Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
EBioMedicine ; 103: 105132, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38677182

RESUMEN

BACKGROUND: SARS-CoV-2 infection is considered as a relapsing inflammatory process with a dysregulation of IL-6 signalling. Classic IL-6 signalling is thought to represent a defence mechanism against pathogens. In contrast, IL-6 trans-signalling has pro-inflammatory effects. In severe COVID-19, therapeutic strategies have focused on global inhibition of IL-6, with controversial results. We hypothesized that specific blockade of IL-6 trans-signalling could inhibit inflammatory response preserving the host defence activity inherent to IL-6 classic signalling. METHODS: To test the role of the specific IL-6 trans-signalling inhibition by sgp130Fc in short- and long-term consequences of COVID-19, we used the established K18-hACE2 transgenic mouse model. Histological as well as immunohistochemical analysis, and pro-inflammatory marker profiling were performed. To investigate IL-6 trans-signalling in human cells we used primary lung microvascular endothelial cells and fibroblasts in the presence/absence of sgp130Fc. FINDINGS: We report that targeting IL-6 trans-signalling by sgp130Fc attenuated SARS-CoV-2-related clinical symptoms and mortality. In surviving mice, the treatment caused a significant decrease in lung damage. In vitro, IL-6 trans-signalling induced strong and persisting JAK1/STAT3 activation in endothelial cells and lung fibroblasts with proinflammatory effects, which were attenuated by sgp130Fc. Our data also suggest that in those cells with scant amounts of IL-6R, the induction of gp130 and IL-6 by IL-6:sIL-6R complex sustains IL-6 trans-signalling. INTERPRETATION: IL-6 trans-signalling fosters progression of COVID-19, and suggests that specific blockade of this signalling mode could offer a promising alternative to mitigate both short- and long-term consequences without affecting the beneficial effects of IL-6 classic signalling. These results have implications for the development of new therapies of lung injury and endotheliopathy in COVID-19. FUNDING: The project was supported by ISCIII, Spain (COV-20/00792 to MB, PI23/01351 to MARH) and the European Commission-Next generation EU (European Union) (Regulation EU 2020/2094), through CSIC's Global Health Platform (PTI Salud Global, SGL2103029 to MB). PID2019-110587RB-I00 (MB) supported by MICIN/AEI/10.13039/501100011033/and PID2022-143034OB-I00 (MB) by MICIN/AEI/10.13039/501100011033/FEDER. MAR-H acknowledges support from ISCIII, Spain and the European Commission-Next generation EU (European Union), through CSIC's Global Health PTI.


Asunto(s)
COVID-19 , Receptor gp130 de Citocinas , Modelos Animales de Enfermedad , Interleucina-6 , Ratones Transgénicos , SARS-CoV-2 , Transducción de Señal , Animales , Interleucina-6/metabolismo , COVID-19/metabolismo , Humanos , Ratones , Transducción de Señal/efectos de los fármacos , Receptor gp130 de Citocinas/metabolismo , Receptor gp130 de Citocinas/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/farmacología , Enzima Convertidora de Angiotensina 2/metabolismo , Pulmón/patología , Pulmón/virología , Pulmón/metabolismo , Células Endoteliales/metabolismo , Tratamiento Farmacológico de COVID-19 , Betacoronavirus , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/virología , Neumonía Viral/patología , Neumonía Viral/metabolismo , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Infecciones por Coronavirus/patología , Receptores de Interleucina-6/metabolismo , Receptores de Interleucina-6/antagonistas & inhibidores , Índice de Severidad de la Enfermedad
2.
Front Microbiol ; 14: 1185504, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37206325

RESUMEN

The emergent human coronavirus SARS-CoV-2 and its resistance to current drugs makes the need for new potent treatments for COVID-19 patients strongly necessary. Dextran sulfate (DS) polysaccharides have long demonstrated antiviral activity against different enveloped viruses in vitro. However, their poor bioavailability has led to their abandonment as antiviral candidates. Here, we report for the first time the broad-spectrum antiviral activity of a DS-based extrapolymeric substance produced by the lactic acid bacterium Leuconostoc mesenteroides B512F. Time of addition assays with SARS-CoV-2 pseudoviruses in in vitro models confirm the inhibitory activity of DSs in the early stages of viral infection (viral entry). In addition, this exopolysaccharide substance also reports broad-spectrum antiviral activity against several enveloped viruses such as SARS-CoV-2, HCoV229E, HSV-1, in in vitro models and in human lung tissue. The toxicity and antiviral capacity of DS from L. mesenteroides was tested in vivo in mouse models which are susceptible to SARS-CoV-2 infection. The described DS, administered by inhalation, a new route of administration for these types of polymers, shows strong inhibition of SARS-CoV-2 infection in vivo, significantly reducing animal mortality and morbidity at non-toxic doses. Therefore, we suggest that it may be considered as a potential candidate for antiviral therapy against SARS-CoV-2.

