Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
EMBO Rep ; 22(5): e51851, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33932076

RESUMEN

Defects in DNA single-strand break repair (SSBR) are linked with neurological dysfunction but the underlying mechanisms remain poorly understood. Here, we show that hyperactivity of the DNA strand break sensor protein Parp1 in mice in which the central SSBR protein Xrcc1 is conditionally deleted (Xrcc1Nes-Cre ) results in lethal seizures and shortened lifespan. Using electrophysiological recording and synaptic imaging approaches, we demonstrate that aberrant Parp1 activation triggers seizure-like activity in Xrcc1-defective hippocampus ex vivo and deregulated presynaptic calcium signalling in isolated hippocampal neurons in vitro. Moreover, we show that these defects are prevented by Parp1 inhibition or deletion and, in the case of Parp1 deletion, that the lifespan of Xrcc1Nes-Cre mice is greatly extended. This is the first demonstration that lethal seizures can be triggered by aberrant Parp1 activity at unrepaired SSBs, highlighting PARP inhibition as a possible therapeutic approach in hereditary neurological disease.


Asunto(s)
Calcio , Proteínas de Unión al ADN , Animales , ADN , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ratones , Neuronas/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Convulsiones/genética
2.
Nature ; 541(7635): 87-91, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-28002403

RESUMEN

XRCC1 is a molecular scaffold protein that assembles multi-protein complexes involved in DNA single-strand break repair. Here we show that biallelic mutations in the human XRCC1 gene are associated with ocular motor apraxia, axonal neuropathy, and progressive cerebellar ataxia. Cells from a patient with mutations in XRCC1 exhibited not only reduced rates of single-strand break repair but also elevated levels of protein ADP-ribosylation. This latter phenotype is recapitulated in a related syndrome caused by mutations in the XRCC1 partner protein PNKP and implicates hyperactivation of poly(ADP-ribose) polymerase/s as a cause of cerebellar ataxia. Indeed, remarkably, genetic deletion of Parp1 rescued normal cerebellar ADP-ribose levels and reduced the loss of cerebellar neurons and ataxia in Xrcc1-defective mice, identifying a molecular mechanism by which endogenous single-strand breaks trigger neuropathology. Collectively, these data establish the importance of XRCC1 protein complexes for normal neurological function and identify PARP1 as a therapeutic target in DNA strand break repair-defective disease.


Asunto(s)
Ataxia Cerebelosa/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Mutación , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Adenosina Difosfato Ribosa/metabolismo , Alelos , Animales , Apraxias/congénito , Apraxias/genética , Ataxia/genética , Axones/patología , Ataxia Cerebelosa/patología , Cerebelo/metabolismo , Cerebelo/patología , Cromatina/metabolismo , Síndrome de Cogan/genética , Roturas del ADN de Cadena Simple , Reparación del ADN/genética , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/deficiencia , Femenino , Humanos , Interneuronas/metabolismo , Interneuronas/patología , Masculino , Ratones , Linaje , Fenotipo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/deficiencia , Poli(ADP-Ribosa) Polimerasa-1/genética , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X
3.
PLoS Biol ; 15(5): e2001264, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28489848

RESUMEN

Stem and differentiated cells frequently differ in their response to DNA damage, which can determine tissue sensitivity. By exploiting insight into the spatial arrangement of subdomains within the adult neural subventricular zone (SVZ) in vivo, we show distinct responses to ionising radiation (IR) between neural stem and progenitor cells. Further, we reveal different DNA damage responses between neonatal and adult neural stem cells (NSCs). Neural progenitors (transit amplifying cells and neuroblasts) but not NSCs (quiescent and activated) undergo apoptosis after 2 Gy IR. This response is cell type- rather than proliferation-dependent and does not appear to be driven by distinctions in DNA damage induction or repair capacity. Moreover, exposure to 2 Gy IR promotes proliferation arrest and differentiation in the adult SVZ. These 3 responses are ataxia telangiectasia mutated (ATM)-dependent and promote quiescent NSC (qNSC) activation, which does not occur in the subdomains that lack progenitors. Neuroblasts arising post-IR derive from activated qNSCs rather than irradiated progenitors, minimising damage compounded by replication or mitosis. We propose that rather than conferring sensitive cell death, apoptosis is a form of rapid cell death that serves to remove damaged progenitors and promote qNSC activation. Significantly, analysis of the neonatal (P5) SVZ reveals that although progenitors remain sensitive to apoptosis, they fail to efficiently arrest proliferation. Consequently, their repopulation occurs rapidly from irradiated progenitors rather than via qNSC activation.


Asunto(s)
Apoptosis , Daño del ADN , Ventrículos Laterales/efectos de la radiación , Células-Madre Neurales/efectos de la radiación , Animales , Animales Recién Nacidos , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Diferenciación Celular , Proliferación Celular/efectos de la radiación , Ratones Endogámicos C57BL , Rayos X
4.
Mol Cell ; 41(1): 33-45, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21211721

RESUMEN

PARP-3 is a member of the ADP-ribosyl transferase superfamily of unknown function. We show that PARP-3 is stimulated by DNA double-strand breaks (DSBs) in vitro and functions in the same pathway as the poly (ADP-ribose)-binding protein APLF to accelerate chromosomal DNA DSB repair. We implicate PARP-3 in the accumulation of APLF at DSBs and demonstrate that APLF promotes the retention of XRCC4/DNA ligase IV complex in chromatin, suggesting that PARP-3 and APLF accelerate DNA ligation during nonhomologous end-joining (NHEJ). Consistent with this, we show that class switch recombination in Aplf(-/-) B cells is biased toward microhomology-mediated end-joining, a pathway that operates in the absence of XRCC4/DNA ligase IV, and that the requirement for PARP-3 and APLF for NHEJ is circumvented by overexpression of XRCC4/DNA ligase IV. These data identify molecular roles for PARP-3 and APLF in chromosomal DNA double-strand break repair reactions.


Asunto(s)
Proteínas Portadoras/fisiología , Proteínas de Ciclo Celular/fisiología , Fosfoproteínas/fisiología , Poli(ADP-Ribosa) Polimerasas/fisiología , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Roturas del ADN de Doble Cadena , Reparación del ADN/fisiología , ADN-(Sitio Apurínico o Apirimidínico) Liasa , Eliminación de Gen , Humanos , Ratones , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa , Proteínas Recombinantes de Fusión/fisiología
5.
Cereb Cortex ; 27(1): 11-23, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28365778

RESUMEN

In mitotic cells, the cyclin-dependent kinase (CDK) subunit protein CKS1 regulates S phase entry by mediating degradation of the CDK inhibitor p27. Although mature neurons lack mitotic CDKs, we found that CKS1 was actively expressed in post-mitotic neurons of the adult hippocampus. Interestingly, Cks1 knockout (Cks1-/-) mice exhibited poor long-term memory, and diminished maintenance of long-term potentiation in the hippocampal circuits. Furthermore, there was neuronal accumulation of cofilin-actin rods or cofilin aggregates, which are associated with defective dendritic spine maturation and synaptic loss. We further demonstrated that it was the increased p27 level that activated cofilin by suppressing the RhoA kinase-mediated inhibitory phosphorylation of cofilin, resulting in the formation of cofilin aggregates in the Cks1-/- neuronal cells. Consistent with reports that the peptidyl-prolyl-isomerase PIN1 competes with CKS1 for p27 binding, we found that inhibition of PIN1 diminished the formation of cofilin aggregates through decreasing p27 levels, thereby activating RhoA and increasing cofilin phosphorylation. Our results revealed that CKS1 is involved in normal glutamatergic synapse development and dendritic spine maturation in adult hippocampus through modulating p27 stability.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Quinasas CDC2-CDC28/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Hipocampo/metabolismo , Trastornos de la Memoria/metabolismo , Memoria a Largo Plazo , Neuronas/metabolismo , Animales , Quinasas CDC2-CDC28/genética , Ciclo Celular , Espinas Dendríticas , Hipocampo/patología , Potenciación a Largo Plazo , Masculino , Trastornos de la Memoria/patología , Ratones , Ratones Noqueados , Agregado de Proteínas , Aprendizaje Espacial
6.
J Cell Sci ; 128(19): 3597-606, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26303202

RESUMEN

The embryonic neural stem cell compartment is characterised by rapid proliferation from embryonic day (E)11 to E16.5, high endogenous DNA double-strand break (DSB) formation and sensitive activation of apoptosis. Here, we ask whether DSBs arise in the adult neural stem cell compartments, the sub-ventricular zone (SVZ) of the lateral ventricles and the sub-granular zone (SGZ) of the hippocampal dentate gyrus, and whether they activate apoptosis. We used mice with a hypomorphic mutation in DNA ligase IV (Lig4(Y288C)), ataxia telangiectasia mutated (Atm(-/-)) and double mutant Atm(-/-)/Lig4(Y288C) mice. We demonstrate that, although DSBs do not arise at a high frequency in adult neural stem cells, the low numbers of DSBs that persist endogenously in Lig4(Y288C) mice or that are induced by low radiation doses can activate apoptosis. A temporal analysis shows that DSB levels in Lig4(Y288C) mice diminish gradually from the embryo to a steady state level in adult mice. The neonatal SVZ compartment of Lig4(Y288C) mice harbours diminished DSBs compared to its differentiated counterpart, suggesting a process selecting against unfit stem cells. Finally, we reveal high endogenous apoptosis in the developing SVZ of wild-type newborn mice.


Asunto(s)
Apoptosis/efectos de la radiación , Roturas del ADN de Doble Cadena/efectos de la radiación , Células-Madre Neurales/efectos de la radiación , Rayos X , Animales , Apoptosis/genética , Células Cultivadas , Femenino , Etiquetado Corte-Fin in Situ , Masculino , Ratones
7.
PLoS Genet ; 9(3): e1003226, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23505375

RESUMEN

Anticancer topoisomerase "poisons" exploit the break-and-rejoining mechanism of topoisomerase II (TOP2) to generate TOP2-linked DNA double-strand breaks (DSBs). This characteristic underlies the clinical efficacy of TOP2 poisons, but is also implicated in chromosomal translocations and genome instability associated with secondary, treatment-related, haematological malignancy. Despite this relevance for cancer therapy, the mechanistic aspects governing repair of TOP2-induced DSBs and the physiological consequences that absent or aberrant repair can have are still poorly understood. To address these deficits, we employed cells and mice lacking tyrosyl DNA phosphodiesterase 2 (TDP2), an enzyme that hydrolyses 5'-phosphotyrosyl bonds at TOP2-associated DSBs, and studied their response to TOP2 poisons. Our results demonstrate that TDP2 functions in non-homologous end-joining (NHEJ) and liberates DSB termini that are competent for ligation. Moreover, we show that the absence of TDP2 in cells impairs not only the capacity to repair TOP2-induced DSBs but also the accuracy of the process, thus compromising genome integrity. Most importantly, we find this TDP2-dependent NHEJ mechanism to be physiologically relevant, as Tdp2-deleted mice are sensitive to TOP2-induced damage, displaying marked lymphoid toxicity, severe intestinal damage, and increased genome instability in the bone marrow. Collectively, our data reveal TDP2-mediated error-free NHEJ as an efficient and accurate mechanism to repair TOP2-induced DSBs. Given the widespread use of TOP2 poisons in cancer chemotherapy, this raises the possibility of TDP2 being an important etiological factor in the response of tumours to this type of agent and in the development of treatment-related malignancy.


Asunto(s)
Roturas del ADN de Doble Cadena , ADN-Topoisomerasas de Tipo II , Inestabilidad Genómica , Hidrolasas Diéster Fosfóricas , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral , Animales , Daño del ADN/genética , Reparación del ADN por Unión de Extremidades/genética , Reparación del ADN/genética , ADN-Topoisomerasas de Tipo II/genética , ADN-Topoisomerasas de Tipo II/metabolismo , ADN-Topoisomerasas de Tipo II/uso terapéutico , Proteínas de Unión al ADN , Ratones , Hidrolasas Diéster Fosfóricas/deficiencia , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo , Recombinación Genética , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/deficiencia , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo
8.
Nucleic Acids Res ; 40(17): 8371-80, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22740648

RESUMEN

The abortive activity of topoisomerases can result in clastogenic and/or lethal DNA damage in which the topoisomerase is covalently linked to the 3'- or 5'-terminus of a DNA strand break. This type of DNA damage is implicated in chromosome translocations and neurological disease and underlies the clinical efficacy of an important class of anticancer topoisomerase 'poisons'. Tyrosyl DNA phosphodiesterase-1 protects cells from abortive topoisomerase I (Top1) activity by hydrolyzing the 3'-phosphotyrosyl bond that links Top1 to a DNA strand break and is currently the only known human enzyme that displays this activity in cells. Recently, we identified a second tyrosyl DNA phosphodiesterase (TDP2; aka TTRAP/EAPII) that possesses weak 3'-tyrosyl DNA phosphodiesterase (3'-TDP) activity, in vitro. Herein, we have examined whether TDP2 contributes to the repair of Top1-mediated DNA breaks by deleting Tdp1 and Tdp2 separately and together in murine and avian cells. We show that while deletion of Tdp1 in wild-type DT40 cells and mouse embryonic fibroblasts decreases DNA strand break repair rates and cellular survival in response to Top1-induced DNA damage, deletion of Tdp2 does not. However, deletion of both Tdp1 and Tdp2 reduces rates of DNA strand break repair and cell survival below that observed in Tdp1-/- cells, suggesting that Tdp2 contributes to cellular 3'-TDP activity in the absence of Tdp1. Consistent with this idea, over-expression of human TDP2 in Tdp1-/-/Tdp2-/-/- DT40 cells increases DNA strand break repair rates and cell survival above that observed in Tdp1-/- DT40 cells, suggesting that Tdp2 over-expression can partially complement the defect imposed by loss of Tdp1. Finally, mice lacking both Tdp1 and Tdp2 exhibit greater sensitivity to Top1 poisons than do mice lacking Tdp1 alone, further suggesting that Tdp2 contributes to the repair of Top1-mediated DNA damage in the absence of Tdp1. In contrast, we failed to detect a contribution for Tdp1 to repair Top2-mediated damage. Together, our data suggest that Tdp1 and Tdp2 fulfil overlapping roles following Top1-induced DNA damage, but not following Top2-induced DNA damage, in vivo.


Asunto(s)
Roturas del ADN , Reparación del ADN , ADN-Topoisomerasas de Tipo I/metabolismo , Hidrolasas Diéster Fosfóricas/fisiología , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/fisiología , Animales , Camptotecina/toxicidad , Células Cultivadas , Proteínas de Unión al ADN , Humanos , Ratones , Ratones Noqueados , Proteínas Nucleares/metabolismo , Hidrolasas Diéster Fosfóricas/genética , Inhibidores de Topoisomerasa I/toxicidad , Factores de Transcripción/metabolismo , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética
9.
J Neurosci ; 31(27): 10088-100, 2011 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-21734301

RESUMEN

The embryonic ventricular and subventricular zones (VZ/SVZ) contain the neuronal stem and progenitor cells and undergo rapid proliferation. The intermediate zone (IZ) contains nonreplicating, differentiated cells. The VZ/SVZ is hypersensitive to radiation-induced apoptosis. Ablation of DNA non-homologous end-joining (NHEJ) proteins, XRCC4 or DNA ligase IV (LigIV), confers ataxia telangiectasia mutated (ATM)-dependent apoptosis predominantly in the IZ. We examine the mechanistic basis underlying these distinct sensitivities using a viable LigIV (Lig4(Y288C)) mouse, which permits an examination of the DNA damage responses in the embryonic and adult brain. Via combined analysis of DNA breakage, apoptosis, and cell-cycle checkpoint control in tissues, we show that apoptosis in the VZ/SVZ and IZ is activated by low numbers of DNA double-strand breaks (DSBs). Unexpectedly, high sensitivity in the VZ/SVZ arises from sensitive activation of ATM-dependent apoptosis plus an ATM-independent process. In contrast, the IZ appears to be hypersensitive to persistent DSBs. NHEJ functions efficiently in both compartments. The VZ/SVZ and IZ regions incur high endogenous DNA breakage, which correlates with VZ proliferation. We demonstrate a functional G(2)/M checkpoint in VZ/SVZ cells and show that it is not activated by low numbers of DSBs, allowing damaged VZ/SVZ cells to transit into the IZ. We propose a novel model in which microcephaly in LIG4 syndrome arises from sensitive apoptotic induction from persisting DSBs in the IZ, which arise from high endogenous breakage in the VZ/SVZ and transit of damaged cells to the IZ. The VZ/SVZ, in contrast, is highly sensitive to acute radiation-induced DSB formation.


Asunto(s)
Ventrículos Cerebrales/citología , Ventrículos Cerebrales/embriología , ADN Ligasas/metabolismo , Desarrollo Embrionario , Neuronas/fisiología , Factores de Edad , Animales , Animales Recién Nacidos , Apoptosis/genética , Apoptosis/fisiología , Apoptosis/efectos de la radiación , Proteínas de la Ataxia Telangiectasia Mutada , Bromodesoxiuridina/metabolismo , Ciclo Celular/genética , Proteínas de Ciclo Celular , Proliferación Celular/efectos de la radiación , Ventrículos Cerebrales/efectos de la radiación , Proteínas Cromosómicas no Histona/metabolismo , Cisteína/genética , Roturas del ADN de Doble Cadena/efectos de la radiación , ADN Ligasa (ATP) , ADN Ligasas/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/efectos de la radiación , Histonas/metabolismo , Etiquetado Corte-Fin in Situ/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Mutación/genética , Células-Madre Neurales/fisiología , Células-Madre Neurales/efectos de la radiación , Neuronas/efectos de la radiación , Proteínas Serina-Treonina Quinasas/deficiencia , Radiación Ionizante , Proteínas de Dominio T Box/metabolismo , Tubulina (Proteína)/metabolismo , Proteínas Supresoras de Tumor/deficiencia , Proteína 1 de Unión al Supresor Tumoral P53 , Tirosina/genética
10.
EMBO J ; 26(22): 4720-31, 2007 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-17914460

RESUMEN

Defective Tyrosyl-DNA phosphodiesterase 1 (TDP1) can cause spinocerebellar ataxia with axonal neuropathy (SCAN1), a neurodegenerative syndrome associated with marked cerebellar atrophy and peripheral neuropathy. Although SCAN1 lymphoblastoid cells show pronounced defects in the repair of chromosomal single-strand breaks (SSBs), it is unknown if this DNA repair activity is important for neurons or for preventing neurodegeneration. Therefore, we generated Tdp1-/- mice to assess the role of Tdp1 in the nervous system. Using both in vitro and in vivo assays, we found that cerebellar neurons or primary astrocytes derived from Tdp1-/- mice display an inability to rapidly repair DNA SSBs associated with Top1-DNA complexes or oxidative damage. Moreover, loss of Tdp1 resulted in age-dependent and progressive cerebellar atrophy. Tdp1-/- mice treated with topotecan, a drug that increases levels of Top1-DNA complexes, also demonstrated significant loss of intestinal and hematopoietic progenitor cells. These data indicate that TDP1 is required for neural homeostasis, and reveal a widespread requisite for TDP1 function in response to acutely elevated levels of Top1-associated DNA strand breaks.


Asunto(s)
Astrocitos/metabolismo , Roturas del ADN de Cadena Simple , Reparación del ADN , Neuronas/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Animales , Antineoplásicos/farmacología , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Camptotecina/farmacología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Cerebelo/citología , Colesterol/sangre , ADN-Topoisomerasas de Tipo I/metabolismo , Femenino , Rayos gamma , Peróxido de Hidrógeno/farmacología , Intestinos/citología , Intestinos/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Hidrolasas Diéster Fosfóricas/deficiencia , Hidrolasas Diéster Fosfóricas/genética , Albúmina Sérica/análisis , Bazo/citología , Bazo/efectos de los fármacos , Bazo/crecimiento & desarrollo , Timo/citología , Timo/efectos de los fármacos , Timo/crecimiento & desarrollo , Topotecan/farmacología
11.
DNA Repair (Amst) ; 8(6): 760-6, 2009 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-19303373

RESUMEN

Ataxia oculomotor apraxia-1 (AOA1) is an autosomal recessive neurodegenerative disease that results from mutations of aprataxin (APTX). APTX associates with the DNA single- and double-strand break repair machinery and is able to remove AMP from 5'-termini at DNA strand breaks in vitro. However, attempts to establish a DNA strand break repair defect in APTX-defective cells have proved conflicting and unclear. We reasoned that this may reflect that DNA strand breaks with 5'-AMP represent only a minor subset of breaks induced in cells, and/or the availability of alternative mechanisms for removing AMP from 5'-termini. Here, we have attempted to increase the dependency of chromosomal single- and double-strand break repair on aprataxin activity by slowing the rate of repair of 3'-termini in aprataxin-defective neural cells, thereby increasing the likelihood that the 5'-termini at such breaks become adenylated and/or block alternative repair mechanisms. To do this, we generated a mouse model in which APTX is deleted together with tyrosyl DNA phosphodiesterase (TDP1), an enzyme that repairs 3'-termini at a subset of single-strand breaks (SSBs), including those with 3'-topoisomerase-1 (Top1) peptide. Notably, the global rate of repair of oxidative and alkylation-induced SSBs was significantly slower in Tdp1(-/-)/Aptx(-/-) double knockout quiescent mouse astrocytes compared with Tdp1(-/-) or Aptx(-/-) single knockouts. In contrast, camptothecin-induced Top1-SSBs accumulated to similar levels in Tdp1(-/-) and Tdp1(-/-)/Aptx(-/-) double knockout astrocytes. Finally, we failed to identify a measurable defect in double-strand break repair in Tdp1(-/-), Aptx(-/-) or Tdp1(-/-)/Aptx(-/-) astrocytes. These data provide direct evidence for a requirement for aprataxin during chromosomal single-strand break repair in primary neural cells lacking Tdp1.


Asunto(s)
Astrocitos/metabolismo , Roturas del ADN de Cadena Simple/efectos de los fármacos , Reparación del ADN/genética , Proteínas de Unión al ADN/fisiología , Neuronas/metabolismo , Proteínas Nucleares/fisiología , Hidrolasas Diéster Fosfóricas/fisiología , Animales , Antineoplásicos Fitogénicos/farmacología , Astrocitos/efectos de los fármacos , Camptotecina/farmacología , Células Cultivadas , ADN-Topoisomerasas de Tipo I/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos
12.
DNA Repair (Amst) ; 76: 70-75, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30822688

RESUMEN

Radiosusceptibility is the sensitivity of a biological organism to ionising radiation (IR)-induced carcinogenesis, an outcome of IR exposure relevant following low doses. The tissue response is strongly influenced by the DNA damage response (DDR) activated in stem and progenitor cells. We previously reported that in vivo exposure to 2 Gy X-rays activates apoptosis, proliferation arrest and premature differentiation in neural progenitor cells (transit amplifying cells and neuroblasts) but not in neural stem cells (NSCs) of the largest neurogenic region of the adult brain, the subventricular zone (SVZ). These responses promote adult quiescent NSC (qNSC) activation after 2 Gy. In contrast, neonatal (P5) SVZ neural progenitors continue proliferating and do not activate qNSCs. Significantly, the human and mouse neonatal brain is radiosusceptible. Here, we examine the response of stem and progenitor cells in the SVZ to low IR doses (50-500 mGy). We observe a linear dose-response for apoptosis but, in contrast, proliferation arrest and neuroblast differentiation require a threshold dose of 200 or 500 mGy, respectively. Importantly, qNSCs were not activated at doses below 500 mGy. Thus, full DDR activation in the neural stem cell compartment in vivo necessitates a threshold dose, which can be considered of significance when evaluating IR-induced cancer risk and dose extrapolation.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/efectos de la radiación , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de la radiación , Animales , Proliferación Celular/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Ratones
13.
DNA Repair (Amst) ; 3(11): 1493-502, 2004 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-15380105

RESUMEN

Ataxia-oculomotor apraxia 1 (AOA1) is an autosomal recessive neurodegenerative disease that is reminiscent of ataxia-telangiectasia (A-T). AOA1 is caused by mutations in the gene encoding aprataxin, a protein whose physiological function is currently unknown. We report here that, in contrast to A-T, AOA1 cell lines exhibit neither radioresistant DNA synthesis nor a reduced ability to phosphorylate downstream targets of ATM following DNA damage, suggesting that AOA1 lacks the cell cycle checkpoint defects that are characteristic of A-T. In addition, AOA1 primary fibroblasts exhibit only mild sensitivity to ionising radiation, hydrogen peroxide, and methyl methanesulphonate (MMS). Strikingly, however, aprataxin physically interacts in vitro and in vivo with the DNA strand break repair proteins XRCC1 and XRCC4. Aprataxin possesses a divergent forkhead associated (FHA) domain that closely resembles the FHA domain present in polynucleotide kinase, and appears to mediate the interactions with CK2-phosphorylated XRCC1 and XRCC4 through this domain. Aprataxin is therefore physically associated with both the DNA single-strand and double-strand break repair machinery, raising the possibility that AOA1 is a novel DNA damage response-defective disease.


Asunto(s)
Apraxias/genética , Apraxias/metabolismo , Ataxia/genética , Ataxia/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ataxia Telangiectasia/genética , Ataxia Telangiectasia/metabolismo , Línea Celular , Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN/química , Humanos , Técnicas In Vitro , Proteínas Nucleares/química , Enfermedades del Nervio Oculomotor/genética , Enfermedades del Nervio Oculomotor/metabolismo , Tolerancia a Radiación , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Técnicas del Sistema de Dos Híbridos , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X
14.
Biomed Environ Sci ; 16(2): 157-62, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12964789

RESUMEN

OBJECTIVE: To establish a cytologic expressing system of rat glutathione S-transferase pi (GST-pi) cDNA for detecting the resistance of HeLa cells to anticancer drugs. METHODS: The assessment was made with various anticancer drugs (adriamycin, mitomycin, cisplatinum and vincristine) that showed different cytotoxicities in transfectant HeLa cells with pSV-GT containing rat GST-pi cDNA (HeLa/pSV-GT) or control pSV-neo (HeLa/pSV-neo). Expression levels of GST-pi mRNA in HeLa/pSV-GT and HeLa/pSV-neo were measured by in situ hybridization using Digoxin-labelled cDNA probe. RESULTS: HeLa/pSV-GT expressed significantly high degree of GST-pi mRNA, whereas both HeLa/pSV-neo and HeLa cells had very low expression. Cytotoxicities of HeLa/pSV-GT and HeLa/pSV-neo with 4 anticancer drugs were measured by MTT assay. Drug concentrations for yielding 50% inhibition (IC50) in HeLa/pSV-GT by adriamycin, mitomycin and cisplatinum were 70.13 microg/mL, 10.95 microg/mL and 16.52 microg/mL, respectively. In contrast, IC50 in HeLa/pSV-neo was 10.34 microg/mL, 7.48 microg/mL and 13.70 microg/mL, respectively. The cytotoxicities of vincristine on both HeLa/pSV-GT and HeLa/pSV-neo were not significantly different. CONCLUSIONS: Our findings suggest that HeLa/pSV-GT containing rat GST-pi cDNA is resistant to some anticancer drugs due to overexpression of GST-pi. Also, HeLa/pSV-GT cell line could serve as a useful cytogenetic model for further research.


Asunto(s)
Antineoplásicos/farmacología , Glutatión Transferasa/biosíntesis , Glutatión Transferasa/farmacología , Transfección , Animales , ADN Complementario , Resistencia a Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Células HeLa , Humanos , Ratas
15.
DNA Repair (Amst) ; 12(5): 356-66, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23518413

RESUMEN

Smc5-6 is a highly conserved protein complex related to cohesin and condensin involved in the structural maintenance of chromosomes. In yeasts the Smc5-6 complex is essential for proliferation and is involved in DNA repair and homologous recombination. siRNA depletion of genes involved in the Smc5-6 complex in cultured mammalian cells results in sensitivity to some DNA damaging agents. In order to gain further insight into its role in mammals we have generated mice mutated in the Smc6 gene. A complete knockout resulted in early embryonic lethality, demonstrating that this gene is essential in mammals. However, mutation of the highly conserved serine-994 to alanine in the ATP hydrolysis motif in the SMC6 C-terminal domain, resulted in mice with a surprisingly mild phenotype. With the neo gene selection marker in the intron following the mutation, resulting in reduced expression of the SMC6 gene, the mice were reduced in size, but fertile and had normal lifespans. When the neo gene was removed, the mice had normal size, but detailed phenotypic analysis revealed minor abnormalities in glucose tolerance, haematopoiesis, nociception and global gene expression patterns. Embryonic fibroblasts derived from the ser994 mutant mice were not sensitive to killing by a range of DNA damaging agents, but they were sensitive to the induction of sister chromatid exchanges induced by ultraviolet light or mitomycin C. They also accumulated more oxidative damage than wild-type cells.


Asunto(s)
Adenosina Trifosfatasas/genética , Proteínas de Ciclo Celular/genética , Mutación Missense , Fenotipo , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Secuencias de Aminoácidos , Animales , Dominio Catalítico , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Fertilidad/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/efectos de la radiación , Genes Esenciales , Intolerancia a la Glucosa/genética , Hematopoyesis/genética , Hidrólisis , Intrones , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitomicina/farmacología , Nocicepción , Intercambio de Cromátides Hermanas/efectos de los fármacos , Intercambio de Cromátides Hermanas/efectos de la radiación , Rayos Ultravioleta
16.
J Biol Chem ; 280(9): 7452-9, 2005 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-15613485

RESUMEN

The T to C substitution at position -175 of the gamma-globin gene has been identified in some individuals with non-deletion hereditary persistence of fetal hemoglobin (HPFH). In this study, the HPFH phenotype was reestablished in transgenic mice carrying the mu'LCRAgamma(-175)psibetadeltabeta construct, which contained a 3.1-kb mu'LCR cassette linked to a 29-kb fragment from the Agamma-to beta-globin gene with the natural chromosome arrangement but with the -175 mutation, which provided evidence for this single mutation as the cause of this form of HPFH. The HPFH phenotype was also reproduced in transgenic mice carrying the mu'LCRAgamma(-173)psibetadeltabeta construct, in which the -175 T to C Agamma gene was substituted with the -173 T to C Agamma gene. In vitro experiments proved that the -175 mutation significantly reduced binding of Oct-1 but not GATA-1, whereas the -173 mutation dramatically decreased binding of GATA-1 but not Oct-1. These results suggest that abrogation of either GATA-1 or Oct-1 binding to this promoter region may result in the HPFH phenotype. An in vivo footprinting assay revealed that either the -175 mutation or the -173 mutation significantly decreased overall protein binding to this promoter region in adult erythrocytes of transgenic mice. We hypothesize that a multiprotein complex containing GATA-1, Oct-1, and other protein factors may contribute to the formation of a repressive chromatin structure that silences gamma-globin gene expression in normal adult erythrocytes. Both the -173 and -175 T to C substitutions may disrupt the complex assembly and result in the reactivation of the gamma-globin gene in adult erythrocytes.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Globinas/genética , Factores de Transcripción/biosíntesis , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Cromatina/genética , Inmunoprecipitación de Cromatina , Cósmidos/metabolismo , ADN/química , ADN/metabolismo , Proteínas de Unión al ADN/química , Eritrocitos/metabolismo , Factores de Unión al ADN Específico de las Células Eritroides , Factor de Transcripción GATA1 , Eliminación de Gen , Regulación de la Expresión Génica , Globinas/biosíntesis , Globinas/metabolismo , Células HeLa , Humanos , Células K562 , Ratones , Ratones Transgénicos , Modelos Genéticos , Datos de Secuencia Molecular , Mutación , Factor 1 de Transcripción de Unión a Octámeros , Fenotipo , Plásmidos/metabolismo , Mutación Puntual , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Unión Proteica , Conformación Proteica , ARN/metabolismo , ARN Mensajero/metabolismo , Factores de Tiempo , Factores de Transcripción/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA