NZ507318A - Compositions comprising mammalian and human REC2 that functions as an ATPase - Google Patents

Compositions comprising mammalian and human REC2 that functions as an ATPase

Info

Publication number
NZ507318A
NZ507318A NZ507318A NZ50731800A NZ507318A NZ 507318 A NZ507318 A NZ 507318A NZ 507318 A NZ507318 A NZ 507318A NZ 50731800 A NZ50731800 A NZ 50731800A NZ 507318 A NZ507318 A NZ 507318A
Authority
NZ
New Zealand
Prior art keywords
leu
ser
glu
ala
gly
Prior art date
Application number
NZ507318A
Inventor
William K Holloman
Michael C Rice
Sheryl T Smith
Zhigang Shu
Eric B Kmiec
Original Assignee
Univ Jefferson
Cornell Res Foundation 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
Application filed by Univ Jefferson, Cornell Res Foundation Inc filed Critical Univ Jefferson
Priority to NZ507318A priority Critical patent/NZ507318A/en
Publication of NZ507318A publication Critical patent/NZ507318A/en

Links

Abstract

A composition comprising a mammalian ATPase is disclosed. The ATPase has 80% identity to amino acid SEQ ID NO: 1, and binds single stranded DNA in the presence of ATP, catalyzing DNA reannealing.

Description

New Zealand Paient Spedficaiion for Paient Number 507318 I 5073 18 NEW ZEALAND PATENTS ACT 1953 COMPLETE SPECIFICATION "MAMMALIAN AND HUMAN REC2" THOMAS JEFFERSON UNIVERSITY, a US corporation, of 1020 Locust Street, Philadelphia, PA 19107 6799, USA and CORNELL RESEARCH FOUNDATION, INC , a US corporation of Suite 105, 20 Thornwood Drive, Ithaca, NY 14850, USA, hereby declare the invention, for which we pray that a patent may be granted to us, and the method by which it is to be performed, to be particularly described in and by the following statement 1 5 !'\!: cllec i"ual—.jhoperty 0fficf ' (the following page is numbered "1a") • qf wz PCT/1B97AH217 MAMMALIAN AND HUMAN REC2 This application claims benefit of the priority of U.S. provisional application Serial No. 60/025,929, filed September 11, 1996. 1. FIELD OF THE INVENTION The present invention concerns the field of molecular genetics and medicine. Particularly, it concerns genes encoding a protein that is involved in homologous recombination and the repair of damaged genomic DNA in mammalian cells. Specifically the invention concerns the gene encoding a mammalian ATP-dependent homologous pairing protein; methods of using the gene to effect gene therapy; methods of using the gene and fragments of the gene to classify a mammalian tissue for medical purposes; and transgenic mice having had one or both alleles of the gene made inoperative. More specifically, the gene of the present invention is the human and murine homologs of the gene termed REC2 previously isolated from the fungus Ustilago maydis. 2. BACKGROUND OF THE INVENTION During the life of every organism the DNA of its cells is constantly subjected to chemical and physical events that cause alterations in its structure, i.e., potential mutations. These potential mutations are recognized by DNA repair enzymes found in the cell because of the mismatch between the strands of the DNA. To prevent the deleterious effects that would occur if these potential mutations became fixed, all organisms have a variety of mechanisms to repair DNA mismatches. In addition, higher animals have evolved mechanisms whereby ceils having highly damaged DNA, undergo a process of programmed death ("apoptosis").
The association between defects in the DNA mismatch repair and apoptosis inducing pathways and the development, progression and response to treatment of oncologic disease is widely recognized, if incompletely understood, by medical scientists. Chung, D.C. & Rustgi, A.K., 1995, Gastroenterology 109:1685-99; Lowe, S.W., et al., 1994, Science 266:807-10. Therefore, there is a continuing need to identify and clone the genes that encode proteins involved in DNA repair and DNA mismatch monitoring. 1<=* Studies with bacteria, fungi and yeast have identified three genetically defined groups of genes involved in mismatch repair processes. The groups are termed, respectively, excision repair group, the error prone repair group and recombination repair group. Mutants in a gene of each group results in a characteristic phenotype. Mutants in the recombination repair group in yeast result in a phenotype having extreme sensitivity to ionizing radiation, a sporulation deficiency, and decreased or absent mitotic recombination. Petes, T.D., et al., 1991, in Broach, J.R., et al., eds, The Molecular Biology of the Yeast Saccharomyces, pp. 407-522 (Cold Spring Harbor Press, 1991).
Several phylogenetically related genes have been identified in the recombination repair group. recA, in £. Coli, Radding, C.M., 1989, Biochim. Biophys. Acta 1008:131-145; RAD51 in 5. cerevisiae, Shinohara, A., 1992, Cell 69:457-470, Aboussekhra, A.R., et al., 1992, Mol. Cell. Biol. 12:3224-3234, Basile, G., et al., 1992, Mol. Cell. Biol. 12:3235-3246; RAD57 in 5. cerevisiae. Gene 105.139-140; REC2 in U. maydis, Bauchwitz, R., & Holloman, W.K., 1990, Gene 96:285-288, Rubin, B.P , et al., 1994, Mol. Cell Biol. 14:6287-6296. A third S. cerevisiae gene DMCl, is related to recA, although mutants of DMCl show defects in cell-cycle progression, recombination and meiosis, but not in recombination repair The phenotype of REC2 defective U. maydis mutants is characterized by extreme sensitivity to ionizing radiation, defective mitotic recombination and interplasmid recombination, and an inability to complete meiosis. Holliday, R., 1967, Mutational Research 4.275-288. UmREC2, the REC2 gene product of U. maydis, has been extensively studied. It is a 781 amino acid ATPase that, in the presence of ATP, catalyzes the pairing of homologous DNA strands in a wide variety of circumstances, e.g., UmREC2 catalyzes the formation of duplex DNA from denatured strands, strand exchange between duplex and single stranded homologous DNA and the formation of a nuclease resistant complex between identical strands. Kmiec, E.B., et al., 1994, Mol.
Cell. Biol. 14:7163-7172. UmREC2 is unique in that it is the only eukaryotic ATPase that forms homolog pairs, an activity it shares with the E. colt enzyme recA.
U.S. patent application. Serial No. 08/373,134, filed January 17, 1995, by W.K. Holloman and E.B. Kmiec discloses REC2 from U. maydis, methods of producing recombinant UmREC2 and methods of its use. Prior to the date of the present invention a 2 fragment of human REC2 cDNA was available from the IMAGE consortium, Lawrence Livermore National Laboratories, as plasmid p153195. Approximately 400 bp of the sequence of p153195 had been made publicly available on dbEST database.
The scientific publication entitled: Isolation of Human and Mouse Genes Based on Homology to REC2, July 1997, Proc. Natl. Acad. Sci. 94, 7417-7422 by Michael C. Rice et al., discloses the sequences of murine and human Rec2, of the human REC2 cDNA. and discloses that irradiation increases the level of hsREC2 transcripts in primary human foreskin fibroblasts. 3. SUMMARY OF THE INVENTION The invention provides nucleic acids encoding mammalian ATP-dependent homologous pairing proteins (a "mammalian recombinase") particularly, the human and murine ATP-dependent homologous pairing protein (hsREC2 and muREC2, respectively). The invention additionally provides DNA clones containing a copy of the mRNA encoding a mammalian recombinase (an "mREC cDNA") and DNA clones containing a copy of the genomic DNA containing an mREC gene or fragments thereof. In further embodiments, the invention concerns a nucleic acid comprising an mREC cDNA linked to a heterologous promoter, i.e., a promoter other than a mammalian recombinase promoter, so that a mammalian recombinase can be expressed or over-expressed in insect and mammalian cells and in bacteria. In one embodiment, the heterologous promoter is the polyhedrin promoter from the baculovirus Autographica cahfornica and the invention provides for an isolated and purified mammalian recombinase, particularly isolated and purified hsREC2.
The invention provides several utilities of said nucleic acids and isolated and purified proteins. In the area of gene therapy and the construction of transgenic animals, the invention provides a method of enhancing homologous recombination between an exogenous nucleic acid and the genome of a cell by introducing a plasmid that expresses ^Qmammalian recombinase into the cell, which method can be used to repair a genetic defect and thereby cure a genetic disease. Alternatively, for the construction of transgenic animals the invention provides a method of enhancing homologous recombination between an exogenous nucleic acid and the genome of a ceil by introducing purified 3 mammalian recombinase into the cell. Alternatively, the invention provides for the construction of a transgenic animal, such as a mouse, having a transgenic mamalian recombinase gene operably linked to a strong promoter so that the recombinase is over expressed in some or all tissues. Such transgenic animals are highly adapted to undergo homologous recombination.
The invention additionally provides.two methods of classifying a sample of human tissue for diagnosis and prognosis: by determining whether the cells of the sample contains two, one or no copies of hsREC2-, and by determining whether the copies of hsREC2 that said cells contain are mutated. For the purpose of diagnosis and classification of tissue samples the invention, firstly, provides paired oligonucleotides that are suitable for the PCR amplification of fragments of hsREC2 and, secondly, identifies a microsatellite DNA sequence, D14S258, that is closely linked to hsREC2.
The invention further provides a transgenic mouse having one or both alleles of muREC2 interrupted and thereby inactivated. The resultant transgenic animals, termed heterozygous and homozygous REC2-knock out mice, respectively, are susceptible to tumorigenesis by chemical carcinogens. REC2-knock-out mice can be used to determine whether their is a significant risk of carcinogenesis associated with a chemical or a process of interest. The reduced level or absence of muREC2 makes REC2-knock-out mice a more sensitive test animal than wild-type.
The invention further provies a method of sensitizing target cells to DNA damage, such as trom y- or UV irradiation or from cytotoxic agents commonly used in oncologic therapy, which comprises causing the expression of high levels of recombinase in the target cell. The expression of such levels causes the cells to more readily undergo apoptosis in response to DNA damage. The invention yet further provides the REC2 promoter a mammalian promoter that is induceable by irradiation or other DNA damaging agents. 4. BRIEF DESCRIPTION OF THE FIGURES Figures 1A-1G. Figures 1A and 1B show the derived amino acid sequence of hsREC2 (SEQ ID NO:l), and Figures 1C and 1D show the nucleic acid sequences of the hsREC2 cDNA coding strand (SEQ ID NO:2) Figures 1E and 1F show the derived amino 4 SUBSTITUTE SHEET (RULE 26) acid sequence of muREC2 (SEQ ID NO:3), and Figure 1G shows the nucleic acid sequences of the hsREC2 cDNA coding strand (SEQ ID NO:4).
Figure 2A-2D. Figure 2A is an annotated amino acid sequence of hsREC2.
Specifically noted are the nuclear localization sequence ("NLS"), A Box and B Box motif sequences, DNA binding sequence and a src-type phosphorylation site CP")- Figure 2B is a cartoon of the annotated sequence, showing in particular the region 80-200 is most closely related to recA. Figures 2C-2D show the sequence homology between hsREC2 and Ustilago maydis REC2. The region of greatest similarity, 43% homology, is in bold. Fipnrp 3 Figure 3 shows the DNA reannealing as a function of added bacu lovi rus-produced hexah i stidyI HsREC2.
Figure 4 Figure 4 is a gel shift assay showing that binding of a hsREC2- thioredoxin fusion protein to ssDNA is ATP or y-SATP dependent Figure 5A-5B. Figure 5A shows the frequency of repair of the Sickle Cell Disease mutation, as a function of added Gs-ISA chimeric repair vector (SC1), in the B-globin genes in a population of EBV-transformed human lymphoblasts in the presence or absence of the hsREC2 expression vector pcHsREC2, pcDNA3 or pcCAT control plasmids or SC1 alone. Figure 5B shows the sequences of SC1,15s and I5A in the region of the Sickle Cell mutation Lower case (a, c, g, and u) indicates 2'-OMe nucleotides.
Figure 6. Figure 6 shows the reannealing of a 123 nt DNA fragment is catalyzed by GST/REC2 fusion protein.
Figures 7A-7F. Figures 7A-7C show the sequence of the hsREC2 promoters and Figures 7D-7F show the sequence of the mu REC2 promoters. The locations of sequences homologous to the sequences of known cis-acting radiation responsive elements in yeast are underlined and the corresponding yeast gene is indicated.
Figures 8A-8H. Figures 8A-8H show FACS histograms of RNase treated, propidium iodide stained, CHO cells that have been transfected with either an hsREC2 expressing plasmid (15C8) or an irrelevant control plasmid (Neo). The DNA content of the cells is displayed in the horizontal axis. The histograms are of unirradiated cells (8A, 8E) or of cells that are 24,48 or 72 hours status post exposure to 15 J/m2 UV irradiation (8B-8D, 8F-8H). The comparison shows that the expression of hsREC2 increases the fraction of irradiated cells having less than the diploid DNA content, which is indicative of SUBSTITUTE SHEET (RULE 26) of apoptosis.
. DETAILED DESCRIPTION OF THE INVENTION As used herein, genes are italicized, e.g., hsREC2, while the corresponding protein is in normal typeface. .1 hsREC2 AND THE STRUCTl JRF OF ITS PRODUCT hsREC2 The results of efforts to obtain hsREC2 cDNA by hybridization under non-stringent conditions with UmREC2 probes were unsatisfactory Efforts were made to isolate a fragment of hsREC2 by PCR amplification using primers that encode pentapeptides based on the UmREC2 sequence. A mixture of four forward primers encoding residues 256-260 of UmREC2, GKTQM (SEQ ID NO:7), was constructed using inosine as the third base for the gly and thr codons and having a 5' noncoding GC dinucleotide, i e , 5'-GC GGI AA(G/A) ACI CA(G/A) ATG-3'. A mixture of eight reverse primers complementary to the sequences that can encode residues 330-334 of UmREC2, YITSG, was synthesized, using inosine in the same way as the forward primers, i.e., 5'-CC ICC G(C/G)(T/A)' IGT IAT (A/G)TA-3' The primers were used to amplify fragments of human genomic and cDNA libraries using the Expand™ system (Boehringer) coupled with two rounds of reamplification. After reamplification the fragments were cloned in pCRII (Invitrogen). Ten different mixtures of primers encoding a total of nine different pentapeptides were used and a total of about 60 fragments were sequenced. One 110 bp fragment from a human kidney cDNA library, hsrl 10, had statistically significant homology with UmREC2.
A computer search of the database dbEST was performed to find clones of cDNAs encoding proteins that have significant homologies with UmREC2 and hsrl 10. The plasmid pi 53195 was identified as having significant homology with UmREC2 and which contained hsrl 10. In one segment of 44 residues of UmREC2 and hsREC2, there was 43% homology between UmREC2 and hsREC2, i.e., 19 of the 44 residues of each sequence were identical. Additionally, there were 8 conservative substitutions. This 1 Only two of the four combinations are complementary to ser codons, however, they are complementary to the ser codons most often used in humans. 6 SUBSTITUTE SHEET (RULE 26) region of high homology corresponds to residues 84-127 of hsREC2 and residues 226-270 of UmREC2. See Figures 2C-2D. Residues 226-270 of UmREC2 is the portion ot UmREC2 that is most highly conserved when compared to recA and related members of the recombination repair group; cf. residues 40-95 of recA, 95-13 of DMC1, residues 100-144 of RAD51, and residues 160-204 of RAD51. See, e.g.. Figure 2, Rubin et al., 1994 supra.
That clone p153195 lacked the 5' end of hsREC2 was determined by the absence of an inframe start codon. The 5' end of hsREC2 cDNA was obtained by PCR amplification of a cDNA library using a forward primer from the cloning vector and nested reverse primers based on pi 53195. An over lap of about 100 bp was identified which contained a unique restriction site that was used to reconstruct the full length hsREC2 cDNA. The sequences of the reconstructed hsREC2 cDNA are given in Figures 1A-1B and the derived sequence of hsREC2 are given in Figures 1C-1D. The hsREC2 cDNA encodes a protein of 350 amino acids, SEQ ID NO:l. The sequence of the hsREC2 cDNA and its complement are SEQ ID NO: 2 and No: 3, respectively. The 5' boundary of pi 53195 was about nt 280 of SEQ ID NO: 2 Comparisons of the hsREC2 sequence with the UmREC2 sequence reveals statistically significant, but distant homologies (p- 2.8x10'5). A similar level of homology is found between hsREC2 and the yeast protein DMCl An expression vector containing the complete hsREC2 cDNA under control of a strong promoter, for example, the cytomegalovirus promoter (pcHsREC2), can be constructed for over-expression of hsREC2 in transfected eukaryotic cells. For the production of purified hsREC2 a vector suitable for the expression of the hsREC2 under control of the baculovirus polyhedrin promoter can be constructed. It is preferred to construct a vector that synthesizes a REC2 fusion protein consisting of a protein or peptide that aids in the purification of the product, such as a hexahistidyl peptide or glutathione S-transferase. Figures 1 E-1 F and 1C show the derived amino acid and nucleic acid sequences of the murine REC2 (muREC2) cDNA .2 HOMOLOC.S OF hsREC.2 The present invention encompasses mammalian homologs ot hsREC2. Nucleic 7 SUBSTITUTE SHEET (RULE 26) PCT7EB97/01217 acids encoding the REC2 from any mammalian species can be identified and isolated by techniques, routine to those skilled in the art, using the sequence information of Figures 1A-1D and/or the hsREC2 cDNA clone. Such routine techniques include use of the hsREC2 cDNA or fragments thereof to probe cDNA and genomic libraries from other mammalian species and use of the sequence data to construct primers for PCR amplification of fragments of mammalian REC2 cDNA. The cloning of hsREC2 and muREC2 genomic DNA (gDNA) is described below.
High levels of transcripts of hsREC2 can be found in heart and skeletal muscle, lung, pancreas, spleen and thymus, and placenta. Moderate or low levels of hsREC transcripts are found in liver, kidney, brain and testes. Thus, the source of mRNA to construct cDNA libraries for obtaining mammalian REC2 clones is not critical. The sequence of residues 83-127, which corresponds to amino acids 226-270 ot UmREC2, is particularly highly conserved and is, therefore, useful in identifying mammalian REC2 homologs.
Mammalian homologs of hsREC2 can be identified by the presence of an amino acid sequence identity of greater than 80% and preferably greater than 90% compared to hsREC2 in the highly conserved portions of the gene, i.e., the portion homologous to residues 83-127 of hsREC2. In a preferred embodiment the mammalian recombinase gene shares greater than 80% sequence identity with hsREC2 gene within the about 130 bp segment that encodes the residues homologous with residues 83-127 of hsREC2 Such mammalian homologs of hsREC2 will also have the above-noted activities ot catalyzing DNA reannealing, ATPase activity and ATP-dependent ssDNA binding activity.
As used herein, a protein having each of these three activities is termed an ATP-dependent homologous pairing protein (a "mammalian recombinase"). A mammalian recombinase having greater than an 80% sequence identity with hsREC2 is termed an "mREC2." Based on the extensive studies of bacterial and yeast homologous recombination proteins, those skilled in the art anticipate that all mammalian recombinases will have greater than 80% amino acid sequence identity with hsREC2, i.e., be an mREC2.
The invention further encompasses fusion proteins comprising a mammalian REC2 protein or fragment thereof, wherein the REC2 fragment displays at least one and 8 SUBSTITUTE SHEET (RULE 26) preferably each of the three above-noted activities to substantially the same extent as the native REC2. Those skilled in the art appreciate that the recombinant production and purification of mammalian proteins in bacterial and insect cell based expression systems is facilitated by the construction of fusion proteins that contain the protein of interest and a second protein that stabilizes the resultant fusion protein and facilitates its purification. Non-limiting examples of fusion proteins include hexahistidyl, Glutathione-S-transterase and thioredoxin fused to the amino terminus of REC2.
In one embodiment, the invention is a composition containing an isolated and purified protein, which is an ATP-dependent homologous pairing protein, i.e., is an ATP-dependent catalyst of DNA reannealing, is an ATPase, and binds ssDNA in the presence of ATP or y-SATP, and which protein comprises a polypeptide of at least 115 amino acids which is substantially identical to a polypeptide found in a mammalian ATP-dependent homologous pairing enzyme. More preferably the isolated and purified protein comprises a polypeptide that is substantially identical to residues 80-200 of hsREC2 In a further embodiment, the isolated and purified protein of the invention comprises the polypeptide which is residues 2-350 of SEQ ID NO-1. As used herein, substantially identical means identical or having at most one conservative substitution per 20 amino acids. As used herein a human protein is an isolated and purified human protein if the composition containing the protein is substantially free of all other normally intracellular human proteins but a defined set of individually identified human proteins; similarly an isolated and purified mammalian protein is free of all other normally intracellular mammalian proteins except for a defined set of individually identified mammalian proteins. As used herein, "a composition which comprises a defined protein substantially free of a named material" means that the weight of the named material in the composition is less than 5% of the weight of the protein in the composition.
The invention further provides an isolated and purified nucleic acid derived from a mammalian species, i.e., derived from a cDNA or gDNA clone, that encodes a protein or fusion protein, having a sequence, which comprises the sequence of a mammalian ATP-dependent homologous pairing protein or a substantially identical sequence. As used herein, an isolated and purified nucleic acid is a nucleic acid isolated and purified free of nucleic acids encoding other mammalian proteins or fragments thereof. As used herein. 9 the sequence of a mammalian ATP-dependent homologous pairing protein means the sequence of a naturally occurring, i.e., wild-type ATP-dependent homologous pairing protein found in a mammal, or of any mutants of wild-type mammalian ATP-dependent homologous pairing protein. In preferred embodiments the nucleic acid of the invention encodes a protein that is greater than 80% sequence identical, or alternatively, more than 90% sequence identical to hsREC2. Those skilled in the art appreciate that the N-terminal and C-terminal one, two or three amino acids can be substituted or deleted without effect and, as used herein, are not considered a part of the sequence unless so specified Those skilled in the art further appreciate that the insertion or deletion of one to four consecutive amino acids during the evolution of homologous proteins is common. Therefore, in the definition of sequence identity between proteins encompasses the introduction of as many as four, one to four residue gaps in one or both sequences to maximize identity.
The isolated and purified nucleic acids of the invention encompass not only cDNA and gDNA clones of mammalian genes encoding a mammalian ATP-dependent homologous pairing protein, but also nucleic acids derived from said cDNA and gDNA clones by site directed mutagenesis. By use of routine PCR techniques, those skilled in the art can make specific, predetermined changes in the sequence of a DNA. Site directed mutagenesis may be conducted by any method. The method of Ho, S.N., et al.. Gene 77.51-59 (herewith incorporated by reference in its entirety), is suitable. According to the method of Ho, overlapping, mutated genome fragments are synthesized in two separate PCR reactions. Of the four primers are used in the two reactions, two are complementary to each other and introduce the desired mutation. The PCR reactions are performed so that the 3' end of the sense strand of one product is complementary to the 3' end of antisense strand of the other. The two PCR products are denatured, mixed and reannealed. The overlapping partial duplex molecules are then extended form a full length dsDNA, amplified in a third PCR reaction, the product isolated and inserted by conventional recombinant techniques into the parent gene. See, also, Liang, Q., et al., 1994, PCR Methods & Applic. 4:269-74; Weiner, M.P. & Costa, G.L., 1994, PCR Methods & Applic. 4:S131-136; Barrettino, D., et al, 1994, Nucleic Acids Research 22:541; Stemmer, W.P., etal., 1992, Biotechniques 13 214-220. By multiple applications of such techniques any desired modifications in the sequence of a cloned DNA can be introduced. Thus, the nucleic acids of the invention are not limited to isolated and purified nucleic acids having naturally occurring sequences, but also include nucleic acids encoding a ATP-dependent homologous pairing protein having substantially the same sequence as a naturally occurring mammalian recombinase.
The compositions of the invention further include compositions comprising not only mammalian recombinases isolated and purified free of mammalian proteins, but also compositions comprising any isolated and purified ATP-dependent homologous pairing protein having substantially the same sequence as a naturally occurring mammalian recombinase.
The hsREC2 sequence contains several sequences that have been identified with specific functions in other proteins. Figure 2A shows the sequence of hsREC2 and indicates the locations of nuclear localization sequence, four sequences associated with recA, namely A box, B box, a src-like phosphorylation site and a DNA binding site.
Those skilled in the art will appreciate that, as was found for UmREC2, not all portions of a mREC2 protein are essential for the in vitro activities that characterize ATP-dependent homologous binding proteins. However, the region of about 120 amino acids from about residue 80 to residue 200, which is recA-like, is essential for these activities. .3 THE USE OF mREC2 AND mREC2 ENCODING GENES TO EFFECT HOMOLOGOUS RECOMBINATION BETWEEN THE GENOME OF A CELL AND AN EXOGENOUS NUCLEIC ACID In one embodiment of the invention, a plasmid that expresses an mREC2 is used to increase the rate of homologous recombination between an exogenous nucleic acid and the genome of a cell. In one embodiment, the exogenous nucleic acid is a chimeric repair vector (CRV), which is an oligonucleotide having mixed ribo- and deoxyribonudeotides. The structure of CRV are disclosed in U.S. patent applications Serial No. 08/353,657, filed December 4, 1994, and Serial No. 08/664,487, filed June 17, 1996, which are hereby incorporated by reference in its entirety. U.S. application Serial No. 08/640,517, entitled "Methods and Compounds for Curing Diseases Caused by Mutations," filed May 1, 1996, by E.B. Kmiec, A. Cole-Strauss and K. Yoon, (the '517 Application), which is hereby incorporated by reference in its entirety, describes the use 11 of CRV to repair mutations that cause diseases. Particularly, the '517 Application concerns the repair of mutations that affect hematopoietic ceils such as the mutation in 6-giobin that causes Sickle Cell Disease.
According to the present invention, the cell having a disease-causing mutation to be repaired (the target cell) is removed from the subject. The target cells are then transfected with a nucleic acid having a promoter operably linked to a nucleic acid encoding a mREC2 (an mREC2 expression vector) such that a mammalian ATP-dependent homologous pairing protein is over-expressed in the target cell. For most types of human cells, the immediate early promoter from cytomegalovirus is suitable. Because the persistent over-expression of a mammalian ATP-dependent homologous pairing protein can effect the growth and differentiation of the target cell, the mREC2 expression vector should be incapable of replication in the target cell. The mREC2 expression vector can be introduced into the target cell by any technique known to those in the field or to be developed. Liposomal compositions such as LIPOFECTIN'™1 and DOTAP1™1 are suitable After transfection with the mREC2 expression vector, the target cells are cultured for twentv four hours and then a CRV designed to repair the disease causing mutation is introduced into the target cells, according to the methods of the '517 Application, and repaired target cells are then reimplanted into the subject. Alternatively, the repaired target cells can be frozen and reimplanted at a clinically opportune time Figure 5A shows the results of the use of an mREC2 expression vector to enhance the effectiveness of a CRV that repairs the mutation that causes Sickle Cell Disease in a human EB-transformed lymphoblastoid cell line. These data show that at a concentration of CRV of about 100 ng/ml, the pretreatment of the target cells with the mREC2 expression vector pcHsREC2, labelled "pchREC2" in Figure 5A, caused an about 5 fold increase, from 12% to 65%, in the percent of repaired copies of S-globin At 250 ng/ml, over 80% of the copies of B-globin were repaired. At higher concentrations of CRV, the differences between pcHsREC2 treated target cells and control target cells become less marked.
The present invention is exemplified by the use of a non-replicating episome to introduce an mREC2 cDNA gene (hsREC2), operably linked to a cytomegalovirus (CMV) promoter, into the target cell and to transiently express mREC2. Alternative embodiments 12 SUBSTITUTE SHEET (RULE 26) of the invention can be produced by introducing the copy of a genomic gene, which can be linked to the homologous mREC2 promoter or, alternatively, modified so that the homologous promoter is replaced by a CMV or other heterologous promoter. Further variants that can be used to increase homologous recombination in different situations include linkage of either mREC2 cDNA or gDNA to tissue specific promoters such as a CD4, immunoglobulin, insulin or globin promoter. By use of tissue specific promoters, transgenic animals, particularly mice, rats and swine can be constructed that overexpress mREC2 in only one particular tissue. In yet a further alternative embodiment the promoter can be an inducible promoter. An inducible promoter particularly suitable for the present invention is a tetracycline inducible promoter, which is described in U.S. Patent No. 5,464,758, which is incorporated by reference in its entirety.
Those skilled in the art will further appreciate that an mREC2 encoding gene can be constructed that contains some but not all introns of the complete mREC2 gDNA.
Such a gene is a mixture of mREC2 gDNA and mREC2 cDNA fragments. As used herein the term "an mREC2 gene" is to be understood to denote, generically, mREC2 cDNA, mREC2 gDNA or a nucleic acid encoding a full length REC2 protein comprising mREC2 gDNA and mREC2 cDNA fragments.
The present invention further encompasses the use of mREC2 expression vectors to facilitate the construction of transgenic animals using cultured embryonic stem cells ("ES cells") according to the method of Capecchi, M.R., 1989, Science 244: 1288 and U.S. Patent 5,487,992, Col. 23-24, which are incorporated by reference in their entirety. A transgenic mouse having a inducible mREC2 gene introduced can be constructed. ES cells from such a transgenic mouse can be obtained and induced to have elevated levels of mREC2. Such cells will more readily undergo homologous recombination with a chimeric mutational vector ("CMutV"), an oligonucleotide having a similar structure and function to those of CRV, that can be used to introduce specific mutations into targeted wild-type genes. By use of CMutV, second and higher generation transgenic animals having further targeted genetic alterations can be constructed.
A further embodiment of the invention concerns the use of isolated and purified mREC2 protein in the construction of transgenic animals. Those skilled in the art of constructing transgenic animals understand that transgenic animals are constructed by 13 direct injection of a nucleic acid into the pronucleus of an ovum according to the method described Brinster, R.L. et al., 1989, Proc. Natl. Acad. Sci 86:7087; see also U.S. Patent No. 4,873,191 to T.E. Wagner and P C. Hoppe, which are hereby incorporated by reference in their entirety. Such direct injection results in the random integration of the injected nucleic acid. As noted above techniques for the introduction of transgenes by homologous recombination have been developed, however, such techniques require a specialized embryonic stem cell line, which is available only for mice, and, in addition require that the genetic alteration be designed so that homologous recombinants can be selected in culture, since the rate of homologous recombination is very low.
Because the use of the present invention in conjunction with CMutV permits a specific alteration to be introduced into a large fraction, e.g., 80%, of the copies of a target gene, those skilled in the art will appreciate that the invention provides a practical technique tor the construction of transgenic animals wherein the function of both alleles of a specifically targeted gene has been deleted ("knocked-out") by homologous recombination using ova directly injected with a REC2 CMutV mixture.
Transgenic animals are constructed according to the invention by injecting a ova pronucleii with mREC2 protein and the CMutV. In a preferred embodiment a mixture of the CMutV and a mREC2 protein is injected into the ova pronucleus. In a preferred embodiment the nucleic acid to be injected is a CMutV that introduces a stop codon or a frameshift mutation into the gene to be knocked out. The concentration of protein to be used is about one molecule of mREC2 protein per between 5,000 base pairs and 50 base pairs of the CMutV, preferably one molecule of mREC2 protein per about 100-500 base pairs of the CMutV. Alternatively, the CMutV can be replaced by a conventional linearized DNA fragment containing homologous regions flanking a mutator region. .4 THF CONSTRUCTION OF muftfO-KNOCK-Ol JT MICE The invention additionally provides transgenic mice that contain inactivated muREC2. Such heterozygous muREC2-knock-out transgenic mice can be constructed by injection of a murine embryonic blastocyst with an embryonic stem cell line (ES cells) that has the appropriate mutation in muREC2 (muREC2ko). The technique of Nichols, J.,et al., 1990, DEVELOPMENT 110:1341-48 can be used. Further teaching regarding the 14 PCT/IB97/D1217 construction of transgenic mice using embryonic stem cell-injected blastocysts can be found in U.S. Patent No. 5,487,992 to Capecchi and Thomas, which is hereby incorporated by reference in its entirety. Homozygous muRfC2-knock-out mice can be obtained by the intercross of heterozygous muR£C2-knock-out mice and selection of offspring that are homozygous for the muREC2ko allele.
Without limitation, a muREC2k° gene can be made in two ways. A CMutV can be constructed according to U.S. patent No. 5,565,350, which is designed to introduce one or more stop codons at different positions within muREC2 (an "muREC2ko chimeric vector"). ES cells line can be treated with the muREC2ko chimeric vector. Preferably several muREC2Ko chimeric vectors, designed to introduce redundant stop codons are used to reduce the reversion rate. After treatment, the ES cells can be cloned and the loss of a functional muREC2 gene confirmed by sequence analysis or by PCR amplification using primers specific for the mutated codons.
Alternatively, a dicistronic targeting construct can be used to introduce a muREC2k° mutation Mountford, P., et al , 1994, Proc. Natl. Acad. Sci. 91:4303-07 More specifically, targeting vector is constructed having a cassette consisting of, in 5* to 3' order, a splice acceptor site, the 500 bp internal ribosome entry site (IRES) from encephalomyocarditis virus (EMCV), a fusion gene Bgeo, that has both B-galactosidase and G418 resistance activity, and an polyadenylation signal from SV40. In the targeting construct, the cassette is inserted, as an example without limitation, between two fragments from the introns 3' and 5' of the second exon of the tnuREC2 gene, wherein the 5' most exon is the first exon, the exon immediately 3' to the 5'-most exon is the second exon etc. The length of the fragments can be preferably between about 500 bp and 5,000 bp.
The linearized targeting construct can be introduced into an ES cells by any technique suitable for the transfection of DNA into ES cells. The muREC2 gene of the transfected ES cells undergoes homologous recombination whereby the cassette replaces the second exon such that the cassette is transcribed from the muREC2 promoter and the Bgeo protein is translated by ribosomes bound to the IRES. ES cells having the cassette integrated into transcriptionally active genes can be selected by exposing the transfected cells to G418 and by histochemical staining to detect galactosidase positive cells.
Typically as many as 70% to 90% of RgalVneo' double transformants have undergone homologous recombination of the targeted gene.
Homozygous muREC2ko mice have an increased susceptibility to mutation caused by chemical and physical agents. Such animals can be used to determine if products are mutagenic and more specifically if such products are carcinogens. Both homozygous and heterozygous muREC2*° mice will also be more susceptible to the development of benign and malignant tumors. These animals can be used to originate tumors of different tissue types for use in biomedical studies. .5 THE CLASSIFICATION OF SAMPLES OF HUMAN TISSUE BY FXAMINATION OF THF hsREC2 GFNFS OF THF SAMP1 F Those skilled in the art appreciate that there is a close connection between the a cell's capacity to remove chemically induced mutations and replication errors from its DNA and the cell's potential to develop the genetic changes that result in the development and progression of malignancies. Altonen, L.A., 1993, Science 260:812-816, Chung, D.C., & Rustgi, A.K., 1995, Gastroenterology 109-1685-99. A cell's capacity to remove mutations and replication errors can be classified by determining, firstly, whether the cell contains the normal, i.e , diploid number of copies of a gene that is essential for DNA mismatch repair and, secondly, by determining whether the copies that are present have been altered, i.e., contain mutations. Cells having a diminished capacity to remove DNA mismatches because of defects in their REC2 are malignant or are more likely to become malignant due to the further accumulation ot mutations.
In one embodiment, the invention consists of classifying a human tissue according to the number of copies of the hsREC2 gene per diploid genome. The reduction of the number to less than two indicates that some cells of the tissue can have a reduced capacity to repair DNA mismatches, because a mutation in the remaining copy would cause the absence of ATP-dependent homologous pairing activity. The number of copies of a gene can be readily determined by quantitative genomic blotting using probes constructed from labelled nucleic acids containing sequences that are fragment of SEQ ID NO:2 or a complement therof. An alternative method of determining the number ot hsREC2 genes per diploid genome in a sample of tissue relies on the fact that the hsREC2 gene is located in bands 14q23-24 and, particularly, that it is tightly linked to the 16 proximal side of the marker D14S258 and also tightly linked to the marker D14S251. The loss of a copy ot a hsREC2 gene in an individual who is heterozygous at a locus linked to the D14S258 marker can be inferred from the loss of the heterozygosity.
An alternative embodiment of the invention consists of classifying a sample of human tissue according to whether or not it contains an unmutated copy of a hsREC2 gene. The hsREC2 gene of the sample and the hsREC2 of a standard tissue can be compared by any technique known to those skilled in the art or to be developed. A sensitive technique suitable for the practice of this embodiment of the invention is single strand conformational polymorphism (SSCP). Orita, M., et al., 1989, Genomics 5:874-879; Hayashi, K., 1991, PCR Methods and Applic. 1:34-38. The technique consists of amplifying a fragment of the gene of interest by PCR; denaturing the fragment and electrophoresing the two denatured single strands under non-denaturing conditions The single strands assume a complex sequence-dependent intrastrand secondary structure that affects the strands electrophoretic mobility. Therefore comparison of an amplified fragment of a hsREC2 gene from a sample of tissue with the amplified fragment from a hsREC2 gene of a standard tissue is a sensitive technique for detecting mutations in the hsREC2 of the sample.
The absence of a copy of an unmutated hsREC2 gene in a sample of tissue indicates that the cells of the tissue have undergone or likely will undergo transformation into a malignant phenotype In a further alternative embodiment of the tissue sample can be classified by Southern blotting of the DNA of the sample. The presence of tissue specific bands in the blot is evidence that at least one copy of the REC2 gene of the sample has undergone a mutational event. In yet a further embodiment of the invention, the tissue sample can be classified by amplifying a fragment of the REC2 gene, by PCR, and analyzing the fragment by sequencing or by electrophoresis to determine if the sequence and length of the amplified fragment is that which can be expected from a normal REC2 gene.
Without limitation, particular types of tissue samples that can be classified according to the invention include tumors which are associated with cytogenetic abnormalities at bands 14q23-24. Such tumor types include renal cell carcinomas and ovarian cancers Mittelman, F., 1994, Catalog of Chromosome Aberrations in Cancer, 17 (Johansson, B. and Mertens,F. eds.) Wiley-Liss, New York, pp 2303-2484. Also suitable for classification according to the method of the invention are tumor types that show a loss of heterozygosity of markers linked to the region 14q23-24. Such tumor types include meningiomas, neuroblastomas, astrocytomas and colon adenomas. Cox, D.W., 1994, Cytogenetic Cell Genet. 66:2-9. Of particular interest is the high rate of breast adenocarcinomas that have been found to have either mutated hsREC2 genes or to have lost heterozygosity of the microsatellite DNA at the closely linked locus D14S258 In addition to the above described methods the embodiments of the invention include a kit comprising a pair of oligonucleotides suitable for use as primers to amplify a fragment of a hsRECl gene, which pair consists of a 5'-primer having a sequence of a fragment of SEQ ID NO:2 and a 3'-primer having a sequence of a fragment of its complement wherein the 3'-primer is complementary to a portion of the sequence of SEQ ID NO.2 that lies 3' of the location of the 5'-primer sequence The length of the 3' and 5'-primers is at least 12 nucleotides and preferably between about 16- and 25-nucleotides and more preferably between 18 and 24 nucleotides. The invention further consists of oligonucleotides having a sequence of a fragment of SEQ ID NO:2 or its complement and a label, which are suitable for hybridization with genomic blots of the hsREC2 gene. Labels include radiolabels such as 32P, fluorescent labels or any label known or to be developed that allows for the specific detection of a nucleic acid sequence The plasmid pcHsREC2, in which the hsREC2 cDNA is operably linked to a CMV immediate early promoter has been deposited on August 20, 1996, in the ATCC, Rockville, MD, and accorded accession No. 97685. The plasmid was deposited under the name "pcHuREC2," but is referred to herein as pcHsREC2 for consistency. The plasmid pcHsREC2 is derived from commercially available plasmid pcDNA3 (Invitrogen, Inc.) and contains a 1.2 Kb insert that encodes hsREC2, which can be removed from pcHsREC2 by cutting with the restriction enzymes Xbal and Kpnl.
EMBL-3-type Aphage clones, designated A5A and A1C, which contain a 12 Kb and 16Kb fragment of the 5' and 3' region of the hsREC2 gene, respectively, were deposited on August 20, 1996, as accession No. 97683 and No. 97682, respectively.
AFIXIl type Aphage clones, designated A5D2a and A7B1a, which contain a 14 Kb 18 and 14.9Kb fragment of the 5* and 3' region of the muREC2 gene of strain 129SVJ, respectively, were deposited on August 22, 1996 and August 20, 1996, as accession No. 97686 and No. 97684, respectively. The inserts of A5D2a and A7Bla are released by cutting with a Notl restriction enzyme. .6 THE REC2 PROMOTER .
The promoters of hsREC2 and muREC2 were cloned. The hs REC2 promoter was cloned by a two step PCR-based promoter walking technique. Briefly, blunt ended genomic fragements are made by digestion with Dral and Sspi, in the first and second step respectively. The restriction fragments are ligated to adapters. A primary PCR amplification is performed using a gene specific primer from the 5" extreme of the gene and an adapter specific primer. A secondary PCR is performed using nested, gene and adapter specific primers. The first step, primary and secondary gene specific primers were 5-CAG ACG GTC ACA CAG CTC TTG TGA TAA-3' (SEQ ID NO:8) and 5'-ACC CAC TCG TTT TAG TTT CTT GCT AC-3 (SEQ ID N09), respectively The second step promoter walking primary and secondary primers were 5-TAG AGA GAG AGA GAG AGC GAG ACA G-3' (SEQ ID NO. 10) and 5'-GTC GAC CAC GCG TGC CCT ATA G-3' (SEQ ID NO: 11), respectively The first step and second step fragments were 0.8 and 0 9 Kb in length respectively The mu REC2 promoter was sequenced by digestion of the clone A5D2a with Xbal. The promoter was found on the largest fragment, of about 7 Kb. The sequences of the hsREC2 and muREC2 are given in Figures 7A-7C and 7D-7F respectively.
The level of REC2 transcripts in cultured human foreskin fibroblasts had been shown to be increased when the cells were exposed to 137Cs irradiation. Several yeast genes have been identified that are radiation induceable and the radiation sensitive cis-acting conrol sequences from the promoters of such genes have been identified. See references cited in footnotes to Tables l-lll. The sequence of the hsREC2 and muREC2 promoters were therefore inspected for the presence of such sequences. Figure 7 demonstrates that numerous such sequences were present. Tables l-lll show the sequence of the yeast UV responsive elements t, their positions in the yeast gene in which they are found and the reference to the acientific publication where they are 19 SUBSTITUTE SHEET (RULE 26) described.
The radiation induceability of the hsREC2 gene was directly assayed using UV radiation and the luciferase reporter gene in transiently transfected HeLa cells. The hsREC2 promoter was operably linked to a luciferase reporter gene and to the SV40 enhancer, placed downstream of the poly A addition signal. Any strong enhancer can be used, e.g., the enhancer from Cytomegalovirus, Hepatitis B Virus, a-fetoprotein, Rous Sarcoma Virus or Simian Virus 40. In this construct hsREC2 promoter was, in the absence of radiation apporoximately as strong a promoter as the SV40 immediate early promoter. When the cells were UV irradiated (35 J/m2 UV) the hsREC2 promter showed an approximate two to three fold increase in activity. See Section 6.8, below A radiation induceable promoter can be used to increase the susceptibility of cells to radiation as, for example, in conjunction with radiation therapy of a cancer. A construct containing a hsREC2 promoter operably linked to a "suicide gene", e.g., herpes thymidine kinase, can be introduced into mitotically active cells using a retrovirus based vector. A tumor can be irradiated and, simultaneously, gancydovir, a DNA antimetabolite prodrug that is converted by herpes thymidine kinase, can be administered.
Those skilled in the art appreciate that the activity of the REC2 promoter can be further localized by testing the activity of the fragment atter deletions having been made. A functional, radiation induceable promoter that is smaller than the fragment of Figure 7 can be found. Accordingly as used herein a human REC2 promoter and a murine REC2 promoter is defined as a DNA having the sequence found in Figures 7A-7C or 7D-7F, respectively, or a fragment thereof, wherein said fragment is a promoter in HeLa cells. The terms hsREC2 promoter and muREC2 promoter refer to DNA molecules having the sequences found in Figures 7A-7C and 7D-7F respectively. A REC2 promoter from any species can be defined analogously. Accordingly, in one embodiment, the invention concerns a composition containing a only a defined number of types of DNA molecules, one of which molecules comprises a REC2 promoter. As used herein such composition is said to comprise an isolated and purified REC2 promoter. In an alternative embodiment, the invention concerns a plasmid having a bacterial origin of replication (henceforth a "cloning plasmid"), which plasmid comprises a mammalian REC2 promoter and SUBSTITUTE SHEET (RULE 26) PCT/1B97/01217 specifically a human or a murine REC2 promoter. Those skilled in the art will further appreciate that the cis-acting radiation sensitive control elements present in the sequences of Figure 7 can identified by systematic testing of fragments having the appropriate deletions Accordingly, there can be REC2 promoters, as defined above, that are less radiation induceable than the hsREC2 promoter. As used herein a mammalian REC2 promoter is said to radiation incduceable if the promoter shows at least a two fold increase in activity and a REC2 promoter is termed "three fold induceable" if it shows a three fold increase when tested under the conditions wherein hsREC2 gives at least a four fold increase In further embodiments the REC2 promoter is operably linked to a enhancer. The present invention is illustrated by use of the SV40 enhancer. Those skilled in the art appreciate that any enhancer that is as strong as the SV40 enhancer can be used. Alternative enhancers include Cytomegalovirus, Hepatitis B Virus, a-fetoprotein, Rous Sarcoma Virus or Simian Virus 40 enhancers.
Table I UASs of Saccharomyces cerevisiae DNA repair genes Gene Location Sequences SEQ ID NO References PHR1 -103 CGAGGAAGCAGT 15 13,14 RAD 2 RAD23 RAD51 RAD 5 4 Consensus -110 -166 -295 -215 -256 CGAGGAAGAAAA GGAGGCATTAAA GGTGGCGAAATT CGTTACCCTAT C GTTAC C CAAT GGAGGARRNANA CTC 16 17 16 19 ,16 21 SUBSTITUTE SHEET (RULE 26) Table II. UASs of Saccharomyces cerevisiae DNA repair genes Gene Location Sequences SEQ ID NO. References Rhp51+ -290 C GTT_C C CTAT 21 11 -260 CCTA_CCCTAA 22 RAD 51 -215 CGTTACCCTAT 23 12 RAD54 -256 CGTTACCCAAT 24 17 RNR3 -429 CGGTTGCCATG 18 Consensus CGTTACCCTAT 26 Table ill URSs of Saccharomyces cerevisiae DNA repair genes Gene Position Sequences SEQ ID NO References MAG -215 GTAGGTCGAA 27 1 PHR1 -103 CGAGGAAGCA 28 2 -109 CGAGGAAGAA 29 2 RAD2 -169 CGTGGAGGCA 1,2,3,4,5 RAD51 -157 CGTGGTGGGA 31 6, 12 DDR4 8 -271 CGAGGATGAC 32 1, " -322 CGTGGTTGAT 33 1,7 RNR2 -374 CGAGGTCGCA 34 8,9 RNR3 -467 CTAGGTAGCA 1, 10 rhp51+ -233 GTAGGTGTTA 36 11 -213 CTAGGTAACA 37 11 RAD16 -309 CATGGTTGCC 3 S 1 Consensus CGTGGTNGAA 39 1 A A CC References to Tables l-lll- 1. Xiao W. et. at., 1993, Mol. Cell. Biol., 13, 7213-7221 2. Sebastian, J. et. al , 1990, Mol. Cell. Biol., 10, 4630-4637 3 Madura, K. et. al., 1986, J Bacterid., 166, 914-923 4. Reynolds, P. et. al., 1985, EMBO J.,.4, 3549-3552 . Siede, W. et. al., 1989, Mol. Microbiol., 3, 1697-1707 6. Basile, G. et. al., 1992, Mol. Cell. Biol., 12, 3235-3246 7. Treger, J. M.. et. al.,1990, Mol. Cell. Biol., 10, 3174-3184 8. Elledge, S.J. et. al., 1989, Mol. Cell. Biol., 9, 5373-5386 9. Hurd, H. K. et. al. , 1989, Mol. Cell. Bio!, 9, 5359-5372 . Yagle, K. et. at., 1990, Mol. Cell. Biol., 10,5553-5557 11. Jang, Y. K.. et. al., May 23,1996, Molecular & General Genetics, 251 (2),167-175, 12. Aboussekhra A. et .al, 1992, Mol. Cell. Biol., 12, 3224-3234 13. Sancar, G. B. ,1985, Nucleic Acids Research, 13,8231-8246 14. Sancar, G. B. et. al., 1995, Nucleic Acids Research, 23, 4320-4328 . Jones, J. S. et. al., 1991, Nucleic Acids Research, 19,893-898 16. Watkins, J. F. et. al., 1993, Mol. Cell. Biol., 13, 7757-7765 17. Cole, G. M. et. al., 1989, Mol. Cell. Biol., 9, 3314-3322 18 Elledge S. J et. al. ,1989a, Mol. Cell. Biol., 7, 4932^940 22 .7 RFO-TRANSFFCTANTS ARE SFNSITIZED TO IRRADIATION One embodiment of the present invention is a plasmid or otherisolated purified DNA molecule in which a mREC2 cDNA is operably linked to a strong promoter, which is preferably a constitutive promoter, e.g., a CMV immediate early promoter. In a further embodiment the invention consists of amammlian cell that is transfected with such plasmid or isolated purified DNA amd which over expresses Rec2. The overexpression of Rec2 causes a mammalian cells to be hypersensitive to DNA damaging agents such as alkylating agents, e.g., cyclophosphamide, Y-ray or UV-irradiation.
Accordingly, the present invention can be used to sensitize a set of cells that can be selectively transfected with a Rec2 expressing plasmid. Such sensitization can be used in conjunction with conventional oncologic chemotherapy or irradiation therapy to treat malignant disease. 6. EXAMPLES 6.1 The production of recombinant hsREC2 protein by baculovirus infection of Autographica cat'rfornica To facilitate the construction of an hsREC2 expression vector, restriction sites for Xhol and Kpnl were appended by PCR amplification to a the hsREC2 cDNA The hsREC2 cDNA starting at nt 71 was amplified using the forward primer 5'-GAG CTCC.AG GC.TArr c ATG GGT AGC AAC AAA C-3' (SEQ ID NO: 14), which placed the Xhol and Kpnl sites (underlined) 5' of the start codon. The recombinant molecule containing the entire coding sequence of hsREC2 cDNA, can be removed using either Xhol or Kpnl and the unique Xbal site located between nt 1270 and 1280 of SEQ ID NO:2.
A vector, pBacGSTSV, for the expression of HsREC2 in baculovirus infected Spodoptera frugiperda (Sf-9) insect cells (ATCC cell line No. CRL1711, Rockville MD), was obtained from Dr. Zailin Yu (Baculovirus Expression Laboratory, Thomas Jefferson University). The vector pVLGS was constructed by the insertion of a fragment encoding a Schistosoma /aponicum glutathione S-transferase polypeptide and a thrombin cleavage site from pGEX-2T (described in Smith & Johnson, GENE 67:31 (1988)), which is hereby incorporated by reference, into the vector into the vector pVL1393. A polyA termination 23 signal sequence was inserted into pVLGS to yield pBacGSTSV. A plasmid containing the 1.2 Kb hsREC2 fragment was cut with Kpnl, the 3' unpaired ends removed with T4 polymerase and the product cut with Xbal. The resultant fragment was inserted into a Smal, Xbal cut pBacGSTSV vector to yield pGST/hsREC2.
Recombinant virus containing the insert from pGST/hsREC2 were isolated in the usual way and Sf-9 cells were infected. Sf-9 cells are grown in SF900II SFM (Gibco/BRL Cat # 10902) or TNM-FH (Gibco/BRL Cat # 11605-011) plus 10% FB5. After between 3-5 days of culture the infected cells are collected, washed in Ca" and Mg*+ free PBS and sonicated in 5ml of PBS plus proteinase inhibitors (ICN Cat # 158837), 1% NP-40, 250 mM NaCI per 5x107 cells. The lysate is cleared by centrifugation at 30,000 xg for 20 minutes. The supernatant is then applied to 0.5 ml of glutathione-agarose resin (Sigma Chem. Co. Cat # G4510) per 5x107 cells. The resin is washed in a buffer of 50 mM Tris-HCI, pH 8 0, 150 mM NaCI and 2.5 mM CaCI2, and the hsREC2 released by treatment with thrombin (Sigma Chem. Co. Cat # T7513) for 2 hours at 23°C in the same buffer. For certain experiments the thrombin is removed bv the technique of Thompson and Davie, 1971, Biochim Biophys Acta 250:210, using an aminocaproyl-p-chlorobenzylmide affinity column (Sigma Chem. Co Cat # A9527) 6.2 Detection of the Enzymatic Properties of hsREC2 protein Baculovirus produced hexahistidylhsREC2 was tested in a DNA reannealing assay as described in Kmiec, E.B., & Holloman, W.K., 1982, Cell 29:367-74 The results, Figure 3, showed that hsREC2 catalyzes the reannealing of denatured DNA. An optimal reaction occurred at about 1 hsREC2 per 50-100 nucleotides.
Further studies to characterize hsREC2 showed that it catalyzes the reaction ATP-ADP + P04. Similar to recA, at ATP concentrations of <100 /jM, there is cooperativity between hsREC2 molecules; the Hill coefficient (1.8) suggests that the functional unit for ATP hydrolysis is at least a dimer. Gel retardation experiments were performed to determine the ATP dependence of hsREC2 binding to ssDNA. The results of these experiments showed that hsREC2 binds ssDNA only in the presence of ATP or its non-hydrolyzable thio analog y-SATP. Figure A. Again the hsREC2 results parallel those of recA. 24 Further examples of specific assays using isolated and purified hsRec2 are as follows: 6.2.1 Binding to Single Stranded DNA A 73 nucleotide single stranded DNA (SS) was "P end labelled using polynucleotide ' kinase. DNA binding was carried out using 0.25 ng of labeled SS in 25 mM Tris-HCI, pH 7.4,10 mM MgCl2,4 mM ATP, and 1 mM DTT and protein. hsRec2-thioredoxin was partially purified on a Thiobond™ column (Invitrogen) and desalted/concentrated using a Microcon 30 spin column (Amicon). Approximately 0.3 fjg protein was added. The reaction mixture was incubated 30 min. at 37°C, following which sucrose was added to facilitate loading onto a polyacrylamide gel. The mixture was loaded onto a 12% nondenaturing gel in 90 mM Tris, 90 mM borate, pH 8.3, 2 mM EDTA for 3 hours at 150 V. The gel was then dried and exposed overnight. Approximately 3% of the label was retarded in the presence of ATP or yS-ADP, while reduced amounts of label were bound in the absence of either of ATP or yS-ADP. 6.2.2 Catalysis of Reannealing of DNA Reannealing of a 123 nucleotide fragment was determined as follows. The single stranded 123 nucleotide (SS) was n? end labelled using polynucleotide kinase. Varying amounts of affinity purified GST-hsRec2 fusion protein was added to 0.5 ng of SS in 25 /jl of 20mM TrisHCI pH 7.5, 10 mM MgCl2,0.5 mM DTT with 5 mM ATP optionally present. Samples were incubated 30 min. at 37°C, followed by phenol/chloroform extraction to stop the reaction, followed by a second 30 min. incubation at 37°C. The reaction mixture was then electrophoresed as in section 6.2.1, above, and autoradiographed. The results, shown in Figure 6, demonstrate that GST-hsREC2 catalyzes the reannealing of the SS in both the presence and absence of ATP. 6.3 Overexpression of hsRBC2 Suppresses UVC4nduced Mutation To determine whether the presence of hsRec2 protects cultured cells from UVC induced mutation a CHO cell line was transfected with a mixture of linearized pcHsREC2 and pCMVneo and a clone resistant to G418 was selected C15C8 hsREC2"). Elevated levels of hsREC2 expression were confirmed by immunoblotting using rabbit antisera raised to baculovirus produced hsRec2 fusion proteins.
SUBSTITUTE SHEET (RULE 26) Mutability was determined as follows. 1.6 x 106 15C8 hsREC2 cells were plated in a 100 mm petri dish and exposed to 0 or between 2.0 and 5.0 J/m2 UV radiation. After 7 days of culture, the remaining cells were exposed to 40 fjM 6-TG. Surviving cells had undergone an inactivation of the HPRT gene. After a further 7-10 days of culture the number ot colonies was counted. The mutation frequency was adjusted for the cloning efficiency of the population which was determined by plating a limiting number of cells without 6-TG.
The results showed that the untransfected, pCMVneo and 15C8 hsREC2 cells had mutation rates of 1.7, 6.2 and 0.4 per million, respectively, without UVC irradiation After UVC radiation the mutation rates observed were, in three experiments, between 94 and 16, 61 and 74, and 3 and 37, per million, for untransfected, pCMV transfected and 15C8 hsREC2 ceils, respectively. Thus, the expression of hsRECZ caused a marked decrease in the susceptibility of CHO cells to UVC induced mutation as well as a drop in the spontaneous mutation frequency. 6.4 Enhanced Repair of B-globin in Cultured, EB-transformed Human Lymphoblasts SC1, a chimeric vector designed to repair the mutation found in Sickle Cell Disease I5-globin, contained two blocks often 2'-0-methyl RNA residues each, flanking an intervening block of five DNA residues, see Figure 5B. When the molecule was folded into the duplex conformation, one strand contained only DNA residues while the other strand contained the RNA/DNA blocks. In this case, the internal sequence is complementary to the 6s globin sequence over a stretch of 25 residues that span the site of the 6s mutation, with the exception of a single base (T) which is in bold and designated with an asterisk. The five DNA residues flanked by RNA residues were centered about the mutant T residue in the coding sequence. Genomic sequences of the l$A, 6\ and closely-related 5-globin genes are also displayed in Figure 3 with the specific site of &s mutation printed in bold.
Lymphoblastoid cells were prepared as follows Heparm-treated blood was obtained from discarded clinical material of a patient with sickle cell disease. Mononuclear cells were prepared from blood (=8 ml) by density gradient centrifugation in Ficoll and 26 infected with Epstein-Barr virus which had been propagated in the marmoset cell line B95-8 (Coriell Institute for Medical Research #GM07404D). Infections were performed with addition of 0.1 mg leucoagglutinin PHA-L in 10 ml RPMI medium supplemented with 20% fetal bovine serum in a T25 flask. Cultures were fed twice a week starting on day 5 and were considered established once 60-70% of the cells remained viable at day 21. The 6A and 6s lymphoblastoid cells were maintained in RPMI medium containing 10% fetal bovine serum.
The EBV-transformed lymphoblastoid cells were transiently transfected with either the vector pcDNA3 or the vector having inserted h$REC2 cDNA (pcHsREC2). Transfection was done using mixtures of 15 pi DOTAP and 2.5 fjg DNA, as detailed below. After transfection the cells were incubated for 24 hours and then treated with varying amounts of SC1.
SC1 was introduced into the above-described lymphoblastoid cells homozygous for the Ss allele as follows. Cells (1 x 10s per ml) were seeded in 1ml of medium in each well of a 24-well tissue culture plate the day prior to the experiment. Transfections were performed by mixing chimeric oligonucleotides in amounts ranging from 0 to 250 ng, with 3 fj\ ot DOTAP (N-t1-(2,3-Dioleoyloxy)propyl]-N,N,N-trimethylammonium methylsulfate, Boehringer-Mannheim) in 20 ml of 20 mM HEPES, pH 7.3, incubated at room temperature for 15 min, and added to the cultured cells. After 6 h the cells were harvested by centrifugation, washed and prepared for PCR amplification following the procedure of E.S. Kawasaki, PCR Protocols, Eds. M.A. Innis, D.H. Celfand, J.J. Sninsky and T.J. White, ppl46-i52, Academic Press, (1990).
Correction of the single base mutation was assessed by taking advantage of well known restriction fragment length polymorphisms resulting from the 6s mutation, R.F. Greeves et al., 1981, Proc. Natl. Acad. Sci. 78:5081;J.C. Chang and Y.W. Kan, 1982, N. Eng. J Med. 307:30; S.H. Orkin et al., ibid., p. 32; J.T. Wilson et al., 1982, Proc. Natl. Acad. Sci. 79:3628. The A to T transversion in the Bs allele results in the loss of a Bsu36l restriction site (CCTGAGG). Thus, the 6s allele can be detected by Southern hybridization analysis of genomic DNA cut with Bsu36l. A 1.2 Kb Bsu36l DNA fragment of the li-globin gene present normally is absent in the 6s allele and is replaced by a diagnostic 1.4 Kb fragment. When genomic DNA recovered from homozygous Gs 27 WO 98/11214 PCT/IB97/01217 lymphoblastoid cells was analyzed by this procedure, the expected 1 4 Kb fragment was observed. However, two fragments were observed in DNA from cells transfected with the SC1 CRV. The presence of the 1.2 Kb fragment in addition to the 1.4 Kb fragment indicates partial correction of the 6s allele had taken place in a dose-dependent fashion.
The results of the experiment are shown in Figure 5A. At 100 ng and 250 ng of SC1 between 65% and 85% of the 6s alleles were mutated to SA alleles in the cells pre-transfected with pcHsREC2, compared to between 10% and 25% in the non pre-transfected cells and negligible levels in the control transfected cells. At levels of SC1 between 25 ng and 50 ng of SC1, no mutations were detected in any of the control cell populations while between 30% and 40% of the Rs alleles were mutated to lsA alleles m the cells pre-transfected with pcHsREC2.
These results show that the over expression of hsREC2 causes marked increase tn the susceptibility of a cell to mutation by a chimeric mutation vector such as SC1 6.5 Identification and Isolation of mREC2 gDNA Clones Genomic blots of human and murine, strain 129 SVJ, DNA were made using Xbal and BamHI digests. Following transfer to Zeta-Probe™ membranes (Bio-Rad) the membranes were prehybridized for 30' at 55°C in 0.25M NaHPO„, pH7 2, 7% SDS, 1 mM EDTA and hybridized overnight with a random primed full length HsREC2 probe Wash was 2X for 20" at 42°C in 0.04M NaHP04, pH7.2, 5% SDS, 1 mM EDTA and 1X each at 42°C and 50°C for 20' in 0.04M NaHP04, pH7.2, 1% SDS, 1 mM EDTA. The results were bands of the following sizes: Human-Xbal 6.0, 4.1, 2.6, 2.0 and 1.5 Kb; Human-BamHI 9.5,8.5, 6.5, 4.6, 1.5 Kb; Murine-Xbal 9.0, 6.0,4.1, 3.5, 1.9, 0.8 Kb; and Murine-BamHI 8 0, 2.7 and 1.8.
To identify and propagate clones containing mREC2 from cDNA or DNA libraries standard techniques for cloning were employed using A-phage libraries. A human genome library in EMBL-3 and a murine genomic library in AFIXII were screened. Phage plaques were transferred to hybridization filters by standard techniques and the filters were probed with radiolabeled hsREC2 cDNA. After hybridization the filters were washed. A wash consisting of twice at 42°C for 20' in 2x SSC, 0.1 % SDS followed by thrice at 50°C for 20' in the same solution was used to isolate murine gDNA clones. To 28 PCT/1B97A>1217 isolate human gDNA clones a the wash procedure was: twice 20 min. at 42°C in 40 mM NaHPO*, pH 7.2, 1 mM EDTA, and 5% SDS; followed by once for 20 min. at 50°C in the same solution except for 1 % SDS.
The 5' and 3' fragments of muREC2 and hsREC2 gDNA were recovered in the following Aphage clones: A5D2a (14 Kb insert, 5' muREC2); A7B1a (14.9 Kb insert, 3' muREC2Y, A5A (12 Kb insert, 5' hsREC2); A1C (16 Kb insert, 3' bsREC2), each of which has been deposited in the ATCC, Bethesda, MD.
Fragments of genomic clones can be used as probes of genomic blots to identify rearrangements deletions or other abnormalities of hsREC2 in tumor cells. Those skilled in the art further appreciate that by routine sequence analysis and comparison with the sequence of SEQ ID NO: 2, the boundaries of the exons and introns of hsREC2 can be identified. Knowing the sequence of at the intron/exon boundaries allows for the construction of PCR suitable for the amplification and analysis of each exon as alternatives to the methods of section 6.6. 6.6 Elevated Incidence of Abnormalities in hsREC2 in Adenocarcinomas of the Breast Samples of 30 primary ductal carcinoma of the breast were analyzed by Southern blot, probed with the hsREC2 cDNA and by a high resolution gel of the PCR product of the microsatellite marker D14S258, which is closely linked to the hsREC2 gene. Ten of the thirty samples gave abnormal results in one of the two assays and 3 showed abnormalities by both assays. In contrast none of 16 samples of primary renal cell carcinoma showed clear abnormalities in a Southern blot. 6.6.1 Loss of Heterozygosity of Microsatellite DNA Linked to hsREC2 The location of hsREC2 was found to be tightly linked to the proximal side of the microsatellite marker D14S258. Because there is extensive polymorphism in the lengths of microsatellite sequences most individuals are heterozygous at the D14S258 locus. Primers specific for unique sequences flanking the polymorphic locus can be used to generate PCR fragments whose length is allele specific. Primers specific for D14S258 were obtained from the Dr. Lincoln Stein, Whitehead Institute, MIT, Cambridge MA. The "5"' primer is 5,-TCACTGCATCTGGAAGCAC-3, (SEQ ID NO:12) and the "3"' primer is 29 S'-CTAACTAAATGGCGAGCATTGAGO' (SEQ ID NO:13). PCR was performed with a genomic DNa concentration of 2.0 ng/pl, a primer concentration of 10.0 fjM, 10.0 (jM dNTP, 500 jjM Tris HC1, pH 9.2, 17.5/yM MgCU, 160j;M (NH«)2SO«, and a polymerase concentration of 0.03 U/pl. Amplification was performed for 35 cycles of 50 seconds each, alternating between 57°C and 94°C, followed by an extension of 7 minutes at 72°C and preceded by an initial heat soak of 5 minutes at 94°C The expected product is about 160-170 nucleotides in length.
A comparison of the products of PCR amplification ot tumor and normal tissue control DNA using the flanking primers can reveal the loss of either or both D14S258 loci, which suggests that the linked hsREC2 has also been lost.
The results of analysis 7 of 30 samples breast tumors showed a complete or partial loss of one allele at locus D14S258.
These results show that instability and loss of a genetic locus tightly linked to the location ot hsREC2 is found in a large fraction of human ductal adenocarcinoma of the breast. 6.6.2 Frequent Rearrangements of hsREC2 Genomic DNA from samples of 16 primary renal and 30 primary breast tumors tumor tissue were digested with either Xbal or BamHl restriction enzymes, electrophoresed in a 0.8% agarose gels and processed for hybridization with labeled random primed copies made from the hsREC2 cDNA. After transfer, Zetaprobe™ blotting membranes were UV crosslinked, prehybridized at 65°C for 20 mm in 0.25M NaHP04, pH 7.4, 7% SDS, ImM EDTA and then hybridized overnight under the same conditions. The membranes were pre-washed once with 40mM NaHPO„, pH 7.2, 5% SDS, ImM EDTA at 42°C for 20 min, then washed repeatedly at 60°C in the same solution, except for 1 % SDS, until background levels were achieved in the periphery of the membrane. The filters were then exposed to film.
Six of the 30 examples of carcinoma of the breast showed rearrangements or abnormalities while none of the 16 samples of renal cell carcinoma showed clear rearrangements 6.7 Construction of a MuREC2ko containing ES ceil line The muREC2 gDNA clone A5D2a contains the first two exons. The second exon is located on 3.6 Kb Eco R1 fragment, approximately 1.2 Kb from the fragment's 5' border. The secnd exon contains a unique Stul site into which was inserted the IRES-(3geo poly A cassette, Mountford, P , et al., 1994, Proc. Natl. Acad. Sci. 91, 4303-4307. .ES cells were cultured on primary mouse embroyo fibroblasts according to standard protocols, Hogan, B., et al., 1996, Manipulating the Mouse Embryo, Cold Spring Harbor Press. Approximately 2x107 ES cells were transfected by electroporation with 25 pg linearized DNA. Selection was begun at 36 hours and continued until day 8 with 250/vg/ml G418 Thirty colonies were isolated and tested by Xbal digest and Southern blot; one colony was found to lack the wild type size Xbal fragment and to have a novel fragment of the predicted size. Transgenic mice are constructed from this ES ceil line by conventional techniques. Ibid. 6.8 The HsREC2 Promoter Is Radiation Induceable A 1 8 Kb fragment immediately 5' to the hsREC2 start codon was cloned. The fragment was tested as a promoter using the luciferase reported gene construct, pGL2, (Promega Cat. No. E1611), luciferase activity was measured using the luciferase reported test kit (Boehringer Mannheim Cat. No 1669 893).
The activity of the promoter is assayed in HeLa cells as follows. The HeLa eels are trypsinized on day -1 and plated at 6.6x10s / 60 mm well in 3.0 ml of DMEM. On day two at -1 h the medium is replaced with serum free medium and the cells are transfected with various quantities of the plasmid with DOSPER at a DNA: DOS PER ratio of! 4. At 5 hour an additional 3.0 ml of medium supplemented with FBS is added; at 24 hours the cells are irradiated with UV light (Stratalinker). Cells are harvested at 48 hours and proteins extracted and assayed. Control experiments done with the same plasmid having the SV40 immediate early promoter in place of the bsREC2 promoter. 31 DNA Added (Micrograms) UV Irradiation (joules/meter2) 3^g 2.4/ug 1-2^g 0.6/ig 0 J/m2 655.6' 4942 27.5 32.8 J/m2 951 5 1287 28.9 28.7 J/m2 1033.6 1398 .8 44.2 J/m2 1134.6 1786 84.89 68 4 1. The corresponding luciferase is 513.9 pSV40-luc-SV40 enhancer at 0 Joules/meter. 2. The corresponding luciferase is 384 pSV40-luc-SV40 enhancer at 0 Joules/meter When the 3" 0.8 Kb of the hsREC2 promoter was tested beginning with nt 869 of SEQ ID NO: 5, it was determined that this 0.8 Kb fragment contains a promoter having reduced activity but which is also shows an about 5 fold induceability with 35 J/m2 UV radiation in HCT 116, which cell line contains a normal p53 gene. The preferred form of the REC2 induceable promoter in HCT 116 is the shortened form starting at nt 869 6.9 The Expression of REC2 Causes increased Radiation Sensitivity UV irradiation induces apoptosis in stable transfectants expressing wild-type HsRec2 but not truncated or full length with an altered tyrosine 163 site. In order to measure the effects of REC2 expression on the rate of UV induced radiation CHO cells were irradiated. During the 24 hour long recovery period following irradiation, more CHO cells expressing wild-type HsRec2 were observed to die than the control cells that expressed an irrelevant or nonfunctional proteins. To determine whether cell death was a result of apoptosis, asynchronous cells were irradiated at a dose of 15 J/m2, and fixed in ethanol at 24, 48 and 72 hours following irradiation. FACS analysis was conducted as folows: Cells were trypsinized, washed once with PBS and fixed in 70% ethanol at least 30 minutes at 4°C Cell pellets were treated with DNase-free Rnase for 30 minutes at 70° C at a final concentration of 0.16 mg/ml and stained in propidium iodide (0.05 mg/ml) 32 for 15 minutes, then stored overnight prior to analysis by FACS. The FACS analysis and determination of the percentage of cells in G1, S and G2 phases (Multicycle Flow program) was carried out in the Cell Cycle Center at the Kimmel Cancer Institute of Thomas Jefferson University. Cells from duplicate cultures were harvested at the same time points, and frozen at -80°C. for DNA isolation. DNA was isolated using a QIAGEN Blood Kit (QIAGEN Inc., Chatsworth, CA) and stored at 4° C. until run on gels. DNA was run o 1 % agarose gels in TAE buffer and stained 30 minutes with a 1:10,000 dilution of SYBR Green I (FMC, Rockland, ME). Gels were then scanned using a Fluorfmager (Molecular Diagnostics, San Diego, CA).
Four cell types were used for analysis; CHO cells containing the empty vector (Neor), CHO cells expressing HsRec2A 103-350 (3D2), HsRec2ala63 (PH4), and the wild-type HsRec2 (15C8) A sub-Gl population was detected at 24, 48, and 72 hours following irradiation for CHO cells expressing the wild-type HsRec2 only. To confirm that apoptosis was occurring, DNA was isolated from cells, and run on a 1 % agarose gel, stained with SYBR Green I and scanned. For each time interval compared, 15C8 exhibited a more pronounced ladder than the other clones. Although there appears to be a small amount of apoptosis for the clone expressing HsRec2ala63 it is considerably lower than for the wild-type HsRec2 clone, and neither the Neor or the transfectants expressing the truncated protein are comparable Therefore, the G1 delay and apoptosis require the wild-type HsRec2, and suggests that perhaps cooperation between a mutant p53 present in CHO cells and Rec2 may be responsible for genome surveillance in these cells.
The results of the FACS analysis of the HsRec2 expressing and the Neor expressing clones are given in Figures 8A-8H. 33 SEQUENCE LISTING (1) GENERAL INFORMATION (i) APPLICANT: Kmiec, Eric B.
Holloman, William K.
Rice, Michael C.
Smith, Sheryl T.
Shu, Zhigang (n) TITLE OF THE INVENTION: Mammalian and Human Rec2 (ill) NUMBER OF SEQUENCES: 39 (IV) CORRESPONDENCE ADDRESS: (A) ADDRESSEE: Kimeragen, Inc.
(B) STREET: 300 Pheasant Run (C) CITY: Newtown (D) STATE: PA (E) COUNTRY: USA (F) ZIP: 18940 (V) COMPUTER READABLE FORM: (A) MEDIUM TYPE: Diskette (B) COMPUTER: IBM Compatible (C) OPERATING SYSTEM: DOS (D) SOFTWARE: FastSEQ for Windows Version 2.0 (vi) CURRENT APPLICATION DATA: (A) APPLICATION NUMBER: (B) FILING DATE: (C) CLASSIFICATION: Ivil) PRIOR APPLICATION DATA: (A) APPLICATION NUMBER: (B) FILING DATE: 34 (vill) ATTORNEY/AGENT INFORMATION: (A) NAME: Hansburg, Daniel (B) REGISTRATION NUMBER: 36156 (C) REFERENCE/DOCKET NUMBER: 7991-010-999 (ix) TELECOMMUNICATION INFORMATION: (A) TELEPHONE: 215-504-4-444 (B) TELEFAX: 215-504-4545 (C) TELEX: (2) INFORMATION FOR SEQ ID NO:l: i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 350 amino acids (B) TYPE: amino acid (C> STRANDEDNESS: single (D! TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:l: Met Gly Ser Lys Lys Leu Lys Arg Val Gly Leu Ser Gin Glu Leu Cys 10 15 Asp Arg Leu Ser Arg His Gin lie Leu Thr Cys Gin Asp Phe Leu Cys 25 30 Leu Ser Pro Leu Glu Leu Met Lys Val Thr Gly Leu Ser Tyr Arg Gly 40 45 Val His Glu Leu Leu Cys Met Val Ser Arg Ala Cys Ala Pro Lys Met 50 55 60 Gin Thr Ala Tyr Gly lie Lys Ala Gin Arg Ser Ala Asp Phe Ser Pro 65 70 75 80 Ala Phe Leu Ser Thr Thr Leu Ser Ala Leu Asp Glu Ala Leu His Gly 85 90 95 Gly Val Ala Cys Gly Ser Leu Thr Glu lie Thr Gly Pro Pro Gly Cys 100 105 110 Gly Lys Thr Gin Phe Cys lie Met Met Ser lie Leu Ala Thr Leu Pro 115 120 125 Thr Asn Met Gly Gly Leu Glu Gly Ala Val Val Tyr lie Asp Thr Glu 13C 135 140 Ser Ala Phe Ser Ala Glu Arg Leu Val Glu lie Ala Glu Ser Arg Phe 145 150 155 160 Pro Arg Tyr Phe Asn Thr Glu Glu Lys Leu Leu Leu Thr Ser Ser Lys 165 170 175 Val His Leu Tyr Arg Glu Leu Thr Cys Asp Glu Val Leu Gin Arg lie 180 185 190 Glu Ser Leu Glu Glu Glu lie lie Ser Lys Gly lie Lys Leu Val lie 195 200 205 Leu Asp Ser Val Ala Ser Val Val Arg Lys Glu Phe Asp Ala Gin Leu 210 215 220 Gin Gly Asn Leu Lys Glu Arg Asn Lys Phe Leu Ala Arg Glu Ala Ser 225 230 235 240 Ser Leu Lys Tyr Leu Ala Glu Glu Phe Ser He Pro Val lie Leu Thr 245 250 255 Asn Gin lie Thr Thr His Leu Ser Gly Ala Leu Ala Ser Gin Ala Asp 260 265 27 Q Leu Val Ser Pro Ala Asp Asp Leu Ser Leu Ser Glu Gly Thr Ser Gly 275 280 285 Ser Ser Cys Val lie Ala Ala Leu Gly Asn Thr Trp Ser His Ser Val 290 295 300 Asn Thr Arg Leu lie Leu Gin Tyr Leu Asp Ser Glu Arg Arg Gin lie 305 310 315 320 Leu He Ala Lys Ser Pro Leu Ala Pro Phe Thr Ser Phe Val Tyr Thr 325 330 335 lie Lys Glu Glu Gly Leu Val Leu Gin Ala Tyr Gly Asn 5er 340 345 350 (2) INFORMATION FOR SEQ ID NO:2: U) SEQUENCE CHARACTERISTICS: (A) LENGTH: 17 97 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single !D> TOPOLOGY: linear (li) MOLECULE TYPE: cDNA (XI) SEQUENCE DESCRIPTION: SEQ ID NO:2: CGGACGCGTG GGCGCGGGGA AACTGTGTAA AGGGTGGGGA AACTTGAAAG TTGGATGCTG 60 CAGACCCGGC ATGGGTAGCA AGAAACTAAA ACGAGTGGGT TTATCACAAG AGCTGTGTGA 120 36 ccgtctgagt agacatcaga tccttacctg tcaggacttt ttatgtcttt ccccactgga 180 gcttat gaag gtgactggtc tgagttatcg aggtgtccat gaacttctat gtatggtcag 240 cagggcctgt gccccaaaga tgcaaacggc ttatgggata aaagcacaaa ggtctgctga 300 ttt ct cac ca gcattcttat ctactaccct ttctgctttg gacgaagccc tgcatggtgg 360 tgtggcttgt ggatccctca cagagattac aggtccacca ggttgtggaa aaactcagtt 420 ttgtataatg atgagcattt tggctacatt acccaccaac atgggaggat tagaaggagc 480 tgtggtgtac attgacacag agtctgcatt tagtgctgaa agactggttg aaatagcaga 540 atcccgtttt cccagatatt ttaacactga agaaaagtta cttttgacaa gtagtaaagt 600 tcatctttat cgggaactca cctgtgatga agttctacaa aggattgaat ctttggaaga 660 agaaattatc tcaaaaggaa ttaaacttgt gattcttgac tctgttgctt ctgtggtcag 720 aaaggagttt gatgcacaac ttcaaggcaa tctcaaagaa agaaacaagt tcttggcaag 780 agaggcatcc tccttgaagt atttggctga ggagttttca atcccagtta tcttgacgaa 840 tcagattaca acccatctga gtggagccct ggcttctcag gcagacctgg tgtctccagc 900 tgatgatttg tccctgtctg aaggcacttc tggatccagc tgtgtgatag ccgcactagg 960 aaatacctgg agtcacagtg tgaatacccg gctgatcctc cagtaccttg attcagagag 1020 aagacagatt cttattgcca agtcccctct ggctcccttc acctcatttg tctacaccat 1080 caaggaggaa ggcctggttc ttcaagccta tggaaattcc tagagacaga taaat gtgca 1140 aacctgttca tcttgccaag aaaaatccgc ttttctgcca cagaaacaaa at att gggaa 1200 agagtcttgt ggtgaaacac ccatcgttct ctgctaaaac atttggttgc tactgtgtag 1260 actcagctta agtcatggaa ttctagagga tgtatctcac aagtaggatc aagaacaagc 1320 ccaacagtaa tctgcatcat aagct gattt gataccatgg cactgacaat gggcactgat 1380 ttgataccat ggcactgaca atgggcacac agggaacagg aaatgggaat gagagcaagg 1440 gttgggttgt gttcgtggaa cacataggtt ttttttttta actttctctt tctaaaatat 1500 ttcattttga tggaggtgaa atttatataa gatgaaatta accattttaa agtaaacaat 1560 tccgtggcaa ctagatatca tgatgtgcaa ccagcatctc tgtctagttc ccaaatattt 1620 catcaccccc aaaagcaaga cccataacca ttatgcaagt gttcctattt ccccctcctc 1680 ccagctcctg ggaaaccacc aatctacttt ttttctatgg ctttacctaa tctggaaatt 1740 tcaaataaat gggatcaaat agtttcccaa aaaaaaaaaa aaaaaaaaaa aaaaaaa 1797 (2) INFORMATION FOR SEQ ID NO: 3: (l) SEQUENCE CHARACTERISTICS: (A) LENGTH: 350 amino acids (B) TYPE: amino acid (C) STRANDEDNESS: single <D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 3: Met Ser Ser Lys Lys Leu Arg Arg Val Gly Leu Ser Pro Glu Leu Cys 37 1 Asp Arg Leu Ser Arg Tyr Leu lie Val Asn Cys Gin His Phe Leu Ser 25 30 Leu Ser Pro Leu Glu Leu Met Lys Val Thr Gly Leu Ser Tyr Arg Gly 40 45 Val His Glu Leu Leu His Thr Val Ser Lys Ala Cys Ala Pro Gin Met 50 55 60 Gin Thr Ala Tyr Glu Leu Lys Thr Arg Arg Ser Ala His Leu Ser Pro 65 70 75 80 Ala Phe Leu Ser Thr Thr Leu Cys Ala Leu Asp Glu Ala Leu His Gly 65 90 95 Gly Val Pro Cys Gly Ser Leu Thr Glu lie Thr Gly Pro Pro Gly Cys 100 105 110 Gly Lys Thr Gin Phe Cys lie Met Met Ser Val Leu Ala Thr Leu Pre 115 120 125 Thr Ser Leu Gly Gly Leu Glu Gly Ala Val Val Tyr He Asp Thr Glu 130 135 140 Ser Ala Phe Thr Ala Glu Arg Leu Val Glu lie Ala Glu Ser Arg Phe 145 150 155 160 Pro Gin Tyr Phe Asn Thr Glu Glu Lys Leu Leu Leu Thr Ser Ser Arg 165 170 175 Val His Leu Cys Arg Glu Leu Thr Cys Glu Gly Leu Leu Gin Arg Leu 180 185 190 Glu Ser Leu Glu Glu Glu lie He Ser Lys Gly Val Lys Leu Val lie 195 200 205 Val Asp Ser lie Ala Ser Val Val Arg Lys Glu Phe Asp Pro Lys Leu 210 215 220 Gin Gly Asn lie Lys Glu Arg Asn Lys Phe Leu Gly Lys Gly Ala Ser 225 230 235 240 Leu Leu Lys Tyr Leu Ala Gly Glu Phe Ser lie Pro Val lie Leu Thr 245 250 255 Asn Gin lie Thr Thr His Leu Ser Gly Ala Leu Pro Ser Gin Ala Asp 260 265 270 Leu Val Ser Pro Ala Asp Asp Leu Ser Leu Ser Glu Gly Thr Ser Gly 275 280 285 Ser Ser Cys Leu Val Ala Ala Leu Gly Asn Thr Trp Gly His Cys Val 290 295 300 Asn Thr Arg Leu lie Leu Gin Tyr Leu Asp Ser Glu Arg Arg Gin lie 305 310 315 320 Leu lie Ala Lys Ser Pro Leu Ala Ala Phe Thr Ser Phe Val Tyr Thr 325 330 335 38 PCT/IB97/D1217 lie Lys Gly Glu Gly Leu Val Leu Gin Gly His Glu Arg Pro 340 345 350 (2) INFORMATION FOR SEQ ID NO: 4: (l) SEQUENCE CHARACTERISTICS: (A) LENGTH: 1525 base pairs (B) TYPE: nucleic acid -{C) STRANDEDNESS: single (D) TOPOLOGY: linear (il) MOLECULE TYPE: cDNA |xi) SEQUENCE DESCRIPTION: SEQ ID NO: 4: GGGAGCCCTG GAAACAT GAG CAGCAAGAAA CTAAGACGAG TGGGTTTATC TCCAGAGCTG 60 TGTGACCGTT TAAGCAGATA CCTGATTGTT AACTGTCAGC ACTTTTTAAG TCTCTCCCCA 120 CTAGAACTTA TGAAAGTGAC TGGCCTGAGT TACAGAGGTG TCCACGAGCT TCTTCATACA 180 GTAAGCAAGG CCTGTGCCCC GCAGATGCAA ACGGCTTATG AGTTAAAGAC ACGAAGGTCT 240 GCACATCTCT CACCGGCATT CCTGTCTACT ACCCTGTGCG CCTTGGATGA AGCATTGCAC 300 GGTGGTGTGC CTTGTGGATC TCTCACAGAG ATTACAGGTC CACCAGGTTG CGGAAAAACT 360 CAGTTTTGCA TAATGAT GAG TGTCTTAGCT ACATTACCTA CCAGCCTGGG AGGATTAGAA 420 GGGGCTGTGG TCTACATCGA CACAGAGT CT GCATTTACTG CTGAGAGACT GGTTGAGATT 480 GCGGAATCTC GTTTTCCACA ATATTTTAAC ACT GAGGAAA AATTGCTTCT GACCAGCAGT 540 AGAGTT CAT C TTTGCCGAGA GCTCACCTGT GAGGGGCTTC TACAAAGGCT TGAGTCTTTG 600 GAGGAAGAGA TCATTTCGAA AGGAGTTAAG CTTGTGATTG TTGACTCCAT TGCTTCTGTG 660 GTCAGAAAGG AGTTTGACCC GAAGCTTCAA GGCAACATCA AAGAAAGGAA CAAGTTCTTG 720 GGCAAAGGAG CGTCCTTACT GAAGTACCTG GCAGGGGAGT TTTCAATCCC AGTTATCTTG 780 ACGAATCAAA TTACGACCCA TCTGAGTGGA GCCCTCCCTT CTCAAGCAGA CCTGGTGTCT 840 CCAGCTGATG ATTTGTCCCT GTCTGAAGGC ACTTCTGGAT CCAGCTGTTT GGTAGCTGCA 900 CTAGGAAACA CATGGGGTCA CTGTGTGAAC ACCCGGCTGA TTCTCCAGTA CCTTGATTCA 960 GAGAGAAGGC AGATTCTCAT TGCCAAGTCT CCTCTGGCTG CCTTCACCTC CTTTGTCTAC 1020 ACCATCAAGG GGGAAGGCCT GGTTCTTCAA GGCCACGAAA GACCATAGGG ATACTGTGAC 1080 CTTTGTCTAG TGCTGATTGC ATGTGACTCA TGAAATGAAA CAGGACTGCG CTGCTTGGAA 1140 AAAGGAAACG GAAGCCAACA TAATGAGGAT TAATTGGTTG GTTGCTGTTG AGGTGGTAAC 1200 AGTGATTTCA GACCCGGAAG GTGAAGATGA AGAAGCCTTT ATCCAGTCTC TGGATGCAGA 1260 GGCTAGGGGC TCCACCACCG TGGGATGTCA GCGGCCATCG TAATAATTTG CACTTACACA 1320 AGCACCTTTC AGCCATGCCC CTCAAAGTGG TTCAGCCACA TTAATTAATT AAAGCCCACA 1380 ATCCCCCTAG GGAGAGCAGG AGGGGGACTA ACAAGATTTG TAATTACAGA AGGGAAAATT 1440 TCCGAATAAA GTATTGTTCC GCCAAAAAAA AAAAAAAAAA AAAAAAAAAA AAAAAAAAAA 1500 AAAAAAAAAA AAAAAAAAAA AAAAA 1525 39 (2) INFORMATION FOR SEQ ID NO:5: (i! SEQUENCE CHARACTERISTICS: (A) LENGTH: 1699 base pairs (3) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (11) MOLECULE TYPE: Other (XI) SEQUENCE DESCRIPTION: SEQ ID NO:5: CGACGGCCCG GGCTGGTATT ATAGCAGGTA TCACTTGGTT TTCTACTGGG GGAAACAAGT 60 CATTGCTAAC AAATTCCCAT GGGAGAGAAA T GAGGAGGAT GTATTTTTGT TTGTGAGAGG 120 TGTGTATGTA TGTATATTGT GTGTGCGTGT GTGTGTGTGT GAGAGAGAGA GATTGATTCA 160 GTCTGA7TCA GAGAATTTAG GTGTTAAATA GAAATTTGGG CCATGGTATT GGAAATAAAC 240 AAATATATAC ATTCTCAGTA TACATATATT TTCATTCCAA AATGTTACTT CTTTTCTGAT 300 AACTATATTG CTTTATTCCC TTGGATCCAT GAAGAGTTCC TGTTTCAGTT CGTTCCAGAG 360 GATACTTCTT TAC CAT CT CA ATGAGATATA CAGCTTCTCC TTTGTATGCA TTAAGAGACT 420 CACAGTAATT CTTTTTTAGC TCTGTGAAGA TAAATCTTTC ATGAGCCTCA TTTACCCCTA 480 GCAAAGTACA ATAGT GAAAT TTAACTGCAT GTGAGAATAT AAGCAGCTAG TGTAATAAAG 540 AACATTTTGG GCCAGGTCTG ATCGCTCATG CCTGTAATCC CAGCACTTTA GGAGGTCAAG 600 GCGAGAGGAT CACTTGAGCC CAGGAGTTCG AGACCAGCTT GGGCAACATG GCAAAACCCT 660 GTCTCTACAA AAAATACAAA AATTGGGCAG GCATGGTGTC GACCCAGTCT CTACAAAAAA 720 TACAAAAATT AGCCAGACAT GGTGGTGCAC GCTTGTGGTC CCAGCTACTT GGGAGGCTGA •780 GGTAGGAGGA TTGCTTGAGC CCAGGAGGGG GAGGTTGCAG TGAGCTGAGA TCGAGCCACT 840 GCACTCCAGC TGGGGTGACA GAGCCAGACC TGTCTCGCTC TCTCTCTCTC TCTATATATA 900 TATATTTAAA AAGAACATTT TAATACTGCA GTGATAAAAT CTCATTTGAT TCAGAAGGTG 960 TGCTCTGACT CCTAGAAAAA GGAAGAGTCA AATATGATTA TGGACTTGCA GTAGAGTGTA 1020 ATGGTTAAGA GGATAGGTTT CAGAATTAGA CTGCCTGGAT TCAAATTCTG GATCAGTTAT 1080 TTATGGTTTC TGGTGACAAT GGACTAGCTA ACTTTTCCAG GCTTTAGTTT TCTCATATGT 1140 AAAAAAGGGG CCAATAATCT ACTTTCCTTC TAGGGCTATT GAGAAGATTA AATGTGATAA 1200 TTTAGATAAG TTTTGGAACA GTGCCTGGTA TGTGGTAGGT GCTCCATAAA TATACCTATT 1260 GCCGTTACAG TGCAATGTAA ATTGTTACAG TGCAATAGAC TTTCTAGTAG TTCTGTTTGG 1320 AAATATGCCT TGAAAGTTAA TTACATTTCC AAATAAAATT TATACATGCA TTGGAACATT 13B0 TTAAGATGCT CTACAAATGT GAAGTGGTAC TATATTCATG TAGTAAATAT CAATTAATTG 1440 TGTGAAATTA TATTTGAGGT TGCCTTGTAG ATTTTCTATG TGCCTGTTTG ACGAACAATT 1500 GTCCCTCCTA TTTAAAACAT TTAAAAAGGT TCTATAGCAT TCCTTTAT CA GTAATATTTT 1560 TAACACAATA TGTTTCATTT TGCATATGGA GAAACTTGAG GAATTTTTAA TTTTGTTTTG 1620 GATAGCCTAT TCACTATCAC TTATGTTATA TTCTGTTGTT TTTTTCATGG TT CTT CTTTT 1660 CTTTGCTGGA TCTGGAGGC 1699 40 (2) INFORMATION FOR SEQ ID NO:6: (l) SEQUENCE CHARACTERISTICS: (A) LENGTH: 2147 base pairs (B) TYPE: nucleic acid (C> STRANDEDNESS: single (D) TOPOLOGY: linear (il) MOLECULE TYPE: Other (xi) SEQUENCE DESCRIPTION: SEQ ID NO:6: TATCTCAGTA GCACGTGCAC ATAGCAACTA CAATACCTGT CACATAAATG TAGTTACTTG 60 AATATATGTC TCTTCATTCT TCAATTGTAA GTATGCAAAA GGGAGGACAT AAGCTTAGCA 120 TAGCATGTGC TTAATATTGG TGAAAGAAAC AAATGAATAG AGAATGTTAT ATTTGGAGAG 180 TTTATATTAT ATTTGGGAGA GTAGGGAAAA AACTTGAAGC CATAAGCAGA ATCGAGGGCA 240 AGTAGTGAGA GTGGTACTGT TAAATCAGAG TGATTATTGC TAAGGTCTTT GTAATTTGGG 300 GTTGTAGGTG TTTTTTGTTT TTGTTGTTTG AGGGTCTGAA TTTATTCGTT ATATGATGTT 360 ATTGCCTGGA ACTACCTTAT CTGAGAAGCA GTAGGCAATA GAGTAGCGTA TAAATGTTGG 420 TAAATTTTCT CTTAAGGAAA CAAATTAT C C TTACAAAATT CCAACTGAAA GAAATAAAGA 480 GAATGTATCT TGGTTTTGTG TGGAGAGAGG GAAGTAGAAG ATGGGGGATG AAGAGAGAGA 540 GGAGGGTTAT TTATTGGGCT ATATATAGT G TTGGTAGTAG GAATCTTAAT TCTTGTGTGT 600 AGTTTTGTTC TTTTGTGTAT AGTTATTGAT TATTACTTTA TTCCATGGGA ATAATGAGTT 660 CCTATTATTT C T GGAGGATA TTTTGCCATT TCGATGAGAC ACACAGCCTC TTCTTTGCTA 720 T GCAATATTA CGAGATTACA ACAGTTCTAA CTCCCTGAAG ACAAATACTT CATGAGTCTC 780 ATTAGCTATC TAAGCTATAG GAAGAGCAGA ATTTAATTCT ACATGGAAAC AGTAAGAAGC 840 TAGTATAATG AAGAATTTTA TTGATATCAC TTGATTGAAA TTTGTTCTGA CTCTTTAGAA 900 AAAGCAAGGG TGAAATAAGA TTTGTGATTC TACAGTAGTA ATGGGTAAGA GGATAGGTCT 960 CAGGACAAAC TGCCTAATGA AACCCTAAAT CTGTTATTTA TTTATTTTCT GATGACAGTG 1020 GGATAACTGA CATTTACACA TTAGCTTTCT CATAT GTAAA AAAGAAATTT TATTTTTATT 1080 ATAGTCTGTC AAGGAATATT AAATATAAGG TTTTGGAGCA TGGTTGATAT TTAGCAGATG 1140 TCTGTTCATT CTTGATCAGT ATAGAGTTGC CACTTGGAAA ATGCATCTTG AAGATTACAT 1200 AACCAGACAA AATTTGTTAG TAACACTCAG TGGTCTTAAG ATGTTATAAG TGACGGGCTA 1260 GTCGTGGTAA TCAACTTGAT ACCTTGACCC TCAGGAGAAG AGGGATTGTC TCCATCGGAT 1320 GGGCCTGTGA GCATATCTGT GGGGACGTTT TTCTTGGACT GC CTAGTTGA TGGAAAAGGG 1380 CTTGGCTCAG TGTCAGTGGT CCTTCTTATG GTGAGCAAGC TGGGGGAAGC GTTGCAGTAA 1440 GCAGTAGTCC TTTGTGGTCT CAGCTTCCTT TTCTTCTCTC TTCTTTCTTT CTTTCTTTCT 1500 TTCTTTCTTT CTTTCTTTCT TTCTTCCTTC CTTCCTTCCT TTTCTCTCTT TCTTTCTTTA 1560 GTTCCGTTCG TTTGTTCATT CGTTCGTTTT TCGAGACAGG GTTTTTCTGT ATAGCCCTGG 1620 CTGTCCTGGA ACT CACTTT G TAGACCAGGC TGTCCTCGAA CTCAGAAATC CGCCTGCCTC 1680 TGCCTCCCTG TGAGTGCTGG AATTAAAGGC ATGCGCCACC CCGCCCGGCT TCTCAGCTTC 1740 41 CATTTCTGTT CAAGCTCTTG CCTTCAGCTC CTGCCTTGGC TTTCTGAGAC AAAGGCATAT 1800 AATCTGTAAG CCAAATCAAA CTTTTCTTCT CAACTTGCTT TTGGCCAGTG TTTTATTACA 1860 GCGACTAAAG GCAAACTAGA CTACTATGTA AATGGGAAGC ACTGTTAAAG T CAAGTAATA 1920 GCAAAAGATT ACATGGCCTG GATTTTTTGA GGTTGCTTAC TTTCTCTGTG TACCCGGTTG 1980 TAAGTGTCTT TCCTACTTTT TTTATTAGCA TTTTTTTTCC ATGTTTTGCT TTGCACATAG 2040 AGAAGTTTGA AGCACTTTAT TTTGTAGGGT GTTTTGTATA ATCTGTCCAC CATCATTTTT 2100 ATTGTTTTCT TATGTTTTTT CAAGATTTCT TTGGGAGCCC TGGAAAC 2147 (2) INFORMATION FOR SEQ ID NO:7: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 5 amino acids (B) TYPE: amino acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ll) MOLECULE TYPE: peptide Ixi) SEQUENCE DESCRIPTION: SEQ ID NO:7: Gly Lys Thr Gin Met 1 5 (2) INFORMATION FOR SEQ ID NO:8: (1) SEQUENCE CHARACTERISTICS: (A) LENGTH: 27 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:8: CAGACGGTCA CACAGCTCTT GTGATAA 27 (2) INFORMATION FOR SEQ ID NO:9: (l) SEQUENCE CHARACTERISTICS: (A) LENGTH: 26 base pairs IB) TYPE: nucleic acid 42 <C> STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:9: ACCCACTCGT TTTAGTTTCT TGCTAC (2) INFORMATION FOR SEQ ID NO:10: (l) SEQUENCE CHARACTERISTICS: (A) LENGTH: 25 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:10 TAGAGAGAGA GAGAGAGCGA GACAG (2) INFORMATION FOR SEQ ID NO:11: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 22 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:11 GTCGACCACG CGTGCCCTAT AG (2) INFORMATION FOR SEQ ID NO:12: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 19 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear 43 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:12. TCACTGCATC TGGAAGCAC (2) INFORMATION FOR SEQ ID NO:13: (i) SEQUENCE CHARACTERISTICS": (A) LENGTH: 23 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single <D> TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO.-13: CTAACTAAAT GGCGAGCATT GAG (2) INFORMATION FOR SEQ ID NO:14: (l) SEQUENCE CHARACTERISTICS: (A) LENGTH: 32 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear 1X1) SEQUENCE DESCRIPTION: SEQ ID NO:14: GAGCTCGAGG GTACCCATGG GTAGCAAGAA AC (2) INFORMATION FOR SEQ ID NO:15: (l) SEQUENCE CHARACTERISTICS: (A) LENGTH: 12 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 15 44 CGAGGAAGCA GT (2) INFORMATION FOR SEQ ID NO:16: il) SEQUENCE CHARACTERISTICS: (A) LENGTH: 12 base pairs (B) TYPE: nucleic acid (C) STRANDEDNES S: s ingl e- (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:16 CGAGGAAGAA AA (2) INFORMATION FOR SEQ ID NO:17: !l) SEQUENCE CHARACTERISTICS: (A) LENGTH: 12 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:17 GGAGGCATTA AA (2! INFORMATION FOR SEQ ID NO:18: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 12 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (XI) SEQUENCE DESCRIPTION: SEQ ID NO:18 GGTGGCGAAA TT 45 (2) INFORMATIOK FOR SEQ ID NO:19: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 11 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single !D) TOPOLOGY: linear liil MOLECULE TYPE: Other (xi) SEQUENCE DESCRIPTION: SEQ ID NO:19: CGTTACCCTA T (2) INFORMATION FOR SEQ ID NO:20: (l) SEQUENCE CHARACTERISTICS: (A) LENGTH: 12 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:20: SGWGGMRRNA NA (2) INFORMATION FOR SEQ ID NO-.21: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 11 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:21 CGTT_CCCTA T (2) INFORMATION FOR SEQ ID NO:22: 46 (l) SEQUENCE CHARACTERISTICS: (A) LENGTH: 11 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION:-SEQ ID NO:22 CCTA_CCCTA A (2) INFORMATION FOR SEQ ID NO:23: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 11 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:23 CGTTACCCTA T (2) INFORMATION FOR SEQ ID NO:24: (l! SEQUENCE CHARACTERISTICS: (A) LENGTH: 11 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:24 CGTTACCCAA T (2) INFORMATION FOR SEQ ID NO:25: (l) SEQUENCE CHARACTERISTICS: (A) LENGTH: 11 base pairs 47 (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:25: CGGTTGCCAT G (2) INFORMATION FOR SEQ ID NO:26: (1) SEQUENCE CHARACTERISTICS: (A) LENGTH: 11 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:26: CGTTACCCTA T (2) INFORMATION FOR SEQ ID NO:27: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 10 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:27 GTAGGTCGAA (2) INFORMATION FOR SEQ ID NO:28: U) SEQUENCE CHARACTERISTICS: (A) LENGTH: 10 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single 48 (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:28 CGAGGAAGCA (2) INFORMATION FOR SEQ ID NO:29: (l) SEQUENCE CHARACTERISTICS: (A) LENGTH: 10 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:29 CGAGGAAGAA (2) INFORMATION FOR SEQ ID NO:30: (l! SEQUENCE CHARACTERISTICS: (A) LENGTH: 10 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single ID) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 30 CGTGGAGGCA (2) INFORMATION FOR SEQ ID NO:31: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 10 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear 49 (Xi) SEQUENCE DESCRIPTION: SEQ ID NO:31: CGTGGTGGGA (2) INFORMATION FOR SEQ ID NO:32: 111 SEQUENCE CHARACTERISTICS: (A) LENGTH: 10 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:32: CGAGGATGAC (2) INFORMATION FOR SEQ ID NO:33: (l) SEQUENCE CHARACTERISTICS: (A) LENGTH: 10 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 33 CGTGGTTGAT (2) INFORMATION FOR SEQ ID NO-.34: (l) SEQUENCE CHARACTERISTICS: (A) LENGTH: 10 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:34 50 CGAGGTCGCA (2) INFORMATION FOR SEQ ID NO:35: (l) SEQUENCE CHARACTERISTICS: (A) LENGTH: 10 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single' (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:35 CTAGGTAGCA (2! INFORMATION FOR SEQ ID NO:36: (i! SEQUENCE CHARACTERISTICS: (Ai LENGTH: 10 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single ID) TOPOLOGY: linear (xil SEQUENCE DESCRIPTION: SEQ ID NO: 36 GTAGGTGTTA (2) INFORMATION FOR SEQ ID NO: 37: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 10 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:37 CTAGGTAACA 51

Claims (6)

  1. WO 98/11214 (2) INFORMATION FOR SEQ ID NO:38: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 10 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:38 CATGGTTGCC (2) INFORMATION FOR SEQ ID NO:39: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 10 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (XI) SEQUENCE DESCRIPTION: SEQ ID NO:39 CGWGGWNGMM 52 CLAIMS ^07318 1 A composition which comprises an ATPase, which composition is substantially free of other normally intracellular mammalian proteins, wherein said ATPase comprises a mammalian ATP-dependent homologous paring protein which: (a) catalyzes DNA reannealing; (b) has ATPase activity, (c) has ATP-dependent single strnaged DNA binding activity; and (d) has greater than 80% sequence identity with SEQ ID NO: 1.
  2. 2 The composition of claim 1, which comprises an ATPase having a sequence which comprises at least 115 amino acids of SEQ ID NO:l or which is substantially identical thereto
  3. 3 The composition of claim 1, wherein said ATPase comprises a sequence that is substantially identical to the sequence of residues 80-200 of SEQ ID NO:l.
  4. 4 The composition of claim 3, in which the ATPase is an mREC2
  5. 5 The composition of claim 4, in which the ATPase is substantially identical to hsREC2.
  6. 6. The composition of claim 5, wherein the sequence of the ATPase comprises amino acids 2-350 of SEQ ID NO 1 Attorneys for THOMAS JEFFERSON UNIVERSITY and CORNELL RESEARCH FOUNATION, INC. INi'ELLZCTUA. PUCV QFrlCE Cr N T 1 0 OCT 2001 36523claims OOl/WI/tt Met Gly Ser Lys Lys Leu Lys Arg Val Gly Leu Ser Gin Glu Leu Cys 1 5 10 15 Asp Arg Leu Ser 20 Arg His Gin lie Leu 25 Thr Cys Gin Asp Phe 30 Leu Cys Leu Ser Pro 35 Leu Glu Leu Met Lys 40 Val Thr Gly Leu Ser 45 Tyr Arg Gly Val His 50 Glu Leu Leu Cys Met 55 Val Ser Arg Ala Cys 60 Ala Pro Lys Met Gin Thr Ala Tyr Gly lie Lys Ala Gin Arg Ser Ala Asp Phe Ser Pro 65 70 75 80 Ala Phe Leu Ser Thr 85 Thr Leu Ser Ala Leu 90 Asp Glu Ala Leu His 95 Gly Gly Val Ala Cys 100 Gly Ser Leu Thr Glu 105 lie Thr Gly Pro Pro 110 Gly Cys Gly Lys Thr Gin Phe Cys lie Met Met Ser lie Leu Ala Thr Leu Pro 115 120 125 Thr Asn 130 Met Gly Gly Leu Glu 135 Gly Ala Val Val Tyr 140 lie Asp Thr Glu Ser Ala Phe Ser Ala Glu Arg Leu Val Glu lie Ala Glu Ser Arg Phe 145 150 155 160 Pro Arg Tyr Phe Asn 165 Thr Glu Glu Lys Leu 170 Leu Leu Thr Ser Ser 175 Lys FIG. 1A Val His Leu Tyr 180 Arg Glu Leu Thr Cys 185 Asp Glu Val Leu Gin 190 Arg lie Glu Ser Leu 195 Glu Glu Glu lie lie 200 Ser Lys Gly lie Lys 205 Leu Val lie Leu Asp 210 Ser Val Ala Ser Val 215 Val Arg Lys Glu Phe 220 Asp Ala Gin Leu Gin Gly Asn Leu Lys Glu Arg Asn Lys Phe Leu Ala Arg Glu Ala Ser 225 230 235 240 Ser Leu Lys Tyr Leu Ala Glu Glu Phe Ser lie Pro Val lie Leu Thr 245 250 255 Asn Gin lie Thr 260 Thr His Leu Ser Gly 265 Ala Leu Ala Ser Gin 270 Ala Asp Leu Val Ser 275 Pro Ala Asp Asp Leu 280 Ser Leu Ser Glu Gly 285 Thr Ser Gly Ser Ser Cys Val lie Ala Ala Leu Gly Asn Thr Trp Ser His Ser Val 290 295 300 Asn Thr Arg Leu lie Leu Gin Tyr Leu Asp Ser Glu Arg Arg Gin lie 305 310 315 320 Leu lie Ala Lys Ser 325 Pro Leu Ala Pro Phe 330 Thr Ser Phe Val Tyr 335 Thr lie Lys Glu Glu 340 Gly Leu Val Leu Gin 345 Ala Tyr Gly Asn Ser 350 FIG.1B CGGACGCGTG GGCGCGGGGA AACTGTGTAA AGGGTGGGGA AACTTGAAAG TTGGATGCTG 60 CAGACCCGGC ATGGGTAGCA AGAAACTAAA ACGAGTGGGT TTATCACAAG AGCTGTGTGA 120 CCGTCTGAGT AGACATCAGA TCCTTACCTG TCAGGACTTT TTATGTCTTT CCCCACTGGA 180 GCTTATGAAG GTGACTGGTC TGAGTTATCG AGGTGTCCAT GAACTTCTAT GTATGGTCAG 240 g CAGGGCCTGT GCCCCAAAGA TGCAAACGGC TTATGGGATA AAAGCACAAA GGTCTGCTGA 300 c» TTTCTCACCA GCATTCTTAT CTACTACCCT TTCTGCTTTG GACGAAGCCC TGCATGGTGG 360 | TGTGGCTTGT GGATCCCTCA CAGAGATTAC AGGTCCACCA GGTTGTGGAA AAACTCAGTT 420 | TTGTATAATG ATGAGCATTT TGGCTACATT ACCCACCAAC ATGGGAGGAT TAGAAGGAGC 480 o, TGTGGTGTAC ATTGACACAG AGTCTGCATT TAGTGCTGAA AGACTGGTTG AAATAGCAGA 540 w m ATCCCGTTTT CCCAGATATT TTAACACTGA AGAAAAGTTA CTTTTGACAA GTAGTAAAGT 600 w 3 TCATCTTTAT CGGGAACTCA CCTGTGATGA AGTTCTACAA AGGATTGAAT CTTTGGAAGA 660 * g AGAAATTATC TCAAAAGGAA TTAAACTTGT GATTCTTGAC TCTGTTGCTT CTGTGGTCAG 720 ft AAAGGAGTTT GATGCACAAC TTCAAGGCAA TCTCAAAGAA AGAAACAAGT TCTTGGCAAG 780 £ AGAGGCATCC TCCTTGAAGT ATTTGGCTGA GGAGTTTTCA ATCCCAGTTA TCTTGACGAA 840 TCAGATTACA ACCCATCTGA GTGGAGCCCT GGCTTCTCAG GCAGACCTGG TGTCTCCAGC 900 FIG. 1C TGATGATTTG AAATACCTGG AAGACAGATT CAAGGAGGAA AACCTGTTCA AGAGTCTTGT ACTCAGCTTA CCAACAGTAA TTGATACCAT GTTGGGTTGT TTCATTTTGA TCCGTGGCAA CATCACCCCC CCAGCTCCTG TCAAATAAAT TCCCTGTCTG AGTCACAGTG CTTATTGCCA GGCCTGGTTC TCTTGCCAAG GGTGAAACAC AGTCATGGAA TCTGCATCAT GGCACTGACA GTTCGTGGAA TGGAGGTGAA CTAGATATCA AAAAGCAAGA GGAAACCACC GGGATCAAAT AAGGCACTTC TGAATACCCG AGTCCCCTCT TTCAAGCCTA AAAAATCCGC CCATCGTTCT TTCTAGAGGA AAGCTGATTT ATGGGCACAC CACATAGGTT ATTTATATAA TGATGTGCAA CCCATAACCA AATCTACTTT AGTTTCCCAA TGGATCCAGC GCTGATCCTC GGCTCCCTTC TGGAAATTCC TTTTCTGCCA CTGCTAAAAC TGTATCTCAC GATACCATGG AGGGAACAGG TTTTTTTTTA GATGAAATTA CCAGCATCTC TTATGCAAGT TTTTCTATGG AAAAAAAAAA TGTGTGATAG CAGTACCTTG ACCTCATTTG TAGAGACAGA CAGAAACAAA ATTTGGTTGC AAGTAGGATC CACTGACAAT AAATGGGAAT ACTTTCTCTT ACCATTTTAA TGTCTAGTTC GTTCCTATTT CTTTACCTAA AAAAAAAAAA CCGCACTAGG ATTCAGAGAG TCTACACCAT TAAATGTGCA ATATTGGGAA TACTGTGTAG AAGAACAAGC GGGCACTGAT GAGAGCAAGG TCTAAAATAT AGTAAACAAT CCAAATATTT CCCCCTCCTC TCTGGAAATT AAAAAAA 960 1020 1060 1140 1200 1260 1320 1380 1440 £ 1500 w 1560 1620 1680 1740 1797 FIG.1D Met Ser Ser Lys Lys Leu Arg Arg Val Gly Leu Ser Pro Glu Leu Cys 1 5 10 15 Asp Arg Leu Ser 20 Arg Tyr Leu lie Val 25 Asn Cys Gin His Phe 30 Leu Ser Leu Ser Pro 35 Leu Glu Leu Met Lys 40 Val Thr Gly Leu Ser 45 Tyr Arg Gly Val His 50 Glu Leu Leu His Thr 55 Val Ser Lys Ala Cys 60 Ala Pro Gin Met Gin Thr Ala Tyr Glu Leu Lys Thr Arg Arg Ser Ala His Leu Ser Pro 65 70 75 80 Ala Phe Leu Ser Thr 85 Thr Leu Cys Ala Leu 90 Asp Glu Ala Leu His 95 Gly Gly Val Pro Cys 100 Gly Ser Leu Thr Glu 105 lie Thr Gly Pro Pro 110 Gly Cys Gly Lys Thr 115 Gin Phe Cys lie Met 120 Met Ser Val Leu Ala 125 Thr Leu Pro Thr Ser 130 Leu Gly Gly Leu Glu 135 Gly Ala Val Val Tyr 140 lie Asp Thr Glu Ser Ala Phe Thr Ala Glu Arg Leu Val Glu lie Ala Glu Ser Arg Phe 145 150 155 160 Pro Gin Tyr Phe Asn Thr Glu Glu Lys Leu Leu Leu Thr Ser Ser Arg 165 170 175 FIG. IE Val His Leu Cys 180 Arg Glu Leu Thr Cys 185 Glu Gly Leu Leu Gin 190 Arg Leu Glu Ser Leu 195 Glu Glu Glu lie lie 200 Ser Lys Gly Val Lys 205 Leu Val lie Val Asp 210 Ser lie Ala Ser Val 215 Val Arg Lys Glu Phe 220 Asp Pro Lys Leu Gin Gly Asn lie Lys Glu Arg Asn Lys Phe Leu Gly Lys Gly Ala Ser 225 230 235 240 Leu Leu Lys Tyr Leu 245 Ala Gly Glu Phe Ser 250 lie Pro Val lie Leu 255 Thr Asn Gin lie Thr 260 Thr His Leu Ser Gly 265 Ala Leu Pro Ser Gin 270 Ala Asp Leu Val Ser 275 Pro Ala Asp Asp Leu 280 Ser Leu Ser Glu Gly 285 Thr Ser Gly Ser Ser 290 Cys Leu Val Ala Ala 295 Leu Gly Asn Thr Trp 300 Gly His Cys Val Asn Thr Arg Leu lie Leu Gin Tyr Leu Asp Ser Glu Arg Arg Gin lie 305 310 315 320 Leu lie Ala Lys Ser 325 Pro Leu Ala Ala Phe 330 Thr Ser Phe Val Tyr 335 Thr lie Lys Gly Glu 340 Gly Leu Val Leu Gin 345 Gly His Glu Arg Pro 350 FIG. IF gggagccctg TGTGACCGTT ctagaactta gtaagcaagg gcacatctct ggtggtgtgc cagttttgca ggggctgtgg gcggaatctc CO c agagttcatc n gaggaagaga 1 gtcagaaagg | ggcaaaggag CO acgaatcaaa £ ccagctgatg 3 ctaggaaaca 30 c gagagaaggc m accatcaagg § ctttgtctag aaaggaaacg agtgatttca ggctaggggc agcacctttc atccccctag tccgaataaa aaaaaaaaaa GAAACATGAG TAAGCAGATA TGAAAGTGAC CCTGTGCCCC CACCGGCATT CTTGTGGATC TAATGATGAG TCTACATCGA GTTTTCCACA TTTGCCGAGA TCATTTCGAA AGTTTGACCC CGTCCTTACT TTACGACCCA ATTTGTCCCT CATGGGGTCA AGATTCTCAT GGGAAGGCCT TGCTGATTGC GAAGCCAACA GACCCGGAAG TCCACCACCG AGCCATGCCC GGAGAGCAGG GTATTGTTCC AAAAAAAAAA CAGCAAGAAA CCTGATTGTT TGGCCTGAGT GCAGATGCAA CCTGTCTACT TCTCACAGAG TGTCTTAGCT CACAGAGTCT ATATTTTAAC GCTCACCTGT AGGAGTTAAG GAAGCTTCAA GAAGTACCTG TCTGAGTGGA GTCTGAAGGC CTGTGTGAAC TGCCAAGTCT GGTTCTTCAA ATGTGACTCA TAATGAGGAT GTGAAGATGA TGGGATGTCA CTCAAAGTGG AGGGGGACTA GCCAAAAAAA AAAAA CTAAGACGAG AACTGTCAGC TACAGAGGTG ACGGCTTATG ACCCTGTGCG ATTACAGGTC ACATTACCTA GCATTTACTG ACTGAGGAAA GAGGGGCTTC CTTGTGATTG GGCAACATCA GCAGGGGAGT GCCCTCCCTT ACTTCTGGAT ACCCGGCTGA CCTCTGGCTG GGCCACGAAA TGAAATGAAA TAATTGGTTG AGAAGCCTTT GCGGCCATCG TTCAGCCACA ACAAGATTTG AAAAAAAAAA FIG. 1G TGGGTTTATC TCCAGAGCTG 60 * o ACTTTTTAAG TCTCTCCCCA 120 TCCACGAGCT TCTTCATACA 180 ** c AGTTAAAGAC ACGAAGGTCT 240 CCTTGGATGA AGCATTGCAC 300 CACCAGGTTG CGGAAAAACT 360 CCAGCCTGGG AGGATTAGAA 420 CTGAGAGACT GGTTGAGATT 480 AATTGCTTCT GACCAGCAGT 540 TACAAAGGCT TGAGTCTTTG 600 TTGACTCCAT TGCTTCTGTG 660 AAGAAAGGAA CAAGTTCTTG 720 TTTCAATCCC AGTTATCTTG 780 CTCAAGCAGA CCTGGTGTCT 840 CCAGCTGTTT GGTAGCTGCA 900 ro 4k TTCTCCAGTA CCTTGATTCA 960 CCTTCACCTC CTTTGTCTAC 1020 GACCATAGGG ATACTGTGAC 1080 CAGGACTGCG CTGCTTGGAA 1140 GTTGCTGTTG AGGTGGTAAC 1200 ATCCAGTCTC TGGATGCAGA 1260 TAATAATTTG CACTTACACA 1320 TTAATTAATT AAAGCCCACA 1380 \ NO 3 N* TAATTACAGA AGGGAAAATT 1440 AAAAAAAAAA AAAAAAAAAA 1500 1525 MGSKKLKRUGLSQELCDRLSBHOILTCQDFLCLSPLELMKUTGLS NLS YBGUHELLCMUSBRCHPKMQTRYGIKflQRSflDFSPRFLSTTLSR 50 LDERLHGGURCGSLTEITGEEGCGKIQFCIMMSILRTLPTNMGGL 100 R BOH EGRUUVIDTESRFSRERLUEIRESBFPRVFNTEEKLLLTSSKUHLV 150 P RELTCDEmORIESLEEEIISKGIKLUlLDSURSUURKEFDROLOG DNR 200 B BOB NLKEBNKFLRRERSSLKVLBEEFSIPUILTNQITrHLSGRLRSQflD 250 LUSPRDDLSLSEGTSGSSCBIRRLGNT11JSHSUNTRLILQVLDSERR 300 QILIRKSPLRPFTSFUYTIKEEGLULQRYGNS* 350 FIG.2A RecA-Like w c o w x m m H 33 c in 8 N- NLS v \ \ uspin—like Motif "A" Motif "BT DNA binding Src—Like Phosphorylation site N> 4s» 1 cm = 33 amino acids FIG.2B 124 i 1 174 48 223 60 273 128 323 137 373 165 173 47 222 79 272 127 322 136 372 164 422 199 LN DAR FAS SCI VP PTQG YDGNF PG AQCF VYDSD AGS DSD ARS SIDAVMHE • •• I ■••• I*** • • » » ♦ •• « I • • l * • •••• ( • ♦ ♦ ♦ «•••• • MGSKKLKR. . . VGLSQELCDRLSRHQILTCQDFLCLSPLELMKVTGLSYR • • • t • DI. ELPSTFCRPQTPQTHDVARDEHHDGYLCDPKVDHASVARDVLSLGRQ . . j | . . . | | | | | . | : GVHELLCMVSRA CAPKMQTAYGIKAQRSADFS • • • | • « RHVFSSGSRELDDLLGGGVRSAVLTELVGESGSGKTQMAIQVCTYAALGL - • :I • • ..||t I III :: I I I«-I..|tI I I I s-I «t-.l-l PAFLSTTLSALDEALHGGVACGSLTEITGPPGCGKTQFCIMMSILATL. . • • • • • VPLSQADDHDKGNNTFQSRTFVRDPIHASTKDDTLSDILQSYGMEPSIGS I * I • * I • • • I • • PTNMGGLEG • • • • • • HRGMGACYITSGGERAAHSIVNRALELASFAINERFDRVYPVCDPTQSSQ I f • • I • I • I -III* • • I • • • I • I * * 1 I * I * . . . . AWYIDTESAFSAfeRLVEIA ESRFPRYF • • ♦ • « DADGRRDALLAKAQQLGRRQALANLHIACVADVEALEHALKYSLPGLIRR . : || .. .: ..|. :|.||..: . . - NTEEKLLLTSSKVHLYRELTCDEV. . LQRIESLEEEI FIG.2C 423 200 473 240 573 264 623 306 669 341 LWSSKRQSGVSREIGWWDNLPALFQQDQAAASDIDSLFQRSKMLVEIA 472 IS KG IKLVILDS VAS WRKE FDAQLQ. GNLKERNKFLAREA 23 9 • * • • * DALKRISAVQWRGASDCGSS AGRAVLVLNH VSDAFGIDKQIARRFVFDSA 522 . . | | . : : . . | : . : . . | | : SSLK YLAEEFSIPVILTNQITTHL 263 1 • • • • « SGLLASIAPTLAEAVGARELDSACASNDVPLRTLEARTAQLGQTWSNLIN 622 II lii I • : • • I : • : : . : : | . | • I • I I • I I h : I SGALASQADLVSPADDLSLSEGTSGSSCV IAALGNTWSHSVN 305 * • • • « VRVFL SKTRARICMRDDQAPACEPVRQNTNQRGTASKSLMNTVRKA 668 I . . I ||.. || | . . | . . TRLILQYLDSERRQILIAKSPLAP FTSFVYTIKEE 340 AWINPFGAT 678 : : | : . : : | . GLVLQAYGNS 350 FIG.2D WO 98/11214 PCT/IB97/012I7 12/24 SUBSTITUTE SHEET (RULE 26) FIG.4 No protein + ATP hRec2 -thio (0.3 pg) + ATP hRec2 -thio (0.3 jig) + ySATP hRec2 -thio (0.3 pg) + no ATP RecA (4.4/jg) + ATP HREC2 Enhanced Conversion vs Controls 01 x in =1 In g c .2 C §! c o u FIG.5A -o- • SCI —4 •rt ■ SCI + pcDNA 2 - SCI + pcCAT • SCI + pchREC2 ng AC CTG ACT CCT GTG GAG AAG TCT GC (5s TG GAC TGA GGA CAC CTC TTC AGA CG u S AC CTG ACT CCT GAG GAG AAG TCT GC 0A 3 TG GAC TGA GGA CTC CTC TTC AGA CG 5 in —• w «* x *•*. m «*> Sj * * 3 T GCGCG ug gac uga ggA CTC Cue uuc aga eg T ST T SCI St T T CGCGC AC CTG ACT CCT GAG GAG AAG TCT GC T pS_pA 3 '5' FIG.5B % 3 VO a fc» -4 WO 98/11214 PCT/IB97/012I7 16/24 Reoneating -ATP + ATP |ilGST-hREC2 1 2 4 8 12 4 8 ds iTl FIG.6 SUBSTITUTE SHEET (RULE 26) s 1 s 1 CGACGGCCCG GGCTGGTATT ATAGCAGGTA TCACTTGGTT TTCTACTGGG 51 GGAAACAAGT CATTGCTAAC AAATTCCCAT GGGAGAGAAA TGAGGAGGAT 101 GTATTTTTGT TTGTGAGAGG TGTGTATGTA TGTATATTGT GTGTGCGTGT <URS> 151 GTGTGTGTGT GAGAGAGAGA GATTGATTCA GTCTGATTCA GAGAATTTAG 201 GTGTTAAATA GAAATTTGGG CCATGGTATT GGAAATAAAC AAATATATAC 251 ATTCTCAGTA TACATATATT TTCATTCCAA AATGTTACTT CTTTTCTGAT 301 AACTATATTG CTTTATTCCC TTGGATCCAT GAAGAGTTCC TGTTTCAGTT 351 CGTTCCAGAG GATACTTCTT TACCATCTCA ATGAGATATA CAGCTTCTCC RHP51-UAS 401 TTTGTATGCA TTAAGAGACT CACAGTAATT CTTTTTTAGC TCTGTGAAGA PHR1-UAS 451 TAAATCTTTC ATGAGCCTCA TTTACCCCTA GCAAAGTACA ATAGTGAAAT RAD23 501 TTAACTGCAT GTGAGAATAT AAGCAGCTAG TGTAATAAAG AACATTTTGG 551 GCCAGGTCTG ATCGCTCATG CCTGTAATCC CAGCACTTTA GGAGGTCAAG 601 GCGAGAGGAT CACTTGAGCQ CAGGAGTTCG AGACCAGCTT GGGCAACATG RAD16-URS 651 GCAAAACCCT GTCTCTACAA AAAATACAAA AATTGGGCAG GCATGGTGTC t K» 0> c o 01 M x _ _ _ 4k y» U M* H* -4 q 701 GACCCAGTCT CTACAAAAAA TACAAAAATT AGCCAGACAT GGTGGTGCAC rad 51-urs 751 GCTTGTGGTC CCAGCTACTT GGGAGGCTGA GGTAGGAGGA TTGCTTGAGC rad 51-urs % 801 CCAGGAGGGG GAGGTTGCAG TGAGCTGAGA TCGAGCCACT GCACTCCAGC RNR3-OAS 851 TGGGGTGACA GAGCCAGACC TGTCTCGCTC TCTCTCTCTC TCTATATATA <PHR1-UES> RPHP51-UAS 901 TATATTTAAA AAGAACATTT TAATACTGCA GTGATAAAAT CTCATTTGAT c 951 TCAGAAGGTG TGCTCTGACT CCTAGAAAAA GGAAGAGTCA AATATGATTA PHR1-URS 1001 TGGACTTGCA GTAGAGTGTA ATGGTTAAGA GGATAGGTTT CAGAATTAGA j» 1051 CTGCCTGGAT TCAAATTCTG GATCAGTTAT TTATGGTTTC TGGTGACAAT 4k ro 1101 ggactagcta acttttccag gctttagttt tctcatatgt aaaaaagggg g rhp51-urs In 1151 ccaataatct actttccttc tagggctatt gagaagatta aatgtgat£a s rhp51-uas np *7 n 1201 tttagataag ttttggaaca gtgcctggta tgtggtaggt gctccataaa rlb./D 1251 tatacctatt gccgttacag tgcaatgtaa attgttacag tgcaatagac rad54-uas 1301 tttctagtag ttctgtttgg aaatatgcct tgaaagttaa ttacatttcc rad54-uas 1351 aaataaaatt tatacatgca ttggaacatt ttaagatgct ctacaaatgt 1401 gaagtggtac tatattcatg tagtaaatat caattaattg tgtgaaatta urs (phr1) i 1451 TATTTGAGGT TGCCTTGTAG ATTTTCTATG TGCCTGTTTG ACGAACAATT RNR3-UAS 1501 GTCCCTCCTA TTTAAAACAT TTAAAAAGGT TCTATAGCAT TCCTTTATCA g 1551 GTAATATTTT TAACACAATA TGTTTCATTT TGCATATGGA GAAACTTGAG o RAD23-UAS =| 1601 GAATTTTTAA TTTTGTTTTG GATAGCCTAT TCACTATCAC TTATGTTATA S PHR1-URS RAD51-UAS TATA BOX rhp51-uas S RNR3-UAS g 1651 TTCTGTTGTT TTTTTCATGG TTCTTCTTTT CTTTGCTGGA TCTGGAGGC £ <PHR1-UAS> (OVERLAP WITH DRE2) M O) FIG.7C 1 TATCTCAGTA GCACGTGCAC ATAGCAACTA CAATACCTGT CACATAAATG RNR3 51 TAGTTACTTG AATATATGTC TCTTCATTCT TCAATTGTAA GTATGCAAAA RAD23 101 GGGAGGACAT AAGCTTAGCA TAGCATGTGC TTAATATTGG TGAAAGAAAC <RAD6> 151 AAATGAATAG AGAATGTTAT ATTTGGAGAG TTTATATTAT ATTTGGGAGA 201 GTAGGGAAAA AACTTGAAGC CATAAGCAGA ATCGAGGGCA AGTAGTGAGA c 251 GTGGTACTGT TAAATCAGAG TGATTATTGC TAAGGTCTTT GTAATTTGGG 19 301 GTTGTAGGTG TTTTTTGTTT TTGTTGTTTG AGGGTCTGAA TTTATTCGTT RHP51 RNR3 g w 351 ATATGATGTT ATTGCCTGGA ACTACCTTAT CTGAGAAGCA GTAGGCAATA FIG.7D 601 AGTTTTGTTC TTTTGTGTAT AGTTATTGAT TATTACTTTA TTCCATGGGA 651 ATAATGAGTT CCTATTATTT CTGGAGGATA TTTTGCCATT TCGATGAGAC 701 ACACAGCCTC TTCTTTGCTA TGCAATATTA CGAGATTACA ACAGTTCTAA ro |" RAD51-UAS * J 401 GAGTAGCGTA TAAATGTTGG TAAATTTTCT CTTAAGGAAA CAAATTATCC c RAD7 K 451 TTACAAAATT CCAACTGAAA GAAATAAAGA GAATGTATCT TGGTTTTGTG B RAD54-UAS 501 TGGAGAGAGG GAAGTAGAAG ATGGGGGATG AAGAGAGAGA GGAGGGTTAT RAD-1 551 TTATTGGGCT ATATATAGTG TTGGTAGTAG GAATCTTAAT TCTTGTGTGT 3 c m M FIG.7E 751 CTCCCTGAAG ACAAATACTT CATGAGTCTC ATTAGCTATC TAAGCTATAG PHR1-1 801 GAAGAGCAGA ATTTAATTCT ACATGGAAAC AGTAAGAAGC TAGTATAATG PHR1-UAS 851 AAGAATTTTA TTGATATCAC TTGATTGAAA TTTGTTCTGA CTCTTTAGAA 901 AAAGCAAGGG TGAAATAAGA TTTGTGATTC TACAGTAGTA ATGGGTAAGA 951 GGATAGGTCT CAGGACAAAC TGCCTAATGA MCOTWT CTGTTATTTA RHP51-UAS 1001 TTTATTTTCT GATGACAGTG GGATAACTGA CATTTACACA TTAGCTTTCT 1051 CATATGTAAA AAAGAAATTT TATTTTTATT ATAGTCTGTC AAGGAATATT RNR3-1 1101 AAATATAAGG TTTTGGAGCA TGGTTGATAT TTAGCAGATG TCTGTTCATT RAD16-1 1151 CTTGATCAGT ATAGAGTTGC CACTTGGAAA ATGCATCTTG AAGATTACAT 1201. AACCAGACAA AATTTGTTAG TAACACTCAG TGGTCTTAAG ATGTTATAAG <RAD16> 1251 TGACGGGCTA GTCGTGGTAA TCAACTTGAT ACCTTGACCC TCAGGAGAAG <DRE2> 1301 AGGGATTGTC TCCATCGGAT GGGCCTGTGA GCATATCTGT GGGGACGTTT 1351 TTCTTGGACT GCCTAGTTGA TGGAAAAGGG CTTGGCTCAG TGTCAGTGGT 1401 CCTTCTTATG GTGAGCAAGC TGGGGGAAGC GTTGCAGTAA GCAGTAGTCC PHR1- -UAS 1451 TTTGTGGTCT CAGCTTCCTT TTCTTCTCTC TTCTTTCTTT CTTTCTTTCT CO c o tN> 01 x m nam fi-1 i\> 1601 1651 1701 1751 1801 1851 1901 1951 2001 2051 2101 TTCTTTCTTT CTTTCTTTCT TTCTTCCTTC CTTCCTTCCT TTTCTCTCTT RHP51-UAS TCTTTCTTTA GTTCCGTTCG TTTGTTCATT CGTTCGTTTT TCGAGACAGG RHP51-UAS <UAS> GTTTTTCTGT ATAGCCCTGG CTGTCCTGGA ACTCACTTTG TAGACCAGGC TGTCCTCGAA CTCAGAAATC CGCCTGCCTC TGCCTCCCTG TGAGTGCTgg RAD2-UAS AATTAAAGGC ATGCGCCACC CCGCCCGGCT TCTCAGCTTC CATTTCTGTT CAAGCTCTTG CCTTCAGCTC CTGCCTTGGC TTTCTGAGAC AAAGGCATAT AATCTGTAAG CCAAATCAAA CTTTTCTTCT CAACTTGCTT TTGGCCAGTG TTTTATTACA GCGACTAAAG GCAAACTAGA CTACTATGTA AATGGGAAGC PHR1 rhp51-2 &CTGTTAAAG TCAAGTAATA GCAAAAGATT ACATGGCCTG GATTTTTTGA RNR2 GGTTGCTTAC TTTCTCTGTG TAtCCGGTTG TAAGTGTCTT TCCTACTTTT RPH51-UAS TTTATTAGCA TTTTTTTTCC ATGTTTTGCT TTGCACATAG AGAAGTTTGA <DRS> AGCACTTTAT TTTGTAGGGT GTTTTGTATA ATCTGTCCAC CATCATTTTT ATTGTTTTCT TATGTTTTTT CAAGATTTCT TTGGGAGCCC TGGAAAC FIG.7F e > . » wo 98/11214 23/24 PCT/IB97/01217 FIG.8A neo 0 J/m2 FIG.8B FIG.8C neo 24hr 15 J/m2 3 O c_> o o FIG.8D neo 72hr 15 J/m2 SUBSTITUTE SHEET (RULE 26) # 1 » 0 WO 98/11214 PCT/IB97/01217 FIG.8E 24/24 15C8 0 J/m2 1508 24hr 15 J/m2 FIG.8F i - 15C8 48hr 15 J/m2 FIG.8G 1 * o - 15C8 72hr 15 J/m2 FIG.8H i - o SUBSTITUTE SHEET (RULE 26)
NZ507318A 2000-10-05 2000-10-05 Compositions comprising mammalian and human REC2 that functions as an ATPase NZ507318A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
NZ507318A NZ507318A (en) 2000-10-05 2000-10-05 Compositions comprising mammalian and human REC2 that functions as an ATPase

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
NZ507318A NZ507318A (en) 2000-10-05 2000-10-05 Compositions comprising mammalian and human REC2 that functions as an ATPase
NZ33464600 2000-10-05

Publications (1)

Publication Number Publication Date
NZ507318A true NZ507318A (en) 2002-07-26

Family

ID=19927170

Family Applications (1)

Application Number Title Priority Date Filing Date
NZ507318A NZ507318A (en) 2000-10-05 2000-10-05 Compositions comprising mammalian and human REC2 that functions as an ATPase

Country Status (1)

Country Link
NZ (1) NZ507318A (en)

Similar Documents

Publication Publication Date Title
US20110088106A1 (en) Compositions and methods for mediating cell cycle progression
EP2009109A1 (en) G protein-related kinase mutants in essential hypertension
KR20220097414A (en) CRISPR and AAV Strategies for X-Linked Combustion Retinal Delaminization Therapy
US5686598A (en) Genes associated with retinal dystrophies
Mouro et al. Cloning, expression, and chromosomal mapping of a human ATPase II gene, member of the third subfamily of P-type ATPases and orthologous to the presumed bovine and murine aminophospholipid translocase
AU744157B2 (en) Novel gene encoding a DNA repair endonuclease and methods of use thereof
AU735296B2 (en) Mammalian and human REC2
US5705380A (en) Identification of a gene encoding TULP2, a retina specific protein
AU780146C (en) Congenic animal models of non-insulin dependent diabetes mellitus
NZ507318A (en) Compositions comprising mammalian and human REC2 that functions as an ATPase
US6642369B1 (en) Nucleic acids involved in the responder phenotype and applications thereof
US20040141994A1 (en) Viral recombinases, related articles, and methods of use thereof
Hamann et al. Consecutive inactivation of both alleles of the gb110 gene has no effect on the proliferation and differentiation of mouse embryonic stem cells
US20090208963A1 (en) Gene Involved in V(D)J recombination and/or DNA repair
WO2001016176A2 (en) Transgenic animal model for neurodegenerative diseases
CA2242382C (en) Compositions and methods for mediating cell cycle progression
JP2003506056A (en) Knockout mouse of tumor suppressor gene anx7
WO1998046726A1 (en) A-myb NULL MUTANT TRANSGENIC ANIMALS AND USES THEREOF

Legal Events

Date Code Title Description
PSEA Patent sealed
RENW Renewal (renewal fees accepted)
RENW Renewal (renewal fees accepted)
ERR Error or correction

Free format text: THE OWNER HAS BEEN CORRECTED TO 241564, THOMAS JEFFERSON UNIVERSITY, 1020 LOCUST STREET, PHILADELPHIA, PA 19107 5587, US; 1304256, CORNELL RESEARCH FOUNDATION, INC., SUITE 105, 20 THORNWOOD DRIVE, ITHACA, NY 14850, US

Effective date: 20140630