3.
Aging Cell ; 22(3): e13771, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36704839

RESUMEN

The enormous societal impact of the ongoing COVID-19 pandemic has been particularly harsh for some social groups, such as the elderly. Recently, it has been suggested that senescent cells could play a central role in pathogenesis by exacerbating the pro-inflammatory immune response against SARS-CoV-2. Therefore, the selective clearance of senescent cells by senolytic drugs may be useful as a therapy to ameliorate the symptoms of COVID-19 in some cases. Using the established COVID-19 murine model K18-hACE2, we demonstrated that a combination of the senolytics dasatinib and quercetin (D/Q) significantly reduced SARS-CoV-2-related mortality, delayed its onset, and reduced the number of other clinical symptoms. The increase in senescent markers that we detected in the lungs in response to SARS-CoV-2 may be related to the post-COVID-19 sequelae described to date. These results place senescent cells as central targets for the treatment of COVID-19, and make D/Q a new and promising therapeutic tool.


Asunto(s)
COVID-19 , Quercetina , Ratones , Humanos , Animales , Quercetina/farmacología , Quercetina/uso terapéutico , Dasatinib/farmacología , Dasatinib/uso terapéutico , SARS-CoV-2 , Senescencia Celular , Senoterapéuticos , Pandemias
4.
Aging (Albany NY) ; 11(24): 12844-12861, 2019 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-31789602

RESUMEN

Cellular senescence is a hallmark of aging, whose onset is linked to a series of both cell and non-cell autonomous processes, leading to several consequences for the organism. To date, several senescence routes have been identified, which play a fundamental role in development, tumor suppression and aging, among other processes. The positive and/or negative effects of senescent cells are directly related to the time that they remain in the organism. Short-term (acute) senescent cells are associated with positive effects; once they have executed their actions, immune cells are recruited to remove them. In contrast, long-term (chronic) senescent cells are associated with disease; they secrete pro-inflammatory and pro-tumorigenic factors in a state known as senescence-associated secretory phenotype (SASP). In recent years, cellular senescence has become the center of attention for the treatment of aging-related diseases. Current therapies are focused on elimination of senescent cell functions in three main ways: i) use of senolytics; ii) inhibition of SASP; and iii) improvement of immune system functions against senescent cells (immunosurveillance). In addition, some anti-cancer therapies are based on the induction of senescence in tumor cells. However, these senescent-like cancer cells must be subsequently cleared to avoid a chronic pro-tumorigenic state. Here is a summary of different scenarios, depending on the therapy used, with a discussion of the pros and cons of each scenario.


Asunto(s)
Senescencia Celular/inmunología , Senescencia Celular/fisiología , Envejecimiento , Animales , Senescencia Celular/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Regulación de la Expresión Génica/fisiología , Humanos , Sistema Inmunológico
5.
Cell Mol Life Sci ; 75(19): 3553-3567, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30030594

RESUMEN

Cellular senescence is the final fate of most cells in response to specific stimuli, but is not the end. Indeed, it is the beginning of a singular life, with multiple side roads leading to diverse effects on the organism. Many studies have been done in the last few years to elucidate the intriguing role of senescent cells in the organism, demonstrating them as the cause of several age-related diseases. However, these cells are also positively implicated in other important pathways, such as embryogenesis and wound healing. It appears that the multiple effects are time-dependent: long-term senescence is mostly implicated in chronic inflammation and disease, whereas in the short term, senescent cells seem to be beneficial, being rapidly targeted by the innate immune system. The influence of senescent cells on their neighbors by paracrine factors, differential activity depending on developmental stage, and duration of the effects make the cellular senescent program a unique spatial-temporal mechanism. During pathological conditions such as progeroid syndromes, this mechanism is deregulated, leading to accelerated onset of some aging-related diseases and a shorter lifespan, among other physiological defects. Here, we review the three primary cell senescence programs described so far (replicative, stress-induced, and developmentally programmed senescence), their onset during development, and their potential roles in diseases with premature aging. Finally, we discuss the role of immune cells in keeping senescence burden below the threshold of disease.


Asunto(s)
Fenómenos Fisiológicos Celulares , Senescencia Celular/fisiología , Envejecimiento/fisiología , Animales , Daño del ADN/fisiología , Desarrollo Embrionario/fisiología , Humanos , Inflamación/etiología , Inflamación/fisiopatología , Cicatrización de Heridas/fisiología
6.
PLoS One ; 7(12): e52976, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23300838

RESUMEN

Cervical cancer is caused by persistent high-risk human papillomavirus (HR-HPV) infection and represents the second most frequent gynecological malignancy in the world. The HPV-16 type accounts for up to 55% of all cervical cancers. The HPV-16 oncoproteins E6 and E7 are necessary for induction and maintenance of malignant transformation and represent tumor-specific antigens for targeted cytotoxic T lymphocyte-mediated immunotherapy. Therapeutic cancer vaccines have become a challenging area of oncology research in recent decades. Among current cancer immunotherapy strategies, virus-like particle (VLP)-based vaccines have emerged as a potent and safe approach. We generated a vaccine (VLP-E7) incorporating a long C-terminal fragment of HPV-16 E7 protein into the infectious bursal disease virus VLP and tested its therapeutic potential in HLA-A2 humanized transgenic mice grafted with TC1/A2 tumor cells. We performed a series of tumor challenge experiments demonstrating a strong immune response against already-formed tumors (complete eradication). Remarkably, therapeutic efficacy was obtained with a single dose without adjuvant and against two injections of tumor cells, indicating a potent and long-lasting immune response.


Asunto(s)
Papillomavirus Humano 16/inmunología , Virus de la Enfermedad Infecciosa de la Bolsa/inmunología , Proteínas E7 de Papillomavirus/inmunología , Vacunas contra Papillomavirus/uso terapéutico , Neoplasias del Cuello Uterino/terapia , Vacunas de Partículas Similares a Virus/uso terapéutico , Animales , Femenino , Ratones , Ratones Transgénicos , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/terapia , Vacunas contra Papillomavirus/inmunología , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/virología , Vacunas de Partículas Similares a Virus/inmunología
7.
Hum Genet ; 124(4): 369-77, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18810497

RESUMEN

Werner syndrome (WS) is an adult onset segmental progeroid syndrome caused by mutations in the WRN gene. The WRN gene encodes a 180 kDa nuclear protein that possesses helicase and exonuclease activities. The absence of WRN protein leads to abnormalities in various DNA metabolic pathways such as DNA repair, replication and telomere maintenance. Individuals with WS generally develop normally until the third decade of life, when premature aging phenotypes and a series of age-related disorders begin to manifest. In Japan, where a founder effect has been described, the frequency of Werner heterozygotes appears to be as high as 1/180 in the general population. Due to the relatively non-specific nature of the symptoms and the lack of awareness of the condition, this disease may be under-diagnosed in other parts of the world. Genetic counseling of WS patients follows the path of other autosomal recessive disorders, with special attention needed for cancer surveillance in relatives. Molecular diagnosis of WS is made by nucleotide sequencing and, in some cases, protein analysis. It is also of potential interest to measure WRN activities in WS patients. More than 50 different disease-causing mutations in the WRN gene have been identified in WS patients from all over the world. All but one of these cases has mutations that result in the premature termination of the protein. Here we describe the clinical, molecular and biochemical characteristics of WS for use by medical professionals in a health care setting. Additional information is available through the International Registry of WS (http://www.wernersyndrome.org).


Asunto(s)
Exodesoxirribonucleasas/genética , RecQ Helicasas/genética , Síndrome de Werner/diagnóstico , Síndrome de Werner/genética , Humanos , Mutación , Polimorfismo Genético , Síndrome de Werner/epidemiología , Helicasa del Síndrome de Werner
8.
Cell Cycle ; 6(21): 2662-8, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17912036

RESUMEN

Although the anti-tumour effects of paclitaxel result mainly from mitotic arrest, recent evidences suggest alternative mechanisms of cytotoxicity. Cell cycle, cell death, and gene expression assays were used to understand the molecular mechanisms of paclitaxel cytotoxicity in breast cancer cells. G(2)/M cell cycle arrest and cell death coincided with the regulation of genes involved in cell death, cell cycle control, microtubule-based processes, oxidative stress, and ubiquitin-proteasome system. Induction of proteasome genes was also correlated with an accumulation of protein for proteasome subunits. Furthermore, a schedule-dependent regulation of paclitaxel-induced cytotoxicity was observed after combining paclitaxel and the proteasome inhibitor MG132. Proteasome inhibition after paclitaxel exposure induced the highest rate of growth inhibition and apoptosis, with no effect on mitotic arrest. These findings give support to clinical combinations of taxanes with proteasome inhibitors, outlining the importance of considering the sequence when designing such regimens.


Asunto(s)
Neoplasias de la Mama/metabolismo , Paclitaxel/efectos adversos , Complejo de la Endopetidasa Proteasomal/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Leupeptinas/farmacología , Paclitaxel/uso terapéutico , Complejo de la Endopetidasa Proteasomal/genética , Inhibidores de Proteasoma
9.
Mech Ageing Dev ; 128(7-8): 423-36, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17624410

RESUMEN

Werner syndrome (WS) is a premature aging and cancer-prone disease caused by loss of the RecQ helicase WRN protein. Cultured WS fibroblasts display high genomic instability and senesce prematurely. Epigenetic inactivation of the WRN gene occurs in numerous tumor types, in which WRN demonstrates tumor suppressor-like activity (Agrelo et al., 2006). However, the role of WRN in tumors that express WRN protein is unknown. Here we report that the inhibition of WRN expression strongly impairs growth of 12 out of 15 cancer cell lines tested. For those cell lines in which WRN depletion induced high cell death, the majority of the surviving proliferative clones exhibited WRN expression. Growth arrest induced by WRN depletion was characterized by an accumulation of cells in the G2/M cell cycle phases and an increase in DNA damage. Importantly, WRN depletion inhibited tumor growth in vivo in SCID mouse xenograft models. Altogether, these findings support a dual role for WRN in tumorigenesis; tumor suppressor-like activity in tumors with WRN inactivation and the promotion of proliferation and survival in tumors that express WRN. These findings suggest a possible therapeutic role for WRN as an anti-cancer target, and highlight the importance of WRN protein status for tumorigenesis and clinical treatments of patients.


Asunto(s)
Neoplasias de la Mama/fisiopatología , Senescencia Celular/fisiología , Neoplasias Ováricas/fisiopatología , RecQ Helicasas/genética , Síndrome de Werner/patología , Síndrome de Werner/fisiopatología , Animales , Neoplasias de la Mama/patología , División Celular/fisiología , Supervivencia Celular/fisiología , Exodesoxirribonucleasas , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor/fisiología , Células HeLa , Humanos , Riñón/citología , Ratones , Ratones SCID , Trasplante de Neoplasias , Neoplasias Ováricas/patología , ARN Interferente Pequeño , RecQ Helicasas/metabolismo , Helicasa del Síndrome de Werner
10.
Proc Natl Acad Sci U S A ; 103(23): 8822-7, 2006 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-16723399

RESUMEN

Werner syndrome (WS) is an inherited disorder characterized by premature onset of aging, genomic instability, and increased cancer incidence. The disease is caused by loss of function mutations of the WRN gene, a RecQ family member with both helicase and exonuclease activities. However, despite its putative tumor-suppressor function, little is known about the contribution of WRN to human sporadic malignancies. Here, we report that WRN function is abrogated in human cancer cells by transcriptional silencing associated with CpG island-promoter hypermethylation. We also show that, at the biochemical and cellular levels, the epigenetic inactivation of WRN leads to the loss of WRN-associated exonuclease activity and increased chromosomal instability and apoptosis induced by topoisomerase inhibitors. The described phenotype is reversed by the use of a DNA-demethylating agent or by the reintroduction of WRN into cancer cells displaying methylation-dependent silencing of WRN. Furthermore, the restoration of WRN expression induces tumor-suppressor-like features, such as reduced colony formation density and inhibition of tumor growth in nude mouse xenograft models. Screening a large collection of human primary tumors (n = 630) from different cell types revealed that WRN CpG island hypermethylation was a common event in epithelial and mesenchymal tumorigenesis. Most importantly, WRN hypermethylation in colorectal tumors was a predictor of good clinical response to the camptothecin analogue irinotecan, a topoisomerase inhibitor commonly used in the clinical setting for the treatment of this tumor type. These findings highlight the importance of WRN epigenetic inactivation in human cancer, leading to enhanced chromosomal instability and hypersensitivity to chemotherapeutic drugs.


Asunto(s)
Envejecimiento Prematuro/genética , Neoplasias del Colon/genética , ADN Helicasas/genética , Epigénesis Genética/genética , Síndrome de Werner/genética , Animales , Camptotecina/análogos & derivados , Camptotecina/farmacología , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Islas de CpG/genética , Metilación de ADN , Exodesoxirribonucleasas , Femenino , Genes Supresores de Tumor , Humanos , Irinotecán , Ratones , Ratones Desnudos , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , RecQ Helicasas , Inhibidores de Topoisomerasa I , Helicasa del Síndrome de Werner
11.
Int J Cancer ; 119(5): 1164-75, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16557594

RESUMEN

The availability of oral precursors of 5-Fluorouracil (5-FU) and its favorable results in treating advanced breast cancer have renewed the interest in the molecular mechanisms underlying its cytotoxicity. We have compared the changes in cell cycle and cell death parameters induced by 2 different concentrations of 5-FU (IC50 and IC80) in the breast adenocarcinoma cell line MCF7. G1/S cell cycle arrest was associated with both concentrations, whereas cell death was mainly induced after IC80 5-FU. These changes were correlated with gene expression assessed by cDNA microarray analysis. Main findings included an overexpression of p53 target genes involved in cell cycle and apoptosis (CDKN1A/p21, TP53INP, TNFRSF6/FAS and BBC3/PUMA), and significant repression of Myc. High dose 5-FU also induced a higher regulation of the mitochondrial death genes APAF1, BAK1 and BCL2, and induction of genes of the ID family. Furthermore, we establish a direct causal relationship between p21, ID1 and ID2 overexpression, increased acetylation of histones H3 and H4 and binding of p53 to their promoters as a result of 5-FU treatment. The relevance of these findings was further studied after interfering p53 expression in MCF7 cells (shp53 cells), showing a lower induction of both, ID1 and ID2 transcripts, after 5-FU when compared with MCF7 shGFP control cells. This molecular characterization of dose- and time-dependent modifications of gene expression after 5-FU treatment should provide a resource for future basic studies addressing the molecular mechanisms of chemotherapy in breast cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Ciclo Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Fluorouracilo/farmacología , Transcripción Genética/efectos de los fármacos , Proteína p53 Supresora de Tumor/efectos de los fármacos , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Immunoblotting , Análisis por Micromatrices , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo
12.
J Biol Chem ; 280(47): 39627-36, 2005 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-16150736

RESUMEN

Naturally occurring mutations in the human RECQ3 gene result in truncated Werner protein (WRN) and manifest as a rare premature aging disorder, Werner syndrome. Cellular and biochemical studies suggest a multifaceted role of WRN in DNA replication, DNA repair, recombination, and telomere maintenance. The RecQ C-terminal (RQC) domain of WRN was determined previously to be the major site of interaction for DNA and proteins. By using site-directed mutagenesis in the WRN RQC domain, we determined which amino acids might be playing a critical role in WRN function. A site-directed mutation at Lys-1016 significantly decreased WRN binding to fork or bubble DNA substrates. Moreover, the Lys-1016 mutation markedly reduced WRN helicase activity on fork, D-loop, and Holliday junction substrates in addition to reducing significantly the ability of WRN to stimulate FEN-1 incision activities. Thus, DNA binding mediated by the RQC domain is crucial for WRN helicase and its coordinated functions. Our nuclear magnetic resonance data on the three-dimensional structure of the wild-type RQC and Lys-1016 mutant proteins display a remarkable similarity in their structures.


Asunto(s)
ADN Helicasas/genética , ADN Helicasas/metabolismo , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Secuencia de Bases , Línea Celular , Secuencia Conservada , ADN/genética , ADN Helicasas/química , Exodesoxirribonucleasas , Humanos , Técnicas In Vitro , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación Missense , Resonancia Magnética Nuclear Biomolecular , Estructura Terciaria de Proteína , RecQ Helicasas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Síndrome de Werner/etiología , Síndrome de Werner/genética , Síndrome de Werner/metabolismo , Helicasa del Síndrome de Werner
13.
Mol Cell Biol ; 25(17): 7625-36, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16107709

RESUMEN

Cockayne syndrome (CS) is a rare genetic disorder characterized as a segmental premature-aging syndrome. The CS group B (CSB) protein has previously been implicated in transcription-coupled repair, transcriptional elongation, and restoration of RNA synthesis after DNA damage. Recently, evidence for a role of CSB in base excision repair of oxidative DNA lesions has accumulated. In our search to understand the molecular function of CSB in this process, we identify a physical and functional interaction between CSB and poly(ADP-ribose) polymerase-1 (PARP-1). PARP-1 is a nuclear enzyme that protects the integrity of the genome by responding to oxidative DNA damage and facilitating DNA repair. PARP-1 binds to single-strand DNA breaks which activate the catalytic ability of PARP-1 to add polymers of ADP-ribose to various proteins. We find that CSB is present at sites of activated PARP-1 after oxidative stress, identify CSB as a new substrate of PARP-1, and demonstrate that poly(ADP-ribosyl)ation of CSB inhibits its DNA-dependent ATPase activity. Furthermore, we find that CSB-deficient cell lines are hypersensitive to inhibition of PARP. Our results implicate CSB in the PARP-1 poly(ADP-ribosyl)ation response after oxidative stress and thus suggest a novel role of CSB in the cellular response to oxidative damage.


Asunto(s)
Síndrome de Cockayne , ADN Helicasas/metabolismo , Estrés Oxidativo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Transporte Activo de Núcleo Celular , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , ADN Helicasas/deficiencia , ADN Helicasas/genética , Enzimas Reparadoras del ADN , Humanos , Mutación/genética , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/genética , Proteínas de Unión a Poli-ADP-Ribosa , Unión Proteica , Especificidad por Sustrato
14.
J Biol Chem ; 280(33): 29494-505, 2005 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-15965237

RESUMEN

The single-stranded DNA-binding protein replication protein A (RPA) interacts with several human RecQ DNA helicases that have important roles in maintaining genomic stability; however, the mechanism for RPA stimulation of DNA unwinding is not well understood. To map regions of Werner syndrome helicase (WRN) that interact with RPA, yeast two-hybrid studies, WRN affinity pull-down experiments and enzyme-linked immunosorbent assays with purified recombinant WRN protein fragments were performed. The results indicated that WRN has two RPA binding sites, a high affinity N-terminal site, and a lower affinity C-terminal site. Based on results from mapping studies, we sought to determine if the WRN N-terminal region harboring the high affinity RPA interaction site was important for RPA stimulation of WRN helicase activity. To accomplish this, we tested a catalytically active WRN helicase domain fragment (WRN(H-R)) that lacked the N-terminal RPA interaction site for its ability to unwind long DNA duplex substrates, which the wild-type enzyme can efficiently unwind only in the presence of RPA. WRN(H-R) helicase activity was significantly reduced on RPA-dependent partial duplex substrates compared with full-length WRN despite the presence of RPA. These results clearly demonstrate that, although WRN(H-R) had comparable helicase activity to full-length WRN on short duplex substrates, its ability to unwind RPA-dependent WRN helicase substrates was significantly impaired. Similarly, a Bloom syndrome helicase (BLM) domain fragment, BLM(642-1290), that lacked its N-terminal RPA interaction site also unwound short DNA duplex substrates similar to wild-type BLM, but was severely compromised in its ability to unwind long DNA substrates that full-length BLM helicase could unwind in the presence of RPA. These results suggest that the physical interaction between RPA and WRN or BLM helicases plays an important role in the mechanism for RPA stimulation of helicase-catalyzed DNA unwinding.


Asunto(s)
Adenosina Trifosfatasas/química , ADN Helicasas/química , Proteínas de Unión al ADN/química , Adenosina Trifosfatasas/fisiología , Sitios de Unión , ADN/química , ADN Helicasas/fisiología , Replicación del ADN , Proteínas de Unión al ADN/fisiología , Exodesoxirribonucleasas , Humanos , RecQ Helicasas , Proteína de Replicación A , Técnicas del Sistema de Dos Híbridos , Helicasa del Síndrome de Werner , Proteínas de Xenopus/fisiología
15.
FASEB J ; 18(15): 1970-2, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15459124

RESUMEN

Werner syndrome (WS) is a rare disease caused by the lack of a functional nuclear WS protein (WRN). WS is characterized by the early onset of premature aging signs and a high incidence of sarcomas. WS diploid fibroblasts have a short life span and extensive genomic instability. Mammalian cells are continuously exposed to reactive oxygen species (ROS), which represent human mutagens and are thought to be a major contributor to the aging process. Hydrogen peroxide (H2O2) is a common ROS intermediate generated by various forms of oxidative stress. In response to H2O2-induced DNA damage, normal human diploid fibroblasts follow a pathway leading to irreversible proliferation arrest and premature senescence. Here we show that in contrast to normal human fibroblasts, WS diploid fibroblasts continue proliferating after extensive H2O2-induced DNA damage and accumulate oxidative DNA lesions. A direct role of WRN in this abnormal cellular response to H2O2 is demonstrated by interfering with WRN expression in normal human fibroblasts. We propose a role for WRN in the detection and/or processing of oxidative DNA lesions and in cellular responses to H2O2 as they relate to some of the phenotypical aspects of WS cells.


Asunto(s)
Daño del ADN , Peróxido de Hidrógeno/toxicidad , Síndrome de Werner/genética , Muerte Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Senescencia Celular , ADN/metabolismo , ADN Helicasas/metabolismo , Exodesoxirribonucleasas , Fibroblastos/efectos de los fármacos , Humanos , Oxidación-Reducción , RecQ Helicasas , Síndrome de Werner/metabolismo , Helicasa del Síndrome de Werner
16.
Nucleic Acids Res ; 32(13): 4003-14, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15292449

RESUMEN

Werner syndrome (WS) is a genetic premature aging disorder in which patients appear much older than their chronological age. The gene mutated in WS encodes a nuclear protein (WRN) which possesses 3'-5' exonuclease and ATPase-dependent 3'-5' helicase activities. The genomic instability associated with WS cells and the biochemical characteristics of WRN suggest that WRN plays a role in DNA metabolic pathways such as transcription, replication, recombination and repair. Recently we have identified poly(ADP-ribose) polymerase-1 (PARP-1) as a new WRN interacting protein. In this paper, we further mapped the interacting domains. We found that PARP-1 bound to the N-terminus of WRN and to the C-terminus containing the RecQ-conserved (RQC) domain. WRN bound to the N-terminus of PARP-1 containing DNA binding and BRCA1 C-terminal (BRCT) domains. We show that unmodified PARP-1 inhibited both WRN exonuclease and helicase activities, and to our knowledge is the only known WRN protein partner that inactivates both of the WRN's catalytic activities suggesting a biologically significant regulation. Moreover, this dual inhibition seems to be specific for PARP-1, as PARP-2 did not affect WRN helicase activity and only slightly inhibited WRN exonuclease activity. The differential effect of PARP-1 and PARP-2 on WRN catalytic activity was not due to differences in affinity for WRN or the DNA substrate. Finally, we demonstrate that the inhibition of WRN by PARP-1 was influenced by the poly(ADP-ribosyl)ation state of PARP-1. The biological relevance of the specific modulation of WRN catalytic activities by PARP-1 are discussed in the context of pathways in which these proteins may function together, namely in the repair of DNA strand breaks.


Asunto(s)
ADN Helicasas/metabolismo , Exonucleasas/metabolismo , Poli(ADP-Ribosa) Polimerasas/fisiología , Sitios de Unión , ADN/química , ADN/metabolismo , ADN Helicasas/química , Exodesoxirribonucleasas , Exonucleasas/química , Células HeLa , Humanos , Modelos Biológicos , Poli(ADP-Ribosa) Polimerasas/química , Poli(ADP-Ribosa) Polimerasas/metabolismo , Estructura Terciaria de Proteína , RecQ Helicasas , Helicasa del Síndrome de Werner
17.
Mech Ageing Dev ; 125(7): 491-6, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15246744

RESUMEN

The leading causes of death for individuals with Werner syndrome (WS) are myocardial infarction (MI) and stroke. The WS gene encodes a nuclear protein with both helicase and exonuclease activities. While individuals with WS have mutations that result in truncated, inactive proteins, several sequence variants have been described in apparently unaffected individuals. Some of these gene polymorphisms encode non-conservative amino acid substitutions, and it is expected that the changes would affect enzyme activity, although this has not been determined. Two research groups have studied the Cys/Arg 1367 polymorphism (located near the nuclear localization signal) in healthy and MI patients. Their results suggest that the Arg allele is protective against MI. We have characterized the Cys (C) and Arg (R) forms of the protein and find no notable difference in helicase and nuclease activities, or in nuclear/cytoplasmic distribution. The frequency of the C/R alleles in healthy individuals and subjects with coronary artery disease (CAD) drawn from the Baltimore Longitudinal Study of Aging (BLSA) was also examined. There was no indication that the R allele was protective against CAD. We conclude that the C/R polymorphism does not affect enzyme function or localization and does not influence CAD incidence in the BLSA cohort.


Asunto(s)
Enfermedad de la Arteria Coronaria/genética , ADN Helicasas/genética , Predisposición Genética a la Enfermedad , Polimorfismo Genético , Población Blanca/genética , Anciano , Anciano de 80 o más Años , Alelos , Sustitución de Aminoácidos , Animales , Núcleo Celular/metabolismo , Células Cultivadas , Estudios de Cohortes , Citoplasma/metabolismo , ADN Helicasas/metabolismo , Exodesoxirribonucleasas , Exonucleasas/genética , Exonucleasas/metabolismo , Frecuencia de los Genes , Humanos , Masculino , Persona de Mediana Edad , RecQ Helicasas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Helicasa del Síndrome de Werner
18.
J Struct Biol ; 146(1-2): 251-9, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15037256

RESUMEN

We have previously shown that the Werner syndrome helicase, WRNp, a member of the RecQ helicase family, forms a tight molecular complex with the p97/Valosin containing protein (VCP), a member of the AAA (ATPases associated with diverse cellular activities) family of proteins. This interaction is disrupted by chemical agents that confer DNA damage, suggesting that VCP plays an important role in the signal-dependent release of WRNp from its nucleolar sequestration site. Here, we characterized the structural requirements for interactions between WRNp and VCP and for the nuclear localization of VCP. We discovered that VCP directly binds to the RQC (RecQ conserved) domain of WRNp, which is a highly conserved motif common to the RecQ helicase family. This interaction is ATP-dependent, suggesting that VCP plays a mechanistic role in releasing WRNp from the nucleolus. Immunohistochemical analysis of various VCP domains and mutated proteins expressed in vitro demonstrated that VCP may contain several hierarchical cellular localization motifs within its domain structure.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/farmacología , Proteínas de Ciclo Celular/metabolismo , ADN Helicasas/metabolismo , Transporte Activo de Núcleo Celular , Animales , Sitios de Unión , Bovinos , Secuencia Conservada , Exodesoxirribonucleasas , Células HeLa , Humanos , Inmunohistoquímica , Sustancias Macromoleculares , Mutación , Fragmentos de Péptidos/metabolismo , Unión Proteica , RecQ Helicasas , Proteína que Contiene Valosina , Helicasa del Síndrome de Werner
19.
J Biol Chem ; 279(20): 21169-76, 2004 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-15026416

RESUMEN

The Werner syndrome and the Nijmegen breakage syndrome are recessive genetic disorders that show increased genomic instability, cancer predisposition, hypersensitivity to mitomycin C and gamma-irradiation, shortened telomeres, and cell cycle defects. The protein mutated in the premature aging disease known as the Werner syndrome is designated WRN and is a member of the RecQ helicase family. The Nbs1 protein is mutated in Nijmegen breakage syndrome individuals and is part of the mammalian Mre11 complex together with the Mre11 and Rad50 proteins. Here, we show that WRN associates with the Mre11 complex via binding to Nbs1 in vitro and in vivo. In response to gamma-irradiation or mitomycin C, WRN leaves the nucleoli and co-localizes with the Mre11 complex in the nucleoplasm. We detect an increased association between WRN and the Mre11 complex after cellular exposure to gamma-irradiation. Small interfering RNA and complementation experiments demonstrated convergence of WRN and Nbs1 in response to gamma-irradiation or mitomycin C. Nbs1 is required for the Mre11 complex promotion of WRN helicase activity. Taken together, these results demonstrate a functional link between the two genetic diseases with partially overlapping phenotypes in a pathway that responds to DNA double strand breaks and interstrand cross-links.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Sitios de Unión , Proteínas de Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/efectos de la radiación , Línea Celular , ADN Helicasas/efectos de los fármacos , ADN Helicasas/genética , ADN Helicasas/efectos de la radiación , Proteínas de Unión al ADN/genética , Exodesoxirribonucleasas , Rayos gamma , Células HeLa , Humanos , Proteína Homóloga de MRE11 , Mitomicina/farmacología , Proteínas Nucleares/efectos de los fármacos , Proteínas Nucleares/genética , Proteínas Nucleares/efectos de la radiación , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/efectos de la radiación , RecQ Helicasas , Proteínas Recombinantes/metabolismo , Valores de Referencia , Transfección , Helicasa del Síndrome de Werner
20.
Mol Cell Biol ; 23(23): 8601-13, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14612404

RESUMEN

A defect in the Werner syndrome protein (WRN) leads to the premature aging disease Werner syndrome (WS). Hallmark features of cells derived from WS patients include genomic instability and hypersensitivity to certain DNA-damaging agents. WRN contains a highly conserved region, the RecQ conserved domain, that plays a central role in protein interactions. We searched for proteins that bound to this region, and the most prominent direct interaction was with poly(ADP-ribose) polymerase 1 (PARP-1), a nuclear enzyme that protects the genome by responding to DNA damage and facilitating DNA repair. In pursuit of a functional interaction between WRN and PARP-1, we found that WS cells are deficient in the poly(ADP-ribosyl)ation pathway after they are treated with the DNA-damaging agents H2O2 and methyl methanesulfonate. After cellular stress, PARP-1 itself becomes activated, but the poly(ADP-ribosyl)ation of other cellular proteins is severely impaired in WS cells. Overexpression of the PARP-1 binding domain of WRN strongly inhibits the poly(ADP-ribosyl)ation activity in H2O2-treated control cell lines. These results indicate that the WRN/PARP-1 complex plays a key role in the cellular response to oxidative stress and alkylating agents, suggesting a role for these proteins in the base excision DNA repair pathway.


Asunto(s)
Daño del ADN , ADN Helicasas/metabolismo , Poli Adenosina Difosfato Ribosa/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Síndrome de Werner/metabolismo , Sitios de Unión , Línea Celular , ADN Helicasas/química , ADN Helicasas/genética , Reparación del ADN , Exodesoxirribonucleasas , Proteínas Fluorescentes Verdes , Células HeLa , Humanos , Peróxido de Hidrógeno/toxicidad , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Sustancias Macromoleculares , Metilmetanosulfonato/toxicidad , Mutación , Estrés Oxidativo , Poli(ADP-Ribosa) Polimerasas/química , Estructura Terciaria de Proteína , RecQ Helicasas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Síndrome de Werner/genética , Helicasa del Síndrome de Werner
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...