Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
PLoS One ; 14(8): e0215882, 2019.
Article in English | MEDLINE | ID: mdl-31386669

ABSTRACT

Babesia bovis establishes persistent infections of long duration in cattle, despite the development of effective anti-disease immunity. One mechanism used by the parasite to achieve persistence is rapid antigenic variation of the VESA1 cytoadhesion ligand through segmental gene conversion (SGC), a phenomenon thought to be a form of homologous recombination (HR). To begin investigation of the enzymatic basis for SGC we initially identified and knocked out the Bbrad51 gene encoding the B. bovis Rad51 ortholog. BbRad51 was found to be non-essential for in vitro growth of asexual-stage parasites. However, its loss resulted in hypersensitivity to methylmethane sulfonate (MMS) and an apparent defect in HR. This defect rendered attempts to complement the knockout phenotype by reinsertion of the Bbrad51 gene into the genome unsuccessful. To circumvent this difficulty, we constructed an artificial chromosome, BbACc3, into which the complete Bbrad51 locus was inserted, for expression of BbRad51 under regulation by autologous elements. Maintenance of BbACc3 makes use of centromeric sequences from chromosome 3 and telomeric ends from chromosome 1 of the B. bovis C9.1 line. A selection cassette employing human dihydrofolate reductase enables recovery of transformants by selection with pyrimethamine. We demonstrate that the BbACc3 platform is stably maintained once established, assembles nucleosomes to form native chromatin, and expands in telomere length over time. Significantly, the MMS-sensitivity phenotype observed in the absence of Bbrad51 was successfully complemented at essentially normal levels. We provide cautionary evidence, however, that in HR-competent parasites BbACc3 can recombine with native chromosomes, potentially resulting in crossover. We propose that, under certain circumstances this platform can provide a useful alternative for the genetic manipulation of this group of parasites, particularly when regulated gene expression under the control of autologous elements may be important.


Subject(s)
Babesia bovis/enzymology , Chromosomes, Artificial/genetics , Gene Knockout Techniques , Rad51 Recombinase/deficiency , Rad51 Recombinase/genetics , Sequence Homology, Nucleic Acid , Babesia bovis/genetics , Centromere/genetics , Gene Expression , Models, Molecular , Phenotype , Protein Conformation , Rad51 Recombinase/chemistry
2.
PLoS Genet ; 15(2): e1007952, 2019 02.
Article in English | MEDLINE | ID: mdl-30716097

ABSTRACT

Meiotic recombination permits exchange of genetic material between homologous chromosomes. The replication protein A (RPA) complex, the predominant ssDNA-binding complex, is required for nearly all aspects of DNA metabolism, but its role in mammalian meiotic recombination remains unknown due to the embryonic lethality of RPA mutant mice. RPA is a heterotrimer of RPA1, RPA2, and RPA3. We find that loss of RPA1, the largest subunit, leads to disappearance of RPA2 and RPA3, resulting in the absence of the RPA complex. Using an inducible germline-specific inactivation strategy, we find that loss of RPA completely abrogates loading of RAD51/DMC1 recombinases to programmed meiotic DNA double strand breaks, thus blocking strand invasion required for chromosome pairing and synapsis. Surprisingly, loading of MEIOB, SPATA22, and ATR to DNA double strand breaks is RPA-independent and does not promote RAD51/DMC1 recruitment in the absence of RPA. Finally, inactivation of RPA reduces crossover formation. Our results demonstrate that RPA plays two distinct roles in meiotic recombination: an essential role in recombinase recruitment at early stages and an important role in promoting crossover formation at later stages.


Subject(s)
Homologous Recombination , Meiosis/genetics , Replication Protein A/metabolism , Animals , Cell Cycle Proteins/metabolism , Chromosome Pairing , Crossing Over, Genetic , DNA Breaks, Double-Stranded , DNA Replication , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Nuclear Proteins/metabolism , Phosphate-Binding Proteins , Protein Stability , Rad51 Recombinase/deficiency , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Replication Protein A/deficiency , Replication Protein A/genetics , Spermatocytes/cytology , Spermatocytes/metabolism
3.
Nucleic Acids Res ; 45(17): 10056-10067, 2017 Sep 29.
Article in English | MEDLINE | ID: mdl-28973445

ABSTRACT

ADP-ribosyltransferases promote repair of DNA single strand breaks and disruption of this pathway by Poly(ADP-ribose) polymerase (PARP) inhibitors (PARPi) is toxic to cells with defects in homologous recombination (HR). Here, we show that this relationship is conserved in the simple eukaryote Dictyostelium and exploit this organism to define mechanisms that drive resistance of the HR-deficient cells to PARPi. Dictyostelium cells disrupted in exonuclease I, a critical factor for HR, are sensitive to PARPi. Deletion of exo1 prevents the accumulation of Rad51 in chromatin induced by PARPi, resulting in DNA damage being channelled through repair by non-homologous end-joining (NHEJ). Inactivation of NHEJ supresses the sensitivity of exo1- cells to PARPi, indicating this pathway drives synthetic lethality and that in its absence alternative repair mechanisms promote cell survival. This resistance is independent of alternate-NHEJ and is instead achieved by re-activation of HR. Moreover, inhibitors of Mre11 restore sensitivity of dnapkcs-exo1- cells to PARPi, indicating redundancy between nucleases that initiate HR can drive PARPi resistance. These data inform on mechanism of PARPi resistance in HR-deficient cells and present Dictyostelium as a convenient genetic model to characterize these pathways.


Subject(s)
ADP Ribose Transferases/physiology , Dictyostelium/enzymology , Drug Resistance/physiology , Homologous Recombination/physiology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/physiology , Protozoan Proteins/physiology , Benzamides/pharmacology , Clone Cells , Cyclin-Dependent Kinase 8/deficiency , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinase 8/physiology , DNA Damage , Dictyostelium/drug effects , Dictyostelium/genetics , Exodeoxyribonucleases/deficiency , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/physiology , Gene Deletion , Indoles/pharmacology , Phthalazines/pharmacology , Piperazines/pharmacology , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , Quinazolines/pharmacology , Rad51 Recombinase/deficiency , Rad51 Recombinase/physiology , Recombinant Proteins/metabolism
4.
Nucleic Acids Res ; 45(8): 4590-4605, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28334891

ABSTRACT

RAD51, a multifunctional protein, plays a central role in DNA replication and homologous recombination repair, and is known to be involved in cancer development. We identified a novel role for RAD51 in innate immune response signaling. Defects in RAD51 lead to the accumulation of self-DNA in the cytoplasm, triggering a STING-mediated innate immune response after replication stress and DNA damage. In the absence of RAD51, the unprotected newly replicated genome is degraded by the exonuclease activity of MRE11, and the fragmented nascent DNA accumulates in the cytosol, initiating an innate immune response. Our data suggest that in addition to playing roles in homologous recombination-mediated DNA double-strand break repair and replication fork processing, RAD51 is also implicated in the suppression of innate immunity. Thus, our study reveals a previously uncharacterized role of RAD51 in initiating immune signaling, placing it at the hub of new interconnections between DNA replication, DNA repair, and immunity.


Subject(s)
DNA Replication , DNA-Binding Proteins/genetics , DNA/genetics , Membrane Proteins/genetics , Rad51 Recombinase/genetics , Recombinational DNA Repair/genetics , Cell Line, Tumor , DNA/immunology , DNA Breaks, Double-Stranded/drug effects , DNA-Binding Proteins/immunology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/immunology , Genes, Reporter , Humans , Hydroxamic Acids/pharmacology , Immunity, Innate , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , MRE11 Homologue Protein , Membrane Proteins/immunology , Pyrimidinones/pharmacology , Rad51 Recombinase/deficiency , Rad51 Recombinase/immunology , Recombinational DNA Repair/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Thiones/pharmacology , Vorinostat , Red Fluorescent Protein
5.
Nucleic Acids Res ; 45(4): 1835-1847, 2017 02 28.
Article in English | MEDLINE | ID: mdl-27924006

ABSTRACT

Homologous recombination (HR) is a DNA double-strand break (DSB) repair pathway that protects the genome from chromosomal instability. RAD51 mediator proteins (i.e. paralogs) are critical for efficient HR in mammalian cells. However, how HR-deficient cells process DSBs is not clear. Here, we utilized a loss-of-function HR-reporter substrate to simultaneously monitor HR-mediated gene conversion and non-conservative mutation events. The assay is designed around a heteroallelic duplication of the Aprt gene at its endogenous locus in isogenic Chinese hamster ovary cell lines. We found that RAD51D-deficient cells had a reduced capacity for HR-mediated gene conversion both spontaneously and in response to I-SceI-induced DSBs. Further, RAD51D-deficiency shifted DSB repair toward highly deleterious single-strand annealing (SSA) and end-joining processes that led to the loss of large chromosomal segments surrounding site-specific DSBs at an exceptionally high frequency. Deletions in the proximity of the break were due to a non-homologous end-joining pathway, while larger deletions were processed via a SSA pathway. Overall, our data revealed that, in addition to leading to chromosomal abnormalities, RAD51D-deficiency resulted in a high frequency of deletions advancing our understanding of how a RAD51 paralog is involved in maintaining genomic stability and how its deficiency may predispose cells to tumorigenesis.


Subject(s)
Genome , Homologous Recombination , Rad51 Recombinase/metabolism , Sequence Deletion , Animals , CHO Cells , Chromosomal Instability , Cricetulus , DNA Breaks, Double-Stranded , DNA Damage , DNA Repair , Deoxyribonucleases, Type II Site-Specific/metabolism , Gene Expression , Gene Knockout Techniques , Genes, Reporter , Mutation , Rad51 Recombinase/deficiency , Rad51 Recombinase/genetics
6.
Free Radic Biol Med ; 101: 424-433, 2016 12.
Article in English | MEDLINE | ID: mdl-27838435

ABSTRACT

Reactive oxygen species (ROS)-mediated DNA adducts as well as DNA strand breaks are highly mutagenic leading to genomic instability and tumorigenesis. DNA damage repair pathways and oxidative stress response signaling have been proposed to be highly associated, but the underlying interaction remains unknown. In this study, we employed mutant strains lacking Rad51, the homolog of E. coli RecA recombinase, and Yap1 or Skn7, two major transcription factors responsive to ROS, to examine genetic interactions between double-strand break (DSB) repair proteins and cellular redox regulators in budding yeast Saccharomyces cerevisiae. Abnormal expression of YAP1 or SKN7 aggravated the mutation rate of rad51 mutants and their sensitivity to DSB- or ROS-generating reagents. Rad51 deficiency exacerbated genome instability in the presence of increased levels of ROS, and the accumulation of DSB lesions resulted in elevated intracellular ROS levels. Our findings suggest that evident crosstalk between DSB repair pathways and ROS signaling proteins contributes to cell survival and maintenance of genome integrity in response to genotoxic stress.


Subject(s)
DNA Repair , DNA, Fungal/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Fungal , Rad51 Recombinase/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Transcription Factors/genetics , Cell Survival , DNA Breaks, Double-Stranded/drug effects , DNA, Fungal/metabolism , DNA-Binding Proteins/metabolism , Genomic Instability , Homologous Recombination , Hydrogen Peroxide/pharmacology , Mutation Rate , Oxidative Stress , Paraquat/pharmacology , Rad51 Recombinase/deficiency , Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism
7.
Nucleic Acids Res ; 44(1): 232-44, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26433224

ABSTRACT

The centromeres of many eukaryotic chromosomes are established epigenetically on potentially variable tandem repeats; hence, these chromosomes are at risk of being acentric. We reported previously that artificially created acentric chromosomes in the fission yeast Schizosaccharomyces pombe can be rescued by end-to-end fusion with functional chromosomes. Here, we show that most acentric/functional chromosome fusion events in S. pombe cells harbouring an acentric chromosome I differed from the non-homologous end-joining-mediated rearrangements that result in deleterious dicentric fusions in normal cells, and were elicited by a previously unidentified homologous recombination (HR) event between chromosome end-associated sequences. The subtelomere repeats associated with the non-fusogenic ends were also destabilized in the surviving cells, suggesting a causal link between general subtelomere destabilization and acentric/functional chromosome fusion. A mutational analysis indicated that a non-canonical HR pathway was involved in the rearrangement. These findings are indicative of a latent mechanism that conditionally induces general subtelomere instability, presumably in the face of accidental centromere loss events, resulting in rescue of the fatal acentric chromosomes by interchromosomal HR.


Subject(s)
Chromosomes, Fungal , Gene Rearrangement , Homologous Recombination , Telomere , Chromosome Mapping , DNA Breaks, Double-Stranded , DNA End-Joining Repair , Fungal Proteins/genetics , Fungal Proteins/metabolism , Genomic Instability , Models, Biological , Rad51 Recombinase/deficiency , Schizosaccharomyces/genetics
8.
Oncogene ; 33(12): 1495-505, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-23604115

ABSTRACT

Homologous recombination (HR), a mechanism to accurately repair DNA in normal cells, is deregulated in cancer. Elevated/deregulated HR is implicated in genomic instability and telomere maintenance, which are critical lifelines of cancer cells. We have previously shown that HR activity is elevated and significantly contributes to genomic instability in Barrett's esophageal adenocarcinoma (BAC). The purpose of this study was to evaluate therapeutic potential of HR inhibition, alone and in combination with telomerase inhibition, in BAC. We demonstrate that telomerase inhibition in BAC cells increases HR activity, RAD51 expression, and association of RAD51 to telomeres. Suppression of HR leads to shorter telomeres as well as markedly reduced genomic instability in BAC cells over time. Combination of HR suppression (whether transgenic or chemical) with telomerase inhibition, causes a significant increase in telomere attrition and apoptotic death in all BAC cell lines tested, relative to either treatment alone. A subset of treated cells also stain positive for ß-galactosidase, indicating senescence. The combined treatment is also associated with decline in S-phase and a strong G2/M arrest, indicating massive telomere attrition. In a subcutaneous tumor model, the combined treatment resulted in the smallest tumors, which were even smaller (P=0.001) than those that resulted from either treatment alone. Even the tumors removed from these mice had significantly reduced telomeres and evidence of apoptosis. We therefore conclude that although telomeres are elongated by telomerase, elevated RAD51/HR assist in their maintenance/stabilization in BAC cells. Telomerase inhibitor prevents telomere elongation but induces RAD51/HR, which contributes to telomere maintenance/stabilization and prevention of apoptosis, reducing the efficacy of treatment. Combining HR inhibition with telomerase renders telomeres more vulnerable to degradation and significantly increases/expedites their attrition, leading to apoptosis. We therefore demonstrate that a therapy targeting HR and telomerase has the potential to prevent both tumor growth and genomic evolution in BAC.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/complications , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Genomic Instability/drug effects , Homologous Recombination/drug effects , Telomerase/antagonists & inhibitors , Telomere/drug effects , Adenocarcinoma/complications , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Antineoplastic Combined Chemotherapy Protocols , Barrett Esophagus/enzymology , Barrett Esophagus/genetics , Barrett Esophagus/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Esophageal Neoplasms/complications , Esophageal Neoplasms/drug therapy , Gene Knockout Techniques , Humans , Male , Mice , Oligonucleotides/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Rad51 Recombinase/deficiency , Rad51 Recombinase/genetics , Telomerase/metabolism , Telomere/genetics
9.
Semin Cell Dev Biol ; 22(8): 898-905, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21821141

ABSTRACT

Chromosomal double-strand breaks (DSBs) have the potential to permanently arrest cell cycle progression and endanger cell survival. They must therefore be efficiently repaired to preserve genome integrity and functionality. Homologous recombination (HR) provides an important error-free mechanism for DSB repair in mammalian cells. In addition to RAD51, the central recombinase activity in mammalian cells, a family of proteins known as the RAD51 paralogs and consisting of five proteins (RAD51B, RAD51C, RAD51D, XRCC2 and XRCC3), play an essential role in the DNA repair reactions through HR. The RAD51 paralogs act to transduce the DNA damage signal to effector kinases and to promote break repair. However, their precise cellular functions are not fully elucidated. Here we discuss recent advances in our understanding of how these factors mediate checkpoint responses and act in the HR repair process. In addition, we highlight potential functional similarities with the BRCA2 tumour suppressor, through the recently reported links between RAD51 paralog deficiencies and tumorigenesis triggered by genome instability.


Subject(s)
Cell Transformation, Neoplastic , DNA Damage , DNA Repair , Rad51 Recombinase/metabolism , Recombination, Genetic/genetics , Signal Transduction , Animals , Humans , Rad51 Recombinase/deficiency
10.
PLoS Genet ; 7(7): e1002148, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21779174

ABSTRACT

RAD51 recombinase polymerizes at the site of double-strand breaks (DSBs) where it performs DSB repair. The loss of RAD51 causes extensive chromosomal breaks, leading to apoptosis. The polymerization of RAD51 is regulated by a number of RAD51 mediators, such as BRCA1, BRCA2, RAD52, SFR1, SWS1, and the five RAD51 paralogs, including XRCC3. We here show that brca2-null mutant cells were able to proliferate, indicating that RAD51 can perform DSB repair in the absence of BRCA2. We disrupted the BRCA1, RAD52, SFR1, SWS1, and XRCC3 genes in the brca2-null cells. All the resulting double-mutant cells displayed a phenotype that was very similar to that of the brca2-null cells. We suggest that BRCA2 might thus serve as a platform to recruit various RAD51 mediators at the appropriate position at the DNA-damage site.


Subject(s)
BRCA2 Protein/genetics , Epistasis, Genetic , Homologous Recombination , Rad51 Recombinase/genetics , Animals , Camptothecin/pharmacology , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Chickens , Chromosome Aberrations/drug effects , Chromosome Aberrations/radiation effects , Cisplatin/pharmacology , Clone Cells , DNA Damage , Epistasis, Genetic/drug effects , Epistasis, Genetic/radiation effects , Gamma Rays , Gene Conversion/drug effects , Gene Conversion/radiation effects , Gene Deletion , Genetic Loci/genetics , Genome/genetics , Homologous Recombination/drug effects , Homologous Recombination/radiation effects , Models, Biological , Phenotype , Phthalazines/pharmacology , Piperazines/pharmacology , Rad51 Recombinase/deficiency
11.
Radiat Res ; 173(1): 27-39, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20041757

ABSTRACT

To test the contribution of homologous recombinational repair (HRR) in repairing DNA damage sites induced by high-energy iron ions, we used (1) HRR-deficient rodent cells carrying a deletion in the RAD51D gene and (2) syngeneic human cells impaired for HRR by RAD51D or RAD51 knockdown using RNA interference. We found that in response to exposure to iron ions, HRR contributed to cell survival in rodent cells and that HRR deficiency abrogated RAD51 focus formation. Complementation of the HRR defect by human RAD51D rescues both enhanced cytotoxicity and RAD51 focus formation. For human cells irradiated with iron ions, cell survival was decreased, and in p53 mutant cells, the levels of mutagenesis were increased when HRR was impaired. Human cells synchronized in S phase exhibited a more pronounced resistance to iron ions compared with cells in G(1) phase, and this increase in radioresistance was diminished by RAD51 knockdown. These results indicate a role for RAD51-mediated DNA repair (i.e. HRR) in removing a fraction of clustered lesions induced by charged-particle radiation. Our results are the first to directly show the requirement for an intact HRR pathway in human cells in ensuring DNA repair and cell survival after exposure to high-energy high-LET radiation.


Subject(s)
DNA Breaks, Double-Stranded/radiation effects , DNA Repair/genetics , Iron/toxicity , Recombination, Genetic , Animals , Base Sequence , CHO Cells , Cell Survival/genetics , Cell Survival/radiation effects , Cricetinae , Cricetulus , G2 Phase/genetics , G2 Phase/radiation effects , Gene Knockdown Techniques , Humans , Mutagenesis/radiation effects , Mutation/radiation effects , Rad51 Recombinase/deficiency , Rad51 Recombinase/genetics , Radiation Tolerance/genetics , S Phase/genetics , S Phase/radiation effects , Thymidine Kinase/genetics , Tumor Suppressor Protein p53/metabolism
12.
Cancer Res ; 69(3): 863-72, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19155299

ABSTRACT

RecA/Rad51 protein family members (Rad51, Rad51b, Rad51c, Rad51d, Xrcc2, and Xrcc3) are essential for DNA repair by homologous recombination, and their role in cancers has been anticipated. Here we provide the first direct evidence for a tumor suppressor function for a member of the Rad51 family. We show that Rad51c deficiency leads to early embryonic lethality, which can be delayed on a Trp53-null background. To uncover the role of Rad51c in tumorigenesis, we have exploited the fact that Rad51c and Trp53 are both closely located on the mouse chromosome 11. We have generated double heterozygous (DH) mice carrying mutant alleles of both genes either on different (DH-trans) or on the same chromosome (DH-cis), the latter allowing for a deletion of wild-type alleles of both genes by loss of heterozygosity. DH-trans mice, in contrast to DH-cis, developed tumors with latency and spectrum similar to Trp53 heterozygous mice. Strikingly, Rad51c mutation in DH-cis mice promoted the development of tumors of specialized sebaceous glands and suppressed tumors characteristic of Trp53 mutation. In addition, DH-cis females developed tumors significantly earlier than any other group.


Subject(s)
Cell Transformation, Neoplastic/genetics , Genes, Tumor Suppressor , Rad51 Recombinase/genetics , Tumor Suppressor Protein p53/genetics , Adenocarcinoma, Sebaceous/genetics , Alleles , Animals , DNA-Binding Proteins , Embryonic Development/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Female , Loss of Heterozygosity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rad51 Recombinase/deficiency , Sebaceous Gland Neoplasms/genetics , Sex Factors
13.
Mol Cancer Ther ; 8(1): 203-13, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19139130

ABSTRACT

RAD51 is a key protein in the homologous recombination (HR) pathway of DNA double-strand break repair, and HR represents a novel target for cancer therapy. Because imatinib (Gleevec) has been reported to reduce RAD51 protein levels, we tested the clonogenic survival for RT112, H1299, PANC1, and PC3 tumor cell lines of varying p53 status and normal GM05757 normal fibroblasts after exposure to single agent imatinib (0-20 micromol/L; 0-72 hours). We also combined imatinib with DNA damaging agents that are toxic to RAD51-deficient cells, including ionizing radiation, gemcitabine, and mitomycin C. We observed decreased nuclear expression and chromatin binding of RAD51 protein following imatinib treatment. Imatinib also resulted in decreased error-free HR as determined by a flow cytometry-based integrated direct repeat-green fusion protein reporter system; this correlated to reduced RAD51 expression. Clonogenic survival experiments revealed increased cell kill for imatinib-treated cells in combination with ionizing radiation, gemcitabine, and mitomycin C, due in part to mitotic catastrophe. In experiments using imatinib and gemcitabine, tumor cell lines were sensitized to a greater extent than normal fibroblasts. This preservation of the therapeutic ratio was confirmed in vivo using PC3 xenograft growth delay and intestinal crypt cell clonogenic assays. HR inhibition may be an additional mechanism of action for the chemosensitization and radiosensitization of solid tumors with imatinib with preservation of the therapeutic ratio.


Subject(s)
Neoplasms/drug therapy , Neoplasms/radiotherapy , Piperazines/pharmacology , Pyrimidines/pharmacology , Radiation Tolerance , Recombination, Genetic/drug effects , Animals , Benzamides , Cell Line, Tumor , Humans , Imatinib Mesylate , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mitosis/drug effects , Neoplasms/pathology , Rad51 Recombinase/deficiency , Rad51 Recombinase/metabolism , Xenograft Model Antitumor Assays
14.
Mutat Res ; 625(1-2): 145-54, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17662313

ABSTRACT

Particulate hexavalent chromium [Cr(VI)] compounds are well-established human carcinogens. Cr(VI)-induced tumors are characterized by chromosomal instability (CIN); however, the mechanisms of this effect are unknown. We investigated the hypothesis that homologous recombination (HR) repair of DNA double-strand breaks protect cells from Cr(VI)-induced CIN by focusing on the XRCC3 and RAD51C genes, which play an important role in cellular resistance to DNA double-strand breaks. We used Chinese hamster cells defective in each HR gene (irs3 for RAD51C and irs1SF for XRCC3) and compared with their wildtype parental and cDNA-complemented controls. We found that the intracellular Cr ion levels varied among the cell lines after particulate chromate treatment. Importantly, accounting for differences in Cr ion levels, we discovered that XRCC3 and RAD51C cells treated with lead chromate had increased cytotoxicity and chromosomal aberrations, relative to wildtype and cDNA-complimented cells. We also observed the emergence of high levels of chromatid exchanges in the two mutant cell lines. For example, 1microg/cm(2) lead chromate induced 20 and 32 exchanges in XRCC3- and RAD51C-deficient cells, respectively, whereas no exchanges were detected in the wildtype and cDNA-complemented cells. These observations suggest that HR protects cells from Cr(VI)-induced CIN, consistent with the ability of particulate Cr(VI) to induce double-strand breaks.


Subject(s)
Chromates/toxicity , Chromosomal Instability/drug effects , DNA Repair , Lead/toxicity , Recombination, Genetic , Animals , CHO Cells , Carcinogens/toxicity , Cell Line , Cricetinae , Cricetulus , DNA Breaks, Double-Stranded/drug effects , DNA Repair/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Humans , Rad51 Recombinase/deficiency , Rad51 Recombinase/genetics
15.
J Gene Med ; 7(8): 1044-52, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15756713

ABSTRACT

BACKGROUND: Every cancer therapy appears to be transiently effective for cancer regression, but cancers gradually transform to be resistant to the therapy. Cancers also develop machineries to resist chemotherapy. Short interfering RNA (siRNA) has been evaluated as an attractive and effective tool for suppressing a target protein by specifically digesting its mRNA. Suppression of the machineries using siRNA may enhance the sensitivity to chemotherapy in cancers when combined with an effective delivery system. METHODS: To enhance the anti-cancer effect of chemotherapy, we transferred siRNA against Rad51 into various human cancer cells using the HVJ (hemagglutinating virus of Japan, Sendai virus) envelope vector in the presence or absence of cis-diamminedichloroplatinum(II) (CDDP, cisplatin). The inhibition of cell growth was assessed by a modified MTT assay, counting cell number, or fluorescence-activated cell sorting (FACS) analysis after Annexin V labeling. The synthetic Rad51 siRNA was also introduced into subcutaneous tumor masses of HeLa cells in SCID mice with or without intraperitoneal injection of CDDP, and tumor growth was monitored. RESULTS: When synthetic Rad51 siRNA was delivered into HeLa cells using the HVJ envelope vector, no Rad51 transcripts were detected on day 2, and Rad51 protein completely disappeared for 4 days after siRNA transfer. When HeLa cells were incubated with 0.02 microg/ml CDDP for 3 h after siRNA transfer, the number of colonies decreased to approximately 10% of that with scrambled siRNA. The sensitivity to CDDP was enhanced in various human cancer cells, but not in normal human fibroblasts. When Rad51 siRNA was delivered into tumors using the HVJ envelope vector, the Rad51 transcript level was reduced to approximately 25%. Rad51 siRNA combined with CDDP significantly inhibited tumor growth when compared to siRNA or CDDP alone. CONCLUSIONS: Rad51 siRNA could enhance the sensitivity to CDDP in cancer cells both in vitro and in vivo. Our results suggest that the combination of CDDP and Rad51 siRNA will be an effective anti-cancer protocol.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , RNA, Small Interfering/genetics , Rad51 Recombinase/deficiency , Rad51 Recombinase/genetics , Sendai virus/genetics , Animals , Antineoplastic Agents/therapeutic use , Cell Count , Cell Line, Tumor , Chick Embryo , Cisplatin/therapeutic use , Fibroblasts , Genetic Vectors , HeLa Cells , Humans , Male , Mice , Mice, SCID , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Tumor Burden
SELECTION OF CITATIONS
SEARCH DETAIL
...