Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Biochim Biophys Acta Mol Cell Res ; 1869(11): 119332, 2022 11.
Article in English | MEDLINE | ID: mdl-35940372

ABSTRACT

Ultraviolet (UV) light irradiation generates pyrimidine dimers on DNA, such as cyclobutane pyrimidine dimers (CPDs) and (6-4) photoproducts. Such dimers distort the high-order DNA structure and prevent transcription and replication. The nucleotide excision repair (NER) system contributes to resolving this type of DNA lesion. There are two pathways that recognize pyrimidine dimers. One acts on transcribed strands of DNA (transcription-coupled NER), and the other acts on the whole genome (global genome-NER; GG-NER). In the latter case, DNA damage-binding protein 2 (DDB2) senses pyrimidine dimers with several histone modification enzymes. We previously reported that histone acetyltransferase binding to ORC1 (HBO1) interacts with DDB2 and facilitates recruitment of the imitation switch chromatin remodeler at UV-irradiated sites via an unknown methyltransferase. Here, we found that the phosphorylated histone methyltransferase mixed lineage leukemia 1 (MLL1) was maintained at UV-irradiated sites in an HBO1-dependent manner. Furthermore, MLL1 catalyzed histone H3K4 methylation and recruited the chromatin remodeler bromodomain adjacent to zinc finger domain 1A (BAZ1A)/ATP-utilizing chromatin assembly and remodeling factor 1 (ACF1). Depletion of MLL1 suppressed BAZ1A accumulation at UV-irradiated sites and inhibited the removal of CPDs. These data indicate that the DDB2-HBO1-MLL1 axis is essential for the recruitment of BAZ1A to facilitate GG-NER.


Subject(s)
Leukemia , Pyrimidine Dimers , Chromatin/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Damage , DNA Repair , Humans , Pyrimidine Dimers/chemistry , Pyrimidine Dimers/metabolism
2.
Mol Cancer Res ; 18(9): 1367-1378, 2020 09.
Article in English | MEDLINE | ID: mdl-32527949

ABSTRACT

Recent studies have demonstrated that lysine acetylation of histones is crucial for nucleotide excision repair (NER) by relaxing the chromatin structure, which facilitates the recruitment of repair factors. However, few studies have focused on the contribution of histone deacetylases (HDAC) to NER. Here, we found that histone H3 Lys14 (H3K14) was deacetylated by HDAC3 after UV irradiation. Depletion of HDAC3 caused defects in cyclobutene pyrimidine dimer excision and sensitized cells to UV irradiation. HDAC3-depleted cells had impaired unscheduled DNA synthesis, but not recovery of RNA synthesis, which indicates that HDAC3 was required for global genome NER. Moreover, xeroderma pigmentosum, complementation group C (XPC) accumulation at the local UV-irradiated area was attenuated in HDAC3-depleted cells. In addition to the delay of XPC accumulation at DNA damage sites, XPC ubiquitylation was inhibited in HDAC3-depleted cells. These results suggest that the deacetylation of histone H3K14 by HDAC3 after UV irradiation contributes to XPC recruitment to DNA lesions to promote global genome NER. IMPLICATIONS: Involvement of histone deacetylation for XPC accumulation after UV irradiation indicates conversion of chromatin structure is essential for nucleotide excision repair in human cancer cells.


Subject(s)
DNA Damage , DNA Repair , DNA-Binding Proteins/metabolism , Histone Deacetylases/metabolism , DNA-Binding Proteins/genetics , HeLa Cells , Histone Deacetylases/genetics , Humans , Ultraviolet Rays/adverse effects
3.
Semin Cancer Biol ; 58: 29-46, 2019 10.
Article in English | MEDLINE | ID: mdl-30922960

ABSTRACT

Cancer cells show various types of mutations and aberrant expression in genes involved in DNA repair responses. These alterations induce genome instability and promote carcinogenesis steps and cancer progression processes. These defects in DNA repair have also been considered as suitable targets for cancer therapies. A most effective target so far clinically demonstrated is "homologous recombination repair defect", such as BRCA1/2 mutations, shown to cause synthetic lethality with inhibitors of poly(ADP-ribose) polymerase (PARP), which in turn is involved in DNA repair as well as multiple physiological processes. Different approaches targeting genomic instability, including immune therapy targeting mismatch-repair deficiency, have also recently been demonstrated to be promising strategies. In these DNA repair targeting-strategies, common issues could be how to optimize treatment and suppress/conquer the development of drug resistance. In this article, we review the extending framework of DNA repair response pathways and the potential impact of exploiting those defects on cancer treatments, including chemotherapy, radiation therapy and immune therapy.


Subject(s)
DNA Repair/genetics , Neoplasms/genetics , Animals , Carcinogenesis/genetics , Genomic Instability/genetics , Humans , Mutation/genetics
5.
Mol Cell Biol ; 37(23)2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28894029

ABSTRACT

DNA replication is frequently perturbed by intrinsic, as well as extrinsic, genotoxic stress. At damaged forks, DNA replication and repair activities require proper coordination to maintain genome integrity. We show here that PARI antirecombinase plays an essential role in modulating the initial response to replication stress in mice. PARI is functionally dormant at replisomes during normal replication, but upon replication stress, it enhances nascent-strand shortening that is regulated by RAD51 and MRE11. PARI then promotes double-strand break induction, followed by new origin firing instead of replication restart. Such PARI function is apparently obstructive to replication but is nonetheless physiologically required for chromosome stability in vivo and ex vivo Of note, Pari-deficient embryonic stem cells exhibit spontaneous chromosome instability, which is attenuated by differentiation induction, suggesting that pluripotent stem cells have a preferential requirement for PARI that acts against endogenous replication stress. PARI is a latent modulator of stalled fork processing, which is required for stable genome inheritance under both endogenous and exogenous replication stress in mice.


Subject(s)
Chromosomal Instability/genetics , DNA Damage/genetics , DNA Repair/genetics , DNA Replication/genetics , DNA-Binding Proteins/genetics , Genomic Instability/genetics , Animals , DNA Breaks, Double-Stranded , DNA-Binding Proteins/metabolism , Humans , Mice
6.
Nat Commun ; 8: 16102, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28719581

ABSTRACT

HBO1, a histone acetyl transferase, is a co-activator of DNA pre-replication complex formation. We recently reported that HBO1 is phosphorylated by ATM and/or ATR and binds to DDB2 after ultraviolet irradiation. Here, we show that phosphorylated HBO1 at cyclobutane pyrimidine dimer (CPD) sites mediates histone acetylation to facilitate recruitment of XPC at the damaged DNA sites. Furthermore, HBO1 facilitates accumulation of SNF2H and ACF1, an ATP-dependent chromatin remodelling complex, to CPD sites. Depletion of HBO1 inhibited repair of CPDs and sensitized cells to ultraviolet irradiation. However, depletion of HBO1 in cells derived from xeroderma pigmentosum patient complementation groups, XPE, XPC and XPA, did not lead to additional sensitivity towards ultraviolet irradiation. Our findings suggest that HBO1 acts in concert with SNF2H-ACF1 to make the chromosome structure more accessible to canonical nucleotide excision repair factors.


Subject(s)
DNA Repair , Histone Acetyltransferases/metabolism , Adenosine Triphosphatases/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Humans , Phosphorylation , Pyrimidine Dimers/metabolism , Transcription Factors/metabolism , Ultraviolet Rays
7.
Oncotarget ; 7(7): 7701-14, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26713604

ABSTRACT

Breast cancer is one of the leading causes of death worldwide, and therefore, new and improved approaches for the treatment of breast cancer are desperately needed. CtIP (RBBP8) is a multifunctional protein that is involved in various cellular functions, including transcription, DNA replication, DNA repair and the G1 and G2 cell cycle checkpoints. CtIP plays an important role in homologous recombination repair by interacting with tumor suppressor protein BRCA1. Here, we analyzed the expression profile of CtIP by data mining using published microarray data sets. We found that CtIP expression is frequently decreased in breast cancer patients, and the patient group with low-expressing CtIP mRNA is associated with a significantly lower survival rate. The knockdown of CtIP in breast cancer MCF7 cells reduced Rad51 foci numbers and enhanced f H2AX foci formation after f-irradiation, suggesting that deficiency of CtIP decreases homologous recombination repair and delays DNA double strand break repair. To explore the effect of CtIP on PARP inhibitor therapy for breast cancer, CtIP-depleted MCF7 cells were treated with PARP inhibitor olaparib (AZD2281) or veliparib (ABT-888). As in BRCA mutated cells, PARP inhibitors showed cytotoxicity to CtIP-depleted cells by preventing cells from repairing DNA damage, leading to decreased cell viability. Further, a xenograft tumor model in mice with MCF7 cells demonstrated significantly increased sensitivity towards PARP inhibition under CtIP deficiency. In summary, this study shows that low level of CtIP expression is associated with poor prognosis in breast cancer, and provides a rationale for establishing CtIP expression as a biomarker of PARP inhibitor response, and consequently offers novel therapeutic options for a significant subset of patients.


Subject(s)
Breast Neoplasms/drug therapy , Carrier Proteins/metabolism , Drug Resistance, Neoplasm , Nuclear Proteins/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Recombinational DNA Repair/genetics , Animals , Apoptosis , BRCA1 Protein/genetics , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carrier Proteins/genetics , Cell Proliferation , Endodeoxyribonucleases , Female , Fluorescent Antibody Technique , Follow-Up Studies , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Fluorescence , Middle Aged , Neoplasm Grading , Nuclear Proteins/genetics , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Methods Mol Biol ; 1114: 25-35, 2014.
Article in English | MEDLINE | ID: mdl-24557895

ABSTRACT

Reverse genetics is gaining importance in the field of modern biological sciences. Gene disruption and the use of siRNAs are the favored techniques for current research. Many researchers, however, are aware that the data from siRNA experiments are frequently inconsistent and that epistatic analysis of multiple genes using siRNAs is barely feasible. In recognition of the drawbacks of the siRNA technique, many researchers, especially in the field of DNA repair, are now introducing multiple genetic disruption techniques using the chicken DT40 cell line into their research. Thus, recent publications increasingly include data utilizing DT40 cells. In this chapter, we describe the current standard methods of multiple genetic manipulation in DT40 cells. We place a particular emphasis on describing the basic concepts and theoretical background of the genetic manipulation of DT40 cells for researchers who are new to such techniques.


Subject(s)
Gene Targeting/methods , Genetic Engineering , Alleles , Animals , Cell Line , Chickens , Gene Expression , Plasmids/genetics , Transgenes
9.
Biochem Biophys Res Commun ; 426(3): 310-6, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22943854

ABSTRACT

SUMO conjugation is a reversible posttranslational modification that regulates protein function. SENP1 is one of the six SUMO-specific proteases present in vertebrate cells and its altered expression is observed in several carcinomas. To characterize SENP1 role in genome integrity, we generated Senp1 knockout chicken DT40 cells. SENP1(-/-) cells show normal proliferation, but are sensitive to spindle poisons. This hypersensitivity correlates with increased sister chromatid separation, mitotic slippage, and apoptosis. To test whether the cohesion defect had a causal relationship with the observed mitotic events, we restored the cohesive status of sister chromatids by introducing the TOP2α(+/-) mutation, which leads to increased catenation, or by inhibiting Plk1 and Aurora B kinases that promote cohesin release from chromosomes during prolonged mitotic arrest. Although TOP2α is SUMOylated during mitosis, the TOP2α(+/-) mutation had no obvious effect. By contrast, inhibition of Plk1 or Aurora B rescued the hypersensitivity of SENP1(-/-) cells to colcemid. In conclusion, we identify SENP1 as a novel factor required for mitotic arrest and cohesion maintenance during prolonged mitotic arrest induced by spindle poisons.


Subject(s)
Endopeptidases/physiology , Genomic Instability , Mitosis/physiology , SUMO-1 Protein/physiology , Spindle Apparatus/physiology , Animals , Antigens, Neoplasm/genetics , Aurora Kinases , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/metabolism , Cell Line , Chickens , Chromosomal Proteins, Non-Histone/deficiency , DNA Breaks, Double-Stranded , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/genetics , Demecolcine/pharmacology , Endopeptidases/genetics , Mitosis/drug effects , Nocodazole/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , SUMO-1 Protein/genetics , Spindle Apparatus/drug effects , Tubulin Modulators/pharmacology , Vinblastine/analogs & derivatives , Vinblastine/pharmacology , Vinorelbine , Cohesins , Polo-Like Kinase 1
10.
DNA Repair (Amst) ; 10(2): 210-24, 2011 Feb 07.
Article in English | MEDLINE | ID: mdl-21130053

ABSTRACT

DPB11/TopBP1 is an essential evolutionarily conserved gene involved in initiation of DNA replication and checkpoint signaling. Here, we show that Saccharomyces cerevisiae Dpb11 forms nuclear foci that localize to sites of DNA damage in G1, S and G2 phase, a recruitment that is conserved for its homologue TopBP1 in Gallus gallus. Damage-induced Dpb11 foci are distinct from Sld3 replication initiation foci. Further, Dpb11 foci are dependent on the checkpoint proteins Mec3 (9-1-1 complex) and Rad24, and require the C-terminal domain of Dpb11. Dpb11 foci are independent of the checkpoint kinases Mec1 and Tel1, and of the checkpoint mediator Rad9. In a site-directed mutagenesis screen, we identify a separation-of-function mutant, dpb11-PF, that is sensitive to DSB-inducing agents yet remains proficient for DNA replication and the S-phase checkpoint at the permissive temperature. The dpb11-PF mutant displays altered rates of heteroallelic and direct-repeat recombination, sensitivity to DSB-inducing drugs as well as delayed kinetics of mating-type switching with a defect in the DNA synthesis step thus implicating Dpb11 in homologous recombination. We conclude that Dpb11/TopBP1 plays distinct roles in replication, checkpoint response and recombination processes, thereby contributing to chromosomal stability.


Subject(s)
Cell Cycle Proteins/physiology , Chromosomal Instability , DNA Replication , Recombination, Genetic , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/genetics , Animals , Cell Cycle Proteins/genetics , Chickens/genetics , DNA Breaks, Double-Stranded , DNA Damage , G2 Phase , Genes, cdc , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Mutagenesis, Site-Directed , S Phase , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae Proteins/genetics
11.
PLoS Genet ; 6(10)2010 Oct 07.
Article in English | MEDLINE | ID: mdl-20949111

ABSTRACT

Replicative DNA polymerases are frequently stalled by DNA lesions. The resulting replication blockage is released by homologous recombination (HR) and translesion DNA synthesis (TLS). TLS employs specialized TLS polymerases to bypass DNA lesions. We provide striking in vivo evidence of the cooperation between DNA polymerase η, which is mutated in the variant form of the cancer predisposition disorder xeroderma pigmentosum (XP-V), and DNA polymerase ζ by generating POLη(-/-)/POLζ(-/-) cells from the chicken DT40 cell line. POLζ(-/-) cells are hypersensitive to a very wide range of DNA damaging agents, whereas XP-V cells exhibit moderate sensitivity to ultraviolet light (UV) only in the presence of caffeine treatment and exhibit no significant sensitivity to any other damaging agents. It is therefore widely believed that Polη plays a very specific role in cellular tolerance to UV-induced DNA damage. The evidence we present challenges this assumption. The phenotypic analysis of POLη(-/-)/POLζ(-/-) cells shows that, unexpectedly, the loss of Polη significantly rescued all mutant phenotypes of POLζ(-/-) cells and results in the restoration of the DNA damage tolerance by a backup pathway including HR. Taken together, Polη contributes to a much wide range of TLS events than had been predicted by the phenotype of XP-V cells.


Subject(s)
DNA Damage , DNA-Directed DNA Polymerase/genetics , Mutation , Animals , Antineoplastic Agents/pharmacology , Base Sequence , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Chickens , Cisplatin/pharmacology , DNA Repair , DNA-Directed DNA Polymerase/metabolism , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , HEK293 Cells , Humans , Methyl Methanesulfonate/pharmacology , Models, Genetic , Molecular Sequence Data , Sequence Homology, Nucleic Acid , Suppression, Genetic , Ultraviolet Rays
13.
Cancer Lett ; 283(1): 1-9, 2009 Sep 28.
Article in English | MEDLINE | ID: mdl-19201084

ABSTRACT

Proteasome-dependent protein degradation is involved in a variety of biological processes, including cell-cycle regulation, apoptosis, and stress-responses. Growing evidence from translational research and clinical trials proved the effectiveness of proteasome inhibitors (PIs) in treating several types of hematological malignancies. Although various key molecules in ubiquitin-dependent cellular processes have been proposed as relevant targets of therapeutic proteasome inhibition, our current understanding is far from complete. Recent rapid progress in DNA repair research has unveiled a crucial role of the ubiquitin-proteasome pathway (UPP) in regulating DNA repair. These findings thus bring up the idea that DNA repair pathways could be effective targets of PIs in mediating their cytotoxicity and enhancing the effect of radiotherapy and some DNA-damaging chemotherapeutic agents, such as cisplatin and camptothecin. In this review, we present the current perspective on the UPP-dependent regulatory mechanisms of DNA repair and discuss their therapeutic potential in the application of PIs to a broad spectrum of human cancers.


Subject(s)
DNA Repair/physiology , Neoplasms/genetics , Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Signal Transduction/physiology , Ubiquitin/metabolism , DNA Repair/drug effects , Humans , Neoplasms/drug therapy , Signal Transduction/drug effects
15.
Proc Natl Acad Sci U S A ; 105(34): 12411-6, 2008 Aug 26.
Article in English | MEDLINE | ID: mdl-18719106

ABSTRACT

Chronic stalling of DNA replication forks caused by DNA damage can lead to genomic instability. Cells have evolved lesion bypass pathways such as postreplication repair (PRR) to resolve these arrested forks. In yeast, one branch of PRR involves proliferating cell nuclear antigen (PCNA) polyubiquitination mediated by the Rad5-Ubc13-Mms2 complex that allows bypass of DNA lesion by a template-switching mechanism. Previously, we identified human SHPRH as a functional homologue of yeast Rad5 and revealed the existence of RAD5-like pathway in human cells. Here we report the identification of HLTF as a second RAD5 homologue in human cells. HLTF, like SHPRH, shares a unique domain architecture with Rad5 and promotes lysine 63-linked polyubiquitination of PCNA. Similar to yeast Rad5, HLTF is able to interact with UBC13 and PCNA, as well as SHPRH; and the reduction of either SHPRH or HLTF expression enhances spontaneous mutagenesis. Moreover, Hltf-deficient mouse embryonic fibroblasts show elevated chromosome breaks and fusions after methyl methane sulfonate treatment. Our results suggest that HLTF and SHPRH are functional homologues of yeast Rad5 that cooperatively mediate PCNA polyubiquitination and maintain genomic stability.


Subject(s)
DNA Helicases/metabolism , DNA Replication , DNA-Binding Proteins/metabolism , Genomic Instability , Polyubiquitin/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Adenosine Triphosphatases , DNA Damage , Humans , Saccharomyces cerevisiae Proteins , Structural Homology, Protein , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination
16.
J Leukoc Biol ; 84(4): 1047-56, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18653461

ABSTRACT

We previously proposed that mouse CD8(+)CD122(+) T cells and human CD57(+) T cells, which increase with age and exhibit potent IFN-gamma production, represent a double-edged sword as they play critical roles in host defense and the lethal IL-12/LPS-induced generalized Shwartzman reaction (GSR). However, our proposal was based solely on comparisons of young and old mice. In this study, we attempted to increase CD8(+)CD122(+) T cells in young mice with exogenous IL-15 and confirm their countervailing functions in young mice. After young mice (6 weeks) were injected with IL-15, they showed significant increases in CD8(+)CD122(+) T cells in the liver and spleen. Liver CD8(+)CD122(+) T cells from IL-15-pretreated mice had a potent capacity to produce IFN-gamma after IL-12 injection or Escherichia coli infection. IL-15-pretreated mice showed increased survival to E. coli infections and enhanced anti-tumor activities against liver metastatic EL4 cells, as well as an exacerbation of the GSR. Correspondingly, liver CD8(+)CD122(+) T cells produced more perforin than CD8(+)CD122(-) T cells in EL4-inoculated mice. Unexpectedly, comparable IL-15 treatment did not induce further increases in CD8(+)CD122(+) T cells in aged mice and did not enhance their defenses against bacterial infection or tumor growth. Interestingly, however, nontreated, aged mice (50 weeks) showed twofold higher IL-15 levels (but not TNF or IFN-gamma) in liver homogenates compared with young mice. Our results further support that CD8(+)CD122(+) T cells, which are increased physiologically or therapeutically by IL-15, are involved in antibacterial immunity, anti-tumor immunity, and the GSR.


Subject(s)
Aging/immunology , Bacterial Infections/immunology , CD8-Positive T-Lymphocytes/immunology , Escherichia coli Infections/immunology , Interleukin-15/therapeutic use , Interleukin-2 Receptor beta Subunit/immunology , Killer Cells, Natural/immunology , Neoplasms, Experimental/immunology , Animals , CD8 Antigens/immunology , Escherichia coli Infections/mortality , Flow Cytometry , Interferon-gamma/immunology , Liver Neoplasms, Experimental/immunology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Survival Analysis , Survivors
17.
J Cell Biol ; 181(7): 1083-93, 2008 Jun 30.
Article in English | MEDLINE | ID: mdl-18591428

ABSTRACT

Gross chromosomal rearrangement (GCR) is a type of genomic instability associated with many cancers. In yeast, multiple pathways cooperate to suppress GCR. In a screen for genes that promote GCR, we identified MPH1, which encodes a 3'-5' DNA helicase. Overexpression of Mph1p in yeast results in decreased efficiency of homologous recombination (HR) as well as delayed Rad51p recruitment to double-strand breaks (DSBs), which suggests that Mph1p promotes GCR by partially suppressing HR. A function for Mph1p in suppression of HR is further supported by the observation that deletion of both mph1 and srs2 synergistically sensitize cells to methyl methanesulfonate-induced DNA damage. The GCR-promoting activity of Mph1p appears to depend on its interaction with replication protein A (RPA). Consistent with this observation, excess Mph1p stabilizes RPA at DSBs. Furthermore, spontaneous RPA foci at DSBs are destabilized by the mph1Delta mutation. Therefore, Mph1p promotes GCR formation by partially suppressing HR, likely through its interaction with RPA.


Subject(s)
Chromosomes, Fungal/genetics , DEAD-box RNA Helicases/metabolism , Recombination, Genetic , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Adenosine Triphosphatases/metabolism , Amino Acid Motifs , DEAD-box RNA Helicases/chemistry , Methyl Methanesulfonate/pharmacology , Mutation/genetics , Protein Binding/drug effects , Recombination, Genetic/drug effects , Replication Protein A/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/chemistry
18.
Methods ; 41(2): 168-76, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17189859

ABSTRACT

Gross chromosomal rearrangements (GCRs), including translocations, deletions, amplifications and aneuploidy are frequently observed in various types of human cancers. Despite their clear importance in carcinogenesis, the molecular mechanisms by which GCRs are generated and held in check are poorly understood. By using a GCR assay, which can measure the rate of accumulation of spontaneous GCRs in Saccharomyces cerevisiae, we have found that many proteins involved in DNA replication, DNA repair, DNA recombination, checkpoints, chromosome remodeling, and telomere maintenance, play crucial roles in GCR metabolism. We describe here the theoretical background and practical procedures of this GCR assay. We will explain the breakpoint structure and DNA damage that lead to GCR formation. We will also summarize the pathways that suppress and enhance GCR formation. Finally, we will briefly describe similar assays developed by others and discuss their potential in studying GCR metabolism.


Subject(s)
Biological Assay/methods , Chromosome Aberrations , Chromosomes, Human, Pair 5/genetics , Neoplasms/genetics , Saccharomyces cerevisiae/genetics , Humans , Mutation , Time Factors
19.
Scand J Infect Dis ; 38(11-12): 1122-5, 2006.
Article in English | MEDLINE | ID: mdl-17148094

ABSTRACT

We describe a 15-month-old girl with acute necrotizing encephalopathy (ANE) associated with HHV-6. Inflammatory cytokines were elevated in the CSF and serum and the number of CD56bright NK cells was significantly increased in the peripheral blood. CD56bright NK cells may be involved in the pathogenesis of ANE by producing inflammatory cytokines.


Subject(s)
Brain Diseases/virology , Herpesvirus 6, Human/pathogenicity , Roseolovirus Infections/virology , Brain Diseases/complications , Brain Diseases/immunology , CD56 Antigen/immunology , Cytokines/blood , Cytokines/cerebrospinal fluid , Female , Herpesvirus 6, Human/immunology , Humans , Immunoglobulin G/cerebrospinal fluid , Infant , Killer Cells, Natural/immunology , Necrosis/virology , Roseolovirus Infections/complications , Roseolovirus Infections/immunology
20.
J Cell Biol ; 175(5): 703-8, 2006 Dec 04.
Article in English | MEDLINE | ID: mdl-17130289

ABSTRACT

Differential modifications of proliferating cell nuclear antigen (PCNA) determine DNA repair pathways at stalled replication forks. In yeast, PCNA monoubiquitination by the ubiquitin ligase (E3) yRad18 promotes translesion synthesis (TLS), whereas the lysine-63-linked polyubiquitination of PCNA by yRad5 (E3) promotes the error-free mode of bypass. The yRad5-dependent pathway is important to prevent genomic instability during replication, although its exact molecular mechanism is poorly understood. This mechanism has remained totally elusive in mammals because of the lack of apparent RAD5 homologues. We report that a putative tumor suppressor gene, SHPRH, is a human orthologue of yeast RAD5. SHPRH associates with PCNA, RAD18, and the ubiquitin-conjugating enzyme UBC13 (E2) and promotes methyl methanesulfonate (MMS)-induced PCNA polyubiquitination. The reduction of SHPRH by stable short hairpin RNA increases sensitivity to MMS and enhances genomic instability. Therefore, the yRad5/SHPRH-dependent pathway is a conserved and fundamental DNA repair mechanism that protects the genome from genotoxic stress.


Subject(s)
DNA Helicases/genetics , Genomic Instability , Polyubiquitin/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitin-Protein Ligases/genetics , Amino Acid Sequence , Cell Line , Conserved Sequence , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Humans , Molecular Sequence Data , Protein Structure, Tertiary , Saccharomyces cerevisiae Proteins/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...