Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Cancer ; 5(1): 66-84, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38151625

RESUMO

Chromosomal instability (CIN) is a hallmark of cancer, caused by persistent errors in chromosome segregation during mitosis. Aggressive cancers like high-grade serous ovarian cancer (HGSOC) and triple-negative breast cancer (TNBC) have a high frequency of CIN and TP53 mutations. Here, we show that inhibitors of the KIF18A motor protein activate the mitotic checkpoint and selectively kill chromosomally unstable cancer cells. Sensitivity to KIF18A inhibition is enriched in TP53-mutant HGSOC and TNBC cell lines with CIN features, including in a subset of CCNE1-amplified, CDK4-CDK6-inhibitor-resistant and BRCA1-altered cell line models. Our KIF18A inhibitors have minimal detrimental effects on human bone marrow cells in culture, distinct from other anti-mitotic agents. In mice, inhibition of KIF18A leads to robust anti-cancer effects with tumor regression observed in human HGSOC and TNBC models at well-tolerated doses. Collectively, our results provide a rational therapeutic strategy for selective targeting of CIN cancers via KIF18A inhibition.


Assuntos
Cinesinas , Neoplasias de Mama Triplo Negativas , Humanos , Animais , Camundongos , Cinesinas/genética , Cinesinas/metabolismo , Mitose/genética , Linhagem Celular , Pontos de Checagem da Fase M do Ciclo Celular
2.
Mutat Res Rev Mutat Res ; 792: 108466, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37643677

RESUMO

Error-corrected Next Generation Sequencing (ecNGS) is rapidly emerging as a valuable, highly sensitive and accurate method for detecting and characterizing mutations in any cell type, tissue or organism from which DNA can be isolated. Recent mutagenicity and carcinogenicity studies have used ecNGS to quantify drug-/chemical-induced mutations and mutational spectra associated with cancer risk. ecNGS has potential applications in genotoxicity assessment as a new readout for traditional models, for mutagenesis studies in 3D organotypic cultures, and for detecting off-target effects of gene editing tools. Additionally, early data suggest that ecNGS can measure clonal expansion of mutations as a mechanism-agnostic early marker of carcinogenic potential and can evaluate mutational load directly in human biomonitoring studies. In this review, we discuss promising applications, challenges, limitations, and key data initiatives needed to enable regulatory testing and adoption of ecNGS - including for advancing safety assessment, augmenting weight-of-evidence for mutagenicity and carcinogenicity mechanisms, identifying early biomarkers of cancer risk, and managing human health risk from chemical exposures.


Assuntos
Sequenciamento de Nucleotídeos em Larga Escala , Mutagênicos , Humanos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Testes de Mutagenicidade , Mutação , Mutagênicos/toxicidade , Carcinógenos/toxicidade , Carcinogênese , Medição de Risco
4.
Regul Toxicol Pharmacol ; 131: 105162, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35331777

RESUMO

Sotorasib (Lumakras™) is a first-in-class, non-genotoxic, small molecule inhibitor of KRAS G12C developed as an anticancer therapeutic for treatment of patients that have a high unmet medical need. Anticancer therapeutics are considered out of scope of ICH M7 guidance for control of mutagenic impurities; however, based on ICH S9 Q&A, mutagenicity assessments are needed for impurities that exceed the qualification threshold, consistent with ICH Q3A/B, and non-mutagenic drugs. Here, we carried out hybrid-based mutagenicity assessment of sotorasib drug substance (DS) impurities using in silico quantitative structure-activity relationship (QSAR) modelling and Ames tests (for in silico positive mutagens). We encountered contradictive mutagenicity results for 2 impurities (Beta-Chloride and PAC). PAC was negative initially by QSAR but positive in a GLP full plate Ames test and Beta-Chloride was positive by QSAR, negative in a non-GLP micro-Ames but positive in a GLP full plate Ames assay. Root cause analyses identified and characterized mutagenic contaminants, 3-chloropropionic acid in batches of Beta-Chloride and 3-chloropropionic acid and Chloro-PAC in batches of PAC, used in initial GLP full-plate Ames tests. Significant reduction of these contaminants in re-purified batches resulted in no induction of mutagenicity in follow-up GLP micro-Ames tests. In summary, root-cause analyses led to accurate mutagenicity assessment for sotorasib DS-associated impurities.


Assuntos
Cloretos , Mutagênicos , Humanos , Mutagênese , Testes de Mutagenicidade/métodos , Mutagênicos/toxicidade , Piperazinas , Piridinas , Pirimidinas
5.
Environ Mol Mutagen ; 63(3): 118-134, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35315142

RESUMO

The Genetic Toxicology Technical Committee (GTTC) of the Health and Environmental Sciences Institute (HESI) is developing adverse outcome pathways (AOPs) that describe modes of action leading to potentially heritable genomic damage. The goal was to enhance the use of mechanistic information in genotoxicity assessment by building empirical support for the relationships between relevant molecular initiating events (MIEs) and regulatory endpoints in genetic toxicology. Herein, we present an AOP network that links oxidative DNA damage to two adverse outcomes (AOs): mutations and chromosomal aberrations. We collected empirical evidence from the literature to evaluate the key event relationships between the MIE and the AOs, and assessed the weight of evidence using the modified Bradford-Hill criteria for causality. Oxidative DNA damage is constantly induced and repaired in cells given the ubiquitous presence of reactive oxygen species and free radicals. However, xenobiotic exposures may increase damage above baseline levels through a variety of mechanisms and overwhelm DNA repair and endogenous antioxidant capacity. Unrepaired oxidative DNA base damage can lead to base substitutions during replication and, along with repair intermediates, can also cause DNA strand breaks that can lead to mutations and chromosomal aberrations if not repaired adequately. This AOP network identifies knowledge gaps that could be filled by targeted studies designed to better define the quantitative relationships between key events, which could be leveraged for quantitative chemical safety assessment. We anticipate that this AOP network will provide the building blocks for additional genotoxicity-associated AOPs and aid in designing novel integrated testing approaches for genotoxicity.


Assuntos
Rotas de Resultados Adversos , Aberrações Cromossômicas/induzido quimicamente , DNA , Humanos , Mutação , Estresse Oxidativo/genética , Medição de Risco
6.
Regul Toxicol Pharmacol ; 125: 104985, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34237378

RESUMO

Apremilast is an oral, selective small molecule inhibitor of phosphodiesterase-4 (PDE4) that has been approved for the treatment of active psoriatic arthritis, moderate to severe plaque psoriasis, and for patients with oral ulcers associated with Behçet's disease. Apremilast modulates the inflammatory cascade in cells by inhibiting PDE4, thus preventing the degradation of cyclic adenosine monophosphate, resulting in the upregulation of interleukin (IL)-10 and the downregulation of proinflammatory cytokines, including IL-23, interferon gamma (IFNγ), and tumor necrosis factor alpha (TNFα). Here, we evaluated the genotoxic and carcinogenic potential of apremilast using Good Laboratory Practice (GLP)-compliant in vitro and in vivo studies. Apremilast was not genotoxic in the genetic toxicology battery, as evaluated for mutagenicity in the Ames test up to concentrations of 5000 µg/plate, clastogenicity in cultured human peripheral blood lymphocytes up to concentrations of 700 ug/mL was in excess of the solubility limit in culture medium and not able to assess; and negative for the induction of micronuclei in the bone marrow micronucleus test in mice up to doses of 2000 mg/kg/day. Furthermore, apremilast did not increase the incidence of tumors in lifetime rat or mouse carcinogenicity studies up to the maximum tolerated dose. In summary, in non-clinical studies, apremilast is not genotoxic and is not carcinogenic.


Assuntos
Inibidores da Fosfodiesterase 4/toxicidade , Talidomida/análogos & derivados , Animais , Medula Óssea/efeitos dos fármacos , Testes de Carcinogenicidade , Feminino , Linfócitos/efeitos dos fármacos , Masculino , Camundongos , Testes para Micronúcleos , Testes de Mutagenicidade , Inibidores da Fosfodiesterase 4/farmacologia , Ratos , Ratos Sprague-Dawley , Talidomida/toxicidade
7.
Environ Mol Mutagen ; 62(5): 293-305, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34089278

RESUMO

A genotoxic carcinogen, N-nitrosodimethylamine (NDMA), was detected as a synthesis impurity in some valsartan drugs in 2018, and other N-nitrosamines, such as N-nitrosodiethylamine (NDEA), were later detected in other sartan products. N-nitrosamines are pro-mutagens that can react with DNA following metabolism to produce DNA adducts, such as O6 -alkyl-guanine. The adducts can result in DNA replication miscoding errors leading to GC>AT mutations and increased risk of genomic instability and carcinogenesis. Both NDMA and NDEA are known rodent carcinogens in male and female rats. The DNA repair enzyme, methylguanine DNA-methyltransferase can restore DNA integrity via the removal of alkyl groups from guanine in an error-free fashion and this can result in nonlinear dose responses and a point of departure or "practical threshold" for mutation at low doses of exposure. Following International recommendations (ICHM7; ICHQ3C and ICHQ3D), we calculated permissible daily exposures (PDE) for NDMA and NDEA using published rodent cancer bioassay and in vivo mutagenicity data to determine benchmark dose values and define points of departure and adjusted with appropriate uncertainty factors (UFs). PDEs for NDMA were 6.2 and 0.6 µg/person/day for cancer and mutation, respectively, and for NDEA, 2.2 and 0.04 µg/person/day. Both PDEs are higher than the acceptable daily intake values (96 ng for NDMA and 26.5 ng for NDEA) calculated by regulatory authorities using simple linear extrapolation from carcinogenicity data. These PDE calculations using a bench-mark approach provide a more robust assessment of exposure limits compared with simple linear extrapolations and can better inform risk to patients exposed to the contaminated sartans.


Assuntos
Adutos de DNA , Exposição Ambiental/análise , Mutação , Nitrosaminas/toxicidade , Poluentes Químicos da Água/toxicidade , Animais , Carcinógenos/toxicidade , Feminino , Masculino , Ratos
8.
Proc Natl Acad Sci U S A ; 117(52): 33414-33425, 2020 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-33318186

RESUMO

The ability to accurately measure mutations is critical for basic research and identifying potential drug and chemical carcinogens. Current methods for in vivo quantification of mutagenesis are limited because they rely on transgenic rodent systems that are low-throughput, expensive, prolonged, and do not fully represent other species such as humans. Next-generation sequencing (NGS) is a conceptually attractive alternative for detecting mutations in the DNA of any organism; however, the limit of resolution for standard NGS is poor. Technical error rates (∼1 × 10-3) of NGS obscure the true abundance of somatic mutations, which can exist at per-nucleotide frequencies ≤1 × 10-7 Using duplex sequencing, an extremely accurate error-corrected NGS (ecNGS) technology, we were able to detect mutations induced by three carcinogens in five tissues of two strains of mice within 31 d following exposure. We observed a strong correlation between mutation induction measured by duplex sequencing and the gold-standard transgenic rodent mutation assay. We identified exposure-specific mutation spectra of each compound through trinucleotide patterns of base substitution. We observed variation in mutation susceptibility by genomic region, as well as by DNA strand. We also identified a primordial marker of carcinogenesis in a cancer-predisposed strain of mice, as evidenced by clonal expansions of cells carrying an activated oncogene, less than a month after carcinogen exposure. These findings demonstrate that ecNGS is a powerful method for sensitively detecting and characterizing mutagenesis and the early clonal evolutionary hallmarks of carcinogenesis. Duplex sequencing can be broadly applied to basic mutational research, regulatory safety testing, and emerging clinical applications.


Assuntos
Carcinogênese/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Mutagênese/genética , Animais , Carcinógenos/toxicidade , Análise por Conglomerados , DNA/genética , Genes ras , Loci Gênicos , Genoma , Humanos , Camundongos Transgênicos , Mutação/genética , Neoplasias/genética , Oncogenes , Fenótipo , Transcrição Gênica
9.
Cancer Epidemiol Biomarkers Prev ; 29(10): 1887-1903, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32152214

RESUMO

The key characteristics (KC) of human carcinogens provide a uniform approach to evaluating mechanistic evidence in cancer hazard identification. Refinements to the approach were requested by organizations and individuals applying the KCs. We assembled an expert committee with knowledge of carcinogenesis and experience in applying the KCs in cancer hazard identification. We leveraged this expertise and examined the literature to more clearly describe each KC, identify current and emerging assays and in vivo biomarkers that can be used to measure them, and make recommendations for future assay development. We found that the KCs are clearly distinct from the Hallmarks of Cancer, that interrelationships among the KCs can be leveraged to strengthen the KC approach (and an understanding of environmental carcinogenesis), and that the KC approach is applicable to the systematic evaluation of a broad range of potential cancer hazards in vivo and in vitro We identified gaps in coverage of the KCs by current assays. Future efforts should expand the breadth, specificity, and sensitivity of validated assays and biomarkers that can measure the 10 KCs. Refinement of the KC approach will enhance and accelerate carcinogen identification, a first step in cancer prevention.See all articles in this CEBP Focus section, "Environmental Carcinogenesis: Pathways to Prevention."


Assuntos
Biomarcadores/metabolismo , Carcinógenos/metabolismo , Neoplasias/diagnóstico , Humanos , Neoplasias/patologia
10.
Environ Mol Mutagen ; 61(8): 770-785, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32078182

RESUMO

Genome instability is a hallmark of most human cancers and is exacerbated following replication stress. However, the effects that drugs/xenobiotics have in promoting genome instability including chromosomal structural rearrangements in normal cells are not currently assessed in the genetic toxicology battery. Here, we show that drug-induced replication stress leads to increased genome instability in vitro using proliferating primary human cells as well as in vivo in rat bone marrow (BM) and duodenum (DD). p53-binding protein 1 (53BP1, biomarker of DNA damage repair) nuclear bodies were increased in a dose-dependent manner in normal proliferating human mammary epithelial fibroblasts following treatment with compounds traditionally classified as either genotoxic (hydralazine) and nongenotoxic (low-dose aphidicolin, duvelisib, idelalisib, and amiodarone). Comparatively, no increases in 53BP1 nuclear bodies were observed in nonproliferating cells. Negative control compounds (mannitol, alosteron, diclofenac, and zonisamide) not associated with cancer risk did not induce 53BP1 nuclear bodies in any cell type. Finally, we studied the in vivo genomic consequences of drug-induced replication stress in rats treated with 10 mg/kg of cyclophosphamide for up to 14 days followed by polymerase chain reaction-free whole genome sequencing (30X coverage) of BM and DD cells. Cyclophosphamide induced chromosomal structural rearrangements at an average of 90 genes, including 40 interchromosomal/intrachromosomal translocations, within 2 days of treatment. Collectively, these data demonstrate that this drug-induced genome instability test (DiGIT) can reveal potential adverse effects of drugs not otherwise informed by standard genetic toxicology testing batteries. These efforts are aligned with the food and drug administration's (FDA's) predictive toxicology roadmap initiative.


Assuntos
Replicação do DNA/efeitos dos fármacos , Genoma/efeitos dos fármacos , Instabilidade Genômica , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Biomarcadores/metabolismo , Aberrações Cromossômicas , Ciclofosfamida/toxicidade , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Sequenciamento Completo do Genoma
11.
Trends Pharmacol Sci ; 39(3): 232-247, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29242029

RESUMO

Cancer risk assessment of therapeutics is plagued by poor translatability of rodent models of carcinogenesis. In order to overcome this fundamental limitation, new approaches are needed that enable us to evaluate cancer risk directly in humans and human-based cellular models. Our enhanced understanding of the mechanisms of carcinogenesis and the influence of human genome sequence variation on cancer risk motivates us to re-evaluate how we assess the carcinogenic risk of therapeutics. This review will highlight new opportunities for applying this knowledge to the development of a battery of human-based in vitro models and biomarkers for assessing cancer risk of novel therapeutics.


Assuntos
Carcinógenos/toxicidade , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/prevenção & controle , Neoplasias/prevenção & controle , Farmacovigilância , Biomarcadores Farmacológicos/análise , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Predisposição Genética para Doença , Humanos
12.
Genes Dev ; 31(8): 816-829, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28487407

RESUMO

DNA replication fork progression can be disrupted at difficult to replicate loci in the human genome, which has the potential to challenge chromosome integrity. This replication fork disruption can lead to the dissociation of the replisome and the formation of DNA damage. To model the events stemming from replisome dissociation during DNA replication perturbation, we used a degron-based system for inducible proteolysis of a subunit of the replicative helicase. We show that MCM2-depleted cells activate a DNA damage response pathway and generate replication-associated DNA double-strand breaks (DSBs). Remarkably, these cells maintain some DNA synthesis in the absence of MCM2, and this requires the MCM8-9 complex, a paralog of the MCM2-7 replicative helicase. We show that MCM8-9 functions in a homologous recombination-based pathway downstream from RAD51, which is promoted by DSB induction. This RAD51/MCM8-9 axis is distinct from the recently described RAD52-dependent DNA synthesis pathway that operates in early mitosis at common fragile sites. We propose that stalled replication forks can be restarted in S phase via homologous recombination using MCM8-9 as an alternative replicative helicase.


Assuntos
Replicação do DNA/genética , DNA/biossíntese , Proteínas de Manutenção de Minicromossomo/metabolismo , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , Ativação Enzimática/genética , Células HCT116 , Recombinação Homóloga/genética , Humanos , Componente 2 do Complexo de Manutenção de Minicromossomo/genética , Componente 2 do Complexo de Manutenção de Minicromossomo/metabolismo , Proteínas de Manutenção de Minicromossomo/genética , Mutação , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Fase S/genética
13.
Mol Cell ; 64(6): 1117-1126, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27984745

RESUMO

Homologous recombination (HR) is necessary to counteract DNA replication stress. Common fragile site (CFS) loci are particularly sensitive to replication stress and undergo pathological rearrangements in tumors. At these loci, replication stress frequently activates DNA repair synthesis in mitosis. This mitotic DNA synthesis, termed MiDAS, requires the MUS81-EME1 endonuclease and a non-catalytic subunit of the Pol-delta complex, POLD3. Here, we examine the contribution of HR factors in promoting MiDAS in human cells. We report that RAD51 and BRCA2 are dispensable for MiDAS but are required to counteract replication stress at CFS loci during S-phase. In contrast, MiDAS is RAD52 dependent, and RAD52 is required for the timely recruitment of MUS81 and POLD3 to CFSs in early mitosis. Our results provide further mechanistic insight into MiDAS and define a specific function for human RAD52. Furthermore, selective inhibition of MiDAS may comprise a potential therapeutic strategy to sensitize cancer cells undergoing replicative stress.


Assuntos
DNA Polimerase III/genética , Replicação do DNA , Proteínas de Ligação a DNA/genética , DNA/genética , Endonucleases/genética , Proteína Rad52 de Recombinação e Reparo de DNA/genética , Reparo de DNA por Recombinação , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Linhagem Celular Tumoral , Sítios Frágeis do Cromossomo , DNA/metabolismo , Dano ao DNA , DNA Polimerase III/metabolismo , Proteínas de Ligação a DNA/metabolismo , Endodesoxirribonucleases/genética , Endodesoxirribonucleases/metabolismo , Endonucleases/metabolismo , Células HeLa , Humanos , Mitose , Osteoblastos/citologia , Osteoblastos/metabolismo , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Estresse Fisiológico
14.
Nature ; 528(7581): 286-90, 2015 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-26633632

RESUMO

Oncogene-induced DNA replication stress has been implicated as a driver of tumorigenesis. Many chromosomal rearrangements characteristic of human cancers originate from specific regions of the genome called common fragile sites (CFSs). CFSs are difficult-to-replicate loci that manifest as gaps or breaks on metaphase chromosomes (termed CFS 'expression'), particularly when cells have been exposed to replicative stress. The MUS81-EME1 structure-specific endonuclease promotes the appearance of chromosome gaps or breaks at CFSs following replicative stress. Here we show that entry of cells into mitotic prophase triggers the recruitment of MUS81 to CFSs. The nuclease activity of MUS81 then promotes POLD3-dependent DNA synthesis at CFSs, which serves to minimize chromosome mis-segregation and non-disjunction. We propose that the attempted condensation of incompletely duplicated loci in early mitosis serves as the trigger for completion of DNA replication at CFS loci in human cells. Given that this POLD3-dependent mitotic DNA synthesis is enhanced in aneuploid cancer cells that exhibit intrinsically high levels of chromosomal instability (CIN(+)) and replicative stress, we suggest that targeting this pathway could represent a new therapeutic approach.


Assuntos
Carcinogênese/genética , Reparo do DNA/fisiologia , Replicação do DNA , Endodesoxirribonucleases/metabolismo , Regulação Neoplásica da Expressão Gênica , Mitose/genética , Estresse Fisiológico/genética , Linhagem Celular Tumoral , Instabilidade Cromossômica , Sítios Frágeis do Cromossomo , Segregação de Cromossomos , DNA Polimerase III/metabolismo , Replicação do DNA/genética , Proteínas de Ligação a DNA/metabolismo , Endodesoxirribonucleases/genética , Endonucleases/metabolismo , Células HCT116 , Células HT29 , Células HeLa , Humanos , Modelos Biológicos , Não Disjunção Genética/genética
15.
DNA Repair (Amst) ; 25: 84-96, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25497329

RESUMO

SUMOylation is a form of post-translational modification involving covalent attachment of SUMO (Small Ubiquitin-like Modifier) polypeptides to specific lysine residues in the target protein. In human cells, there are four SUMO proteins, SUMO1-4, with SUMO2 and SUMO3 forming a closely related subfamily. SUMO2/3, in contrast to SUMO1, are predominantly involved in the cellular response to certain stresses, including heat shock. Substantial evidence from studies in yeast has shown that SUMOylation plays an important role in the regulation of DNA replication and repair. Here, we report a proteomic analysis of proteins modified by SUMO2 in response to DNA replication stress in S phase in human cells. We have identified a panel of 22 SUMO2 targets with increased SUMOylation during DNA replication stress, many of which play key functions within the DNA replication machinery and/or in the cellular response to DNA damage. Interestingly, POLD3 was found modified most significantly in response to a low dose aphidicolin treatment protocol that promotes common fragile site (CFS) breakage. POLD3 is the human ortholog of POL32 in budding yeast, and has been shown to act during break-induced recombinational repair. We have also shown that deficiency of POLD3 leads to an increase in RPA-bound ssDNA when cells are under replication stress, suggesting that POLD3 plays a role in the cellular response to DNA replication stress. Considering that DNA replication stress is a source of genome instability, and that excessive replication stress is a hallmark of pre-neoplastic and tumor cells, our characterization of SUMO2 targets during a perturbed S-phase should provide a valuable resource for future functional studies in the fields of DNA metabolism and cancer biology.


Assuntos
DNA Polimerase III/metabolismo , Reparo do DNA , Replicação do DNA , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação , Células Cultivadas , Sítios Frágeis do Cromossomo , DNA/biossíntese , DNA/metabolismo , DNA Polimerase III/genética , DNA de Cadeia Simples/metabolismo , Humanos , Inibidores da Síntese de Ácido Nucleico/farmacologia , Proteômica , Proteína de Replicação A/metabolismo , Fase S , Estresse Fisiológico/genética
16.
Hum Mol Genet ; 23(23): 6163-76, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-24986922

RESUMO

Genome instability, epigenetic remodelling and structural chromosomal rearrangements are hallmarks of cancer. However, the coordinated epigenetic effects of constitutional chromosomal rearrangements that disrupt genes associated with congenital neurodevelopmental diseases are poorly understood. To understand the genetic-epigenetic interplay at breakpoints of chromosomal translocations disrupting CG-rich loci, we quantified epigenetic modifications at DLGAP4 (SAPAP4), a key post-synaptic density 95 (PSD95) associated gene, truncated by the chromosome translocation t(8;20)(p12;q11.23), co-segregating with cerebellar ataxia in a five-generation family. We report significant epigenetic remodelling of the DLGAP4 locus triggered by the t(8;20)(p12;q11.23) translocation and leading to dysregulation of DLGAP4 expression in affected carriers. Disruption of DLGAP4 results in monoallelic hypermethylation of the truncated DLGAP4 promoter CpG island. This induced hypermethylation is maintained in somatic cells of carriers across several generations in a t(8;20) dependent-manner however, is erased in the germ cells of the translocation carriers. Subsequently, chromatin remodelling of the locus-perturbed monoallelic expression of DLGAP4 mRNAs and non-coding RNAs in haploid cells having the translocation. Our results provide new mechanistic insight into the way a balanced chromosomal rearrangement associated with a neurodevelopmental disorder perturbs allele-specific epigenetic mechanisms at breakpoints leading to the deregulation of the truncated locus.


Assuntos
Ataxia Cerebelar/genética , Montagem e Desmontagem da Cromatina , Epigênese Genética , Proteínas do Tecido Nervoso/genética , Cromossomos Humanos Par 8/genética , Ilhas de CpG , Metilação de DNA , Feminino , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Proteínas do Tecido Nervoso/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Proteínas Associadas SAP90-PSD95 , Translocação Genética
17.
Trends Cell Biol ; 24(5): 321-7, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24361091

RESUMO

Fragile sites are conserved loci predisposed to form breaks in metaphase chromosomes. The inherent instability of these loci is associated with chromosomal rearrangements in cancers and is a feature of cells from patients with chromosomal instability syndromes. One class of fragile sites, the common fragile sites (CFSs), have previously been shown to recruit several DNA repair proteins after the completion of bulk DNA synthesis in the cell, probably indicative of their inability to complete timely DNA replication. CFS loci are also prone to trigger mitotic non-disjunction of sister chromatids, leading to the formation of ultra-fine anaphase bridges (UFBs) and micronuclei. We discuss recent developments in the CFS field; in particular, the role of DNA structure-specific endonucleases in promoting cleavage at CFSs.


Assuntos
Sítios Frágeis do Cromossomo , Proteínas de Ligação a DNA/fisiologia , Endodesoxirribonucleases/fisiologia , Endonucleases/fisiologia , Animais , Reparo do DNA , Distúrbios no Reparo do DNA/genética , Replicação do DNA , Expressão Gênica , Instabilidade Genômica , Humanos
18.
Nat Cell Biol ; 15(8): 1001-7, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23811685

RESUMO

Fragile sites are chromosomal loci with a propensity to form gaps or breaks during early mitosis, and their instability is implicated as being causative in certain neurological disorders and cancers. Recent work has demonstrated that the so-called common fragile sites (CFSs) often impair the faithful disjunction of sister chromatids in mitosis. However, the mechanisms by which CFSs express their fragility, and the cellular factors required to suppress CFS instability, remain largely undefined. Here, we report that the DNA structure-specific nuclease MUS81-EME1 localizes to CFS loci in early mitotic cells, and promotes the cytological appearance of characteristic gaps or breaks observed at CFSs in metaphase chromosomes. These data indicate that CFS breakage is an active, MUS81-EME1-dependent process, and not a result of inadvertent chromatid rupturing during chromosome condensation. Moreover, CFS cleavage by MUS81-EME1 promotes faithful sister chromatid disjunction. Our findings challenge the prevailing view that CFS breakage is a nonspecific process that is detrimental to cells, and indicate that CFS cleavage actually promotes genome stability.


Assuntos
Sítios Frágeis do Cromossomo/genética , Proteínas de Ligação a DNA/metabolismo , Endonucleases/metabolismo , Regulação da Expressão Gênica , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Segregação de Cromossomos , Endodesoxirribonucleases/metabolismo , Imunofluorescência , Instabilidade Genômica , Humanos , Reação em Cadeia da Polimerase
19.
Nucleic Acids Res ; 40(14): 6660-72, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22510267

RESUMO

The finished human genome-assemblies comprise several hundred un-sequenced euchromatic gaps, which may be rich in long polypurine/polypyrimidine stretches. Human chromosome 20 (chr 20) currently has three unfinished gaps remaining on its q-arm. All three gaps are within gene-dense regions and/or overlap disease-associated loci, including the DLGAP4 locus. In this study, we sequenced ∼ 99% of all three unfinished gaps on human chr 20, determined their complete genomic sizes and assessed epigenetic profiles using a combination of Sanger sequencing, mate pair paired-end high-throughput sequencing and chromatin, methylation and expression analyses. We found histone 3 trimethylated at Lysine 27 to be distributed across all three gaps in immortalized B-lymphocytes. In one gap, five novel CpG islands were predominantly hypermethylated in genomic DNA from peripheral blood lymphocytes and human cerebellum. One of these CpG islands was differentially methylated and paternally hypermethylated. We found all chr 20 gaps to comprise structured non-coding RNAs (ncRNAs) and to be conserved in primates. We verified expression for 13 candidate ncRNAs, some of which showed tissue specificity. Four ncRNAs expressed within the gap at DLGAP4 show elevated expression in the human brain. Our data suggest that unfinished human genome gaps are likely to comprise numerous functional elements.


Assuntos
Cromossomos Humanos Par 20/química , Cromossomos Humanos Par 20/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Sequência Conservada , Ilhas de CpG , Metilação de DNA , Histonas/metabolismo , Humanos , Camundongos , Dados de Sequência Molecular , RNA não Traduzido/genética , Análise de Sequência de DNA
20.
Epigenetics ; 7(4): 326-34, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22419065

RESUMO

Most pathogenic mitochondrial DNA (mtDNA) mutations induce defects in mitochondrial oxidative phosphorylation (OXPHOS). However, phenotypic effects of these mutations show a large degree of variation depending on the tissue affected. These differences are difficult to reconcile with OXPHOS as the sole pathogenic factor suggesting that additional mechanisms contribute to lack of genotype and clinical phenotype correlationship. An increasing number of studies have identified a possible effect on the epigenetic landscape of the nuclear genome as a consequence of mitochondrial dysfunction. In particular, these studies demonstrate reversible or irreversible changes in genomic DNA methylation profiles of the nuclear genome. Here we review how mitochondria damage checkpoint (mitocheckpoint) induces epigenetic changes in the nucleus. Persistent pathogenic mutations in mtDNA may also lead to epigenetic changes causing genomic instability in the nuclear genome. We propose that "mitocheckpoint" mediated epigenetic and genetic changes may play key roles in phenotypic variation related to mitochondrial diseases or host of human diseases in which mitochondrial defect plays a primary role.


Assuntos
DNA Mitocondrial/metabolismo , Epigênese Genética , Mitocôndrias/genética , Doenças Mitocondriais/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Montagem e Desmontagem da Cromatina , Metilação de DNA , DNA Mitocondrial/genética , Predisposição Genética para Doença , Genoma Mitocondrial , Instabilidade Genômica , Humanos , Mitocôndrias/metabolismo , Doenças Mitocondriais/metabolismo , Mutação , Neoplasias/genética , Neoplasias/metabolismo , Fosforilação Oxidativa , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...