Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Stem Cells ; 31(3): 511-25, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23097336

RESUMEN

Daily, cells incur tens of thousands of DNA lesions caused by endogenous processes. Due to their long-lived nature, adult stem cells may be particularly susceptible to the negative impact of this constant genotoxic stress. Indeed, in murine models of DNA repair deficiencies, there is accumulation of DNA damage in hematopoietic stem cells and premature loss of function. Herein, we demonstrate that mice expressing reduced levels of ERCC1-XPF DNA repair endonuclease (Ercc1-/Δ mice) spontaneously display a progressive decline in the number and function of hematopoietic stem/progenitor cells (HSPCs). This was accompanied by increased cell death, expression of senescence markers, reactive oxygen species, and DNA damage in HSPC populations, illustrating cell autonomous mechanisms that contribute to loss of function. In addition, the bone marrow microenvironment of Ercc1-/Δ mice was not permissive for the engraftment of transplanted normal stem cells. Bones from Ercc1-/Δ mice displayed excessive osteoclastic activity, which alters the microenvironment in a way that is unfavorable to HSPC maintenance. This was accompanied by increased proinflammatory cytokines in the bone marrow of Ercc1-/Δ mice. These data provide novel evidence that spontaneous, endogenous DNA damage, if not repaired, promotes progressive attrition of adult stem cells via both cell autonomous and nonautonomous mechanisms.


Asunto(s)
Reparación del ADN , Células Madre Hematopoyéticas/fisiología , Animales , Procesos de Crecimiento Celular/fisiología , Microambiente Celular/genética , Daño del ADN , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Endonucleasas/deficiencia , Endonucleasas/genética , Endonucleasas/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Mutación
2.
Hepatology ; 55(2): 609-21, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21953681

RESUMEN

UNLABELLED: The liver changes with age, leading to an impaired ability to respond to hepatic insults and increased incidence of liver disease in the elderly. Therefore, there is critical need for rapid model systems to study aging-related liver changes. One potential opportunity is murine models of human progerias or diseases of accelerated aging. Ercc1(-/Δ) mice model a rare human progeroid syndrome caused by inherited defects in DNA repair. To determine whether hepatic changes that occur with normal aging occur prematurely in Ercc1(-/Δ) mice, we systematically compared liver from 5-month-old progeroid Ercc1(-/Δ) mice to old (24-36-month-old) wild-type (WT) mice. Both displayed areas of necrosis, foci of hepatocellular degeneration, and acute inflammation. Loss of hepatic architecture, fibrosis, steatosis, pseudocapillarization, and anisokaryosis were more dramatic in Ercc1(-/Δ) mice than in old WT mice. Liver enzymes were significantly elevated in serum of Ercc1(-/Δ) mice and old WT mice, whereas albumin was reduced, demonstrating liver damage and dysfunction. The regenerative capacity of Ercc1(-/Δ) liver after partial hepatectomy was significantly reduced. There was evidence of increased oxidative damage in Ercc1(-/Δ) and old WT liver, including lipofuscin, lipid hydroperoxides and acrolein, as well as increased hepatocellular senescence. There was a highly significant correlation in genome-wide transcriptional changes between old WT and 16-week-old, but not 5-week-old, Ercc1(-/Δ) mice, emphasizing that the Ercc1(-/Δ) mice acquire an aging profile in early adulthood. CONCLUSION: There are strong functional, regulatory, and histopathological parallels between accelerated aging driven by a DNA repair defect and normal aging. This supports a role for DNA damage in driving aging and validates a murine model for rapidly testing hypotheses about causes and treatment for aging-related hepatic changes.


Asunto(s)
Envejecimiento/fisiología , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Endonucleasas/genética , Hígado/fisiopatología , Progeria/fisiopatología , Envejecimiento/patología , Animales , Senescencia Celular , Reparación del ADN , Perfilación de la Expresión Génica , Hígado/metabolismo , Hígado/patología , Ratones , Estrés Oxidativo , Progeria/genética , Progeria/metabolismo , Progeria/patología
3.
Nature ; 444(7122): 1038-43, 2006 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-17183314

RESUMEN

XPF-ERCC1 endonuclease is required for repair of helix-distorting DNA lesions and cytotoxic DNA interstrand crosslinks. Mild mutations in XPF cause the cancer-prone syndrome xeroderma pigmentosum. A patient presented with a severe XPF mutation leading to profound crosslink sensitivity and dramatic progeroid symptoms. It is not known how unrepaired DNA damage accelerates ageing or its relevance to natural ageing. Here we show a highly significant correlation between the liver transcriptome of old mice and a mouse model of this progeroid syndrome. Expression data from XPF-ERCC1-deficient mice indicate increased cell death and anti-oxidant defences, a shift towards anabolism and reduced growth hormone/insulin-like growth factor 1 (IGF1) signalling, a known regulator of lifespan. Similar changes are seen in wild-type mice in response to chronic genotoxic stress, caloric restriction, or with ageing. We conclude that unrepaired cytotoxic DNA damage induces a highly conserved metabolic response mediated by the IGF1/insulin pathway, which re-allocates resources from growth to somatic preservation and life extension. This highlights a causal contribution of DNA damage to ageing and demonstrates that ageing and end-of-life fitness are determined both by stochastic damage, which is the cause of functional decline, and genetics, which determines the rates of damage accumulation and decline.


Asunto(s)
Daño del ADN , Progeria/genética , Progeria/fisiopatología , Somatotrofos/metabolismo , Envejecimiento/genética , Envejecimiento/fisiología , Animales , Línea Celular , Reparación del ADN , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Endonucleasas/deficiencia , Endonucleasas/genética , Regulación de la Expresión Génica , Hormona del Crecimiento/antagonistas & inhibidores , Hormona del Crecimiento/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/metabolismo , Hígado/metabolismo , Ratones , Síndrome
4.
PLoS Genet ; 4(8): e1000161, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18704162

RESUMEN

Mutant dwarf and calorie-restricted mice benefit from healthy aging and unusually long lifespan. In contrast, mouse models for DNA repair-deficient progeroid syndromes age and die prematurely. To identify mechanisms that regulate mammalian longevity, we quantified the parallels between the genome-wide liver expression profiles of mice with those two extremes of lifespan. Contrary to expectation, we find significant, genome-wide expression associations between the progeroid and long-lived mice. Subsequent analysis of significantly over-represented biological processes revealed suppression of the endocrine and energy pathways with increased stress responses in both delayed and premature aging. To test the relevance of these processes in natural aging, we compared the transcriptomes of liver, lung, kidney, and spleen over the entire murine adult lifespan and subsequently confirmed these findings on an independent aging cohort. The majority of genes showed similar expression changes in all four organs, indicating a systemic transcriptional response with aging. This systemic response included the same biological processes that are triggered in progeroid and long-lived mice. However, on a genome-wide scale, transcriptomes of naturally aged mice showed a strong association to progeroid but not to long-lived mice. Thus, endocrine and metabolic changes are indicative of "survival" responses to genotoxic stress or starvation, whereas genome-wide associations in gene expression with natural aging are indicative of biological age, which may thus delineate pro- and anti-aging effects of treatments aimed at health-span extension.


Asunto(s)
Envejecimiento Prematuro/genética , Longevidad/genética , Progeria/genética , Envejecimiento Prematuro/metabolismo , Animales , Restricción Calórica , Enanismo/genética , Perfilación de la Expresión Génica , Humanos , Riñón/fisiopatología , Hígado/fisiopatología , Pulmón/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Progeria/metabolismo , Bazo/fisiopatología
5.
Cell Metab ; 18(3): 403-15, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-24011075

RESUMEN

Lipodystrophies represent a group of heterogeneous disorders characterized by loss of fat tissue. However, the underlying mechanisms remain poorly understood. Using mice carrying an ERCC1-XPF DNA repair defect systematically or in adipocytes, we show that DNA damage signaling triggers a chronic autoinflammatory response leading to fat depletion. Ercc1-/- and aP2-Ercc1F/- fat depots show extensive gene expression similarities to lipodystrophic Pparγ(ldi/+) animals, focal areas of ruptured basement membrane, the reappearance of primary cilia, necrosis, fibrosis, and a marked decrease in adiposity. We find that persistent DNA damage in aP2-Ercc1F/- fat depots and in adipocytes ex vivo triggers the induction of proinflammatory factors by promoting transcriptionally active histone marks and the dissociation of nuclear receptor corepressor complexes from promoters; the response is cell autonomous and requires ataxia telangiectasia mutated (ATM). Thus, persistent DNA damage-driven autoinflammation plays a causative role in adipose tissue degeneration, with important ramifications for progressive lipodystrophies and natural aging.


Asunto(s)
Tejido Adiposo/metabolismo , Daño del ADN , Adipocitos/citología , Adipocitos/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Células Cultivadas , Citocinas/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Endonucleasas/deficiencia , Endonucleasas/genética , Endonucleasas/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Histonas/metabolismo , Ratones , Ratones Noqueados , PPAR gamma/genética , PPAR gamma/metabolismo , Progeria/metabolismo , Progeria/patología , Recombinasa Rad51/metabolismo , Transcriptoma
6.
PLoS One ; 7(11): e48560, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23185265

RESUMEN

Heightened production of collagen and other matrix proteins underlies the fibrotic phenotype of systemic sclerosis (SSc). Roscovitine is an inhibitor of cyclin-dependent kinases that promote cell cycling (CDK1, 2), neuronal development (CDK5) and control transcription (CDK7,9). In an in vivo glomerulonephritis model, roscovitine treatment decreased mesangial cell proliferation and matrix proteins [1]. We investigated whether roscovitine could regulate fibrotic protein production directly rather than through cell cycling. Our investigations revealed that roscovitine coordinately inhibited the expression of collagen, fibronectin, and connective tissue growth factor (CTGF) in normal and SSc fibroblasts. This effect occurred on a transcriptional basis and did not result from roscovitine-mediated cell cycle inhibition. Roscovitine-mediated suppression of matrix proteins could not be reversed by the exogenous profibrotic cytokines TGF-ß or IL-6. To our knowledge, we are the first to report that roscovitine modulates matrix protein transcription. Roscovitine may thus be a viable treatment option for SSc and other fibrosing diseases.


Asunto(s)
Fibroblastos/patología , Purinas/uso terapéutico , Esclerodermia Sistémica/tratamiento farmacológico , Esclerodermia Sistémica/patología , Animales , Colágeno/genética , Colágeno/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Fibroblastos/efectos de los fármacos , Fibronectinas/genética , Fibronectinas/metabolismo , Fibrosis , Humanos , Interleucina-6/biosíntesis , Ratones , Células 3T3 NIH , Fosforilación/efectos de los fármacos , Roscovitina , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba/efectos de los fármacos
7.
DNA Repair (Amst) ; 10(7): 781-91, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21612988

RESUMEN

ERCC1-XPF is a structure-specific endonuclease required for nucleotide excision repair, interstrand crosslink repair, and the repair of some double-strand breaks. Mutations in ERCC1 or XPF cause xeroderma pigmentosum, XFE progeroid syndrome or cerebro-oculo-facio-skeletal syndrome, characterized by increased risk of cancer, accelerated aging and severe developmental abnormalities, respectively. This review provides a comprehensive overview of the health impact of ERCC1-XPF deficiency, based on these rare diseases and mouse models of them. This offers an understanding of the tremendous health impact of DNA damage derived from environmental and endogenous sources.


Asunto(s)
Trastornos por Deficiencias en la Reparación del ADN/genética , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Endonucleasas/metabolismo , Animales , ADN/genética , ADN/metabolismo , ADN/efectos de la radiación , Daño del ADN , Trastornos por Deficiencias en la Reparación del ADN/metabolismo , Trastornos por Deficiencias en la Reparación del ADN/fisiopatología , Proteínas de Unión al ADN/genética , Endonucleasas/genética , Regulación de la Expresión Génica , Genotipo , Humanos , Ratones , Ratones Noqueados , Mutación , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/fisiopatología , Enfermedades Raras/genética , Enfermedades Raras/metabolismo , Enfermedades Raras/fisiopatología , Telómero/genética , Telómero/metabolismo , Rayos Ultravioleta
8.
Mech Ageing Dev ; 132(8-9): 437-42, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21596054

RESUMEN

Peripheral neuropathy is a common aging-related degenerative disorder that interferes with daily activities and leads to increased risk of falls and injury in the elderly. The etiology of most aging-related peripheral neuropathy is unknown. Inherited defects in several genome maintenance mechanisms cause tissue-specific accelerated aging, including neurodegeneration. We tested the hypothesis that a murine model of XFE progeroid syndrome, caused by reduced expression of ERCC1-XPF DNA repair endonuclease, develops peripheral neuropathy. Nerve conduction studies revealed normal nerve function in young adult (8 week) Ercc1(-/Δ) mice, but significant abnormalities in 20 week-old animals. Morphologic and ultrastructural analysis of the sciatic nerve from mutant mice revealed significant alterations at 20 but not 8 weeks of age. We conclude that Ercc1(-/Δ) mice have accelerated spontaneous peripheral neurodegeneration that mimics aging-related disease. This provides strong evidence that DNA damage can drive peripheral neuropathy and offers a rapid and novel model to test therapies.


Asunto(s)
Envejecimiento/metabolismo , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Endonucleasas/metabolismo , Enfermedades del Sistema Nervioso Periférico/enzimología , Progeria/enzimología , Envejecimiento/genética , Envejecimiento/patología , Animales , Proteínas de Unión al ADN/genética , Endonucleasas/genética , Masculino , Ratones , Ratones Noqueados , Enfermedades del Sistema Nervioso Periférico/genética , Enfermedades del Sistema Nervioso Periférico/patología , Progeria/genética , Progeria/patología
9.
Mol Cell Biol ; 28(16): 5082-92, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18541667

RESUMEN

ERCC1-XPF endonuclease is required for nucleotide excision repair (NER) of helix-distorting DNA lesions. However, mutations in ERCC1 or XPF in humans or mice cause a more severe phenotype than absence of NER, prompting a search for novel repair activities of the nuclease. In Saccharomyces cerevisiae, orthologs of ERCC1-XPF (Rad10-Rad1) participate in the repair of double-strand breaks (DSBs). Rad10-Rad1 contributes to two error-prone DSB repair pathways: microhomology-mediated end joining (a Ku86-independent mechanism) and single-strand annealing. To determine if ERCC1-XPF participates in DSB repair in mammals, mutant cells and mice were screened for sensitivity to gamma irradiation. ERCC1-XPF-deficient fibroblasts were hypersensitive to gamma irradiation, and gammaH2AX foci, a marker of DSBs, persisted in irradiated mutant cells, consistent with a defect in DSB repair. Mutant mice were also hypersensitive to irradiation, establishing an essential role for ERCC1-XPF in protecting against DSBs in vivo. Mice defective in both ERCC1-XPF and Ku86 were not viable. However, Ercc1(-/-) Ku86(-/-) fibroblasts were hypersensitive to gamma irradiation compared to single mutants and accumulated significantly greater chromosomal aberrations. Finally, in vitro repair of DSBs with 3' overhangs led to large deletions in the absence of ERCC1-XPF. These data support the conclusion that, as in yeast, ERCC1-XPF facilitates DSB repair via an end-joining mechanism that is Ku86 independent.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Endonucleasas/metabolismo , Animales , Antígenos Nucleares/metabolismo , Línea Celular Transformada , Supervivencia Celular/efectos de la radiación , Senescencia Celular/efectos de la radiación , Aberraciones Cromosómicas/efectos de la radiación , Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN/efectos de la radiación , Proteínas de Unión al ADN/deficiencia , Pérdida del Embrión/metabolismo , Embrión de Mamíferos/citología , Endonucleasas/deficiencia , Fibroblastos/enzimología , Fibroblastos/efectos de la radiación , Inestabilidad Genómica/efectos de la radiación , Células HeLa , Histonas/metabolismo , Humanos , Autoantígeno Ku , Ratones , Plásmidos/genética , Radiación Ionizante , Análisis de Secuencia de ADN
10.
Am J Hum Genet ; 80(3): 457-66, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17273966

RESUMEN

Nucleotide excision repair (NER) is a genome caretaker mechanism responsible for removing helix-distorting DNA lesions, most notably ultraviolet photodimers. Inherited defects in NER result in profound photosensitivity and the cancer-prone syndrome xeroderma pigmentosum (XP) or two progeroid syndromes: Cockayne and trichothiodystrophy syndromes. The heterodimer ERCC1-XPF is one of two endonucleases required for NER. Mutations in XPF are associated with mild XP and rarely with progeria. Mutations in ERCC1 have not been reported. Here, we describe the first case of human inherited ERCC1 deficiency. Patient cells showed moderate hypersensitivity to ultraviolet rays and mitomycin C, yet the clinical features were very severe and, unexpectedly, were compatible with a diagnosis of cerebro-oculo-facio-skeletal syndrome. This discovery represents a novel complementation group of patients with defective NER. Further, the clinical severity, coupled with a relatively mild repair defect, suggests novel functions for ERCC1.


Asunto(s)
Encéfalo/anomalías , Anomalías Craneofaciales/genética , Reparación del ADN/genética , Proteínas de Unión al ADN/deficiencia , Endonucleasas/deficiencia , Anomalías del Ojo/genética , Anomalías Múltiples/genética , Animales , Células Cultivadas , Proteínas de Unión al ADN/genética , Discapacidades del Desarrollo/genética , Endonucleasas/genética , Resultado Fatal , Femenino , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/efectos de la radiación , Genotipo , Humanos , Lactante , Recién Nacido , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Piel/citología , Síndrome
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA