Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Blood Cancer J ; 14(1): 16, 2024 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-38253636

RESUMO

Plk1-interacting checkpoint helicase (PICH) is a DNA translocase involved in resolving ultrafine anaphase DNA bridges and, therefore, is important to safeguard chromosome segregation and stability. PICH is overexpressed in various human cancers, particularly in lymphomas such as Burkitt lymphoma, which is caused by MYC translocations. To investigate the relevance of PICH in cancer development and progression, we have combined novel PICH-deficient mouse models with the Eµ-Myc transgenic mouse model, which recapitulates B-cell lymphoma development. We have observed that PICH deficiency delays the onset of MYC-induced lymphomas in Pich heterozygous females. Moreover, using a Pich conditional knockout mouse model, we have found that Pich deletion in adult mice improves the survival of Eµ-Myc transgenic mice. Notably, we show that Pich deletion in healthy adult mice is well tolerated, supporting PICH as a suitable target for anticancer therapies. Finally, we have corroborated these findings in two human Burkitt lymphoma cell lines and we have found that the death of cancer cells was accompanied by chromosomal instability. Based on these findings, we propose PICH as a potential therapeutic target for Burkitt lymphoma and for other cancers where PICH is overexpressed.


Assuntos
Linfoma de Burkitt , Adulto , Feminino , Animais , Humanos , Camundongos , Linfoma de Burkitt/genética , Linhagem Celular , Instabilidade Cromossômica , Modelos Animais de Doenças , Camundongos Knockout , Camundongos Transgênicos , DNA
2.
Cell Cycle ; 23(1): 92-113, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38234243

RESUMO

The Fragile Histidine Triad Diadenosine Triphosphatase (FHIT) gene is located in the Common Fragile Site FRA3B and encodes an enzyme that hydrolyzes the dinucleotide Ap3A. Although FHIT loss is one of the most frequent copy number alterations in cancer, its relevance for cancer initiation and progression remains unclear. FHIT is frequently lost in cancers from the digestive tract, which is compatible with being a cancer driver event in these tissues. However, FHIT loss could also be a passenger event due to the inherent fragility of the FRA3B locus. Moreover, the physiological relevance of FHIT enzymatic activity and the levels of Ap3A is largely unclear. We have conducted here a systematic pan-cancer analysis of FHIT status in connection with other mutations and phenotypic alterations, and we have critically discussed our findings in connection with the literature to provide an overall view of FHIT implications in cancer.


Assuntos
Hidrolases Anidrido Ácido , Proteínas de Neoplasias , Neoplasias , Humanos , Hidrolases Anidrido Ácido/genética , Sítios Frágeis do Cromossomo , Proteínas de Neoplasias/genética , Neoplasias/genética
3.
Trends Genet ; 39(6): 505-519, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36894374

RESUMO

ATRX (alpha-thalassemia mental retardation X-linked) is one of the most frequently mutated tumor suppressor genes in human cancers, especially in glioma, and recent findings indicate roles for ATRX in key molecular pathways, such as the regulation of chromatin state, gene expression, and DNA damage repair, placing ATRX as a central player in the maintenance of genome stability and function. This has led to new perspectives about the functional role of ATRX and its relationship with cancer. Here, we provide an overview of ATRX interactions and molecular functions and discuss the consequences of its impairment, including alternative lengthening of telomeres and therapeutic vulnerabilities that may be exploited in cancer cells.


Assuntos
Cromatina , Glioma , Humanos , Cromatina/genética , DNA Helicases/genética , Proteína Nuclear Ligada ao X/genética , Homeostase do Telômero/genética , Glioma/genética , Telômero
4.
Cancers (Basel) ; 14(7)2022 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-35406561

RESUMO

High-grade glioma, including anaplastic astrocytoma and glioblastoma (GBM) patients, have a poor prognosis due to the lack of effective treatments. Therefore, the development of new therapeutic strategies to treat these gliomas is urgently required. Given that high-grade gliomas frequently harbor mutations in the SNF2 family chromatin remodeler ATRX, we performed a screen to identify FDA-approved drugs that are toxic to ATRX-deficient cells. Our findings reveal that multi-targeted receptor tyrosine kinase (RTK) and platelet-derived growth factor receptor (PDGFR) inhibitors cause higher cellular toxicity in high-grade glioma ATRX-deficient cells. Furthermore, we demonstrate that a combinatorial treatment of RTKi with temozolomide (TMZ)-the current standard of care treatment for GBM patients-causes pronounced toxicity in ATRX-deficient high-grade glioma cells. Our findings suggest that combinatorial treatments with TMZ and RTKi may increase the therapeutic window of opportunity in patients who suffer high-grade gliomas with ATRX mutations. Thus, we recommend incorporating the ATRX status into the analyses of clinical trials with RTKi and PDGFRi.

5.
Aging (Albany NY) ; 12(7): 5612-5624, 2020 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-32253367

RESUMO

Replication Stress (RS) is a type of DNA damage generated at the replication fork, characterized by single-stranded DNA (ssDNA) accumulation, and which can be caused by a variety of factors. Previous studies have reported elevated RS levels in aged cells. In addition, mouse models with a deficient RS response show accelerated aging. However, the relevance of endogenous or physiological RS, compared to other sources of genomic instability, for the normal onset of aging is unknown. We have performed long term survival studies of transgenic mice with extra copies of the Chk1 and/or Rrm2 genes, which we previously showed extend the lifespan of a progeroid ATR-hypomorphic model suffering from high levels of RS. In contrast to their effect in the context of progeria, the lifespan of Chk1, Rrm2 and Chk1/Rrm2 transgenic mice was similar to WT littermates in physiological settings. Most mice studied died due to tumors -mainly lymphomas- irrespective of their genetic background. Interestingly, a higher but not statistically significant percentage of transgenic mice developed tumors compared to WT mice. Our results indicate that supraphysiological protection from RS does not extend lifespan, indicating that RS may not be a relevant source of genomic instability on the onset of normal aging.


Assuntos
Quinase 1 do Ponto de Checagem/genética , Dano ao DNA , Longevidade/genética , Ribonucleosídeo Difosfato Redutase/genética , Animais , Quinase 1 do Ponto de Checagem/metabolismo , Replicação do DNA , Camundongos , Camundongos Transgênicos , Ribonucleosídeo Difosfato Redutase/metabolismo
6.
Nucleic Acids Res ; 47(15): 8004-8018, 2019 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-31180492

RESUMO

Common fragile sites (CFSs) are conserved genomic regions prone to break under conditions of replication stress (RS). Thus, CFSs are hotspots for rearrangements in cancer and contribute to its chromosomal instability. Here, we have performed a global analysis of proteins that recruit to CFSs upon mild RS to identify novel players in CFS stability. To this end, we performed Chromatin Immunoprecipitation (ChIP) of FANCD2, a protein that localizes specifically to CFSs in G2/M, coupled to mass spectrometry to acquire a CFS interactome. Our strategy was validated by the enrichment of many known regulators of CFS maintenance, including Fanconi Anemia, DNA repair and replication proteins. Among the proteins identified with unknown functions at CFSs was the chromatin remodeler ATRX. Here we demonstrate that ATRX forms foci at a fraction of CFSs upon RS, and that ATRX depletion increases the occurrence of chromosomal breaks, a phenotype further exacerbated under mild RS conditions. Accordingly, ATRX depletion increases the number of 53BP1 bodies and micronuclei, overall indicating that ATRX is required for CFS stability. Overall, our study provides the first proteomic characterization of CFSs as a valuable resource for the identification of novel regulators of CFS stability.


Assuntos
Sítios Frágeis do Cromossomo , Instabilidade Genômica , Proteoma/metabolismo , Proteômica/métodos , Proteína Nuclear Ligada ao X/metabolismo , Quebra Cromossômica , Reparo do DNA , Replicação do DNA/genética , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Células HeLa , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Proteoma/genética , Interferência de RNA , Espectrometria de Massas em Tandem , Proteína Nuclear Ligada ao X/genética
7.
PLoS One ; 13(12): e0209202, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30566531

RESUMO

The specific role of polyamines in the testis physiology is not fully understood. Antizymes (OAZs) and antizyme inhibitors (AZINs) are modulators of ornithine decarboxylase (ODC), a key enzyme in polyamine biosynthesis and polyamine uptake. Although the three known OAZs are expressed in the testis, only OAZ3 is testis specific and has been proven to have an essential role in male fertility. Regarding the two existing AZINs, AZIN2 is the most abundantly expressed member in this gonad. Whereas previous studies suggested that AZIN2 might participate in mouse spermatogenesis, immunohistological analysis of human testicular sections revealed that AZIN2 is also detected in the steroidogenic Leydig cells but not in the germinal epithelium. In the present study, we found a close ontogenic similarity in the mRNA levels of OAZs and AZINs between mice and rats, but an opposite expression pattern of ODC activity. Further analysis of AZIN2 and OAZ3 in the testis of mice with different alterations in spermatogenesis and fertility, induced either genetically or pharmacologically, corroborated that both AZIN2 and OAZ3 are mainly expressed in the haploid germinal cells. Finally, by using transgenic mice with a truncated Azin2 gene fused to the bacterial lacZ gene, we studied the expression of Azin2 in testes, epididymides and spermatozoa. AZIN2 was detected in spermatids and spermatozoa, as well as in Leydig cells, and in epithelial epidydimal cells. Azin2 knock-out male mice were fertile; however, they showed marked decreases in testicular putrescine and plasma and testicular testosterone levels, and a dramatic reduction in the sperm motility. These results suggest an important role for AZIN2 in testicular cells by modulating polyamine concentrations, testosterone synthesis and sperm function. Overall, our data corroborate the relevance of polyamine regulation in testis functions, where both AZIN2 and OAZ3 play fundamental roles.


Assuntos
Proteínas de Transporte/metabolismo , Poliaminas/metabolismo , Motilidade dos Espermatozoides/fisiologia , Testículo/enzimologia , Testosterona/metabolismo , Animais , Gonadotropina Coriônica/administração & dosagem , Gonadotropina Coriônica/metabolismo , Epididimo/enzimologia , Epididimo/crescimento & desenvolvimento , Células Epiteliais/enzimologia , Expressão Gênica , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Embrionárias de Células Germinativas/metabolismo , Ratos Sprague-Dawley , Espermatozoides/metabolismo , Neoplasias Testiculares/metabolismo , Testículo/crescimento & desenvolvimento
8.
Cell Rep ; 24(12): 3274-3284, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30232008

RESUMO

PICH is a DNA translocase necessary for the resolution of ultrafine anaphase DNA bridges and to ensure the fidelity of chromosomal segregation. Here, we report the generation of an animal model deficient for PICH that allowed us to investigate its physiological relevance. Pich KO mice lose viability during embryonic development due to a global accumulation of DNA damage. However, despite the presence of chromosomal instability, extensive p53 activation, and increased apoptosis throughout the embryo, Pich KO embryos survive until day 12.5 of embryonic development. The absence of p53 failed to improve the viability of the Pich KO embryos, suggesting that the observed developmental defects are not solely due to p53-induced apoptosis. Moreover, Pich-deficient mouse embryonic fibroblasts exhibit chromosomal instability and are resistant to RASV12/E1A-induced transformation. Overall, our data indicate that PICH is essential to preserve chromosomal integrity in rapidly proliferating cells and is therefore critical during embryonic development and tumorigenesis.


Assuntos
Instabilidade Cromossômica , Desenvolvimento Embrionário/genética , Animais , Apoptose , Células Cultivadas , Dano ao DNA , DNA Helicases/metabolismo , Camundongos , Proteína Supressora de Tumor p53/metabolismo
10.
Transgenic Res ; 26(3): 429-434, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28105543

RESUMO

The generation of genetically engineered mouse models (GEMMs), including knock-out (KO) and knock-in (KI) models, often requires genomic screening of many mouse ES cell (mESC) clones by Southern blot. The use of large targeting constructs facilitates the recombination of exogenous DNA in a specific genomic locus, but limits the detection of its correct genomic integration by standard PCR methods. Genomic Long Range PCR (LR-PCR), using primers adjacent to the homology arms, has been used as an alternative to radioactive-based Southern blot screenings. However, LR-PCRs are often difficult and render many false positive and false negative results. Here, we propose an alternative screening method based on the detection of a genetic modification at the mRNA level, which we successfully optimized in two mouse models. This screening method consists of a reverse-transcription PCR (RT-PCR) using primers that match exons flanking the targeting construct. The detection of the expected modification in this PCR product confirms the integration at the correct genomic location and shows that the mutant mRNA is expressed. This is a simple and sensitive strategy to screen locus-specific recombination of targeting constructs which can also be useful to screen KO and KI mutant mice or cell lines including those generated by CRISPR/Cas9.


Assuntos
Células-Tronco Embrionárias/fisiologia , Recombinação Genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Animais , Southern Blotting , Genes BRCA1 , Camundongos Transgênicos
11.
Oncotarget ; 7(37): 58759-58767, 2016 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-27577084

RESUMO

Ewing sarcomas (ES) are pediatric bone tumors that arise from a driver translocation, most frequently EWS/FLI1. Current ES treatment involves DNA damaging agents, yet the basis for the sensitivity to these therapies remains unknown. Oncogene-induced replication stress (RS) is a known source of endogenous DNA damage in cancer, which is suppressed by ATR and CHK1 kinases. We here show that ES suffer from high endogenous levels of RS, rendering them particularly dependent on the ATR pathway. Accordingly, two independent ATR inhibitors show in vitro toxicity in ES cell lines as well as in vivo efficacy in ES xenografts as single agents. Expression of EWS/FLI1 or EWS/ERG oncogenic translocations sensitizes non-ES cells to ATR inhibitors. Our data shed light onto the sensitivity of ES to genotoxic agents, and identify ATR inhibitors as a potential therapy for Ewing Sarcomas.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Ósseas/metabolismo , Sarcoma de Ewing/metabolismo , Animais , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Proteínas Mutadas de Ataxia Telangiectasia/genética , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/genética , Linhagem Celular Tumoral , Dano ao DNA , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos SCID , RNA Interferente Pequeno/genética , Proteína EWS de Ligação a RNA/genética , Sarcoma de Ewing/tratamento farmacológico , Sarcoma de Ewing/genética , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Nat Struct Mol Biol ; 23(4): 270-7, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26950370

RESUMO

Post-translational modification of proteins by ubiquitin (Ub) and Ub-like modifiers regulates DNA replication. We have previously shown that chromatin around replisomes is rich in SUMO and poor in Ub, whereas mature chromatin exhibits an opposite pattern. How this SUMO-rich, Ub-poor environment is maintained at sites of DNA replication in mammalian cells remains unexplored. Here we identify USP7 as a replisome-enriched SUMO deubiquitinase that is essential for DNA replication. By acting on SUMO and SUMOylated proteins, USP7 counteracts their ubiquitination. Inhibition or genetic deletion of USP7 leads to the accumulation of Ub on SUMOylated proteins, which are displaced away from replisomes. Our findings provide a model explaining the differential accumulation of SUMO and Ub at replication forks and identify an essential role of USP7 in DNA replication that should be considered in the development of USP7 inhibitors as anticancer agents.


Assuntos
Replicação do DNA , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Ubiquitina Tiolesterase/metabolismo , Proteases Específicas de Ubiquitina/metabolismo , Dano ao DNA , Reparo do DNA , Células HCT116 , Células HeLa , Humanos , Modelos Moleculares , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/análise , Sumoilação , Ubiquitina Tiolesterase/análise , Peptidase 7 Específica de Ubiquitina , Proteases Específicas de Ubiquitina/análise , Ubiquitinação
13.
Cell ; 167(5): 1264-1280.e18, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-28084216

RESUMO

Granulomas are immune cell aggregates formed in response to persistent inflammatory stimuli. Granuloma macrophage subsets are diverse and carry varying copy numbers of their genomic information. The molecular programs that control the differentiation of such macrophage populations in response to a chronic stimulus, though critical for disease outcome, have not been defined. Here, we delineate a macrophage differentiation pathway by which a persistent Toll-like receptor (TLR) 2 signal instructs polyploid macrophage fate by inducing replication stress and activating the DNA damage response. Polyploid granuloma-resident macrophages formed via modified cell divisions and mitotic defects and not, as previously thought, by cell-to-cell fusion. TLR2 signaling promoted macrophage polyploidy and suppressed genomic instability by regulating Myc and ATR. We propose that, in the presence of persistent inflammatory stimuli, pathways previously linked to oncogene-initiated carcinogenesis instruct a long-lived granuloma-resident macrophage differentiation program that regulates granulomatous tissue remodeling.


Assuntos
Dano ao DNA , Granuloma/imunologia , Macrófagos/imunologia , Mycobacterium tuberculosis/imunologia , Animais , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Diferenciação Celular , Proliferação de Células , Humanos , Inflamação/imunologia , Lipoproteínas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Mitose , Proteínas Proto-Oncogênicas c-myc/metabolismo , Receptor 2 Toll-Like
14.
Nat Commun ; 6: 8548, 2015 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-26456157

RESUMO

Replicative stress during embryonic development influences ageing and predisposition to disease in adults. A protective mechanism against replicative stress is provided by the licensing of thousands of origins in G1 that are not necessarily activated in the subsequent S-phase. These 'dormant' origins provide a backup in the presence of stalled forks and may confer flexibility to the replication program in specific cell types during differentiation, a role that has remained unexplored. Here we show, using a mouse strain with hypomorphic expression of the origin licensing factor mini-chromosome maintenance (MCM)3 that limiting origin licensing in vivo affects the functionality of hematopoietic stem cells and the differentiation of rapidly-dividing erythrocyte precursors. Mcm3-deficient erythroblasts display aberrant DNA replication patterns and fail to complete maturation, causing lethal anemia. Our results indicate that hematopoietic progenitors are particularly sensitive to replication stress, and full origin licensing ensures their correct differentiation and functionality.


Assuntos
Replicação do DNA , Eritropoese , Células-Tronco Hematopoéticas/fisiologia , Componente 3 do Complexo de Manutenção de Minicromossomo/metabolismo , Animais , Quinase 1 do Ponto de Checagem , Dano ao DNA , Suscetibilidade a Doenças , Embrião de Mamíferos/fisiologia , Desenvolvimento Embrionário , Feminino , Genes Letais , Neoplasias Hematológicas , Fígado/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Componente 3 do Complexo de Manutenção de Minicromossomo/genética , Proteínas Quinases/metabolismo
15.
Nat Commun ; 6: 8036, 2015 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-26292731

RESUMO

The generation of induced pluripotent stem cells (iPSC) from adult somatic cells is one of the most remarkable discoveries in recent decades. However, several works have reported evidence of genomic instability in iPSC, raising concerns on their biomedical use. The reasons behind the genomic instability observed in iPSC remain mostly unknown. Here we show that, similar to the phenomenon of oncogene-induced replication stress, the expression of reprogramming factors induces replication stress. Increasing the levels of the checkpoint kinase 1 (CHK1) reduces reprogramming-induced replication stress and increases the efficiency of iPSC generation. Similarly, nucleoside supplementation during reprogramming reduces the load of DNA damage and genomic rearrangements on iPSC. Our data reveal that lowering replication stress during reprogramming, genetically or chemically, provides a simple strategy to reduce genomic instability on mouse and human iPSC.


Assuntos
Proliferação de Células/fisiologia , Reprogramação Celular/fisiologia , Instabilidade Genômica/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Estresse Fisiológico/fisiologia , Animais , Linhagem Celular , Quinase 1 do Ponto de Checagem , DNA/genética , Fibroblastos/fisiologia , Regulação da Expressão Gênica/fisiologia , Humanos , Camundongos , Camundongos Transgênicos , Hibridização de Ácido Nucleico , Plasmídeos , Mutação Puntual , Proteínas Quinases/genética , Proteínas Quinases/metabolismo
16.
Mol Cell Biol ; 35(17): 2910-7, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26077802

RESUMO

The ribonucleotide reductase (RNR) complex, composed of a catalytic subunit (RRM1) and a regulatory subunit (RRM2), is thought to be a rate-limiting enzymatic complex for the production of nucleotides. In humans, the Rrm1 gene lies at 11p15.5, a tumor suppressor region, and RRM1 expression in cancer has been shown to predict responses to chemotherapy. Nevertheless, whether RRM1 is essential in mammalian cells and what the effects of its haploinsufficiency are remain unknown. To model RNR function in mice we used a mutation previously described in Saccharomyces cerevisiae (Rnr1-W688G) which, despite being viable, leads to increased interaction of the RNR complex with its allosteric inhibitor Sml1. In contrast to yeast, homozygous mutant mice carrying the Rrm1 mutation (Rrm1(WG/WG)) are not viable, even at the earliest embryonic stages. Proteomic analyses failed to identify proteins that specifically bind to the mutant RRM1 but revealed that, in mammals, the mutation prevents RRM1 binding to RRM2. Despite the impact of the mutation, Rrm1(WG/+) mice and cells presented no obvious phenotype, suggesting that the RRM1 protein exists in excess. Our work reveals that binding of RRM1 to RRM2 is essential for mammalian cells and provides the first loss-of-function model of the RNR complex for genetic studies.


Assuntos
Desenvolvimento Embrionário/genética , Ribonucleosídeo Difosfato Redutase/metabolismo , Ribonucleotídeo Redutases/metabolismo , Animais , Linhagem Celular , Células-Tronco Embrionárias/citologia , Técnicas de Introdução de Genes , Células HEK293 , Histonas/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mucinas/metabolismo , Proteínas Musculares/metabolismo , Mutação/genética , Peptídeos/metabolismo , Ligação Proteica/genética , Ribonucleosídeo Difosfato Redutase/antagonistas & inibidores , Ribonucleosídeo Difosfato Redutase/genética , Ribonucleotídeo Redutases/antagonistas & inibidores , Ribonucleotídeo Redutases/genética , Fator Trefoil-2
17.
Methods Mol Biol ; 1267: 413-37, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25636482

RESUMO

Damaged DNA has a profound impact on mammalian health and overall survival. In addition to being the source of mutations that initiate cancer, the accumulation of toxic amounts of DNA damage can cause severe developmental diseases and accelerate aging. Therefore, understanding how cells respond to DNA damage has become one of the most intense areas of biomedical research in the recent years. However, whereas most mechanistic studies derive from in vitro or in cellulo work, the impact of a given mutation on a living organism is largely unpredictable. For instance, why BRCA1 mutations preferentially lead to breast cancer whereas mutations compromising mismatch repair drive colon cancer is still not understood. In this context, evaluating the specific physiological impact of mutations that compromise genome integrity has become crucial for a better dimensioning of our knowledge. We here describe the various technologies that can be used for modeling mutations in mice and provide a review of the genes and pathways that have been modeled so far in the context of DNA damage responses.


Assuntos
Dano ao DNA , Modelos Animais , Animais , Marcação de Genes , Técnicas de Transferência de Genes , Instabilidade Genômica , Humanos , Camundongos , Fenótipo
18.
Cell Cycle ; 13(12): 1980-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24762558

RESUMO

Stable Fos expression in the osteoblast lineage results in the development of osteosarcomas (OS) in mice, yet the underlying mechanisms are poorly understood. Using a genetic system in which Fos expression can be induced in osteoblasts in a doxycycline-dependent manner and through subsequent RNA sequencing and gene set enrichment analysis, we were able to identify novel transcriptional targets of Fos in osteoblasts. These included a distinct activation of cellular response toward replication stress (RS), exemplified by a Fos-dependent induction of the RS-suppressing Chk1 kinase. Importantly, Fos expression protects osteoblasts from RS and DNA damage likely through upregulation of Chk1 and facilitates transformation by Ras/E1A oncogenes. These data reveal a novel function of Fos in safeguarding genome stability during replication, which is particularly relevant in conditions of oncogene-induced S-phase entry.


Assuntos
Replicação do DNA/fisiologia , Proteínas Oncogênicas v-fos/metabolismo , Osteoblastos/metabolismo , Proteínas Quinases/metabolismo , Animais , Células Cultivadas , Quinase 1 do Ponto de Checagem , Dano ao DNA/fisiologia , Técnicas de Inativação de Genes , Camundongos , Proteínas Oncogênicas v-fos/genética , Proteína Supressora de Tumor p53/metabolismo
19.
Cell Rep ; 6(4): 670-83, 2014 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-24508461

RESUMO

The Anaphase-promoting complex/cyclosome (APC/C) cofactor Cdh1 modulates cell proliferation by targeting multiple cell-cycle regulators for ubiquitin-dependent degradation. Lack of Cdh1 results in structural and numerical chromosome aberrations, a hallmark of genomic instability. By using a proteomic approach in Cdh1-null cells and mouse tissues, we have identified kinesin Eg5 and topoisomerase 2α as Cdh1 targets involved in the maintenance of genomic stability. These proteins are ubiquitinated and degraded through specific KEN and D boxes in a Cdh1-dependent manner. Whereas Cdh1-null cells display partial resistance to Eg5 inhibitors such as monastrol, lack of Cdh1 results in a dramatic sensitivity to Top2α poisons as a consequence of increased levels of trapped Top2α-DNA complexes. Chemical inhibition of the APC/C in cancer cells results in increased sensitivity to Top2α poisons. This work identifies in vivo targets of the mammalian APC/C-Cdh1 complex and reveals synthetic lethal interactions of relevance in anticancer treatments.


Assuntos
Proteínas Cdh1/metabolismo , Proteoma/metabolismo , Pirimidinas/farmacologia , Tionas/farmacologia , Inibidores da Topoisomerase II/farmacologia , Animais , Antígenos de Neoplasias/química , Antígenos de Neoplasias/metabolismo , Sítios de Ligação , Proteínas Cdh1/genética , DNA Topoisomerases Tipo II/química , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Instabilidade Genômica , Células HEK293 , Células HeLa , Humanos , Cinesinas/química , Cinesinas/metabolismo , Camundongos , Ligação Proteica , Ubiquitinação , Xenopus
20.
Cell Cycle ; 12(12): 1948-54, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23676215

RESUMO

Replication stress (RS) is a source of DNA damage that has been linked to cancer and aging, which is suppressed by the ATR kinase. In mice, reduced ATR levels in a model of the ATR-Seckel syndrome lead to RS and accelerated aging. Similarly, ATR-Seckel embryonic fibroblasts (MEF) accumulate RS and undergo cellular senescence. We previously showed that senescence of ATR-Seckel MEF cannot be rescued by p53-deletion. Here, we show that the genetic ablation of the INK4a/Arf locus fully rescues senescence on ATR mutant MEF, but also that induced by other conditions that generate RS such as low doses of hydroxyurea or ATR inhibitors. In addition, we show that a persistent exposure to RS leads to increased levels of INK4a/Arf products, revealing that INK4a/ARF behaves as a bona fide RS checkpoint. Our data reveal an unknown role for INK4a/ARF in limiting the expansion of cells suffering from persistent replication stress, linking this well-known tumor suppressor to the maintenance of genomic integrity.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Animais , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/genética , Dano ao DNA/genética , Dano ao DNA/fisiologia , Replicação do DNA/genética , Replicação do DNA/fisiologia , Immunoblotting , Imuno-Histoquímica , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA