Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 49(17): 9886-9905, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34469544

RESUMO

Telomere maintenance is essential to preserve genomic stability and involves telomere-specific proteins, DNA replication and repair proteins. Lamins are key components of the nuclear envelope and play numerous roles, including maintenance of the nuclear integrity, regulation of transcription, and DNA replication. Elevated levels of lamin B1, one of the major lamins, have been observed in some human pathologies and several cancers. Yet, the effect of lamin B1 dysregulation on telomere maintenance remains unknown. Here, we unveil that lamin B1 overexpression drives telomere instability through the disruption of the shelterin complex. Indeed, lamin B1 dysregulation leads to an increase in telomere dysfunction-induced foci, telomeric fusions and telomere losses in human cells. Telomere aberrations were preceded by mislocalizations of TRF2 and its binding partner RAP1. Interestingly, we identified new interactions between lamin B1 and these shelterin proteins, which are strongly enhanced at the nuclear periphery upon lamin B1 overexpression. Importantly, chromosomal fusions induced by lamin B1 in excess were rescued by TRF2 overexpression. These data indicated that lamin B1 overexpression triggers telomere instability through a mislocalization of TRF2. Altogether our results point to lamin B1 as a new interacting partner of TRF2, that is involved in telomere stability.


Assuntos
Lamina Tipo B/metabolismo , Complexo Shelterina/metabolismo , Telômero/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Células Cultivadas , Humanos , Lamina Tipo B/química , Proteínas de Ligação a Telômeros/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/química
2.
Chem Res Toxicol ; 30(8): 1629-1640, 2017 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-28657713

RESUMO

Telomeres protect the ends of chromosomes against illegitimate recombination and repair. They can be targets for G-quadruplex ligands and platinum complexes due to their repeated G-rich sequences. Protection of telomeres is ensured by a complex of six proteins, including TRF2, which inhibits the DNA damage response pathway. We analyzed telomere modifications induced in cancer cells by the experimental hybrid platinum complex, Pt-MPQ, comprising both an ethylene diamine monofunctional platinum complex and a G-quadruplex recognition moiety (MPQ). Pt-MPQ promotes the displacement of two telomeric proteins (TRF2 and TRF1) from telomeres, as well as the formation of telomere damage and telomere sister losses, whereas the control compound MPQ does not. This suggests that the platinum moiety potentiates the targeting of the G-quadruplex ligand to telomeres, opening a new perspective for telomere biology and anticancer therapy. Interestingly, the chemotherapy drug cisplatin, which has no specific affinity for G-quadruplex structures, partially induces the TRF2 delocalization from telomeres but produces less telomeric DNA damage, suggesting that this TRF2 displacement could be independent of G-quadruplex recognition.


Assuntos
Complexos de Coordenação/toxicidade , Quadruplex G/efeitos dos fármacos , Platina/química , Telômero/efeitos dos fármacos , Acridinas/toxicidade , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Cisplatino/toxicidade , Dano ao DNA/efeitos dos fármacos , Humanos , Ligantes , Microscopia de Fluorescência , Compostos Organoplatínicos/toxicidade , Telômero/metabolismo , Encurtamento do Telômero/efeitos dos fármacos , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo
3.
Nucleic Acids Res ; 43(2): 904-16, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25539913

RESUMO

The prion protein (PrP) is highly conserved and ubiquitously expressed, suggesting that it plays an important physiological function. However, despite decades of investigation, this role remains elusive. Here, by using animal and cellular models, we unveil a key role of PrP in the DNA damage response. Exposure of neurons to a genotoxic stress activates PRNP transcription leading to an increased amount of PrP in the nucleus where it interacts with APE1, the major mammalian endonuclease essential for base excision repair, and stimulates its activity. Preventing the induction of PRNP results in accumulation of abasic sites in DNA and impairs cell survival after genotoxic treatment. Brains from Prnp(-/-) mice display a reduced APE1 activity and a defect in the repair of induced DNA damage in vivo. Thus, PrP is required to maintain genomic stability in response to genotoxic stresses.


Assuntos
Reparo do DNA , Príons/metabolismo , Animais , Encéfalo/enzimologia , Linhagem Celular , Núcleo Celular/química , Sobrevivência Celular , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Humanos , Metanossulfonato de Metila/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Mutagênicos/toxicidade , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteínas Priônicas , Príons/análise , Príons/biossíntese , Príons/genética , Ativação Transcricional
4.
Nucleic Acids Res ; 42(9): 5616-32, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24598253

RESUMO

The repair of toxic double-strand breaks (DSB) is critical for the maintenance of genome integrity. The major mechanisms that cope with DSB are: homologous recombination (HR) and classical or alternative nonhomologous end joining (C-NHEJ versus A-EJ). Because these pathways compete for the repair of DSB, the choice of the appropriate repair pathway is pivotal. Among the mechanisms that influence this choice, deoxyribonucleic acid (DNA) end resection plays a critical role by driving cells to HR, while accurate C-NHEJ is suppressed. Furthermore, end resection promotes error-prone A-EJ. Increasing evidence define Poly(ADP-ribose) polymerase 3 (PARP3, also known as ARTD3) as an important player in cellular response to DSB. In this work, we reveal a specific feature of PARP3 that together with Ku80 limits DNA end resection and thereby helps in making the choice between HR and NHEJ pathways. PARP3 interacts with and PARylates Ku70/Ku80. The depletion of PARP3 impairs the recruitment of YFP-Ku80 to laser-induced DNA damage sites and induces an imbalance between BRCA1 and 53BP1. Both events result in compromised accurate C-NHEJ and a concomitant increase in DNA end resection. Nevertheless, HR is significantly reduced upon PARP3 silencing while the enhanced end resection causes mutagenic deletions during A-EJ. As a result, the absence of PARP3 confers hypersensitivity to anti-tumoral drugs generating DSB.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Reparo do DNA por Junção de Extremidades , Poli(ADP-Ribose) Polimerases/fisiologia , Reparo de DNA por Recombinação , Antígenos Nucleares/metabolismo , Antineoplásicos/farmacologia , Proteína BRCA1/metabolismo , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais , Etoposídeo/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Autoantígeno Ku , Proteínas Nucleares/metabolismo , Processamento de Proteína Pós-Traducional , Transporte Proteico , Proteína de Replicação A/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
5.
Int J Cancer ; 136(7): 1546-58, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25175359

RESUMO

Glioblastoma multiforme is the most aggressive primary tumor of the central nervous system. Glioma stem cells (GSCs), a small population of tumor cells with stem-like properties, are supposedly responsible for glioblastoma multiforme relapse after current therapies. In approximately thirty percent of glioblastoma multiforme tumors, telomeres are not maintained by telomerase but through an alternative mechanism, termed alternative lengthening of telomere (ALT), suggesting potential interest in developing specific therapeutic strategies. However, no preclinical model of ALT glioma was available until the isolation of TG20 cells from a human ALT glioma. Herein, we show that TG20 cells exhibit a high level of telomeric recombination but a stable karyotype, indicating that their telomeres retain their protective function against chromosomal instability. TG20 cells possess all of the characteristic features of GSCs: the expression of neural stem cell markers, the generation of intracerebral tumors in NOD-SCID-IL2Rγ (NSG) mice as well as in nude mice, and the ability to sustain serial intracerebral transplantations without expressing telomerase, demonstrating the stability of the ALT phenotype in vivo. Furthermore, we also demonstrate that 360B, a G-quadruplex ligand of the pyridine derivative series that impairs telomere replication and mitotic progression in cancer cells, prevents the development of TG20 tumors. Together, our results show that intracerebral grafts of TG20 cells in immunodeficient mice constitute an efficient preclinical model of ALT glioblastoma multiforme and that G-quadruplex ligands are a potential therapy for this specific type of tumor.


Assuntos
Glioma/genética , Telômero/genética , Adulto , Animais , Linhagem Celular Tumoral , Metilação de DNA , Modelos Animais de Doenças , Quadruplex G , Regulação da Expressão Gênica , Glioma/metabolismo , Xenoenxertos , Humanos , Subunidade gama Comum de Receptores de Interleucina/genética , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Fenótipo , Troca de Cromátide Irmã , Telomerase/genética , Telomerase/metabolismo , Telômero/metabolismo , Homeostase do Telômero
6.
Stem Cells ; 32(12): 3257-65, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25098224

RESUMO

Neurogenesis decreases during aging causing a progressive cognitive decline but it is still controversial whether proliferation defects in neurogenic niches result from a loss of neural stem cells or from an impairment of their progression through the cell cycle. Using an accurate fluorescence-activated cell sorting technique, we show that the pool of neural stem cells is maintained in the subventricular zone of middle-aged mice while they have a reduced proliferative potential eventually leading to the subsequent decrease of their progeny. In addition, we demonstrate that the G1 phase is lengthened during aging specifically in activated stem cells, but not in transit-amplifying cells, and directly impacts on neurogenesis. Finally, we report that inhibition of TGFß signaling restores cell cycle progression defects in stem cells. Our data highlight the significance of cell cycle dysregulation in stem cells in the aged brain and provide an attractive foundation for the development of anti-TGFß regenerative therapies based on stimulating endogenous neural stem cells.


Assuntos
Envelhecimento/fisiologia , Encéfalo/citologia , Diferenciação Celular/fisiologia , Fase G1 , Neurogênese/fisiologia , Células-Tronco/citologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Proliferação de Células/fisiologia , Fase G1/genética , Camundongos Endogâmicos C57BL , Nicho de Células-Tronco/fisiologia
7.
Hum Mol Genet ; 21(1): 121-35, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21968513

RESUMO

Fanconi anemia (FA) is a human rare genetic disorder characterized by congenital defects, bone marrow (BM) failure and predisposition to leukemia. The progressive aplastic anemia suggests a defect in the ability of hematopoietic stem cells (HSC) to sustain hematopoieis. We have examined the role of the nuclear FA core complex gene Fancg in the functionality of HSC. In Fancg-/- mice, we observed a decay of long-term HSC and multipotent progenitors that account for the reduction in the LSK compartment containing primitive hematopoietic cells. Fancg-/- lymphoid and myeloid progenitor cells were also affected, and myeloid progenitors show compromised in vitro functionality. HSC from Fancg-/- mice failed to engraft and to reconstitute at short and long term the hematopoiesis in a competitive transplantation assay. Fancg-/- LSK cells showed a loss of quiescence, an impaired migration in vitro in response to the chemokine CXCL12 and a defective homing to the BM after transplantation. Finally, the expression of several key genes involved in self-renewal, quiescence and migration of HSC was dysregulated in Fancg-deficient LSK subset. Collectively, our data reveal that Fancg should play a role in the regulation of physiological functions of HSC.


Assuntos
Proteína do Grupo de Complementação G da Anemia de Fanconi/deficiência , Anemia de Fanconi/fisiopatologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Animais , Medula Óssea/metabolismo , Movimento Celular , Quimiocina CXCL12/metabolismo , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteína do Grupo de Complementação G da Anemia de Fanconi/genética , Feminino , Hematopoese , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
8.
Proc Natl Acad Sci U S A ; 108(7): 2783-8, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21270334

RESUMO

The ADP ribosyl transferase [poly(ADP-ribose) polymerase] ARTD3(PARP3) is a newly characterized member of the ARTD(PARP) family that catalyzes the reaction of ADP ribosylation, a key posttranslational modification of proteins involved in different signaling pathways from DNA damage to energy metabolism and organismal memory. This enzyme shares high structural similarities with the DNA repair enzymes PARP1 and PARP2 and accordingly has been found to catalyse poly(ADP ribose) synthesis. However, relatively little is known about its in vivo cellular properties. By combining biochemical studies with the generation and characterization of loss-of-function human and mouse models, we describe PARP3 as a newcomer in genome integrity and mitotic progression. We report a particular role of PARP3 in cellular response to double-strand breaks, most likely in concert with PARP1. We identify PARP3 as a critical player in the stabilization of the mitotic spindle and in telomere integrity notably by associating and regulating the mitotic components NuMA and tankyrase 1. Both functions open stimulating prospects for specifically targeting PARP3 in cancer therapy.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Quebras de DNA de Cadeia Dupla , Instabilidade Genômica/genética , Mitose/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Fuso Acromático/fisiologia , Difosfato de Adenosina/metabolismo , Animais , Antígenos Nucleares/metabolismo , Western Blotting , Linhagem Celular Tumoral , Ensaio de Unidades Formadoras de Colônias , Ensaio Cometa , Primers do DNA/genética , Técnica Indireta de Fluorescência para Anticorpo , Instabilidade Genômica/fisiologia , Humanos , Imunoprecipitação , Hibridização in Situ Fluorescente , Espectrometria de Massas , Camundongos , Camundongos Knockout , Microscopia de Vídeo , Mitose/fisiologia , Proteínas Associadas à Matriz Nuclear/metabolismo , Poli(ADP-Ribose) Polimerases/deficiência , Tanquirases/metabolismo
9.
Stem Cells ; 30(3): 537-47, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22162343

RESUMO

The cyclin-dependent kinase inhibitor p21(waf1/cip) mediates the p53-dependent G1/S checkpoint, which is generally considered to be a critical requirement to maintain genomic stability after DNA damage. We used staggered 5-ethynyl-2'deoxyuridine/5-bromo-2'-deoxyuridine double-labeling in vivo to investigate the cell cycle progression and the role of p21(waf1/cip) in the DNA damage response of neural stem and progenitor cells (NSPCs) after exposure of the developing mouse cortex to ionizing radiation. We observed a radiation-induced p21-dependent apoptotic response in migrating postmitotic cortical cells. However, neural stem and progenitor cells (NSPCs) did not initiate a p21(waf1/cip1) -dependent G1/S block and continued to enter S-phase at a similar rate to the non-irradiated controls. The G1/S checkpoint is not involved in the mechanisms underlying the faithful transmission of the NSPC genome and/or the elimination of critically damaged cells. These processes typically involve intra-S and G2/M checkpoints that are rapidly activated after irradiation. p21 is normally repressed in neural cells during brain development except at the G1 to G0 transition. Lack of activation of a G1/S checkpoint and apoptosis of postmitotic migrating cells after DNA damage appear to depend on the expression of p21 in neural cells, since substantial cell-to-cell variations are found in the irradiated cortex. This suggests that repression of p21 during brain development prevents the induction of the G1/S checkpoint after DNA damage.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/deficiência , Dano ao DNA , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos da radiação , Células-Tronco Neurais/fisiologia , Animais , Apoptose , Núcleo Celular/metabolismo , Proliferação de Células/efeitos da radiação , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Embrião de Mamíferos/efeitos da radiação , Feminino , Instabilidade Genômica/efeitos da radiação , Ventrículos Laterais/metabolismo , Ventrículos Laterais/patologia , Ventrículos Laterais/efeitos da radiação , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/efeitos da radiação , Neuroglia/fisiologia , Neuroglia/efeitos da radiação , Gravidez , Pontos de Checagem da Fase S do Ciclo Celular/efeitos da radiação , Estatísticas não Paramétricas
10.
Cell Mol Life Sci ; 69(4): 629-40, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21773671

RESUMO

Functional telomeres are protected from non-homologous end-joining (NHEJ) and homologous recombination (HR) DNA repair pathways. Replication is a critical period for telomeres because of the requirement for reconstitution of functional protected telomere conformations, a process that involves DNA repair proteins. Using knockdown of DNA-PKcs and Rad51 expression in three different cell lines, we demonstrate the respective involvement of NHEJ and HR in the formation of telomere aberrations induced by the G-quadruplex ligand 360A during or after replication. HR contributed to specific chromatid-type aberrations (telomere losses and doublets) affecting the lagging strand telomeres, whereas DNA-PKcs-dependent NHEJ was responsible for sister telomere fusions as a direct consequence of G-quadruplex formation and/or stabilization induced by 360A on parental telomere G strands. NHEJ and HR activation at telomeres altered mitotic progression in treated cells. In particular, NHEJ-mediated sister telomere fusions were associated with altered metaphase-anaphase transition and anaphase bridges and resulted in cell death during mitosis or early G1. Collectively, these data elucidate specific molecular and cellular mechanisms triggered by telomere targeting by the G-quadruplex ligand 360A, leading to cancer cell death.


Assuntos
Apoptose , Proteína Quinase Ativada por DNA/metabolismo , Quadruplex G , Mitose/genética , Proteínas Nucleares/metabolismo , Piridinas/farmacologia , Quinolinas/farmacologia , Rad51 Recombinase/metabolismo , Telômero , Anáfase , Linhagem Celular , Reparo do DNA por Junção de Extremidades , Proteína Quinase Ativada por DNA/antagonistas & inibidores , Proteína Quinase Ativada por DNA/genética , Recombinação Homóloga , Humanos , Ligantes , Metáfase , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Rad51 Recombinase/antagonistas & inibidores , Rad51 Recombinase/genética , Telômero/metabolismo , Telômero/patologia
11.
Cell Mol Life Sci ; 69(17): 2933-49, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22460582

RESUMO

DNA ligase I (LigI) plays a central role in the joining of strand interruptions during replication and repair. In our current study, we provide evidence that DNA ligase III (LigIII) and XRCC1, which form a complex that functions in single-strand break repair, are required for the proliferation of mammalian LigI-depleted cells. We show from our data that in cells with either dysfunctional LigI activity or depleted of this enzyme, both LigIII and XRCC1 are retained on the chromatin and accumulate at replication foci. We also demonstrate that the LigI and LigIII proteins cooperate to inhibit sister chromatid exchanges but that only LigI prevents telomere sister fusions. Taken together, these results suggest that in cells with dysfunctional LigI, LigIII contributes to the ligation of replication intermediates but not to the prevention of telomeric instability.


Assuntos
DNA Ligases/fisiologia , Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Mitose/fisiologia , Telômero/química , Animais , Western Blotting , Sobrevivência Celular , Células Cultivadas , Cromatina/genética , Ensaio de Unidades Formadoras de Colônias , Dano ao DNA/genética , DNA Ligase Dependente de ATP , DNA Ligases/antagonistas & inibidores , Reparo do DNA , Replicação do DNA , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Embrião de Mamíferos/enzimologia , Fibroblastos/enzimologia , Imunofluorescência , Teste de Complementação Genética , Humanos , Hibridização in Situ Fluorescente , Camundongos , Camundongos Knockout , Proteínas de Ligação a Poli-ADP-Ribose , RNA Interferente Pequeno/genética , Troca de Cromátide Irmã/genética , Telômero/genética , Proteína 1 Complementadora Cruzada de Reparo de Raio-X , Proteínas de Xenopus
12.
Oncol Rep ; 49(1)2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36367190

RESUMO

Cancer stem cells (CSCs) serve an essential role in failure of conventional antitumor therapy. In breast cancer, CD24­/low/CD44+ phenotype and high aldehyde dehydrogenase activity are associated with CSC subtypes. Furthermore, CD24­/low/CD44+ pattern is also characteristic of mesenchymal cells generated by epithelial­mesenchymal transition (EMT). CD24 is a surface marker expressed in numerous types of tumor, however, its biological functions and role in cancer progression and treatment resistance remain poorly documented. Loss of CD24 expression in breast cancer cells is associated with radiation resistance and control of oxidative stress. Reactive oxygen species (ROS) mediate the effects of anticancer drugs as well as ionizing radiation; therefore, the present study investigated if CD24 mediates radiation­ and chemo­resistance of breast cancer cells. Using a HMLE breast cancer cell model, CD24 expression has been artificially modulated and it was observed that loss of CD24 expression induced stemness properties associated with acquisition of a hybrid E/M phenotype. CD24­/low cells were more radiation­ and chemo­resistant than CD24+ cells. The resistance was associated with lower levels of ROS; CD24 controlled ROS levels via regulation of mitochondrial function independently of antioxidant activity. Together, these results suggested a key role of CD24 in de­differentiation of breast cancer cells and promoting acquisition of therapeutic resistance properties.


Assuntos
Antígeno CD24 , Neoplasias , Antígeno CD24/genética , Antígeno CD24/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Células-Tronco Neoplásicas/metabolismo , Transição Epitelial-Mesenquimal , Diferenciação Celular , Linhagem Celular Tumoral , Neoplasias/patologia
13.
Cell Rep ; 42(4): 112342, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37027298

RESUMO

XLF/Cernunnos is a component of the ligation complex used in classical non-homologous end-joining (cNHEJ), a major DNA double-strand break (DSB) repair pathway. We report neurodevelopmental delays and significant behavioral alterations associated with microcephaly in Xlf-/- mice. This phenotype, reminiscent of clinical and neuropathologic features in humans deficient in cNHEJ, is associated with a low level of apoptosis of neural cells and premature neurogenesis, which consists of an early shift of neural progenitors from proliferative to neurogenic divisions during brain development. We show that premature neurogenesis is related to an increase in chromatid breaks affecting mitotic spindle orientation, highlighting a direct link between asymmetric chromosome segregation and asymmetric neurogenic divisions. This study reveals thus that XLF is required for maintaining symmetric proliferative divisions of neural progenitors during brain development and shows that premature neurogenesis may play a major role in neurodevelopmental pathologies caused by NHEJ deficiency and/or genotoxic stress.


Assuntos
Enzimas Reparadoras do DNA , Proteínas de Ligação a DNA , Humanos , Animais , Camundongos , Enzimas Reparadoras do DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Reparo do DNA , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Encéfalo/metabolismo
14.
Chromosoma ; 120(3): 309-19, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21359527

RESUMO

Interstitial telomeric sequences (ITSs) in hamster cells are hot spots for spontaneous and induced chromosome aberrations (CAs). Most data on ITS instability to date have been obtained in DNA repair-proficient cells. The classical non-homologous end joining repair pathway (C-NHEJ), which is the principal double strand break (DSB) repair mechanism in mammalian cells, is thought to restore the morphologically correct chromosome structure. The production of CAs thus involves DNA-PKcs-independent repair pathways. In our current study, we investigated the participation of DNA-PKcs from the C-NHEJ pathway in the repair of spontaneous or radiation-induced DSBs in ITSs using wild-type and DNA-PKcs mutant Chinese hamster ovary cells. Our data demonstrate that DNA-PKcs stabilizes spontaneous DSBs within ITSs from the chromosome 9 long arm, leading to the formation of terminal deletions. In addition, we show that DNA-PKcs-dependent C-NHEJ is employed following radiation-induced DSBs in other ITSs and restores morphologically correct chromosomes, whereas DNA-PKcs independent mechanisms co-exist in DNA-PKcs proficient cells leading to an excess of CAs within ITSs.


Assuntos
Aberrações Cromossômicas , Quebras de DNA de Cadeia Dupla , Reparo do DNA/genética , Proteína Quinase Ativada por DNA/metabolismo , Telômero/genética , Animais , Células CHO , Cricetinae , Cricetulus
15.
EMBO J ; 27(5): 770-81, 2008 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-18239686

RESUMO

Although brain development abnormalities and brain cancer predisposition have been reported in some Fanconi patients, the possible role of Fanconi DNA repair pathway during neurogenesis is unclear. We thus addressed the role of fanca and fancg, which are involved in the activation of Fanconi pathway, in neural stem and progenitor cells during brain development and adult neurogenesis. Fanca(-/-) and fancg(-/-) mice presented with microcephalies and a decreased neuronal production in developing cortex and adult brain. Apoptosis of embryonic neural progenitors, but not that of postmitotic neurons, was increased in the neocortex of fanca(-/-) and fancg(-/-) mice and was correlated with chromosomal instability. In adult Fanconi mice, we showed a reduced proliferation of neural progenitor cells related to apoptosis and accentuated neural stem cells exhaustion with ageing. In addition, embryonic and adult Fanconi neural stem cells showed a reduced capacity to self-renew in vitro. Our study demonstrates a critical role for Fanconi pathway in neural stem and progenitor cells during developmental and adult neurogenesis.


Assuntos
Encéfalo/citologia , Proteína do Grupo de Complementação A da Anemia de Fanconi/deficiência , Proteína do Grupo de Complementação G da Anemia de Fanconi/deficiência , Neurônios/citologia , Células-Tronco/citologia , Animais , Apoptose , Encéfalo/embriologia , Proliferação de Células , Aberrações Cromossômicas , Reparo do DNA , Desenvolvimento Embrionário , Anemia de Fanconi , Proteína do Grupo de Complementação A da Anemia de Fanconi/genética , Proteína do Grupo de Complementação G da Anemia de Fanconi/genética , Feminino , Camundongos , Camundongos Knockout , Gravidez
16.
Stem Cells ; 29(3): 440-51, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21425407

RESUMO

Cancer stem cells are increasingly recognized as major therapeutic targets. We report here the isolation of glioma stem cells (GSCs) maintaining telomere length through a telomerase-independent mechanism known as alternative lengthening of telomeres (ALTs). TG20 cells were isolated from a glioblastoma multiforme, which had the ALT phenotype. They have no detectable telomerase activity and extremely long and heterogeneous telomeres colocalizing with promyelocytic leukemia bodies. The cancer stem cell potential of TG20 cells was confirmed based on their expression of neural stem cell markers, their capacity of in vitro long-term proliferation and to form intracranial tumors in immune-deficient mice. Interestingly, we found that both in vitro and in vivo TG20 cells were significantly more resistant to ionizing radiation than GSCs with telomerase activity. Analysis of DNA damage foci, DNA double-strand breaks repair, and chromosome instability suggest that radiation resistance was related to interference of ALT pathway with DNA damage response. Therefore, our data show for the first time that the ALT pathway can confer to cancer stem cells the capacity to sustain long-term proliferation as telomerase activity and importantly may also affect treatment efficiency. TG20 cells are thus the first cellular model of GSCs displaying ALT and should prove to be useful for the development of specific treatment strategies.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Células-Tronco Neoplásicas/metabolismo , Telômero/metabolismo , Adulto , Idoso , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Feminino , Glioma/genética , Glioma/metabolismo , Humanos , Hibridização in Situ Fluorescente , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Pessoa de Meia-Idade , Células-Tronco Neoplásicas/patologia , Telômero/genética , Transplante Heterólogo , Células Tumorais Cultivadas
17.
Nucleic Acids Res ; 38(9): 2955-63, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20147462

RESUMO

Telomere maintenance is essential to preserve genomic stability and involves several telomere-specific proteins as well as DNA replication and repair proteins. The kinase ATR, which has a crucial function in maintaining genome integrity from yeast to human, has been shown to be involved in telomere maintenance in several eukaryotic organisms, including yeast, Arabidopsis and Drosophila. However, its role in telomere maintenance in mammals remains poorly explored. Here, we report by using telomere-fluorescence in situ hybridization (Telo-FISH) on metaphase chromosomes that ATR deficiency causes telomere instability both in primary human fibroblasts from Seckel syndrome patients and in HeLa cells. The telomere aberrations resulting from ATR deficiency (i.e. sister telomere fusions and chromatid-type telomere aberrations) are mainly generated during and/or after telomere replication, and involve both leading and lagging strand telomeres as shown by chromosome orientation-FISH (CO-FISH). Moreover, we show that ATR deficiency strongly sensitizes cells to the G-quadruplex ligand 360A, enhancing sister telomere fusions and chromatid-type telomere aberrations involving specifically the lagging strand telomeres. Altogether, these data reveal that ATR plays a critical role in telomere maintenance during and/or after telomere replication in human cells.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Telômero/química , Adolescente , Adulto , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Células Cultivadas , Criança , Pré-Escolar , Aberrações Cromossômicas , Feminino , Fibroblastos/química , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Piridinas/farmacologia , Quinolinas/farmacologia , Telômero/efeitos dos fármacos
18.
Elife ; 102021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34519267

RESUMO

We developed an Xrcc4M61R separation of function mouse line to overcome the embryonic lethality of Xrcc4-deficient mice. XRCC4M61R protein does not interact with Xlf, thus obliterating XRCC4-Xlf filament formation while preserving the ability to stabilize DNA ligase IV. X4M61R mice, which are DNA repair deficient, phenocopy the Nhej1-/- (known as Xlf -/-) setting with a minor impact on the development of the adaptive immune system. The core non-homologous end-joining (NHEJ) DNA repair factor XRCC4 is therefore not mandatory for V(D)J recombination aside from its role in stabilizing DNA ligase IV. In contrast, Xrcc4M61R mice crossed on Paxx-/-, Nhej1-/-, or Atm-/- backgrounds are severely immunocompromised, owing to aborted V(D)J recombination as in Xlf-Paxx and Xlf-Atm double Knock Out (DKO) settings. Furthermore, massive apoptosis of post-mitotic neurons causes embryonic lethality of Xrcc4M61R -Nhej1-/- double mutants. These in vivo results reveal new functional interplays between XRCC4 and PAXX, ATM and Xlf in mouse development and provide new insights into the understanding of the clinical manifestations of human XRCC4-deficient condition, in particular its absence of immune deficiency.


Assuntos
Reparo do DNA por Junção de Extremidades , Proteínas de Ligação a DNA/genética , Linfócitos/imunologia , Mutação de Sentido Incorreto , Imunodeficiência Combinada Severa/genética , Recombinação V(D)J , Animais , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Encéfalo/embriologia , Encéfalo/metabolismo , DNA Ligase Dependente de ATP/genética , DNA Ligase Dependente de ATP/metabolismo , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Predisposição Genética para Doença , Humanos , Linfócitos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Neurônios/patologia , Fenótipo , Imunodeficiência Combinada Severa/imunologia , Imunodeficiência Combinada Severa/metabolismo
19.
Environ Int ; 147: 106295, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33341586

RESUMO

The last decades have seen increased concern about the possible effects of low to moderate doses of ionizing radiation (IR) exposure on cognitive function. An interdisciplinary group of experts (biologists, epidemiologists, dosimetrists and clinicians) in this field gathered together in the framework of the European MELODI workshop on non-cancer effects of IR to summarise the state of knowledge on the topic and elaborate research recommendations for future studies in this area. Overall, there is evidence of cognitive effects from low IR doses both from biology and epidemiology, though a better characterization of effects and understanding of mechanisms is needed. There is a need to better describe the specific cognitive function or diseases that may be affected by radiation exposure. Such cognitive deficit characterization should consider the human life span, as effects might differ with age at exposure and at outcome assessment. Measurements of biomarkers, including imaging, will likely help our understanding on the mechanism of cognitive-related radiation induced deficit. The identification of loci of individual genetic susceptibility and the study of gene expression may help identify individuals at higher risk. The mechanisms behind the radiation induced cognitive effects are not clear and are likely to involve several biological pathways and different cell types. Well conducted research in large epidemiological cohorts and experimental studies in appropriate animal models are needed to improve the understanding of radiation-induced cognitive effects. Results may then be translated into recommendations for clinical radiation oncology and imaging decision making processes.


Assuntos
Exposição à Radiação , Lesões por Radiação , Animais , Biomarcadores , Cognição , Humanos , Exposição à Radiação/efeitos adversos , Lesões por Radiação/epidemiologia , Radiação Ionizante
20.
Cell Mol Life Sci ; 66(19): 3219-34, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19672559

RESUMO

Poly(ADP-ribose) polymerase-1 (Parp-1) and the protein deacetylase SirT1 are two of the most effective NAD(+)-consuming enzymes in the cell with key functions in genome integrity and chromatin-based pathways. Here, we examined the in vivo crosstalk between both proteins. We observed that the double disruption of both genes in mice tends to increase late post-natal lethality before weaning consistent with important roles of both proteins in genome integrity during mouse development. We identified increased spontaneous telomeric abnormalities associated with decreased cell growth in the absence of either SirT1 or SirT1 and Parp-1 in mouse cells. In contrast, the additional disruption of Parp-1 rescued the abnormal pericentric heterochromatin, the nucleolar disorganization and the mitotic defects observed in SirT1-deficient cells. Together, these findings are in favor of key functions of both proteins in cellular response to DNA damage and in the modulation of histone modifications associated with constitutive heterochromatin integrity.


Assuntos
Cromatina/fisiologia , Genoma , Poli(ADP-Ribose) Polimerases/metabolismo , Sirtuínas/metabolismo , Animais , Nucléolo Celular/ultraestrutura , Proliferação de Células , Células Cultivadas , Cromatina/metabolismo , Dano ao DNA , Instabilidade Genômica , Histonas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitose/fisiologia , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/genética , Sirtuína 1 , Sirtuínas/genética , Telômero/genética , Telômero/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA