be detected by changes in the mRNA. Alternatively, the DNA Ligase III gene can be used as a reference to identify individuals expressing a decreased level of DNA Ligase III protein, e.g., by Northern blotting. [0106]
  • 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. [0107]
  • 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. [0108]
  • 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. [0109]
  • Fluorescence in situ hybridization (FISH) of a cDNA clone 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 example, 2,000 bp is good, 4,000 is better, and more than 4,000 is probably not necessary to get good results a reasonable percentage of the time. For a review of this technique, see Verna et al., Human Chromosomes: a Manual of Basic Techniques, Pergamon Press, New York (1988). [0110]
  • Detailed analysis of 19 individual chromosomes using a combination of fractional length measurements and fluorescent binding combined with high-resolution image analysis indicated that Human DNA Ligase III is located within bands 17q11.2-12. [0111]
  • 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). The gene of the present invention has been mapped to chromosome 13q33-34. [0112]
  • 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. [0113]
  • 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). [0114]
  • 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. [0115]
  • 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. hi 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. [0116]
  • 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). [0117]
  • 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. [0118]
  • 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. [0119]
  • In order to facilitate understanding of the following examples certain frequently occurring methods and/or terms will be described. [0120]
  • “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. [0121]
  • “Digestion” of DNA refers to catalytic cleavage of the DNA with a restriction enzymes 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 μg of plasmid or DNA fragment is used with about 2 units of enzyme in about 20 μl of buffer solution. For the purpose of isolating DNA fragments for plasmid construction, typically 5 to 50 μg 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. [0122]
  • 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). [0123]
  • “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. [0124]
  • “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 of T4 DNA ligase (“ligase”) per 0.5 μg of approximately equimolar amounts of the DNA fragments to be ligated. [0125]
  • Unless otherwise stated, transformation was performed as described in the method of Graham, F. and Van der Eb, A., Virology, 52:456-457 (1973). [0126]
  • EXAMPLE 1
  • Bacterial Expression and Purification of DNA Ligase III [0127]
  • The DNA sequence encoding DNA Ligase III, ATCC # 97052, is initially amplified using PCR oligonucleotide primers corresponding to the 5′ and 3∝[0128] 0 end sequences of the processed DNA Ligase III gene. The 5′ oligonucleotide primer has the sequence 5′ CGCGGATCCATGGCTGAGCAACGGTTCTG 3′ (SEQ ID NO:3) contains a Bam HI restriction enzyme site (underlined) followed by 20 nucleotides of DNA Ligase III coding sequence starting from the presumed terminal amino acid of the processed protein codon. The 3′ sequence 5′ GCGTCTAGACTAGCAGGGAGCTACCAG 3′ (SEQ ID NO:4) contains complementary sequences to a XbaI site (underlined) and is followed by 18 nucleotides of DNA Ligase III at C-terminal of DNA Ligase III. The restriction enzyme sites correspond to the restriction enzyme sites on the bacterial expression vector pQE-9 (Qiagen, Inc. Chatsworth, Calif.). pQE-9 encodes antibiotic resistance (Ampr), a bacterial origin of replication (ori), an IPTG-regulatable promoter operator (P/O), a ribosome binding site (RBS), a 6-His tag and restriction enzyme sites. pQE-9 is then digested with Bam HI and Pst I. The amplified sequences are ligated into pQE-9 and inserted in frame with the sequence encoding for the histidine tag and the RBS. The ligation mixture is then used to transform E. coli strain M15/rep 4 (Qiagen, Inc.) under the trademark M15/rep 4 by the procedure described in Sambrook, J. et al., Molecular Cloning: A Laboratory Manual, Cold Spring Laboratory Press, (1989). M15/rep4 contains multiple copies of the plasmid pREP4, which expresses the lacI repressor and also confers kanamycin resistance (Kanr). Transformants are identified by their ability to grow on LB plates and ampicillin/kanamycin resistant colonies are selected. Plasmid DNA is isolated and confirmed by restriction analysis. Clones containing the desired constructs are grown overnight (O/N) in liquid culture in LB media supplemented with both Amp (100 ug/ml) and Kan (25 ug/ml). The O/N culture is used to inoculate a large culture at a ratio of 1:100 to 1:250. The cells are grown to an optical density 600 (O.D.600) of between 0.4 and 0.6. IPTG (“Isopropyl-B-D-thiogalacto pyranoside”) is then added to a final concentration of 1 mM. IPTG induces by inactivating the lacI repressor, clearing the P/O leading to increased gene expression. Cells are grown an extra 3 to 4 hours. Cells are then harvested by centrifugation. The cell pellet is solubilized in the chaotropic agent 6 Molar Guanidine HCl. After clarification, solubilized protein extract is purified from this solution by chromatography on a Nickel-Chelate column under conditions that allow for tight binding by proteins containing the 6-His tag (Hochuli, E. et al., J. Chromatography 411:177-184 (1984)) and eluted from the column in 6 molar guanidine HCl pH 5.0 and for the purpose of renaturation adjusted to 3 molar guanidine HCl, 100 mM sodium phosphate, 10 mmolar glutathione (reduced) and 2 mmolar glutathione (oxidized). After incubation in this solution for 12 hours the protein is dialyzed to 10 mmolar sodium phosphate.
  • EXAMPLE 2
  • Cloning and Expression of DNA Ligase III using the Baculovirus Expression System [0129]
  • A DNA sequence encoding full length DNA Ligase III protein, ATCC # 97052, is amplified using PCR oligonucleotide primers corresponding to the 5′ and 3′ sequences of the gene: [0130]
  • The 5′ primer has the sequence 5′ CGCGAATCCATGGCTGAGCAACGGTTCTG 3′ (SEQ ID NO:5) and contains a BamHI restriction enzyme site (in bold) followed first by 20 nucleotides of N-terminal sequence (the initiation codon for translation “ATG” is underlined). [0131]
  • The 3′ primer has the sequence 5′ GCGTCTAGACTAGCAGGGAGCTACCAG 3′ (SEQ ID NO:6) and contains the cleavage site for the restriction endonuclease XbaI (in bold) and 18 nucleotides complementary to the C-terminal sequence of the DNA Ligase III gene. The amplified sequences were isolated from a 1% agarose gel using a commercially available kit (“Geneclean,” BIO 101 Inc., La Jolla, Calif.). The fragment was then digested with the endonucleases BamHI and XbaI and then purified again on a 1% agarose gel. This fragment is designated F2. [0132]
  • The vector pA2 (modification of pVL941 vector, discussed below) is used for the expression of the DNA Ligase II protein using the baculovirus expression system (for review see: Summers, M. D. and Smith, G. E. 1987, A manual of methods for baculovirus vectors and insect cell culture procedures, Texas Agricultural Experimental Station Bulletin No. 1555). This expression vector contains the strong polyhedrin promoter of the [0133] Autographa califormica nuclear polyhidrosis virus (AcMNPV) followed by the recognition sites for the restriction endonucleases BamHI and XbaI. The polyadenylation site of the simian virus (SV)40 is used for efficient polyadenylation. For an easy selection of recombinant viruses the beta-galactosidase gene from E. coli is inserted in the same orientation as the polyhedrin promoter followed by the polyadenylation signal of the polyhedrin gene. The polyhedrin sequences are flanked at both sides by viral sequences for the cell-mediated homologous recombination of co-transfected wild-type viral DNA. Many other baculovirus vectors could be used in place of pRG1 such as pAc373, pVL941 and pAcIM1 (Luckow, V. A. and Summers, M. D., Virology, 170:31-39).
  • The plasmid is digested with the restriction enzymes BamHI and XbaI and then dephosphorylated using calf intestinal phosphatase by procedures known in the art. The DNA is then isolated from a 1% agarose gel using the commercially available kit (“Geneclean” BIO 101 Inc., La Jolla, Calif.). This vector DNA is designated V2. [0134]
  • Fragment F2 and the dephosphorylated plasmid V2 were ligated with T4 DNA ligase. [0135] E. coli HB101 cells are then transformed and bacteria identified that contained the plasmid (pBac DNA Ligase III) with the DNA Ligase III gene using the enzymes BamHI and XbaI. The sequence of the cloned fragment is confirmed by DNA sequencing.
  • 5 μg of the plasmid pBac DNA Ligase III was co-transfected with 1.0 μg of a commercially available linearized baculovirus (“BaculoGold baculovirus DNA”, Pharmingen, San Diego, Calif.) using the lipofection method (Felgner et al. Proc. Natl. Acad. Sci. USA, 84:7413-7417 (1987)). [0136]
  • 1 μg of BaculoGold™ virus DNA and 5 μg of the plasmid pBac DNA Ligase III are mixed in a sterile well of a microtiter plate containing 50 μl of serum free Grace's medium (Life Technologies Inc., Gaithersburg, Md.). Afterwards 10 μl Lipofectin plus 90 μl Grace's medium are added, mixed and incubated for 15 minutes at room temperature. Then the transfection mixture is added drop-wise to the Sf9 insect cells (ATCC CRL 1711) seeded in a 35 mm tissue culture plate with 1 ml Grace's medium without serum. The plate is rocked back and forth to mix the newly added solution. The plate is then incubated for 5 hours at 27° C. After 5 hours the transfection solution is removed from the plate and 1 ml of Grace's insect medium supplemented with 10% fetal calf serum is added. The plate is put back into an incubator and cultivation continued at 27° C. for four days. [0137]
  • After four days the supematant is collected and a plaque assay performed similar as described by Summers and Smith (supra). As a modification an agarose gel with “Blue Gal” (Life Technologies Inc., Gaithersburg) is used which allows an easy isolation of blue stained plaques. (A detailed description of a “plaque assay” can also be found in the user's guide for insect cell culture and baculovirology distributed by Life Technologies Inc., Gaithersburg, page 9-10). [0138]
  • Four days after the serial dilution, the viruses are added to the cells and blue stained plaques are picked with the tip of an Eppendorf pipette. The agar containing the recombinant viruses is then resuspended in an Eppendorf tube containing 200 μl of Grace's medium. The agar is removed by a brief centrifugation and the supernatant containing the recombinant baculovirus is used to infect Sf9 cells seeded in 35 mm dishes. Four days later the supernatants of these culture dishes are harvested and then stored at 4° C. [0139]
  • Sf9 cells are grown in Grace's medium supplemented with 10% heat-inactivated FBS. The cells are infected with the recombinant baculovirus V-DNA Ligase III at a multiplicity of infection (MOI) of 2. Six hours later the medium is removed and replaced with SF900 III medium minus methionine and cysteine (Life Technologies Inc., Gaithersburg). 42 hours later 5 μCi of [0140] 35S-methionine and 5 μCi 35S cysteine (Amersham) are added. The cells are further incubated for 16 hours before they are harvested by centrifugation and the labeled proteins visualized by SDS-PAGE and autoradiography.
  • EXAMPLE 3
  • Expression of Recombinant DNA Ligase III in COS Cells [0141]
  • The expression of plasmid, DNA Ligase III HA is derived from a vector pcDNAI/Amp (Invitrogen) containing: 1) SV40 origin of replication, 2) ampicillin resistance gene, 3) [0142] E. coli replication origin, 4) CMV promoter followed by a polylinker region, a SV40 intron and polyadenylation site. A DNA fragment encoding the entire DNA Ligase III precursor and a HA tag fused in frame to its 3′ end was cloned into the polylinker region of the vector, therefore, the recombinant protein expression is directed under the CMV promoter. The HA tag correspond to an epitope derived from the influenza hemagglutinin protein as previously described (I. Wilson, H. Niman, R. Heighten, A Cherenson, M. Connolly, and R. Lerner, Cell 37:767 (1984)). The infusion of HA tag to the target protein allows detection of the recombinant protein with an antibody that recognizes the HA epitope.
  • The plasmid construction strategy is described as follows: [0143]
  • The DNA sequence encoding DNA Ligase III, ATCC # 97052, is constructed by PCR using two primers: the 5′ primer 5′ CGC{overscore (GAATCC)}ATGGCTGAGCAACGGTTCTG 3′ (SEQ ID NO:7) contains an BamHI site (underlined) followed by 20 nucleotides of DNA Ligase III coding sequence starting from the initiation codon; the 3′ sequence 5′ GCG{overscore (TCTAGA)}TCAAGCGTAGTCTGGGACGTCGTATGGGTAGCAGGGAGCTACCA GTC 3′ (SEQ ID NO:8) contains complementary sequences to an Xbal site (underlined), translation stop codon, HA tag and the last 17 nucleotides of the DNA Ligase III coding sequence (not including the stop codon). Therefore, the PCR product contains an BamHI site, DNA Ligase III coding sequence followed by HA tag fused in frame, a translation termination stop codon next to the HA tag, and an XbaI site. The PCR amplified DNA fragment and the vector, pcDNAI/Amp, are digested with BamHI and XbaI restriction enzyme and ligated. The ligation mixture is transformed into [0144] E. coli strain SURE (Stratagene Cloning Systems, La Jolla, Calif.) the transformed culture is plated on ampicillin media plates and resistant colonies are selected. Plasmid DNA is isolated from transformants and examined by restriction analysis for the presence of the correct fragment. For expression of the recombinant DNA Ligase III, COS cells are transfected with the expression vector by DEAE-DEXTRAN method (J. Sambrook, E. Fritsch, T. Maniatis, Molecular Cloning: A Laboratory Manual, Cold Spring Laboratory Press, (1989)). The expression of the DNA Ligase III HA protein is detected by radiolabeling and immunoprecipitation method (E. Harlow, D. Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, (1988)). Cells are labeled for 8 hours with 35S-cysteine two days post transfection. Culture media are then collected and cells are lysed with detergent (RIPA buffer (150 mM NaCl, 0.1% SDS, 1% NP-40, 0.5% DOC, 50 mM Tris, pH 7.5) (Wilson, I. et al., Id. 37:767 (1984)). Both cell lysate and culture media are precipitated with a HA specific monoclonal antibody. Proteins precipitated are analyzed on 15% SDS-PAGE gels.
  • EXAMPLE 4
  • Expression Pattern of DNA Ligase III in Human Tissue [0145]
  • Northern blot analysis may be performed to examine the levels of expression of DNA Ligase III in human tissues. Total cellular RNA samples are isolated with RNAzol™ B system (Biotecx Laboratories, Inc. Houston, Tex.) About 15 μg of total RNA isolated from each human tissue specified is separated on 1% agarose gel and blotted onto a nylon filter (Sambrook, Fritsch, and Maniatis, Molecular Cloning, Cold Spring Harbor Press, (1989)). The labeling reaction is done according to the Stratagene Prime-It kit with 50 ng DNA fragment. The labeled DNA is purified with a Select-G-50 column (5 Prime-3 Prime, Inc. Boulder, Colo.). The filter containing the particular RNA blot is then hybridized with radioactive labeled full length DNA Ligase III gene at 1,000,000 cpm/ml in 0.5 M NaPO[0146] 4, pH 7.4 and 7% SDS overnight at 65° C. After wash twice at room temperature and twice at 60° C. with 0.5×SSC, 0.1% SDS, the filter is then exposed at −70° C. overnight with an intensifying screen. The message RNA for DNA Ligase II is abundant in the testis, prostate, heart, and thymus.
  • EXAMPLE 5
  • In vitro Transcription/Translation of cDNA Clones [0147]
  • Putative full-length cDNA clone was subcloned as follows: DNA ligase III was subcloned as a Sal I/Not I restriction fragment into the multiple cloning sire of pSPORT (Life Technologies), with the 5′ end proximal to the T7 promoter; the DNA ligase III plasmid constructs (1 μg) was linearized with either Not I or Xho I (New England Biolabs), downstream of the cDNA insert, then transcribed and capped at 36° C. for 30 minutes with T7 polymerase and the mCAP RNA capping kit (Stratagene). The reactions were terminated by incubation with 10 units RNase-free DNase at 37° C. for 5 minutes. Following phenol/chloroform extraction and ethanol precipitation, the in vitro transcription products were resuspended in 20 [0148] μl 10 mM Tris-HCl/1 mM EDTA, pH 8.0 (TE). The transcript (0 to 5 μl, made up to a final volume of 5 μl with water) was translated in 20 μl rabbit reticulocyte lysate (Amersham) at 30° C. for 90 minutes. In order to radiolabel the product of in vitro translation, reaction was supplemented with 20 μCi [35S]methionine (3000 Ci mmol−1, Amersham). Translations were terminated by incubation with 5 μl of 400 ml−1 RNase A/50 mM EDTA at 37° C. for 15 minutes (30 μl final volume). Samples (5 μl) of translations carried out in the presence of [35S]methionine were analyzed by electrophoresis in SDS-7.5% polyacrylamide gels and autoradiography. Non-radiolabeled translation products were assayed for ability to form protein-adenylate complexes after removal of ATP by chromatography through spun 1 ml columns of Sephadex G50 (Pharmacia) equilibrated with TE.
  • EXAMPLE 6
  • DNA Ligase Assays [0149]
  • 5 μl samples from in vitro translations were adenylylated in reaction mixtures (30 μl) containing 60 mM Tris HCl (pH 8.0), 10 mM MgCl[0150] 2, 50 μg ml−1 BSA, 5 mM DTT and 1 μCi [α-32P] ATP (3000 Ci mmol−1, Amersham) at 20° C. for 10 minutes and then analyzed by electrophoresis in SDS-7.5% polyacrylamide gels and autoradiography. In order to monitor transfer of [32P]AMP from protein-adenylate to a nicked DNA substrate, 5 μl samples from adenylylation reactions were incubated for further time periods with or without the addition of 500 ng non-radiolabeled oligo(dT) 16-poly(dA), as described previously. The ability to transfer [32P]AMP from enzyme-adenylate to the hybrid substrates, oligo(dT)-poly(rA) or oligo(rA)-poly(dT), differentiates DNA ligase I, II and III. However, both these latter substrates were rapidly degraded by an RNase H activity upon incubation in the reticulocyte lysate, even when mixtures were used directly without termination of translation reactions by addition of RNase A.
  • EXAMPLE 7
  • Expression of DNA Ligase III via Gene Therapy [0151]
  • Fibroblasts are obtained from a subject by skin biopsy. The resulting tissue is placed in tissue-culture medium and separated into small pieces. Small chunks of the tissue are placed on a wet surface of a tissue culture flask, approximately ten pieces are placed in each flask. The flask is turned upside down, closed tight and left at room temperature over night. After 24 hours at room temperature, the flask is inverted and the chunks of tissue remain fixed to the bottom of the flask and fresh media (e.g., Ham's F12 media, with 10% FBS, penicillin and streptomycin, is added. This is then incubated at 37° C. for approximately one week. At this time, fresh media is added and subsequently changed every several days. After an additional two weeks in culture, a monolayer of fibroblasts emerge. The monolayer is trypsinized and scaled into larger flasks. [0152]
  • Moloney murine leukemia virus is digested and treated with calf intestinal phosphatase. The linear vector is fractionated on agarose gel and purified, using glass beads. The DNA Ligase III cDNA (see FIGS. [0153] 1A-1J) is isolated and the ends of this fragment are treated with DNA polymerase in order to fill in the recessed ends and create blunt ends.
  • Equal quantities of the Moloney murine leukemia virus linear backbone and the gene are added together, in the presence of T4 DNA ligase. The resulting mixture is maintained under conditions appropriate for ligation of the two fragments. The ligation mixture was used to transform bacteria HB101, which were then plated onto agar-containing kanamycin for the purpose of confirming that the vector had the DNA Ligase III gene properly inserted. [0154]
  • PE501 packaging cells are grown in tissue culture to confluent density in Dulbecco's Modified Eagles Medium (DMEM) with 10% calf serum (CS), penicillin and streptomycin. The Moloney murine leukemia virus vector containing the gene is then added to the media and the packaging cells are transduced with the vector. The packaging cells now produce infectious viral particles containing the DNA Ligase III gene. [0155]
  • Fresh media is added to the transduced producer cells, and subsequently the media is harvested from a 10 cm plate of confluent producer cells. The spent media, containing the infectious viral particles, is filtered through a millipore filter to remove detached producer cells and this media is then used to infect fibroblast cells. Media is removed from a sub-confluent plate of fibroblasts and quickly replaced with the media from the producer cells. [0156]
  • The engineered fibroblasts are then injected into the into a host, for example, a rat, either alone or after having been grown to confluence on cytodex 3 microcarrier beads. The fibroblasts now produce the protein product and the biological actions of DNA Ligase III are conveyed to the host. [0157]
  • 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, the invention may be practiced otherwise than as particularly described. [0158]
  • 1 11 1 3417 DNA Homo sapiens CDS (334)..(3102) 1 ccacgcgtcc ggcagcctgt atgagcaagt gccgaggcct acggtgagcg ccggagccgg 60 agaggcagct atatgtcttt ggctttcaag atcttctttc cacaaaccct ccgtgcactc 120 agccgaaaag aactgtgcct attccgaaaa catcactggc gtgatgtaag acaattcagc 180 cagtggtcag aaacagatct gcttcatgga catcccctct tcctgagaag aaagcctgtt 240 ctatcattcc agggaagcca tctaagatca cgtgccacct accttgtttt cttgccaggg 300 ttgcatgtgg gactctgcag tggcccctgt gag atg gct gag caa cgg ttc tgt 354 Met Ala Glu Gln Arg Phe Cys 1 5 gtg gac tat gcc aag cgt ggc aca gct ggc tgc aaa aaa tgc aag gaa 402 Val Asp Tyr Ala Lys Arg Gly Thr Ala Gly Cys Lys Lys Cys Lys Glu 10 15 20 aag att gtg aag ggc gta tgc cga att ggc aaa gtg gtg ccc aat ccc 450 Lys Ile Val Lys Gly Val Cys Arg Ile Gly Lys Val Val Pro Asn Pro 25 30 35 ttc tca gag tct ggg ggt gat atg aaa gag tgg tac cac att aaa tgc 498 Phe Ser Glu Ser Gly Gly Asp Met Lys Glu Trp Tyr His Ile Lys Cys 40 45 50 55 atg ttt gag aaa cta gag cgg gcc cgg gcc acc aca aaa aaa atc gag 546 Met Phe Glu Lys Leu Glu Arg Ala Arg Ala Thr Thr Lys Lys Ile Glu 60 65 70 gac ctc aca gag ctg gaa ggc tgg gaa gag ctg gaa gat aat gag aag 594 Asp Leu Thr Glu Leu Glu Gly Trp Glu Glu Leu Glu Asp Asn Glu Lys 75 80 85 gaa cag ata acc cag cac att gca gat ctg tct tct aag gca gca ggt 642 Glu Gln Ile Thr Gln His Ile Ala Asp Leu Ser Ser Lys Ala Ala Gly 90 95 100 aca cca aag aag aaa gct gtt gtc cag gct aag ttg aca acc act ggc 690 Thr Pro Lys Lys Lys Ala Val Val Gln Ala Lys Leu Thr Thr Thr Gly 105 110 115 cag gtg act tct cca gtg aaa ggc gcc tca ttt gtc acc agt acc aat 738 Gln Val Thr Ser Pro Val Lys Gly Ala Ser Phe Val Thr Ser Thr Asn 120 125 130 135 ccc cgg aaa ttt tct ggc ttt tca gcc aag ccc aac aac tct ggg gaa 786 Pro Arg Lys Phe Ser Gly Phe Ser Ala Lys Pro Asn Asn Ser Gly Glu 140 145 150 gcc ccc tcg agc ccc acc cct aag aga agt ctg tct tca agc aaa tgt 834 Ala Pro Ser Ser Pro Thr Pro Lys Arg Ser Leu Ser Ser Ser Lys Cys 155 160 165 gac ccc agg cat aag gac tgt ctg cta cgg gag ttt cga aag tta tgc 882 Asp Pro Arg His Lys Asp Cys Leu Leu Arg Glu Phe Arg Lys Leu Cys 170 175 180 gcc atg gtg gcc gat aat cct agc tac aac acg aag acc cag atc atc 930 Ala Met Val Ala Asp Asn Pro Ser Tyr Asn Thr Lys Thr Gln Ile Ile 185 190 195 cag gac ttc ctt cgg aaa ggc tca gca gga gat ggt ttc cac ggt gat 978 Gln Asp Phe Leu Arg Lys Gly Ser Ala Gly Asp Gly Phe His Gly Asp 200 205 210 215 gtg tac cta aca gtg aag ctg ctg ctg cca gga gtc att aag act gtt 1026 Val Tyr Leu Thr Val Lys Leu Leu Leu Pro Gly Val Ile Lys Thr Val 220 225 230 tac aac ttg aac gat aag cag att gtg aag ctt ttc agt cgc att ttt 1074 Tyr Asn Leu Asn Asp Lys Gln Ile Val Lys Leu Phe Ser Arg Ile Phe 235 240 245 aac tgc aac cca gat gat atg gca cgg gac cta gag cag ggt gac gtg 1122 Asn Cys Asn Pro Asp Asp Met Ala Arg Asp Leu Glu Gln Gly Asp Val 250 255 260 tca gag aca atc aga gtc ttc ttt gag cag agc aag tct ttc ccc cca 1170 Ser Glu Thr Ile Arg Val Phe Phe Glu Gln Ser Lys Ser Phe Pro Pro 265 270 275 gct gcc aag agc ctc ctt acc atc cag gaa gtg gat gag ttc ctt ctg 1218 Ala Ala Lys Ser Leu Leu Thr Ile Gln Glu Val Asp Glu Phe Leu Leu 280 285 290 295 cgg ctg tcc aag ctc acc aag gag gat gag cag caa cag gcc cta cag 1266 Arg Leu Ser Lys Leu Thr Lys Glu Asp Glu Gln Gln Gln Ala Leu Gln 300 305 310 gac att gcc tcc agg tgt aca gcc aat gac ctt aaa tgc atc atc agg 1314 Asp Ile Ala Ser Arg Cys Thr Ala Asn Asp Leu Lys Cys Ile Ile Arg 315 320 325 ttg atc aaa cat gat ctg aag atg aac tca ggt gca aaa cat gtg tta 1362 Leu Ile Lys His Asp Leu Lys Met Asn Ser Gly Ala Lys His Val Leu 330 335 340 gac gcc ctt gac ccc aat gcc tat gaa gcc ttc aaa gcc tcg cgc aac 1410 Asp Ala Leu Asp Pro Asn Ala Tyr Glu Ala Phe Lys Ala Ser Arg Asn 345 350 355 ctg cag gat gtg gtg gag cgg gtc ctt cac aac gcg cag gag gtg gag 1458 Leu Gln Asp Val Val Glu Arg Val Leu His Asn Ala Gln Glu Val Glu 360 365 370 375 aag gag ccg ggc cag aga cga gct ctg agc gtc cag gcc tcg ctg atg 1506 Lys Glu Pro Gly Gln Arg Arg Ala Leu Ser Val Gln Ala Ser Leu Met 380 385 390 aca cct gtg cag ccc atg ttg gcg gag gcc tgc aag tcc gtt gag tat 1554 Thr Pro Val Gln Pro Met Leu Ala Glu Ala Cys Lys Ser Val Glu Tyr 395 400 405 gca atg aag aaa tgt ccc aat ggc atg ttc tct gag atc aag tac gat 1602 Ala Met Lys Lys Cys Pro Asn Gly Met Phe Ser Glu Ile Lys Tyr Asp 410 415 420 gga gag cga gtc cag gtg cat aag aat gga gac cac ttc agc tac ttc 1650 Gly Glu Arg Val Gln Val His Lys Asn Gly Asp His Phe Ser Tyr Phe 425 430 435 agc cgc agt ctc aag ccc gtc ctt cct cac aag gtg gcc cac ttt aag 1698 Ser Arg Ser Leu Lys Pro Val Leu Pro His Lys Val Ala His Phe Lys 440 445 450 455 gac tac att ccc cag gct ttt cct ggg ggc cac agc atg atc ttg gat 1746 Asp Tyr Ile Pro Gln Ala Phe Pro Gly Gly His Ser Met Ile Leu Asp 460 465 470 tct gaa gtg ctt ctg att gac aac aag aca ggc aaa cca ctg ccc ttt 1794 Ser Glu Val Leu Leu Ile Asp Asn Lys Thr Gly Lys Pro Leu Pro Phe 475 480 485 ggg act ctg gga gta cac aag aaa gca gcc ttc cag gat gct aat gtc 1842 Gly Thr Leu Gly Val His Lys Lys Ala Ala Phe Gln Asp Ala Asn Val 490 495 500 tgc ctg ttt gtt ttt gat tgt atc tac ttt aat gat gtc agc ttg atg 1890 Cys Leu Phe Val Phe Asp Cys Ile Tyr Phe Asn Asp Val Ser Leu Met 505 510 515 gac aga cct ctg tgt gag cgg cgg aag ttt ctt cat gac aac atg gtt 1938 Asp Arg Pro Leu Cys Glu Arg Arg Lys Phe Leu His Asp Asn Met Val 520 525 530 535 gaa att cca aac cgg atc atg ttc tca gaa atg aag cga gtc aca aaa 1986 Glu Ile Pro Asn Arg Ile Met Phe Ser Glu Met Lys Arg Val Thr Lys 540 545 550 gct ttg gac ttg gct gac atg ata acc cgg gtg atc cag gag gga ttg 2034 Ala Leu Asp Leu Ala Asp Met Ile Thr Arg Val Ile Gln Glu Gly Leu 555 560 565 gag ggg ctg gtg ctg aag gat gtg aag ggt aca tat gag cct ggg aag 2082 Glu Gly Leu Val Leu Lys Asp Val Lys Gly Thr Tyr Glu Pro Gly Lys 570 575 580 cgg cac tgg ctg aaa gtg aag aaa gac tat ttg aac gag ggg gcc atg 2130 Arg His Trp Leu Lys Val Lys Lys Asp Tyr Leu Asn Glu Gly Ala Met 585 590 595 gcc gac aca gct gac ctg gtg gtc ctt gga gcc ttc tat ggg caa ggg 2178 Ala Asp Thr Ala Asp Leu Val Val Leu Gly Ala Phe Tyr Gly Gln Gly 600 605 610 615 agc aaa ggc ggc atg atg tca atc ttc ctc atg ggc tgc tac gac cct 2226 Ser Lys Gly Gly Met Met Ser Ile Phe Leu Met Gly Cys Tyr Asp Pro 620 625 630 ggc agc cag aag tgg tgc aca gtc acc aag tgt gca gga ggc cat gat 2274 Gly Ser Gln Lys Trp Cys Thr Val Thr Lys Cys Ala Gly Gly His Asp 635 640 645 gat gcc acg ctt gcc cgc ctg cag aat gaa cta gac atg gtg aag atc 2322 Asp Ala Thr Leu Ala Arg Leu Gln Asn Glu Leu Asp Met Val Lys Ile 650 655 660 agc aag gac ccc agc aaa ata ccc agc tgg ttg aag gtc aac aag atc 2370 Ser Lys Asp Pro Ser Lys Ile Pro Ser Trp Leu Lys Val Asn Lys Ile 665 670 675 tac tat cct gac ttc atc gtc cca gac cca aag aaa gct gcc gtg tgg 2418 Tyr Tyr Pro Asp Phe Ile Val Pro Asp Pro Lys Lys Ala Ala Val Trp 680 685 690 695 gag atc aca ggg gct gaa ttc tcc aaa tcg gag gct cat aca gct gac 2466 Glu Ile Thr Gly Ala Glu Phe Ser Lys Ser Glu Ala His Thr Ala Asp 700 705 710 ggg atc tcc atc cga ttc cct cgc tgc acc cga atc cga gat gat aag 2514 Gly Ile Ser Ile Arg Phe Pro Arg Cys Thr Arg Ile Arg Asp Asp Lys 715 720 725 gac tgg aaa tct gcc act aac ctt ccc caa ctc aag gaa ctg tac cag 2562 Asp Trp Lys Ser Ala Thr Asn Leu Pro Gln Leu Lys Glu Leu Tyr Gln 730 735 740 ttg tcc aag gag aag gca gac ttc act gta gtg gct gga gat gag ggg 2610 Leu Ser Lys Glu Lys Ala Asp Phe Thr Val Val Ala Gly Asp Glu Gly 745 750 755 agc tcc act aca ggg ggt agc agt gaa gag aat aag ggt ccc tca ggg 2658 Ser Ser Thr Thr Gly Gly Ser Ser Glu Glu Asn Lys Gly Pro Ser Gly 760 765 770 775 tct gct gtg tcc cgc aag gcc ccc agc aag ccc tca gcc agt acc aag 2706 Ser Ala Val Ser Arg Lys Ala Pro Ser Lys Pro Ser Ala Ser Thr Lys 780 785 790 aaa gca gaa ggg aag ctg agt aac tcc aac agc aaa gat ggc aac atg 2754 Lys Ala Glu Gly Lys Leu Ser Asn Ser Asn Ser Lys Asp Gly Asn Met 795 800 805 cag act gca aag cct tcc gct atg aag gtg ggg gag aag ctg gcc aca 2802 Gln Thr Ala Lys Pro Ser Ala Met Lys Val Gly Glu Lys Leu Ala Thr 810 815 820 aag tct tct cca gtg aaa gta ggg gag aag cgg aaa gct gct gat gag 2850 Lys Ser Ser Pro Val Lys Val Gly Glu Lys Arg Lys Ala Ala Asp Glu 825 830 835 acg ctg tgc caa aca aag gta ttg ctg gac atc ttc act ggg gtg cgg 2898 Thr Leu Cys Gln Thr Lys Val Leu Leu Asp Ile Phe Thr Gly Val Arg 840 845 850 855 ctt tac ttg cca ccc tcc aca cca gac ttc agc cgt ctc aga cgc tac 2946 Leu Tyr Leu Pro Pro Ser Thr Pro Asp Phe Ser Arg Leu Arg Arg Tyr 860 865 870 ttt gtg gca ttc gac ggg gac ctg gta cag gaa ttt gat atg act tca 2994 Phe Val Ala Phe Asp Gly Asp Leu Val Gln Glu Phe Asp Met Thr Ser 875 880 885 gcc acg cac gtg ctg ggt agc agg gac aag aac cct gcg gcc cag cag 3042 Ala Thr His Val Leu Gly Ser Arg Asp Lys Asn Pro Ala Ala Gln Gln 890 895 900 gtc tcc cca gag tgg att tgg gca tgt atc cgg aaa cgg aga ctg gta 3090 Val Ser Pro Glu Trp Ile Trp Ala Cys Ile Arg Lys Arg Arg Leu Val 905 910 915 gct ccc tgc tag gtttgctgtc ttccctctcc ctcaggccat actctccttt 3142 Ala Pro Cys 920 accatactat tggactggac tcaggctgga ggcagataga cacagtatag ggggaatggg 3202 cttgcttctc ccaaacccac cagttctcca ctgtctcttc tggaccagga attagttgct 3262 gtgggtgcca cagctgaagt cagtttgtct tgctggttta aatagatctt tcagagctgg 3322 gtgctgggtt tgccatcttt ttgttttctt tgaaaagcag cttagttacc ctttttataa 3382 ataaaatatc ttgcagttaa aaaaaaaaaa aaaaa 3417 2 922 PRT Homo sapiens ACT_SITE (421)..(421) Active site lysine 2 Met Ala Glu Gln Arg Phe Cys Val Asp Tyr Ala Lys Arg Gly Thr Ala 1 5 10 15 Gly Cys Lys Lys Cys Lys Glu Lys Ile Val Lys Gly Val Cys Arg Ile 20 25 30 Gly Lys Val Val Pro Asn Pro Phe Ser Glu Ser Gly Gly Asp Met Lys 35 40 45 Glu Trp Tyr His Ile Lys Cys Met Phe Glu Lys Leu Glu Arg Ala Arg 50 55 60 Ala Thr Thr Lys Lys Ile Glu Asp Leu Thr Glu Leu Glu Gly Trp Glu 65 70 75 80 Glu Leu Glu Asp Asn Glu Lys Glu Gln Ile Thr Gln His Ile Ala Asp 85 90 95 Leu Ser Ser Lys Ala Ala Gly Thr Pro Lys Lys Lys Ala Val Val Gln 100 105 110 Ala Lys Leu Thr Thr Thr Gly Gln Val Thr Ser Pro Val Lys Gly Ala 115 120 125 Ser Phe Val Thr Ser Thr Asn Pro Arg Lys Phe Ser Gly Phe Ser Ala 130 135 140 Lys Pro Asn Asn Ser Gly Glu Ala Pro Ser Ser Pro Thr Pro Lys Arg 145 150 155 160 Ser Leu Ser Ser Ser Lys Cys Asp Pro Arg His Lys Asp Cys Leu Leu 165 170 175 Arg Glu Phe Arg Lys Leu Cys Ala Met Val Ala Asp Asn Pro Ser Tyr 180 185 190 Asn Thr Lys Thr Gln Ile Ile Gln Asp Phe Leu Arg Lys Gly Ser Ala 195 200 205 Gly Asp Gly Phe His Gly Asp Val Tyr Leu Thr Val Lys Leu Leu Leu 210 215 220 Pro Gly Val Ile Lys Thr Val Tyr Asn Leu Asn Asp Lys Gln Ile Val 225 230 235 240 Lys Leu Phe Ser Arg Ile Phe Asn Cys Asn Pro Asp Asp Met Ala Arg 245 250 255 Asp Leu Glu Gln Gly Asp Val Ser Glu Thr Ile Arg Val Phe Phe Glu 260 265 270 Gln Ser Lys Ser Phe Pro Pro Ala Ala Lys Ser Leu Leu Thr Ile Gln 275 280 285 Glu Val Asp Glu Phe Leu Leu Arg Leu Ser Lys Leu Thr Lys Glu Asp 290 295 300 Glu Gln Gln Gln Ala Leu Gln Asp Ile Ala Ser Arg Cys Thr Ala Asn 305 310 315 320 Asp Leu Lys Cys Ile Ile Arg Leu Ile Lys His Asp Leu Lys Met Asn 325 330 335 Ser Gly Ala Lys His Val Leu Asp Ala Leu Asp Pro Asn Ala Tyr Glu 340 345 350 Ala Phe Lys Ala Ser Arg Asn Leu Gln Asp Val Val Glu Arg Val Leu 355 360 365 His Asn Ala Gln Glu Val Glu Lys Glu Pro Gly Gln Arg Arg Ala Leu 370 375 380 Ser Val Gln Ala Ser Leu Met Thr Pro Val Gln Pro Met Leu Ala Glu 385 390 395 400 Ala Cys Lys Ser Val Glu Tyr Ala Met Lys Lys Cys Pro Asn Gly Met 405 410 415 Phe Ser Glu Ile Lys Tyr Asp Gly Glu Arg Val Gln Val His Lys Asn 420 425 430 Gly Asp His Phe Ser Tyr Phe Ser Arg Ser Leu Lys Pro Val Leu Pro 435 440 445 His Lys Val Ala His Phe Lys Asp Tyr Ile Pro Gln Ala Phe Pro Gly 450 455 460 Gly His Ser Met Ile Leu Asp Ser Glu Val Leu Leu Ile Asp Asn Lys 465 470 475 480 Thr Gly Lys Pro Leu Pro Phe Gly Thr Leu Gly Val His Lys Lys Ala 485 490 495 Ala Phe Gln Asp Ala Asn Val Cys Leu Phe Val Phe Asp Cys Ile Tyr 500 505 510 Phe Asn Asp Val Ser Leu Met Asp Arg Pro Leu Cys Glu Arg Arg Lys 515 520 525 Phe Leu His Asp Asn Met Val Glu Ile Pro Asn Arg Ile Met Phe Ser 530 535 540 Glu Met Lys Arg Val Thr Lys Ala Leu Asp Leu Ala Asp Met Ile Thr 545 550 555 560 Arg Val Ile Gln Glu Gly Leu Glu Gly Leu Val Leu Lys Asp Val Lys 565 570 575 Gly Thr Tyr Glu Pro Gly Lys Arg His Trp Leu Lys Val Lys Lys Asp 580 585 590 Tyr Leu Asn Glu Gly Ala Met Ala Asp Thr Ala Asp Leu Val Val Leu 595 600 605 Gly Ala Phe Tyr Gly Gln Gly Ser Lys Gly Gly Met Met Ser Ile Phe 610 615 620 Leu Met Gly Cys Tyr Asp Pro Gly Ser Gln Lys Trp Cys Thr Val Thr 625 630 635 640 Lys Cys Ala Gly Gly His Asp Asp Ala Thr Leu Ala Arg Leu Gln Asn 645 650 655 Glu Leu Asp Met Val Lys Ile Ser Lys Asp Pro Ser Lys Ile Pro Ser 660 665 670 Trp Leu Lys Val Asn Lys Ile Tyr Tyr Pro Asp Phe Ile Val Pro Asp 675 680 685 Pro Lys Lys Ala Ala Val Trp Glu Ile Thr Gly Ala Glu Phe Ser Lys 690 695 700 Ser Glu Ala His Thr Ala Asp Gly Ile Ser Ile Arg Phe Pro Arg Cys 705 710 715 720 Thr Arg Ile Arg Asp Asp Lys Asp Trp Lys Ser Ala Thr Asn Leu Pro 725 730 735 Gln Leu Lys Glu Leu Tyr Gln Leu Ser Lys Glu Lys Ala Asp Phe Thr 740 745 750 Val Val Ala Gly Asp Glu Gly Ser Ser Thr Thr Gly Gly Ser Ser Glu 755 760 765 Glu Asn Lys Gly Pro Ser Gly Ser Ala Val Ser Arg Lys Ala Pro Ser 770 775 780 Lys Pro Ser Ala Ser Thr Lys Lys Ala Glu Gly Lys Leu Ser Asn Ser 785 790 795 800 Asn Ser Lys Asp Gly Asn Met Gln Thr Ala Lys Pro Ser Ala Met Lys 805 810 815 Val Gly Glu Lys Leu Ala Thr Lys Ser Ser Pro Val Lys Val Gly Glu 820 825 830 Lys Arg Lys Ala Ala Asp Glu Thr Leu Cys Gln Thr Lys Val Leu Leu 835 840 845 Asp Ile Phe Thr Gly Val Arg Leu Tyr Leu Pro Pro Ser Thr Pro Asp 850 855 860 Phe Ser Arg Leu Arg Arg Tyr Phe Val Ala Phe Asp Gly Asp Leu Val 865 870 875 880 Gln Glu Phe Asp Met Thr Ser Ala Thr His Val Leu Gly Ser Arg Asp 885 890 895 Lys Asn Pro Ala Ala Gln Gln Val Ser Pro Glu Trp Ile Trp Ala Cys 900 905 910 Ile Arg Lys Arg Arg Leu Val Ala Pro Cys 915 920 3 29 DNA Artificial Sequence Contains a Bam HI restriction enzyme site 3 cgcggatcca tggctgagca acggttctg 29 4 27 DNA Artificial Sequence Contains complementary sequences to a XbaI site 4 gcgtctagac tagcagggag ctaccag 27 5 29 DNA Artificial Sequence Contains a BamHI restriction enzyme site 5 cgcgaatcca tggctgagca acggttctg 29 6 29 DNA Artificial Sequence Contains the cleavage site for the restriction endonuclease XbaI 6 cgcgaatcca tggctgagca acggttctg 29 7 29 DNA Artificial Sequence Contains a BamHI site 7 cgcgaatcca tggctgagca acggttctg 29 8 56 DNA Artificial Sequence Contains complementary sequences to an XbaI site (underlined), translation stop codon, and an HA tag 8 gcgtctagat caagcgtagt ctgggacgtc gtatgggtag cagggagcta ccagtc 56 9 21 PRT Homo sapiens 9 Lys Cys Pro Asn Gly Met Phe Ser Glu Ile Lys Tyr Asp Gly Glu Arg 1 5 10 15 Val Gln Val His Lys 20 10 8 PRT Homo sapiens MISC_FEATURE (1)..(1) Xaa is any amino acid 10 Xaa Lys Xaa Asp Gly Xaa Arg Xaa 1 5 11 8 PRT Homo sapiens MISC_FEATURE (2)..(2) Xaa is any amino acid 11 Glu Xaa Lys Tyr Asp Gly Xaa Arg 1 5

Claims (49)

What is claimed is:
1. An antibody or portion thereof that specifically binds to a protein whose sequence consists of an amino acid sequence selected from the group consisting of:
(a) amino acids 1 to 922 of SEQ ID NO:2;
(b) amino acids 2 to 922 of SEQ ID NO:2;
(c) an antigenic fragment of the amino acid sequence of SEQ ID NO:2;
(d) amino acids 1 to 922 of SEQ ID NO:2, wherein the amino acid sequence has one to five conservative substitutions;
(e) at least 30 contiguous amino acid residues of SEQ ID NO:2; and
(f) at least 50 contiguous amino acid residues of SEQ ID NO:2.
2. The antibody or portion thereof of
claim 1
, wherein the amino acid sequence consists of amino acid sequence (a).
3. The antibody or portion thereof of
claim 1
, wherein the amino acid sequence consists of amino acid sequence (b).
4. The antibody or portion thereof of
claim 1
, wherein the amino acid sequence consists of amino acid sequence (c).
5. The antibody or portion thereof of
claim 1
, wherein the amino acid sequence consists of amino acid sequence (d).
6. The antibody or portion thereof of
claim 1
, wherein the amino acid sequence consists of amino acid sequence (e).
7. The antibody or portion thereof of
claim 1
, wherein the amino acid sequence consists of amino acid sequence (f).
8. The antibody or portion thereof of
claim 1
which is a monoclonal antibody.
9. The antibody or portion thereof of
claim 1
which is a polyclonal antibody.
10. The antibody or portion thereof of
claim 1
which is a chimeric antibody.
11. The antibody or portion thereof of
claim 1
which is a humanized antibody.
12. The antibody or portion thereof of
claim 1
which is a single chain antibody.
13. The antibody or portion thereof of
claim 1
which is an Fab fragment.
14. A pharmaceutical composition comprising the antibody or portion thereof of
claim 1
and a pharmaceutically acceptable carrier.
15. The pharmaceutical composition of
claim 14
, wherein the antibody or portion thereof is a monoclonal antibody.
16. The pharmaceutical composition of
claim 14
, wherein the antibody or portion thereof is a polyclonal antibody.
17. The pharmaceutical composition of
claim 14
, wherein the antibody or portion thereof is a chimeric antibody.
18. The pharmaceutical composition of
claim 14
, wherein the antibody or portion thereof is a humanized antibody.
19. The pharmaceutical composition of
claim 14
, wherein the antibody or portion thereof is a single chain antibody.
20. The pharmaceutical composition of
claim 14
, wherein the antibody or portion thereof is an Fab fragment.
21. The pharmaceutical composition of
claim 14
, wherein the antibody or portion thereof is a humanized monoclonal antibody.
22. A hybridoma cell line that produces a monoclonal antibody or portion thereof that specifically binds to a protein whose sequence consists of an amino acid sequence selected from the group consisting of:
(a) amino acids 1 to 922 of SEQ ID NO:2;
(b) amino acids 2 to 922 of SEQ ID NO:2;
(c) an antigenic fragment of the amino acid sequence of SEQ ID NO:2;
(d) amino acids 1 to 922 of SEQ ID NO:2, wherein the amino acid sequence has one to five conservative substitutions;
(e) at least 30 contiguous amino acid residues of SEQ ID NO:2; and
(f) at least 50 contiguous amino acid residues of SEQ ID NO:2.
23. The hybridoma cell line of
claim 22
, wherein the antibody or portion thereof is humanized.
24. An antibody or portion thereof produced by immunizing an animal with a protein whose sequence consists of an amino acid sequence selected from the group consisting of:
(a) amino acids 1 to 922 of SEQ ID NO:2;
(b) amino acids 2 to 922 of SEQ ID NO:2;
(c) an antigenic fragment of the amino acid sequence of SEQ ID NO:2;
(d) amino acids 1 to 922 of SEQ ID NO:2, wherein the amino acid sequence has one to five conservative substitutions;
(e) at least 30 contiguous amino acid residues of SEQ ID NO:2; and
(f) at least 50 contiguous amino acid residues of SEQ ID NO:2;
wherein said antibody or portion thereof specifically binds to said protein.
25. An antibody or portion thereof that specifically binds to a protein whose sequence consists of an amino acid sequence selected from the group consisting of:
(a) the full-length protein encoded by the human cDNA contained in ATCC Deposit No. 97052;
(b) the full-length protein, excluding the N-terminal methionine, encoded by the human cDNA contained in ATCC Deposit No. 97052;
(c) a mature protein encoded by the human cDNA contained in ATCC Deposit No. 97052;
(d) the full-length protein encoded by the human cDNA contained in ATCC Deposit No. 97052, wherein the amino acid sequence has one to five conservative substitutions;
(e) an antigenic fragment of a protein encoded by the human cDNA contained in ATCC Deposit No. 97052;
(f) at least 30 contiguous amino acid residues of a protein encoded by the human cDNA contained in ATCC Deposit No. 97052; and
(g) at least 50 contiguous amino acid residues of a protein encoded by the human cDNA contained in ATCC Deposit No. 97052.
26. The antibody or portion thereof of
claim 25
, wherein the amino acid sequence consists of amino acid sequence (a).
27. The antibody or portion thereof of
claim 25
, wherein the amino acid sequence consists of amino acid sequence (b).
28. The antibody or portion thereof of
claim 25
, wherein the amino acid sequence consists of amino acid sequence (c).
29. The antibody or portion thereof of
claim 25
, wherein the amino acid sequence consists of amino acid sequence (d).
30. The antibody or portion thereof of
claim 25
, wherein the amino acid sequence consists of amino acid sequence (e).
31. The antibody or portion thereof of
claim 25
, wherein the amino acid sequence consists of amino acid sequence (f).
32. The antibody or portion thereof of
claim 25
, wherein the amino acid sequence consists of amino acid sequence (g).
33. The antibody or portion thereof of
claim 25
which is a monoclonal antibody.
34. The antibody or portion thereof of
claim 25
which is a polyclonal antibody.
35. The antibody or portion thereof of
claim 25
which is a chimeric antibody.
36. The antibody or portion thereof of
claim 25
which is a humanized antibody.
37. The antibody or portion thereof of
claim 25
which is a single chain antibody.
38. The antibody or portion thereof of
claim 25
which is an Fab fragment.
39. A pharmaceutical composition comprising the antibody or portion thereof of
claim 25
and a pharmaceutically acceptable carrier.
40. The pharmaceutical composition of
claim 39
, wherein the antibody or portion thereof is a monoclonal antibody.
41. The pharmaceutical composition of
claim 39
, wherein the antibody or portion thereof is a polyclonal antibody.
42. The pharmaceutical composition of
claim 39
, wherein the antibody or portion thereof is a chimeric antibody.
43. The pharmaceutical composition of
claim 39
, wherein the antibody or portion thereof is a humanized antibody.
44. The pharmaceutical composition of
claim 39
, wherein the antibody or portion thereof is a single chain antibody.
45. The pharmaceutical composition of
claim 39
, wherein the antibody or portion thereof is an Fab fragment.
46. The pharmaceutical composition of
claim 39
, wherein the antibody or portion thereof is a humanized monoclonal antibody.
47. A hybridoma cell line that produces a monoclonal antibody or portion thereof that specifically binds to a protein whose sequence consists of an amino acid sequence selected from the group consisting of:
(a) the full-length protein encoded by the human cDNA contained in ATCC Deposit No. 97052;
(b) the full-length protein, excluding the N-terminal methionine, encoded by the human cDNA contained in ATCC Deposit No. 97052;
(c) a mature protein encoded by the human cDNA contained in ATCC Deposit No. 97052;
(d) the full-length protein encoded by the human cDNA contained in ATCC Deposit No. 97052, wherein the amino acid sequence has one to five conservative substitutions;
(e) an antigenic fragment of a protein encoded by the human cDNA contained in ATCC Deposit No. 97052;
(f) at least 30 contiguous amino acid residues of a protein encoded by the human cDNA contained in ATCC Deposit No. 97052; and
(g) at least 50 contiguous amino acid residues of a protein encoded by the human cDNA contained in ATCC Deposit No. 97052.
48. The hybridoma cell line of
claim 47
, wherein the antibody or portion thereof is humanized.
49. An antibody or portion thereof produced by immunizing an animal with a protein whose sequence consists of an amino acid sequence selected from the group consisting of:
(a) the full-length protein encoded by the human cDNA contained in ATCC Deposit No. 97052;
(b) the full-length protein, excluding the N-terminal methionine, encoded by the human cDNA contained in ATCC Deposit No. 97052;
(c) a mature protein encoded by the human cDNA contained in ATCC Deposit No. 97052;
(d) the full-length protein encoded by the human cDNA contained in ATCC Deposit No. 97052, wherein the amino acid sequence has one to five conservative substitutions;
(e) an antigenic fragment of a protein encoded by the human cDNA contained in ATCC Deposit No. 97052;
(f) at least 30 contiguous amino acid residues of a protein encoded by the human cDNA contained in ATCC Deposit No. 97052; and
(g) at least 50 contiguous amino acid residues of a protein encoded by the human cDNA contained in ATCC Deposit No. 97052;
wherein said antibody or portion thereof specifically binds to said protein.
US09/879,228 1995-03-31 2001-06-13 Human DNA ligase III Abandoned US20010041350A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US09/879,228 US20010041350A1 (en) 1995-03-31 2001-06-13 Human DNA ligase III
US10/464,768 US20030211582A1 (en) 1995-03-31 2003-06-19 Human DNA Ligase III

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
PCT/US1995/003939 WO1996030524A1 (en) 1995-03-31 1995-03-31 Human dna ligase iii
US08/464,402 US5858705A (en) 1995-06-05 1995-06-05 Polynucleotides encoding human DNA ligase III and methods of using these polynucleotides
US09/054,775 US6284504B1 (en) 1995-03-31 1998-04-03 Human DNA ligase III
US09/879,228 US20010041350A1 (en) 1995-03-31 2001-06-13 Human DNA ligase III

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/054,775 Division US6284504B1 (en) 1995-03-31 1998-04-03 Human DNA ligase III

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/464,768 Continuation US20030211582A1 (en) 1995-03-31 2003-06-19 Human DNA Ligase III

Publications (1)

Publication Number Publication Date
US20010041350A1 true US20010041350A1 (en) 2001-11-15

Family

ID=23843814

Family Applications (4)

Application Number Title Priority Date Filing Date
US08/464,402 Expired - Lifetime US5858705A (en) 1995-03-31 1995-06-05 Polynucleotides encoding human DNA ligase III and methods of using these polynucleotides
US09/054,775 Expired - Fee Related US6284504B1 (en) 1995-03-31 1998-04-03 Human DNA ligase III
US09/879,228 Abandoned US20010041350A1 (en) 1995-03-31 2001-06-13 Human DNA ligase III
US10/464,768 Abandoned US20030211582A1 (en) 1995-03-31 2003-06-19 Human DNA Ligase III

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US08/464,402 Expired - Lifetime US5858705A (en) 1995-03-31 1995-06-05 Polynucleotides encoding human DNA ligase III and methods of using these polynucleotides
US09/054,775 Expired - Fee Related US6284504B1 (en) 1995-03-31 1998-04-03 Human DNA ligase III

Family Applications After (1)

Application Number Title Priority Date Filing Date
US10/464,768 Abandoned US20030211582A1 (en) 1995-03-31 2003-06-19 Human DNA Ligase III

Country Status (1)

Country Link
US (4) US5858705A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030133939A1 (en) * 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5853977A (en) * 1996-07-12 1998-12-29 Schering Corporation Mammalian TNF-α convertases
US6461869B1 (en) * 1999-07-20 2002-10-08 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Purging leukemia cells from hematopoietic stem cells
US20030207297A1 (en) * 1999-10-13 2003-11-06 Hubert Koster Methods for generating databases and databases for identifying polymorphic genetic markers
US20030190644A1 (en) * 1999-10-13 2003-10-09 Andreas Braun Methods for generating databases and databases for identifying polymorphic genetic markers
EP2088209B1 (en) 1999-10-13 2017-05-31 Sequenom, Inc. Methods for generating databases for identifying polymorphic genetic markers
CN1325961A (en) * 2000-05-26 2001-12-12 上海博德基因开发有限公司 Polypeptide-DNA ligase 21 and polynucleotide for coding it
CA2484676A1 (en) * 2002-05-03 2003-11-13 Sequenom, Inc. Kinase anchor protein muteins, peptides thereof, and related methods
EP1613723B1 (en) * 2002-11-27 2013-05-15 Sequenom, Inc. Fragmentation-based methods for sequence variation detection and discovery
US20050009053A1 (en) * 2003-04-25 2005-01-13 Sebastian Boecker Fragmentation-based methods and systems for de novo sequencing
US9394565B2 (en) * 2003-09-05 2016-07-19 Agena Bioscience, Inc. Allele-specific sequence variation analysis
US7608394B2 (en) 2004-03-26 2009-10-27 Sequenom, Inc. Methods and compositions for phenotype identification based on nucleic acid methylation
CA2561381C (en) * 2004-03-26 2015-05-12 Sequenom, Inc. Base specific cleavage of methylation-specific amplification products in combination with mass analysis
AU2005284980A1 (en) * 2004-09-10 2006-03-23 Sequenom, Inc. Methods for long-range sequence analysis of nucleic acids
US11093547B2 (en) * 2018-06-19 2021-08-17 Intel Corporation Data storage based on encoded DNA sequences

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8907468D0 (en) * 1989-04-03 1989-05-17 Smith Geoffrey L Vaccinia vectors,vaccinia genes and expression products thereof

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030133939A1 (en) * 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins

Also Published As

Publication number Publication date
US6284504B1 (en) 2001-09-04
US5858705A (en) 1999-01-12
US20030211582A1 (en) 2003-11-13

Similar Documents

Publication Publication Date Title
EP0792370B1 (en) Interleukin-1 beta converting enzyme like apoptosis protease-3 and 4
US6284504B1 (en) Human DNA ligase III
US6709653B1 (en) Antibodies specific for human inositol monophosphatase H1
US6602984B1 (en) Human elastase IV
US5747312A (en) Human AlkB polypeptide
US6552174B2 (en) Human MutT2 antibodies
US6455274B1 (en) Human DNA Ligase IV
US6403310B1 (en) Human inositol monophosphatase H1
US6432708B1 (en) Human choline acetyltransferase
US5914258A (en) Human deoxycytidine kinase 2
US6849436B2 (en) Human hypoxanthine-(guanine) phosphoribosyl transferase-2
US6630443B2 (en) Human amine transporter antibodies
AU687484B2 (en) Human dna ligase IV
WO1996030524A1 (en) Human dna ligase iii
US6255077B1 (en) Human DNA topoisomerase I α
US5859200A (en) Human amine transporter
WO1996021724A1 (en) Human deoxycytidine kinase 2
JP2002204697A (en) Human dna ligase iv

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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