Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Free Radic Biol Med ; 138: 23-32, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31029787

RESUMO

AIMS: Oxidative stress is known to induce early replicative senescence. Senescence has been proposed to work as a barrier to immortalization and tumor development. Here, we aimed to evaluate the impact of the loss of peroxisome proliferator activated receptor γ co-activator 1α (PGC-1α), a master regulator of oxidative metabolism and mitochondrial reactive oxygen species (ROS) generation, on replicative senescence and immortalization in mouse embryonic fibroblasts (MEFs). RESULTS: We found that primary MEFs lacking PGC-1α showed higher levels of ROS than wild-type MEFs at all cell passages tested. The elevated production of ROS was associated with higher levels of oxidative DNA damage and the increased formation of DNA double-strand breaks. Evaluation of the induction of DNA repair systems in response to γ-radiation indicated that the loss of PGC-1α also resulted in a small but significant reduction in their activity. DNA damage induced the early activation of senescence markers, including an increase in the number of ß-galactosidase-positive cells, the induction of p53 phosphorylation, and the increase in p16 and p19 protein. These changes were, however, not sufficient to reduce proliferation rates of PGC-1α-deficient MEFs at any cell passage tested. Moreover, PGC-1α-deficient cells escaped replicative senescence. INNOVATION & CONCLUSION: PGC-1α plays an important role in the control of cellular senescence and immortalization.


Assuntos
Senescência Celular/efeitos da radiação , Reparo do DNA , DNA/genética , Fibroblastos/efeitos da radiação , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Animais , Biomarcadores/metabolismo , Proliferação de Células/genética , Proliferação de Células/efeitos da radiação , Senescência Celular/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p19/genética , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , DNA/metabolismo , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Embrião de Mamíferos , Fibroblastos/citologia , Fibroblastos/metabolismo , Raios gama , Regulação da Expressão Gênica , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Mitocôndrias/efeitos da radiação , Estresse Oxidativo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/deficiência , Fosforilação/efeitos da radiação , Espécies Reativas de Oxigênio/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
2.
DNA Repair (Amst) ; 54: 40-45, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28460268

RESUMO

Non-homologous end joining (NHEJ) is the main mechanism for double strand break (DSB) DNA repair. The error-prone DNA polymerase mu (Polµ) is involved in immunoglobulin variable region rearrangement and in general, NHEJ in non-lymphoid cells. Deletion of NHEJ factors in P53-/- mice, which are highly prone to development of T cell lymphoma, generally increases cancer incidence and shifts the tumor spectrum towards aggressive pro-B lymphoma. In contrast, Polµ deletion increased sarcoma incidence, proportionally reducing pro-B lymphoma development on the P53-deficient background. Array comparative genomic hybridization (aCGH) analyses showed DNA copy number alterations in both P53-/- and Polµ-/-P53-/- lymphomas. Our results also indicate that the increase in sarcoma incidence in Polµ-/-P53-/- mice could be associated with Cdk4 and Kub3 amplification and overexpression. These results identify a role for Polµ in the prevention of sarcomagenesis on a murine P53-deficient background, in contrast to most other NHEJ factors.


Assuntos
Carcinogênese , Reparo do DNA por Junção de Extremidades , DNA Polimerase Dirigida por DNA/genética , Sarcoma/metabolismo , Proteína Supressora de Tumor p53/genética , Animais , Proteínas de Transporte/genética , Quinase 4 Dependente de Ciclina/genética , DNA/metabolismo , Variações do Número de Cópias de DNA , Amplificação de Genes , Deleção de Genes , Instabilidade Genômica , Linfoma/genética , Linfoma/metabolismo , Linfoma/patologia , Camundongos , Camundongos Knockout , Sarcoma/genética , Sarcoma/patologia , Regulação para Cima
3.
EMBO Mol Med ; 6(6): 835-48, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24859981

RESUMO

Gene targeting is progressively becoming a realistic therapeutic alternative in clinics. It is unknown, however, whether this technology will be suitable for the treatment of DNA repair deficiency syndromes such as Fanconi anemia (FA), with defects in homology-directed DNA repair. In this study, we used zinc finger nucleases and integrase-defective lentiviral vectors to demonstrate for the first time that FANCA can be efficiently and specifically targeted into the AAVS1 safe harbor locus in fibroblasts from FA-A patients. Strikingly, up to 40% of FA fibroblasts showed gene targeting 42 days after gene editing. Given the low number of hematopoietic precursors in the bone marrow of FA patients, gene-edited FA fibroblasts were then reprogrammed and re-differentiated toward the hematopoietic lineage. Analyses of gene-edited FA-iPSCs confirmed the specific integration of FANCA in the AAVS1 locus in all tested clones. Moreover, the hematopoietic differentiation of these iPSCs efficiently generated disease-free hematopoietic progenitors. Taken together, our results demonstrate for the first time the feasibility of correcting the phenotype of a DNA repair deficiency syndrome using gene-targeting and cell reprogramming strategies.


Assuntos
Reprogramação Celular , Proteína do Grupo de Complementação A da Anemia de Fanconi/genética , Anemia de Fanconi/genética , Anemia de Fanconi/terapia , Marcação de Genes , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/metabolismo , Terapia Genética/métodos , Hematopoese , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo
4.
PLoS One ; 9(4): e93074, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24691161

RESUMO

Polµ is an error-prone PolX polymerase that contributes to classical NHEJ DNA repair. Mice lacking Polµ (Polµ(-/-)) show altered hematopoiesis homeostasis and DSB repair and a more pronounced nucleolytic resection of some V(D)J junctions. We previously showed that Polµ(-/-) mice have increased learning capacity at old ages, suggesting delayed brain aging. Here we investigated the effect of Polµ(-/-) deficiency on liver aging. We found that old Polµ(-/-) mice (>20 month) have greater liver regenerative capacity compared with wt animals. Old Polµ(-/-) liver showed reduced genomic instability and increased apoptosis resistance. However, Polµ(-/-) mice did not show an extended life span and other organs (e.g., heart) aged normally. Our results suggest that Polµ deficiency activates transcriptional networks that reduce constitutive apoptosis, leading to enhanced liver repair at old age.


Assuntos
Envelhecimento/patologia , DNA Polimerase Dirigida por DNA/deficiência , Fígado/patologia , Estresse Oxidativo , Animais , Instabilidade Genômica , Fígado/fisiopatologia , Testes de Função Hepática , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Modelos Biológicos , Miocárdio/patologia , Fenótipo , Troca de Cromátide Irmã
5.
Stem Cells ; 31(8): 1726-30, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23630174

RESUMO

Although there is an increasing interest in defining the role of DNA damage response mechanisms in cell reprogramming, the relevance of proteins participating in nonhomologous end joining (NHEJ), a major mechanism of DNA double-strand breaks repair, in this process remains to be investigated. Herein, we present data related to the reprogramming of primary mouse embryonic fibroblasts (MEF) from severe combined immunodeficient (Scid) mice defective in DNA-PKcs, a key protein for NHEJ. Reduced numbers of induced pluripotent stem cell (iPSC) colonies were generated from Scid cells using reprogramming lentiviral vectors (LV), being the reprogramming efficiency fourfold to sevenfold lower than that observed in wt cells. Moreover, these Scid iPSC-like clones were prematurely lost or differentiated spontaneously. While the Scid mutation neither reduce the proliferation rate nor the transduction efficacy of fibroblasts transduced with reprogramming LV, both the expression of SA-ß-Gal and of P16/INK(4a) senescence markers were highly increased in Scid versus wt MEFs during the reprogramming process, accounting for the reduced reprogramming efficacy of Scid MEFs. The use of improved Sleeping Beauty transposon/transposase systems allowed us, however, to isolate DNA-PKcs-deficient iPSCs which preserved their parental genotype and hypersensitivity to ionizing radiation. This new disease-specific iPSC model would be useful to understand the physiological consequences of the DNA-PKcs mutation during development and would help to improve current cell and gene therapy strategies for the disease.


Assuntos
Reprogramação Celular/genética , Reparo do DNA por Junção de Extremidades , Células-Tronco Pluripotentes Induzidas/fisiologia , Animais , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Transfecção
6.
PLoS One ; 8(1): e53243, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23301049

RESUMO

A definitive consequence of the aging process is the progressive deterioration of higher cognitive functions. Defects in DNA repair mechanisms mostly result in accelerated aging and reduced brain function. DNA polymerase µ is a novel accessory partner for the non-homologous end-joining DNA repair pathway for double-strand breaks, and its deficiency causes reduced DNA repair. Using associative learning and long-term potentiation experiments, we demonstrate that Polµ(-/-) mice, however, maintain the ability to learn at ages when wild-type mice do not. Expression and biochemical analyses suggest that brain aging is delayed in Polµ(-/-) mice, being associated with a reduced error-prone DNA oxidative repair activity and a more efficient mitochondrial function. This is the first example in which the genetic ablation of a DNA-repair function results in a substantially better maintenance of learning abilities, together with fewer signs of brain aging, in old mice.


Assuntos
Envelhecimento , Encéfalo/fisiologia , DNA Polimerase Dirigida por DNA/metabolismo , Aprendizagem , Potenciação de Longa Duração/genética , Animais , Comportamento Animal , Encéfalo/metabolismo , Condicionamento Clássico , DNA/genética , Reparo do DNA , DNA Polimerase Dirigida por DNA/genética , Hipocampo/metabolismo , Locomoção , Masculino , Camundongos , Camundongos Knockout , Estresse Oxidativo , Fenótipo , Reprodutibilidade dos Testes , Temperatura
7.
Springerplus ; 1(1): 63, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23487597

RESUMO

Cardiac healing, which follows myocardial infarction, is a complex process guided by intricate interactions among different components. Some resident cell populations with a potential role in cardiac healing have already been described in cardiac tissues. These non-cardiomyocyte cell subsets, globally described as cardiac pluripotent/progenitor cells (CPCs), are able to differentiate into all three major cardiac cell lineages (endothelial, smooth muscle and cardiomyocyte cells) in experimental settings. Nevertheless, physiological cardiac healing results in a fibrous scar, which remains to be fully modelled experimentally. Since a role for complement anaphylatoxins (C3a and C5a) has been described in several regeneration/repair processes, we examined the effects that C3a and C5a exert on a defined population of CPCs. We found that C3a and C5a are able to enhance CPC migration and proliferation. In vitro studies showed that this effect is linked to activation of telomerase mRNA and partial preservation of telomere length, in an NFκB-dependent manner. In addition, anaphylatoxin signalling modulates the CPC phenotype, increasing myofibroblast differentiation and reducing endothelial and cardiac gene expression. These findings may denote that C3a and C5a are able to maintain/increase the cardiac stem cell pool within the heart, whilst simultaneously facilitating and modulating resident cell differentiation. We found that this modulation was directed towards scar forming cells, which increased fibroblast/myofibroblast generation and suggests that both these anaphylatoxins could play a relevant role in the damage-coupled activation of resident cells, and regulation of the cardiac healing process after injury.

8.
PLoS Genet ; 5(2): e1000389, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19229323

RESUMO

Polymerase micro (Polmicro) is an error-prone, DNA-directed DNA polymerase that participates in non-homologous end-joining (NHEJ) repair. In vivo, Polmicro deficiency results in impaired Vkappa-Jkappa recombination and altered somatic hypermutation and centroblast development. In Polmicro(-/-) mice, hematopoietic development was defective in several peripheral and bone marrow (BM) cell populations, with about a 40% decrease in BM cell number that affected several hematopoietic lineages. Hematopoietic progenitors were reduced both in number and in expansion potential. The observed phenotype correlates with a reduced efficiency in DNA double-strand break (DSB) repair in hematopoietic tissue. Whole-body gamma-irradiation revealed that Polmicro also plays a role in DSB repair in non-hematopoietic tissues. Our results show that Polmicro function is required for physiological hematopoietic development with an important role in maintaining early progenitor cell homeostasis and genetic stability in hematopoietic and non-hematopoietic tissues.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , DNA Polimerase Dirigida por DNA/metabolismo , Hematopoese , Animais , Células Cultivadas , DNA Polimerase Dirigida por DNA/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/enzimologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout
9.
Free Radic Biol Med ; 46(3): 387-96, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19038329

RESUMO

Lymphomas adapt to their environment by undergoing a complex series of biochemical changes that are currently not well understood. To better define these changes, we examined the gene expression and gene ontology profiles of thymic lymphomas from a commonly used model of carcinogenesis, the p53(-/-) mouse. These tumors show a highly significant upregulation of mitochondrial biogenesis, mitochondrial protein translation, mtDNA copy number, reactive oxygen species, antioxidant defenses, proton transport, ATP synthesis, hypoxia response, and glycolysis, indicating a fundamental change in the bioenergetic profile of the transformed T cell. Our results suggest that T cell tumorigenesis involves a simultaneous upregulation of mitochondrial biogenesis, mitochondrial respiration, and glycolytic activity. These processes would allow cells to adapt to the stressful tumor environment by facilitating energy production and thereby promote tumor growth. Understanding these adaptations is likely to result in improved therapeutic strategies for this tumor type.


Assuntos
DNA Mitocondrial/análise , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Linfoma de Células T/genética , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Complexo de Proteínas da Cadeia de Transporte de Elétrons/genética , Perfilação da Expressão Gênica , Glicólise/genética , Linfoma de Células T/metabolismo , Linfoma de Células T/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/genética , Estresse Oxidativo/genética , Consumo de Oxigênio , Proteínas Proto-Oncogênicas c-myc/genética , Timo/metabolismo , Timo/patologia , Proteína Supressora de Tumor p53/genética
10.
Aging Cell ; 6(2): 179-88, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17286610

RESUMO

The nematode Caenorhabditis elegans has become one of the most widely used model systems for the study of aging, yet very little is known about how C. elegans age. The development of the worm, from egg to young adult has been completely mapped at the cellular level, but such detailed studies have not been extended throughout the adult lifespan. Numerous single gene mutations, drug treatments and environmental manipulations have been found to extend worm lifespan. To interpret the mechanism of action of such aging interventions, studies to characterize normal worm aging, similar to those used to study worm development are necessary. We have used 4',6'-diamidino-2-phenylindole hydrochloride staining and quantitative polymerase chain reaction to investigate the integrity of nuclei and quantify the nuclear genome copy number of C. elegans with age. We report both systematic loss of nuclei or nuclear DNA, as well as dramatic age-related changes in nuclear genome copy number. These changes are delayed or attenuated in long-lived daf-2 mutants. We propose that these changes are important pathobiological characteristics of aging nematodes.


Assuntos
Envelhecimento/genética , Caenorhabditis elegans/genética , Núcleo Celular/genética , Genoma Helmíntico , Animais , Caenorhabditis elegans/fisiologia , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas , DNA de Helmintos/metabolismo , Dosagem de Genes , Genes de Helmintos , Masculino
11.
Blood ; 108(12): 3928-37, 2006 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16912231

RESUMO

The chemokine CXCL12 influences self-renewal and differentiation of hematopoietic stem cell precursors in bone marrow by directing them toward specific stromalcell components. CXCL12 up-regulates members of the SOCS family through JAK/STAT activation, a mechanism that attenuates chemokine responses. SOCS expression may thus modulate retention of hematopoietic precursors (Sca-1(+) c-Kit(+)Lin(-) cells) in bone marrow. We show that in bovine growth hormone transgenic mice and in growth hormone-treated mice, SOCS up-regulation correlated with a large number of Sca-1(+) c-Kit(+)Lin(-) cells in blood. Retroviral transduction of SOCSs blocked in vitro migration of Sca-1(+)c-Kit(+)Lin(-) cells, as well as their capacity to reconstitute lethally irradiated mice. Furthermore, in lethally irradiated mice reconstituted with bone marrow infected by a tetracycline-regulated, SOCS-expressing lentiviral vector, doxycycline treatment promoted rapid, extensive precursor mobilization to the periphery. The results indicate that by blocking CXCR4-mediated functions, SOCSs modulate hematopoietic precursor cell retention in bone marrow, and suggest the therapeutic interest of SOCS manipulation in several pathologic situations.


Assuntos
Movimento Celular , Células-Tronco Hematopoéticas/metabolismo , Receptores CXCR4/metabolismo , Proteínas Supressoras da Sinalização de Citocina/biossíntese , Regulação para Cima , Animais , Antígenos de Diferenciação/biossíntese , Medula Óssea/metabolismo , Transplante de Medula Óssea , Bovinos , Linhagem Celular , Movimento Celular/genética , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Hormônio do Crescimento/genética , Hormônio do Crescimento/metabolismo , Mobilização de Células-Tronco Hematopoéticas/métodos , Humanos , Camundongos , Camundongos Transgênicos , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/genética , Proteínas Supressoras da Sinalização de Citocina/genética , Transplante Autólogo
12.
PLoS Genet ; 2(1): e10, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16450009

RESUMO

RNA interference (RNAi) has been used increasingly for reverse genetics in invertebrates and mammalian cells, and has the potential to become an alternative to gene knockout technology in mammals. Thus far, only RNA polymerase III (Pol III)-expressed short hairpin RNA (shRNA) has been used to make shRNA-expressing transgenic mice. However, widespread knockdown and induction of phenotypes of gene knockout in postnatal mice have not been demonstrated. Previous studies have shown that Pol II synthesizes micro RNAs (miRNAs)-the endogenous shRNAs that carry out gene silencing function. To achieve efficient gene knockdown in mammals and to generate phenotypes of gene knockout, we designed a construct in which a Pol II (ubiquitin C) promoter drove the expression of an shRNA with a structure that mimics human miRNA miR-30a. Two transgenic lines showed widespread and sustained shRNA expression, and efficient knockdown of the target gene Sod2. These mice were viable but with phenotypes of SOD2 deficiency. Bigenic heterozygous mice generated by crossing these two lines showed nearly undetectable target gene expression and phenotypes consistent with the target gene knockout, including slow growth, fatty liver, dilated cardiomyopathy, and premature death. This approach opens the door of RNAi to a wide array of well-established Pol II transgenic strategies and offers a technically simpler, cheaper, and quicker alternative to gene knockout by homologous recombination for reverse genetics in mice and other mammalian species.


Assuntos
RNA Polimerase II/genética , RNA/genética , Superóxido Dismutase/deficiência , Superóxido Dismutase/genética , Regiões 3' não Traduzidas/genética , Animais , Sequência de Bases , Primers do DNA , Feminino , Fertilização , Regulação Enzimológica da Expressão Gênica , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Conformação de Ácido Nucleico , Óvulo/fisiologia , Fenótipo , Reação em Cadeia da Polimerase , RNA/química , Interferência de RNA
13.
Breast ; 14(4): 256-63, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16085231

RESUMO

To investigate the dysregulating effect of excess oxidative stress on mammary gland development, mammary anlage from newborn female mice with normal (+/+) or absent (null, -/-) manganese superoxide dismutase (SOD2) were excised and implanted under the renal capsule of normal host female nude mice with/without concurrent estrogen supplementation. After 30 days the transplanted glands were excised for wholemount, microscopic and immunohistochemical evaluation. In contrast to the normal growth and maturation of transplanted SOD2+/+ glands, SOD2-/- glands showed arrested development, reduced ductal outgrowth and branching, and absent lumen. These hypomorphic SOD2-/- ducts contained hyperplastic epithelium with increased Ki-67 labelling, loss of E-cadherin expression, and disorganized p63 and cytokeratin (K)-14 expressing basal and myoepithelial components. Estrogen treatment failed to upregulate progesterone receptor or normalize development. These findings suggest that excess oxidative stress from loss of SOD2 function can arrest mammary gland maturation and induce hyperplastic epithelium with early premalignant features.


Assuntos
Caderinas/biossíntese , Glândulas Mamárias Animais/enzimologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Estresse Oxidativo , Superóxido Dismutase/metabolismo , Animais , Transformação Celular Neoplásica , Feminino , Hiperplasia , Camundongos , Mitocôndrias/enzimologia , Espécies Reativas de Oxigênio
14.
Oncogene ; 23(3): 735-43, 2004 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-14737108

RESUMO

The absence of functional p53 has complex consequences on the cellular responses to cytotoxic drugs. Here, we have examined the role of p53 in the response to 5-aza-2'-deoxycytidine (5-aza-dC or decitabine). Primary mouse embryonic fibroblasts deficient for p53 undergo apoptosis after treatment with 5-aza-dC. When compared with other demethylating drugs or chemotherapeutic treatments, 5-aza-dC showed the highest selectivity ratio for triggering apoptosis in p53-deficient cells relative to wild-type cells. Moreover, the apoptotic efficacy of 5-aza-dC is proprietary of p53-deficient cells, not being observed in cells lacking other cell-cycle regulators, such as p19ARF, p16INK4a, p21(CIP1/WAF1), E2F-1, or E2F-2. Interestingly, treatment with 5-aza-dC results in the same degree of global genomic hypomethylation in wild-type and p53-null cells. However, wild-type cells activate p53 and arrest at G2/M, whereas p53-null cells accumulate severe chromosomal aberrations and undergo apoptosis. Significantly, the impact of p53-deficiency on the response to 5-aza-dC is not exclusive of primary non-neoplastic cells, but it is also present in neoplastically transformed cells. Finally, treatment of mice bearing genetically defined tumors with nontoxic doses of 5-aza-dC results in therapeutical responses only on tumors lacking p53, but not on tumors lacking p19ARF. Together, our results put forward the hypothesis that the absence of p53 may determine a higher chemotherapeutic index for 5-aza-dC.


Assuntos
Apoptose/efeitos dos fármacos , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Apoptose/fisiologia , Decitabina , Fase G1/efeitos dos fármacos , Camundongos , Mitose/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
15.
Nat Cell Biol ; 5(8): 741-7, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12855956

RESUMO

Most mammalian cells do not divide indefinitely, owing to a process termed replicative senescence. In human cells, replicative senescence is caused by telomere shortening, but murine cells senesce despite having long stable telomeres. Here, we show that the phenotypes of senescent human fibroblasts and mouse embryonic fibroblasts (MEFs) differ under standard culture conditions, which include 20% oxygen. MEFs did not senesce in physiological (3%) oxygen levels, but underwent a spontaneous event that allowed indefinite proliferation in 20% oxygen. The proliferation and cytogenetic profiles of DNA repair-deficient MEFs suggested that DNA damage limits MEF proliferation in 20% oxygen. Indeed, MEFs accumulated more DNA damage in 20% oxygen than 3% oxygen, and more damage than human fibroblasts in 20% oxygen. Our results identify oxygen sensitivity as a critical difference between mouse and human cells, explaining their proliferative differences in culture, and possibly their different rates of cancer and ageing.


Assuntos
Divisão Celular/fisiologia , Senescência Celular/fisiologia , Fibroblastos/fisiologia , Oxigênio/metabolismo , Animais , Células Cultivadas , Ensaio Cometa , Dano ao DNA , Reparo do DNA , Fibroblastos/citologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
16.
Blood ; 99(8): 2767-75, 2002 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11929765

RESUMO

Telomere length must be tightly regulated in highly proliferative tissues, such as the lymphohematopoietic system. Under steady-state conditions, the levels and functionality of hematopoietic-committed or multipotent progenitors were not affected in late-generation telomerase-deficient mice (mTerc(-/-)) with critically short telomeres. Evaluation of self-renewal potential of mTerc(-/-) day-12 spleen colony-forming units demonstrated no alteration as compared with wildtype progenitors. However, the replating ability of mTerc(-/-) granulocyte-macrophage CFUs (CFU-GMs) was greatly reduced as compared with wildtype CFU-GMs, indicating a diminished capacity of late-generation mTerc(-/-) committed progenitors when forced to proliferate. Long-term bone marrow cultures of mTerc(-/-) bone marrow (BM) cells show a reduction in proliferative capacity; this defect can be mainly attributed to the hematopoietic, not to the stromal, mTerc(-/-) cells. In serial and competitive transplantations, mTerc(-/-) BM stem cells show reduced long-term repopulating capacity, concomitant with an increase in genetic instability compared with wildtype cells. Nevertheless, in competitive transplantations late-generation mTerc(-/-) precursors can occasionally overcome this proliferative impairment and reconstitute irradiated recipients. In summary, our results demonstrate that late-generation mTerc(-/-) BM cells with short telomeres, although exhibiting reduced proliferation ability and reduced long-term repopulating capacity, can still reconstitute myeloablated animals maintaining stem cell function.


Assuntos
Hematopoese , Células-Tronco Hematopoéticas/enzimologia , Telomerase/fisiologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/enzimologia , Transplante de Medula Óssea , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Divisão Celular , Análise Citogenética , Feminino , Células-Tronco Hematopoéticas/classificação , Células-Tronco Hematopoéticas/citologia , Masculino , Camundongos , Camundongos Knockout , RNA/genética , Telomerase/deficiência , Telomerase/genética , Translocação Genética
17.
Hum Mol Genet ; 11(4): 439-44, 2002 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11854176

RESUMO

Fanconi anemia (FA) is a rare genetic disease characterized by chromosome instability, progressive pancytopenia and cancer susceptibility. Telomeres are intimately related to chromosome stability and play an important role in organismal viability at the hematological level. Since previous works suggested an accelerated shortening of telomeres in FA, we have studied several markers of telomere integrity and function in FA patients and age-matched controls to get insights into the mechanisms and consequences of telomere erosion in FA. A higher frequency of extra-chromosomic TTAGGG signals and of chromosome ends with undetectable TTAGGG repeats was observed in FA cells by fluorescence in situ hybridization (FISH), suggesting intensive breakage at telomeric sequences. This was proven by measuring the frequency of excess of telomeric signals per cell, which was 2.8-fold higher in FA. Consistent with previous reports, quantitative FISH analysis showed an accelerated telomere shortening of 0.68 kb in FA, which occurred concurrently in both chromosome arms in a similar magnitude. Our data therefore suggest that the telomere erosion in FA is caused by a higher rate of breakage at TTAGGG sequences in vivo in differentiated cells, in addition to mere replicative shortening during lymphocyte proliferation. Consistent with impaired telomeres in FA patients, we observed a >10-fold increase in chromosome end fusions in FA compared to normal controls. This observation was independent of TRF2, a telomere binding factor that protects human telomeres from end fusions, since immunohistochemistry studies in FA cell lines and corrected counterparts by retrovirus-mediated transfer of FANCA and FANCD2 cDNA showed that a functional FA pathway is not required for telomere binding of TRF2.


Assuntos
Aberrações Cromossômicas , Anemia de Fanconi/genética , Telômero , Adolescente , Adulto , Criança , Pré-Escolar , Proteínas de Ligação a DNA/metabolismo , Feminino , Humanos , Masculino , Ligação Proteica , Telômero/patologia , Proteína 2 de Ligação a Repetições Teloméricas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA