Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros

Base de dados
Tipo de documento
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.
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
3.
Int J Cancer ; 136(4): 982-8, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24975135

RESUMO

The hematopoietic growth factor granulocyte colony-stimulating factor (G-CSF) has a role in proliferation, differentiation and migration of the myeloid lineage and in mobilizing hematopoietic stem and progenitor cells into the bloodstream. However, G-CSF has been newly characterized as a neurotrophic factor in the brain. We recently uncovered that autonomic nerve development in the tumor microenvironment participates actively in prostate tumorigenesis and metastasis. Here, we found that G-CSF constrains cancer to grow and progress by, respectively, supporting the survival of sympathetic nerve fibers in 6-hydroxydopamine-sympathectomized mice and also, promoting the aberrant outgrowth of parasympathetic nerves in transgenic or xenogeneic prostate tumor models. This provides insight into how neurotrophic growth factors may control tumor neurogenesis and may lead to new antineurogenic therapies for prostate cancer.


Assuntos
Axônios/fisiologia , Carcinogênese/metabolismo , Fator Estimulador de Colônias de Granulócitos/fisiologia , Neoplasias da Próstata/metabolismo , Fibras Adrenérgicas/patologia , Fibras Adrenérgicas/fisiologia , Animais , Axônios/patologia , Sobrevivência Celular , Células HL-60 , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Transplante de Neoplasias , Fatores de Crescimento Neural/fisiologia , Próstata/inervação , Neoplasias da Próstata/patologia
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
Nucleic Acids Res ; 36(5): 1741-54, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18263609

RESUMO

Telomeres are known to prevent chromosome ends from being recognized as DNA double-strand breaks. Conversely, many DNA damage response proteins, including ATM, are thought to participate to telomere maintenance. However, the precise roles of ATM at telomeres remain unclear due to its multiple functions in cell checkpoints and apoptosis. To gain more insights into the role of ATM in telomere maintenance, we determined the effects of the G-quadruplex ligand 360A in various cell lines lacking functional ATM. We showed, by using Fluorescence in situ hybridization (FISH) and Chromosome Orientation-FISH using telomere PNA probes, that 360A induced specific telomere aberrations occurring during or after replication, mainly consisting in sister telomere fusions and also recombinations that involved preferentially the lagging strand telomeres. We demonstrate that ATM reduced telomere instability independently of apoptosis induction. Our results suggest thus that ATM has a direct role in preventing inappropriate DNA repair at telomeres, which could be related to its possible participation to the formation of protected structures at telomeres.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Reparo do DNA , Proteínas de Ligação a DNA/fisiologia , Quadruplex G/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/fisiologia , Piridinas/toxicidade , Quinolinas/toxicidade , Telômero/química , Proteínas Supressoras de Tumor/fisiologia , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia , Ciclo Celular , Proteínas de Ciclo Celular/antagonistas & inibidores , Aberrações Cromossômicas , Dano ao DNA , Proteínas de Ligação a DNA/antagonistas & inibidores , Células HeLa , Humanos , Ligantes , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Transdução de Sinais , Troca de Cromátide Irmã/efeitos dos fármacos , Telômero/efeitos dos fármacos , Proteínas Supressoras de Tumor/antagonistas & inibidores
12.
Sci Rep ; 10(1): 18742, 2020 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-33128011

RESUMO

Human glioblastoma (GBM) is the most common primary malignant brain tumor. A minor subpopulation of cancer cells, known as glioma stem-like cells (GSCs), are thought to play a major role in tumor relapse due to their stem cell-like properties, their high resistance to conventional treatments and their high invasion capacity. We show that ionizing radiation specifically enhances the motility and invasiveness of human GSCs through the stabilization and nuclear accumulation of the hypoxia-inducible factor 1α (HIF1α), which in turn transcriptionally activates the Junction-mediating and regulatory protein (JMY). Finally, JMY accumulates in the cytoplasm where it stimulates GSC migration via its actin nucleation-promoting activity. Targeting JMY could thus open the way to the development of new therapeutic strategies to improve the efficacy of radiotherapy and prevent glioma recurrence.


Assuntos
Glioblastoma/metabolismo , Glioblastoma/patologia , Glioma/metabolismo , Glioma/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteínas Nucleares/metabolismo , Transativadores/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Movimento Celular/efeitos da radiação , Núcleo Celular/metabolismo , Núcleo Celular/efeitos da radiação , Citoplasma/metabolismo , Citoplasma/efeitos da radiação , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Proteínas Nucleares/genética , Radiação Ionizante , Transdução de Sinais/genética , Transdução de Sinais/efeitos da radiação , Transativadores/genética
14.
Oncotarget ; 10(7): 773-784, 2019 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-30774779

RESUMO

Some cancer cells elongate their telomeres through the ALT (alternative lengthening of telomeres) pathway, which is based on homologous recombination for the addition of telomere repeats without telomerase activity. General control non-derepressible 5 (GCN5) and P300/CBP-associated factor (PCAF), two homologous lysine acetyltransferases, exert opposite effects on the ALT pathway, inhibiting or favoring it respectively. Here we show that ALT cells are particularly sensitive to the inhibition of acetyltransferases activities using Anacardic Acid (AA). AA treatment recapitulates the effect of PCAF knockdown on several ALT features, suggesting that AA decreased the ALT mechanism through the inhibition of lysine transferase activity of PCAF, but not that of GCN5. Furthermore, AA specifically sensitizes human ALT cells to radiation as compared to telomerase-positive cells suggesting that the inhibition of lysine acetyltransferases activity may be used to increase the radiotherapy efficiency against ALT cancers.

15.
Cell Death Differ ; 26(9): 1615-1630, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30442946

RESUMO

PARP3 has been shown to be a key driver of TGFß-induced epithelial-to-mesenchymal transition (EMT) and stemness in breast cancer cells, emerging as an attractive therapeutic target. Nevertheless, the therapeutic value of PARP3 inhibition has not yet been assessed. Here we investigated the impact of the absence of PARP3 or its inhibition on the tumorigenicity of BRCA1-proficient versus BRCA1-deficient breast cancer cell lines, focusing on the triple-negative breast cancer subtype (TNBC). We show that PARP3 knockdown exacerbates centrosome amplification and genome instability and reduces survival of BRCA1-deficient TNBC cells. Furthermore, we engineered PARP3-/- BRCA1-deficient or BRCA1-proficient TNBC cell lines using the CRISPR/nCas9D10A gene editing technology and demonstrate that the absence of PARP3 selectively suppresses the growth, survival and in vivo tumorigenicity of BRCA1-deficient TNBC cells, mechanistically via effects associated with an altered Rictor/mTORC2 signaling complex resulting from enhanced ubiquitination of Rictor. Accordingly, PARP3 interacts with and ADP-ribosylates GSK3ß, a positive regulator of Rictor ubiquitination and degradation. Importantly, these phenotypes were rescued by re-expression of a wild-type PARP3 but not by a catalytic mutant, demonstrating the importance of PARP3's catalytic activity. Accordingly, reduced survival and compromised Rictor/mTORC2 signaling were also observed using a cell-permeable PARP3-specific inhibitor. We conclude that PARP3 and BRCA1 are synthetic lethal and that targeting PARP3's catalytic activity is a promising therapeutic strategy for BRCA1-associated cancers via the Rictor/mTORC2 signaling pathway.


Assuntos
Proteína BRCA1/genética , Proteínas de Ciclo Celular/genética , Poli(ADP-Ribose) Polimerases/genética , Proteína Companheira de mTOR Insensível à Rapamicina/genética , Neoplasias de Mama Triplo Negativas/genética , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Transição Epitelial-Mesenquimal/genética , Feminino , Xenoenxertos , Humanos , Alvo Mecanístico do Complexo 2 de Rapamicina/genética , Camundongos , Transdução de Sinais , Fator de Crescimento Transformador beta/genética , Neoplasias de Mama Triplo Negativas/patologia
16.
Stem Cell Reports ; 11(2): 565-577, 2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-29983386

RESUMO

Deciphering the mechanisms that regulate the quiescence of adult neural stem cells (NSCs) is crucial for the development of therapeutic strategies based on the stimulation of their endogenous regenerative potential in the damaged brain. We show that LeXbright cells sorted from the adult mouse subventricular zone exhibit all the characteristic features of quiescent NSCs. Indeed, they constitute a subpopulation of slowly dividing cells that is able to enter the cell cycle to regenerate the irradiated niche. Comparative transcriptomic analyses showed that they express hallmarks of NSCs but display a distinct molecular signature from activated NSCs (LeX+EGFR+ cells). Particularly, numerous membrane receptors are expressed on quiescent NSCs. We further revealed a different expression pattern of Syndecan-1 between quiescent and activated NSCs and demonstrated its role in the proliferation of activated NSCs. Our data highlight the central role of the stem cell microenvironment in the regulation of quiescence in adult neurogenic niches.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Ciclo Celular , Diferenciação Celular , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Nicho de Células-Tronco , Células-Tronco Adultas/efeitos da radiação , Ciclo Celular/genética , Ciclo Celular/efeitos da radiação , Diferenciação Celular/genética , Diferenciação Celular/efeitos da radiação , Metabolismo Energético , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Células-Tronco Neurais/efeitos da radiação , Neurogênese , Estresse Oxidativo , Transdução de Sinais , Nicho de Células-Tronco/genética , Nicho de Células-Tronco/efeitos da radiação
17.
Nucleic Acids Res ; 33(13): 4182-90, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16052031

RESUMO

The G-overhangs of telomeres are thought to adopt particular conformations, such as T-loops or G-quadruplexes. It has been suggested that G-quadruplex structures could be stabilized by specific ligands in a new approach to cancer treatment consisting in inhibition of telomerase, an enzyme involved in telomere maintenance and cell immortality. Although the formation of G-quadruplexes was demonstrated in vitro many years ago, it has not been definitively demonstrated in living human cells. We therefore investigated the chromosomal binding of a tritiated G-quadruplex ligand, 3H-360A (2,6-N,N'-methyl-quinolinio-3-yl)-pyridine dicarboxamide [methyl-3H]. We verified the in vitro selectivity of 3H-360A for G-quadruplex structures by equilibrium dialysis. We then showed by binding experiments with human genomic DNA that 3H-360A has a very potent selectivity toward G-quadruplex structures of the telomeric 3'-overhang. Finally, we performed autoradiography of metaphase spreads from cells cultured with 3H-360A. We found that 3H-360A was preferentially bound to chromosome terminal regions of both human normal (peripheral blood lymphocytes) and tumor cells (T98G and CEM1301). In conclusion, our results provide evidence that a specific G-quadruplex ligand interacts with the terminal ends of human chromosomes. They support the hypothesis that G-quadruplex ligands induce and/or stabilize G-quadruplex structures at telomeres of human cells.


Assuntos
Cromossomos Humanos/química , DNA/metabolismo , Piridinas/metabolismo , Quinolinas/metabolismo , Telômero/química , Sítios de Ligação , Linhagem Celular Tumoral , Células Cultivadas , Cromossomos Humanos/metabolismo , DNA/química , Quadruplex G , Guanina/química , Humanos , Ligantes , Linfócitos/ultraestrutura , Metáfase , Piridinas/química , Quinolinas/química , Telômero/metabolismo
18.
Oncotarget ; 8(16): 26269-26280, 2017 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-28412741

RESUMO

Cancer cells can use a telomerase-independent mechanism, known as alternative lengthening of telomeres (ALT), to elongate their telomeres. General control non-derepressible 5 (GCN5) and P300/CBP-associated factor (PCAF) are two homologous acetyltransferases that are mutually exclusive subunits in SAGA-like complexes. Here, we reveal that down regulation of GCN5 and PCAF had differential effects on some phenotypic characteristics of ALT cells. Our results suggest that GCN5 is present at telomeres and opposes telomere recombination, in contrast to PCAF that may indirectly favour them in ALT cells.


Assuntos
Estudos de Associação Genética , Homeostase do Telômero/genética , Telômero/genética , Fatores de Transcrição de p300-CBP/genética , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Expressão Gênica , Técnicas de Silenciamento de Genes , Instabilidade Genômica , Humanos , Corpos de Inclusão Intranuclear/genética , Corpos de Inclusão Intranuclear/metabolismo , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patologia , Ligação Proteica , Troca de Cromátide Irmã , Translocação Genética
19.
Oncogene ; 24(4): 541-51, 2005 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-15608689

RESUMO

Hepatoblasts are bipotent progenitors of both hepatocytes and cholangiocytes. The lack of stable in vitro culture systems for such cells makes it necessary to generate liver progenitor cell lines by means of immortalization. In this study, we describe the long-term behaviour of a clone of simian foetal hepatic progenitor cells immortalized by Simian virus 40 (SV40) large T-antigen (T-Ag) flanked by loxP sites. Immortalization was associated with the re-expression of telomerase activity, which decreased at late passages (population doubling 120) after more than a year in culture. This decrease was concomitant to telomere shortening and karyotypic instability. However, the chromosomes carrying the p53 gene remained intact and long-term immortalized progenitor cells maintained contact inhibition and proliferative properties. They also displayed the features of a normal bipotent phenotype. We constructed a retroviral vector expressing an inducible Cre recombinase and transferred it into the immortalized progenitors. Activation of the Cre recombinase by 4-hydroxy-tamoxifen induced SV40 T-Ag excision, leading to the death of cells expressing Cre recombinase. Immortalized progenitors at late passages stopped growing and eventually disappeared after transplantation into the livers of immunocompromised mice. These cells provide a novel model to study hepatic differentiation and carcinogenesis.


Assuntos
Antígenos Virais de Tumores/genética , Antígenos Virais de Tumores/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Vírus 40 dos Símios/genética , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/genética , Linhagem Celular Transformada , Proliferação de Células , Células Cultivadas , Cromossomos de Mamíferos/metabolismo , Haplorrinos , Cariotipagem , Camundongos , Camundongos Knockout , Telomerase/metabolismo , Telômero/metabolismo , Fatores de Tempo
20.
Oncogene ; 24(18): 2917-28, 2005 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-15735722

RESUMO

Telomerase represents a relevant target for cancer therapy. Molecules able to stabilize the G-quadruplex (G4), a structure adopted by the 3'-overhang of telomeres, are thought to inhibit telomerase by blocking its access to telomeres. We investigated the cellular effects of four new 2,6-pyridine-dicarboxamide derivatives displaying strong selectivity for G4 structures and strong inhibition of telomerase in in vitro assays. These compounds inhibited cell proliferation at very low concentrations and then induced a massive apoptosis within a few days in a dose-dependent manner in cultures of three telomerase-positive glioma cell lines, T98G, CB193 and U118-MG. They had also antiproliferative effects in SAOS-2, a cell line in which telomere maintenance involves an alternative lengthening of telomeres (ALT) mechanism. We show that apoptosis was preceded by multiple alterations of the cell cycle: activation of S-phase checkpoints, dramatic increase of metaphase duration and cytokinesis defects. These effects were not associated with telomere shortening, but they were directly related to telomere instability involving telomere end fusion and anaphase bridge formation. Pyridine-based G-quadruplex ligands are therefore promising agents for the treatment of various tumors including malignant gliomas.


Assuntos
Apoptose/fisiologia , Instabilidade Genômica/fisiologia , Telômero/genética , Telômero/metabolismo , Ciclo Celular , Divisão Celular/genética , Divisão Celular/imunologia , Glioma/tratamento farmacológico , Humanos , Ligantes , Piridinas/farmacologia , Telomerase/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA