Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 50(5): 2681-2699, 2022 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-35189637

RESUMEN

Cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) is activated in cells with defective DNA damage repair and signaling (DDR) factors, but a direct role for DDR factors in regulating cGAS activation in response to micronuclear DNA is still poorly understood. Here, we provide novel evidence that Nijmegen breakage syndrome 1 (NBS1) protein, a well-studied DNA double-strand break (DSB) sensor-in coordination with Ataxia Telangiectasia Mutated (ATM), a protein kinase, and Carboxy-terminal binding protein 1 interacting protein (CtIP), a DNA end resection factor-functions as an upstream regulator that prevents cGAS from binding micronuclear DNA. When NBS1 binds to micronuclear DNA via its fork-head-associated domain, it recruits CtIP and ATM via its N- and C-terminal domains, respectively. Subsequently, ATM stabilizes NBS1's interaction with micronuclear DNA, and CtIP converts DSB ends into single-strand DNA ends; these two key events prevent cGAS from binding micronuclear DNA. Additionally, by using a cGAS tripartite system, we show that cells lacking NBS1 not only recruit cGAS to a major fraction of micronuclear DNA but also activate cGAS in response to these micronuclear DNA. Collectively, our results underscore how NBS1 and its binding partners prevent cGAS from binding micronuclear DNA, in addition to their classical functions in DDR signaling.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas Supresoras de Tumor , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Ciclo Celular/metabolismo , ADN/genética , Daño del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Supresoras de Tumor/genética
2.
Proc Natl Acad Sci U S A ; 118(34)2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34417314

RESUMEN

The inability of adult mammalian cardiomyocytes to proliferate underpins the development of heart failure following myocardial injury. Although the newborn mammalian heart can spontaneously regenerate for a short period of time after birth, this ability is lost within the first week after birth in mice, partly due to increased mitochondrial reactive oxygen species (ROS) production which results in oxidative DNA damage and activation of DNA damage response. This increase in ROS levels coincides with a postnatal switch from anaerobic glycolysis to fatty acid (FA) oxidation by cardiac mitochondria. However, to date, a direct link between mitochondrial substrate utilization and oxidative DNA damage is lacking. Here, we generated ROS-sensitive fluorescent sensors targeted to different subnuclear compartments (chromatin, heterochromatin, telomeres, and nuclear lamin) in neonatal rat ventricular cardiomyocytes, which allowed us to determine the spatial localization of ROS in cardiomyocyte nuclei upon manipulation of mitochondrial respiration. Our results demonstrate that FA utilization by the mitochondria induces a significant increase in ROS detection at the chromatin level compared to other nuclear compartments. These results indicate that mitochondrial metabolic perturbations directly alter the nuclear redox status and that the chromatin appears to be particularly sensitive to the prooxidant effect of FA utilization by the mitochondria.


Asunto(s)
Ácidos Grasos/metabolismo , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Línea Celular , Proliferación Celular , Daño del ADN , Ratones , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo
3.
Nature ; 541(7636): 222-227, 2017 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-27798600

RESUMEN

The adult mammalian heart is incapable of regeneration following cardiomyocyte loss, which underpins the lasting and severe effects of cardiomyopathy. Recently, it has become clear that the mammalian heart is not a post-mitotic organ. For example, the neonatal heart is capable of regenerating lost myocardium, and the adult heart is capable of modest self-renewal. In both of these scenarios, cardiomyocyte renewal occurs via the proliferation of pre-existing cardiomyocytes, and is regulated by aerobic-respiration-mediated oxidative DNA damage. Therefore, we reasoned that inhibiting aerobic respiration by inducing systemic hypoxaemia would alleviate oxidative DNA damage, thereby inducing cardiomyocyte proliferation in adult mammals. Here we report that, in mice, gradual exposure to severe systemic hypoxaemia, in which inspired oxygen is gradually decreased by 1% and maintained at 7% for 2 weeks, results in inhibition of oxidative metabolism, decreased reactive oxygen species production and oxidative DNA damage, and reactivation of cardiomyocyte mitosis. Notably, we find that exposure to hypoxaemia 1 week after induction of myocardial infarction induces a robust regenerative response with decreased myocardial fibrosis and improvement of left ventricular systolic function. Genetic fate-mapping analysis confirms that the newly formed myocardium is derived from pre-existing cardiomyocytes. These results demonstrate that the endogenous regenerative properties of the adult mammalian heart can be reactivated by exposure to gradual systemic hypoxaemia, and highlight the potential therapeutic role of hypoxia in regenerative medicine.


Asunto(s)
Corazón/crecimiento & desarrollo , Hipoxia/metabolismo , Miocardio/citología , Miocardio/metabolismo , Regeneración , Medicina Regenerativa/métodos , Animales , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Proliferación Celular , Respiración de la Célula , Daño del ADN , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Mitosis , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Función Ventricular Izquierda
4.
J Biol Chem ; 295(32): 11144-11160, 2020 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-32540968

RESUMEN

Defective DNA damage response (DDR) signaling is a common mechanism that initiates and maintains the cellular senescence phenotype. Dysfunctional telomeres activate DDR signaling, genomic instability, and cellular senescence, but the links among these events remains unclear. Here, using an array of biochemical and imaging techniques, including a highly regulatable CRISPR/Cas9 strategy to induce DNA double strand breaks specifically in the telomeres, ChIP, telomere immunofluorescence, fluorescence in situ hybridization (FISH), micronuclei imaging, and the telomere shortest length assay (TeSLA), we show that chromosome mis-segregation due to imperfect DDR signaling in response to dysfunctional telomeres creates a preponderance of chromatin fragments in the cytosol, which leads to a premature senescence phenotype. We found that this phenomenon is caused not by telomere shortening, but by cyclic GMP-AMP synthase (cGAS) recognizing cytosolic chromatin fragments and then activating the stimulator of interferon genes (STING) cytosolic DNA-sensing pathway and downstream interferon signaling. Significantly, genetic and pharmacological manipulation of cGAS not only attenuated immune signaling, but also prevented premature cellular senescence in response to dysfunctional telomeres. The findings of our study uncover a cellular intrinsic mechanism involving the cGAS-mediated cytosolic self-DNA-sensing pathway that initiates premature senescence independently of telomere shortening.


Asunto(s)
Senescencia Celular/genética , Ligasas/metabolismo , Nucleótidos Cíclicos/metabolismo , Telómero , Ciclo Celular , Roturas del ADN de Doble Cadena , Humanos , Transducción de Señal
5.
Nature ; 523(7559): 226-30, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-26098368

RESUMEN

Although the adult mammalian heart is incapable of meaningful functional recovery following substantial cardiomyocyte loss, it is now clear that modest cardiomyocyte turnover occurs in adult mouse and human hearts, mediated primarily by proliferation of pre-existing cardiomyocytes. However, fate mapping of these cycling cardiomyocytes has not been possible thus far owing to the lack of identifiable genetic markers. In several organs, stem or progenitor cells reside in relatively hypoxic microenvironments where the stabilization of the hypoxia-inducible factor 1 alpha (Hif-1α) subunit is critical for their maintenance and function. Here we report fate mapping of hypoxic cells and their progenies by generating a transgenic mouse expressing a chimaeric protein in which the oxygen-dependent degradation (ODD) domain of Hif-1α is fused to the tamoxifen-inducible CreERT2 recombinase. In mice bearing the creERT2-ODD transgene driven by either the ubiquitous CAG promoter or the cardiomyocyte-specific α myosin heavy chain promoter, we identify a rare population of hypoxic cardiomyocytes that display characteristics of proliferative neonatal cardiomyocytes, such as smaller size, mononucleation and lower oxidative DNA damage. Notably, these hypoxic cardiomyocytes contributed widely to new cardiomyocyte formation in the adult heart. These results indicate that hypoxia signalling is an important hallmark of cycling cardiomyocytes, and suggest that hypoxia fate mapping can be a powerful tool for identifying cycling cells in adult mammals.


Asunto(s)
Miocardio/citología , Miocitos Cardíacos/citología , Proteínas Recombinantes de Fusión/metabolismo , Animales , Hipoxia de la Célula , Proliferación Celular/genética , Femenino , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Recombinasas/genética , Recombinasas/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo
6.
Int J Mol Sci ; 21(21)2020 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-33142765

RESUMEN

Radiation therapy (RT), an integral component of curative treatment for many malignancies, can be administered via an increasing array of techniques. In this review, we summarize the properties and application of different types of RT, specifically, conventional therapy with x-rays, stereotactic body RT, and proton and carbon particle therapies. We highlight how low-linear energy transfer (LET) radiation induces simple DNA lesions that are efficiently repaired by cells, whereas high-LET radiation causes complex DNA lesions that are difficult to repair and that ultimately enhance cancer cell killing. Additionally, we discuss the immunogenicity of radiation-induced tumor death, elucidate the molecular mechanisms by which radiation mounts innate and adaptive immune responses and explore strategies by which we can increase the efficacy of these mechanisms. Understanding the mechanisms by which RT modulates immune signaling and the key players involved in modulating the RT-mediated immune response will help to improve therapeutic efficacy and to identify novel immunomodulatory drugs that will benefit cancer patients undergoing targeted RT.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Inmunidad Celular/inmunología , Factores Inmunológicos , Neoplasias/radioterapia , Animales , Inestabilidad Genómica , Humanos , Inmunidad Celular/efectos de la radiación , Neoplasias/inmunología , Neoplasias/patología
7.
Nucleic Acids Res ; 45(8): 4590-4605, 2017 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-28334891

RESUMEN

RAD51, a multifunctional protein, plays a central role in DNA replication and homologous recombination repair, and is known to be involved in cancer development. We identified a novel role for RAD51 in innate immune response signaling. Defects in RAD51 lead to the accumulation of self-DNA in the cytoplasm, triggering a STING-mediated innate immune response after replication stress and DNA damage. In the absence of RAD51, the unprotected newly replicated genome is degraded by the exonuclease activity of MRE11, and the fragmented nascent DNA accumulates in the cytosol, initiating an innate immune response. Our data suggest that in addition to playing roles in homologous recombination-mediated DNA double-strand break repair and replication fork processing, RAD51 is also implicated in the suppression of innate immunity. Thus, our study reveals a previously uncharacterized role of RAD51 in initiating immune signaling, placing it at the hub of new interconnections between DNA replication, DNA repair, and immunity.


Asunto(s)
Replicación del ADN , Proteínas de Unión al ADN/genética , ADN/genética , Proteínas de la Membrana/genética , Recombinasa Rad51/genética , Reparación del ADN por Recombinación/genética , Línea Celular Tumoral , ADN/inmunología , Roturas del ADN de Doble Cadena/efectos de los fármacos , Proteínas de Unión al ADN/inmunología , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/inmunología , Genes Reporteros , Humanos , Ácidos Hidroxámicos/farmacología , Inmunidad Innata , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Proteína Homóloga de MRE11 , Proteínas de la Membrana/inmunología , Pirimidinonas/farmacología , Recombinasa Rad51/deficiencia , Recombinasa Rad51/inmunología , Reparación del ADN por Recombinación/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Tionas/farmacología , Vorinostat , Proteína Fluorescente Roja
8.
Semin Cell Dev Biol ; 58: 14-25, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26849909

RESUMEN

Cardiovascular disease and cancer are the two leading causes of morbidity and mortality worldwide. As advancements in radiation therapy (RT) have significantly increased the number of cancer survivors, the risk of radiation-induced cardiovascular disease (RICD) in this group is a growing concern. Recent epidemiological data suggest that accidental or occupational exposure to low dose radiation, in addition to therapeutic ionizing radiation, can result in cardiovascular complications. The progression of radiation-induced cardiotoxicity often takes years to manifest but is also multifaceted, as the heart may be affected by a variety of pathologies. The risk of cardiovascular disease development in RT cancer survivors has been known for 40 years and several risk factors have been identified in the last two decades. However, most of the early work focused on clinical symptoms and manifestations, rather than understanding cellular processes regulating homeostatic processes of the cardiovascular system in response to radiation. Recent studies have suggested that a different approach may be needed to refute the risk of cardiovascular disease following radiation exposure. In this review, we will focus on how different radiation types and doses may induce cardiovascular complications, highlighting clinical manifestations and the mechanisms involved in the pathophysiology of radiation-induced cardiotoxicity. We will finally discuss how current and future research on heart development and homeostasis can help reduce the incidence of RICD.


Asunto(s)
Corazón/efectos de la radiación , Radiación Ionizante , Animales , Enfermedades Cardiovasculares/etiología , Daño del ADN , Humanos , Factores de Riesgo , Transducción de Señal/efectos de la radiación
9.
Nucleic Acids Res ; 44(18): 8842-8854, 2016 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-27568005

RESUMEN

Defects in kinetochore-microtubule (KT-MT) attachment and the spindle assembly checkpoint (SAC) during cell division are strongly associated with chromosomal instability (CIN). CIN has been linked to carcinogenesis, metastasis, poor prognosis and resistance to cancer therapy. We previously reported that the DAB2IP is a tumor suppressor, and that loss of DAB2IP is often detected in advanced prostate cancer (PCa) and is indicative of poor prognosis. Here, we report that the loss of DAB2IP results in impaired KT-MT attachment, compromised SAC and aberrant chromosomal segregation. We discovered that DAB2IP directly interacts with Plk1 and its loss inhibits Plk1 kinase activity, thereby impairing Plk1-mediated BubR1 phosphorylation. Loss of DAB2IP decreases the localization of BubR1 at the kinetochore during mitosis progression. In addition, the reconstitution of DAB2IP enhances the sensitivity of PCa cells to microtubule stabilizing drugs (paclitaxel, docetaxel) and Plk1 inhibitor (BI2536). Our findings demonstrate a novel function of DAB2IP in the maintenance of KT-MT structure and SAC regulation during mitosis which is essential for chromosomal stability.


Asunto(s)
Puntos de Control del Ciclo Celular , Inestabilidad Cromosómica , Cinetocoros/metabolismo , Microtúbulos/metabolismo , Huso Acromático/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Animales , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Aberraciones Cromosómicas , Segregación Cromosómica , Técnicas de Inactivación de Genes , Humanos , Ratones , Mitosis/efectos de los fármacos , Mitosis/genética , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN , Moduladores de Tubulina/farmacología , Proteínas Activadoras de ras GTPasa/genética , Quinasa Tipo Polo 1
10.
Int J Mol Sci ; 19(11)2018 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-30400178

RESUMEN

Werner Syndrome (WS) is an autosomal recessive disorder characterized by the premature development of aging features. Individuals with WS also have a greater predisposition to rare cancers that are mesenchymal in origin. Werner Syndrome Protein (WRN), the protein mutated in WS, is unique among RecQ family proteins in that it possesses exonuclease and 3' to 5' helicase activities. WRN forms dynamic sub-complexes with different factors involved in DNA replication, recombination and repair. WRN binding partners either facilitate its DNA metabolic activities or utilize it to execute their specific functions. Furthermore, WRN is phosphorylated by multiple kinases, including Ataxia telangiectasia mutated, Ataxia telangiectasia and Rad3 related, c-Abl, Cyclin-dependent kinase 1 and DNA-dependent protein kinase catalytic subunit, in response to genotoxic stress. These post-translational modifications are critical for WRN to function properly in DNA repair, replication and recombination. Accumulating evidence suggests that WRN plays a crucial role in one or more genome stability maintenance pathways, through which it suppresses cancer and premature aging. Among its many functions, WRN helps in replication fork progression, facilitates the repair of stalled replication forks and DNA double-strand breaks associated with replication forks, and blocks nuclease-mediated excessive processing of replication forks. In this review, we specifically focus on human WRN's contribution to replication fork processing for maintaining genome stability and suppressing premature aging. Understanding WRN's molecular role in timely and faithful DNA replication will further advance our understanding of the pathophysiology of WS.


Asunto(s)
Replicación del ADN , Helicasa del Síndrome de Werner/metabolismo , Animales , Reparación del ADN , Humanos , Fosforilación , Estabilidad Proteica , Proteolisis , Helicasa del Síndrome de Werner/química
13.
Proc Natl Acad Sci U S A ; 109(2): 443-8, 2012 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-22184222

RESUMEN

The concept of DNA "repair centers" and the meaning of radiation-induced foci (RIF) in human cells have remained controversial. RIFs are characterized by the local recruitment of DNA damage sensing proteins such as p53 binding protein (53BP1). Here, we provide strong evidence for the existence of repair centers. We used live imaging and mathematical fitting of RIF kinetics to show that RIF induction rate increases with increasing radiation dose, whereas the rate at which RIFs disappear decreases. We show that multiple DNA double-strand breaks (DSBs) 1 to 2 µm apart can rapidly cluster into repair centers. Correcting mathematically for the dose dependence of induction/resolution rates, we observe an absolute RIF yield that is surprisingly much smaller at higher doses: 15 RIF/Gy after 2 Gy exposure compared to approximately 64 RIF/Gy after 0.1 Gy. Cumulative RIF counts from time lapse of 53BP1-GFP in human breast cells confirmed these results. The standard model currently in use applies a linear scale, extrapolating cancer risk from high doses to low doses of ionizing radiation. However, our discovery of DSB clustering over such large distances casts considerable doubts on the general assumption that risk to ionizing radiation is proportional to dose, and instead provides a mechanism that could more accurately address risk dose dependency of ionizing radiation.


Asunto(s)
Neoplasias de la Mama/genética , Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Relación Dosis-Respuesta en la Radiación , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Cultivadas , Reparación del ADN/efectos de la radiación , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Cinética , Modelos Biológicos , Medición de Riesgo , Proteína 1 de Unión al Supresor Tumoral P53
14.
Proc Natl Acad Sci U S A ; 108(20): 8293-8, 2011 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-21527720

RESUMEN

Clustered DNA damage induced by ionizing radiation is refractory to repair and may trigger carcinogenic events for reasons that are not well understood. Here, we used an in situ method to directly monitor induction and repair of clustered DNA lesions in individual cells. We showed, consistent with biophysical modeling, that the kinetics of loss of clustered DNA lesions was substantially compromised in human fibroblasts. The unique spatial distribution of different types of DNA lesions within the clustered damages, but not the physical location of these damages within the subnuclear domains, determined the cellular ability to repair the damage. We then examined checkpoint arrest mechanisms and yield of gross chromosomal aberrations. Induction of nonrepairable clustered damage affected only G2 accumulation but not the early G2/M checkpoint. Further, cells that were released from the G2/M checkpoint with unrepaired clustered damage manifested a spectrum of chromosome aberrations in mitosis. Difficulties associated with clustered DNA damage repair and checkpoint release before the completion of clustered DNA damage repair appear to promote genome instability that may lead to carcinogenesis.


Asunto(s)
Rotura Cromosómica , Daño del ADN , Fibroblastos/ultraestructura , Ciclo Celular , División Celular , Células Cultivadas , Reparación del ADN , Fase G2 , Humanos , Distribución Tisular
15.
Nucleic Acids Res ; 39(13): 5474-88, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21421565

RESUMEN

DNA damage and consequent mutations initiate the multistep carcinogenic process. Differentiated cells have a reduced capacity to repair DNA lesions, but the biological impact of unrepaired DNA lesions in differentiated lung epithelial cells is unclear. Here, we used a novel organotypic human lung three-dimensional (3D) model to investigate the biological significance of unrepaired DNA lesions in differentiated lung epithelial cells. We showed, consistent with existing notions that the kinetics of loss of simple double-strand breaks (DSBs) were significantly reduced in organotypic 3D culture compared to kinetics of repair in two-dimensional (2D) culture. Strikingly, we found that, unlike simple DSBs, a majority of complex DNA lesions were irreparable in organotypic 3D culture. Levels of expression of multiple DNA damage repair pathway genes were significantly reduced in the organotypic 3D culture compared with those in 2D culture providing molecular evidence for the defective DNA damage repair in organotypic culture. Further, when differentiated cells with unrepaired DNA lesions re-entered the cell cycle, they manifested a spectrum of gross-chromosomal aberrations in mitosis. Our data suggest that downregulation of multiple DNA repair pathway genes in differentiated cells renders them vulnerable to DSBs, promoting genome instability that may lead to carcinogenesis.


Asunto(s)
Rotura Cromosómica , Roturas del ADN de Doble Cadena , Diferenciación Celular , Línea Celular , Aberraciones Cromosómicas , Reparación del ADN/genética , Regulación hacia Abajo , Células Epiteliales/metabolismo , Células Epiteliales/efectos de la radiación , Humanos , Imagenología Tridimensional , Hierro/toxicidad , Cinética , Transferencia Lineal de Energía , Pulmón/citología , Técnicas de Cultivo de Órganos
16.
J Biol Chem ; 285(33): 25699-707, 2010 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-20516064

RESUMEN

Werner syndrome (WS) is a rare progeroid disorder characterized by genomic instability, increased cancer incidence, and early onset of a variety of aging pathologies. WS is unique among early aging syndromes in that affected individuals are developmentally normal, and phenotypic onset is in early adulthood. The protein defective in WS (WRN) is a member of the large RecQ family of helicases but is unique among this family in having an exonuclease. RecQ helicases form multimers, but the mechanism and consequence of multimerization remain incompletely defined. Here, we identify a novel heptad repeat coiled coil region between the WRN nuclease and helicase domains that facilitates multimerization of WRN. We mapped a novel and unique DNA-dependent protein kinase phosphorylation site proximal to the WRN multimerization region. However, phosphorylation at this site affected neither exonuclease activity nor multimeric state. We found that WRN nuclease is stimulated by DNA-dependent protein kinase independently of kinase activity or WRN nuclease multimeric status. In addition, WRN nuclease multimerization significantly increased nuclease processivity. We found that the novel WRN coiled coil domain is necessary for multimerization of the nuclease domain and sufficient to multimerize with full-length WRN in human cells. Importantly, correct homomultimerization is required for WRN function in vivo as overexpression of this multimerization domain caused increased sensitivity to camptothecin and 4-nitroquinoline 1-oxide similar to that in cells lacking functional WRN protein.


Asunto(s)
Exodesoxirribonucleasas/metabolismo , Exonucleasas/metabolismo , RecQ Helicasas/metabolismo , 4-Nitroquinolina-1-Óxido/farmacología , Western Blotting , Camptotecina/farmacología , Cromatografía en Gel , Daño del ADN/efectos de los fármacos , Exodesoxirribonucleasas/química , Exodesoxirribonucleasas/genética , Exonucleasas/genética , Células HeLa , Humanos , Inmunoprecipitación , Fosforilación , Reacción en Cadena de la Polimerasa , Multimerización de Proteína/genética , Multimerización de Proteína/fisiología , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , RecQ Helicasas/química , RecQ Helicasas/genética , Helicasa del Síndrome de Werner
17.
Nat Struct Mol Biol ; 13(5): 414-22, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16622405

RESUMEN

WRN is unique among the five human RecQ DNA helicases in having a functional exonuclease domain (WRN-exo) and being defective in the premature aging and cancer-related disorder Werner syndrome. Here, we characterize WRN-exo crystal structures, biochemical activity and participation in DNA end joining. Metal-ion complex structures, active site mutations and activity assays reveal a nuclease mechanism mediated by two metal ions. The DNA end-binding Ku70/80 complex specifically stimulates WRN-exo activity, and structure-based mutational inactivation of WRN-exo alters DNA end joining in human cells. We furthermore establish structural and biochemical similarities of WRN-exo to DnaQ-family replicative proofreading exonucleases, describing WRN-specific adaptations consistent with double-stranded DNA specificity and functionally important conformational changes. These results indicate WRN-exo is a human DnaQ family member and support DnaQ-like proofreading activities stimulated by Ku70/80, with implications for WRN functions in age-related pathologies and maintenance of genomic integrity.


Asunto(s)
ADN Helicasas/química , ADN Helicasas/metabolismo , ADN/genética , ADN/metabolismo , Animales , Sitios de Unión , Secuencia Conservada , Cristalografía por Rayos X , ADN Helicasas/clasificación , ADN Helicasas/genética , Exodesoxirribonucleasas , Humanos , Metales/química , Metales/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Pliegue de Proteína , RecQ Helicasas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Helicasa del Síndrome de Werner
18.
Mutat Res ; 711(1-2): 87-99, 2011 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-21126526

RESUMEN

Low-linear energy transfer (LET) radiation (i.e., γ- and X-rays) induces DNA double-strand breaks (DSBs) that are rapidly repaired (rejoined). In contrast, DNA damage induced by the dense ionizing track of high-atomic number and energy (HZE) particles is slowly repaired or is irreparable. These unrepaired and/or misrepaired DNA lesions may contribute to the observed higher relative biological effectiveness for cell killing, chromosomal aberrations, mutagenesis, and carcinogenesis in HZE particle irradiated cells compared to those treated with low-LET radiation. The types of DNA lesions induced by HZE particles have been characterized in vitro and usually consist of two or more closely spaced strand breaks, abasic sites, or oxidized bases on opposing strands. It is unclear why these lesions are difficult to repair. In this review, we highlight the potential of a new technology allowing direct visualization of different types of DNA lesions in human cells and document the emerging significance of live-cell imaging for elucidation of the spatio-temporal characterization of complex DNA damage. We focus on the recent insights into the molecular pathways that participate in the repair of HZE particle-induced DSBs. We also discuss recent advances in our understanding of how different end-processing nucleases aid in repair of DSBs with complicated ends generated by HZE particles. Understanding the mechanism underlying the repair of DNA damage induced by HZE particles will have important implications for estimating the risks to human health associated with HZE particle exposure.


Asunto(s)
Reparación del ADN , ADN/efectos de la radiación , Transferencia Lineal de Energía , Daño del ADN , Proteínas de Unión al ADN , Desoxirribonucleasas , Endonucleasas , Exodesoxirribonucleasas/fisiología , Humanos , Inmunoensayo , Microscopía Fluorescente , Proteínas Nucleares/metabolismo , RecQ Helicasas/fisiología , Helicasa del Síndrome de Werner
19.
Nucleic Acids Res ; 37(12): 3912-23, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19401436

RESUMEN

DNA double-strand breaks (DSBs) are a serious threat to genome stability and cell viability. Although biological effects of low levels of radiation are not clear, the risks of low-dose radiation are of societal importance. Here, we directly monitored induction and repair of single DSBs and quantitatively analyzed the dynamics of interaction of DNA repair proteins at individual DSB sites in living cells using 53BP1 fused to yellow fluorescent protein (YFP-53BP1) as a surrogate marker. The number of DSBs formed was linear with dose from 5 mGy to 1 Gy. The DSBs induced by very low radiation doses (5 mGy) were repaired with efficiency similar to repair of DSBs induced at higher doses. The YFP-53BP1 foci are dynamic structures: 53BP1 rapidly and reversibly interacted at these DSB sites. The time frame of recruitment and affinity of 53BP1 for DSB sites were indistinguishable between low and high doses, providing mechanistic evidence for the similar DSB repair after low- and high-dose radiation. These findings have important implications for estimating the risk associated with low-dose radiation exposure on human health.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Rayos gamma , Proteínas Bacterianas/análisis , Proteínas Bacterianas/genética , Línea Celular , Recuperación de Fluorescencia tras Fotoblanqueo , Humanos , Cinética , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/genética , Dosis de Radiación , Proteínas Recombinantes de Fusión/análisis
20.
J Hepatocell Carcinoma ; 8: 1169-1179, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34595139

RESUMEN

Localized hepatocellular carcinoma (HCC) that is unresectable and non-transplantable can be treated by several liver-directed therapies. External beam radiation therapy (EBRT) is an increasingly accepted and widely utilized treatment modality in this setting. Accelerated charged particles such as proton beam therapy (PBT) and carbon ion radiation therapy (CIRT) offer technological advancements over conventional photon radiotherapy. In this review, we summarize the distinct advantages of CIRT use for HCC treatment, focusing on physical and biological attributes, and outline dosimetric and treatment planning caveats. Based on these considerations, we posit that HCC may be among the best indications for use of CIRT, as it allows for maximizing tumoricidal doses to the target volume while minimizing the dose to the organs at risk.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA