Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Oncol (Dordr) ; 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38668817

RESUMEN

PURPOSE: Acquired resistance to immune checkpoint blockers (ICBs) is a major barrier in cancer treatment, emphasizing the need for innovative strategies. Dectin-1 (gene Clec7a) is a C-type lectin receptor best known for its ability to recognize ß-glucan-rich structures in fungal cell walls. While Dectin-1 is expressed in myeloid cells and tumor cells, its significance in cancer remains the subject of controversy. METHODS: Using Celc7a-/- mice and curdlan administration to stimulate Dectin-1 signaling, we explored its impact. VISTA KO mice were employed to assess VISTA's role, and bulk RNAseq analyzed curdlan effects on neutrophils. RESULTS: Our findings reveal myeloid cells as primary Dectin-1 expressing cells in the tumor microenvironment (TME), displaying an activated phenotype. Strong Dectin-1 co-expression/co-localization with VISTA and PD-L1 in TME myeloid cells was observed. While Dectin-1 deletion lacked protective effects, curdlan stimulation significantly curtailed B16-F10 tumor progression. RNAseq and pathway analyses supported curdlan's role in triggering a cascade of events leading to increased production of pro-inflammatory mediators, potentially resulting in the recruitment and activation of immune cells. Moreover, we identified a heterogeneous subset of Dectin-1+ effector T cells in the TME. Similar to mice, human myeloid cells are the prominent cells expressing Dectin-1 in cancer patients. CONCLUSION: Our study proposes Dectin-1 as a potential adjunctive target with ICBs, orchestrating a comprehensive engagement of innate and adaptive immune responses in melanoma. This innovative approach holds promise for overcoming acquired resistance to ICBs in cancer treatment, offering avenues for further exploration and development.

2.
J Biol Chem ; 300(3): 105709, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38309501

RESUMEN

Double-strand breaks (DSBs) are DNA lesions that pose a significant threat to genomic stability. The repair of DSBs by the homologous recombination (HR) pathway is preceded by DNA end resection, the 5' to 3' nucleolytic degradation of DNA away from the DSB. We and others previously identified a role for RNF138, a really interesting new gene finger E3 ubiquitin ligase, in stimulating DNA end resection and HR. Yet, little is known about how RNF138's function is regulated in the context of DSB repair. Here, we show that RNF138 is phosphorylated at residue T27 by cyclin-dependent kinase (CDK) activity during the S and G2 phases of the cell cycle. We also observe that RNF138 is ubiquitylated constitutively, with ubiquitylation occurring in part on residue K158 and rising during the S/G2 phases. Interestingly, RNF138 ubiquitylation decreases upon genotoxic stress. By mutating RNF138 at residues T27, K158, and the previously identified S124 ataxia telangiectasia mutated phosphorylation site (Han et al., 2016, ref. 22), we find that post-translational modifications at all three positions mediate DSB repair. Cells expressing the T27A, K158R, and S124A variants of RNF138 are impaired in DNA end resection, HR activity, and are more sensitive to ionizing radiation compared to those expressing wildtype RNF138. Our findings shed more light on how RNF138 activity is controlled by the cell during HR.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Ubiquitina-Proteína Ligasas , Recombinación Homóloga , Fosforilación , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Humanos , Células HEK293
3.
Cells ; 12(14)2023 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-37508568

RESUMEN

Detailing the connection between homeostatic functions of enzymatic families and eventual progression into tumorigenesis is crucial to our understanding of anti-cancer therapies. One key enzyme group involved in this process is the Poly (ADP-ribose) polymerase (PARP) family, responsible for an expansive number of cellular functions, featuring members well established as regulators of DNA repair, genomic stability and beyond. Several PARP inhibitors (PARPi) have been approved for clinical use in a range of cancers, with many more still in trials. Unfortunately, the occurrence of resistance to PARPi therapy is growing in prevalence and requires the introduction of novel counter-resistance mechanisms to maintain efficacy. In this review, we summarize the updated understanding of the vast homeostatic functions the PARP family mediates and pin the importance of PARPi therapies as anti-cancer agents while discussing resistance mechanisms and current up-and-coming counter-strategies for countering such resistance.


Asunto(s)
Antineoplásicos , Neoplasias , Humanos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Poli(ADP-Ribosa) Polimerasas/genética , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Carcinogénesis , Transformación Celular Neoplásica , Poli(ADP-Ribosa) Polimerasa-1/uso terapéutico
4.
STAR Protoc ; 4(1): 101917, 2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36520630

RESUMEN

Here, we present a chromatin-immunoprecipitation-based protocol to quantify the recruitment of proteins adjacent to site-specific DNA double-strand breaks (DSBs), such as proteins involved in DSB repair. We describe steps to induce DSBs in U2OS osteosarcoma cells stably expressing the restriction endonucleases FokI or AsiSI. We then detail the procedures of chromatin isolation and immunoprecipitation, followed by protein elution and quantitative-PCR-based quantification of DNA. This protocol cannot be used on DSBs generated at random loci by DNA damaging agents. For complete details on the use and execution of this protocol, please refer to Fitieh et al. (2022).1.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Humanos , Reparación del ADN/genética , Cromatina/genética , ADN/metabolismo , Inmunoprecipitación de Cromatina
5.
Int J Mol Sci ; 23(10)2022 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-35628498

RESUMEN

Multiple Myeloma (MM) is a B cell malignancy marked by genomic instability that arises both through pathogenesis and during disease progression. Despite recent advances in therapy, MM remains incurable. Recently, it has been reported that DNA repair can influence genomic changes and drug resistance in MM. The dysregulation of DNA repair function may provide an alternative explanation for genomic instability observed in MM cells and in cells derived from MM patients. This review provides an overview of DNA repair pathways with a special focus on their involvement in MM and discusses the role they play in MM progression and drug resistance. This review highlights how unrepaired DNA damage due to aberrant DNA repair response in MM exacerbates genomic instability and chromosomal abnormalities, enabling MM progression and drug resistance.


Asunto(s)
Mieloma Múltiple , Aberraciones Cromosómicas , Daño del ADN/genética , Reparación del ADN/genética , Inestabilidad Genómica , Humanos , Mieloma Múltiple/genética
6.
Redox Biol ; 52: 102300, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35430547

RESUMEN

Solid tumours are often poorly oxygenated, which confers resistance to standard treatment modalities. Targeting hypoxic tumours requires compounds, such as nitroimidazoles (NIs), equipped with the ability to reach and become activated within diffusion limited tumour niches. NIs become selectively entrapped in hypoxic cells through bioreductive activation, and have shown promise as hypoxia directed therapeutics. However, little is known about their mechanism of action, hindering the broader clinical usage of NIs. Iodoazomycin arabinofuranoside (IAZA) and fluoroazomycin arabinofuranoside (FAZA) are clinically validated 2-NI hypoxic radiotracers with excellent tumour uptake properties. Hypoxic cancer cells have also shown preferential susceptibility to IAZA and FAZA treatment, making them ideal candidates for an in-depth study in a therapeutic setting. Using a head and neck cancer model, we show that hypoxic cells display higher sensitivity to IAZA and FAZA, where the drugs alter cell morphology, compromise DNA replication, slow down cell cycle progression and induce replication stress, ultimately leading to cytostasis. Effects of IAZA and FAZA on target cellular macromolecules (DNA, proteins and glutathione) were characterized to uncover potential mechanism(s) of action. Covalent binding of these NIs was only observed to cellular proteins, but not to DNA, under hypoxia. While protein levels remained unaffected, catalytic activities of NI target proteins, such as the glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and the detoxification enzyme glutathione S-transferase (GST) were significantly curtailed in response to drug treatment under hypoxia. Intraperitoneal administration of IAZA was well-tolerated in mice and produced early (but transient) growth inhibition of subcutaneous mouse tumours.


Asunto(s)
Neoplasias de Cabeza y Cuello , Nitroimidazoles , Animales , Hipoxia de la Célula , Línea Celular Tumoral , Hipoxia/tratamiento farmacológico , Ratones , Nitroimidazoles/farmacología
7.
Cell Rep ; 38(12): 110536, 2022 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-35320715

RESUMEN

BMI-1 is an essential regulator of transcriptional silencing during development. Recently, the role of BMI-1 in the DNA damage response has gained much attention, but the exact mechanism of how BMI-1 participates in the process is unclear. Here, we establish a role for BMI-1 in the repair of DNA double-strand breaks by homologous recombination (HR), where it promotes DNA end resection. Mechanistically, BMI-1 mediates DNA end resection by facilitating the recruitment of CtIP, thus allowing RPA and RAD51 accumulation at DNA damage sites. Interestingly, treatment with transcription inhibitors rescues the DNA end resection defects of BMI-1-depleted cells, suggesting BMI-1-dependent transcriptional silencing mediates DNA end resection. Moreover, we find that H2A ubiquitylation at K119 (H2AK119ub) promotes end resection. Taken together, our results identify BMI-1-mediated transcriptional silencing and promotion of H2AK119ub deposition as essential regulators of DNA end resection and thus the progression of HR.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN por Recombinación , Índice de Masa Corporal , ADN , Reparación del ADN por Unión de Extremidades , Reparación del ADN , Endodesoxirribonucleasas/metabolismo , Recombinación Homóloga
8.
STAR Protoc ; 3(4): 101861, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36595899

RESUMEN

DNA end resection is a critical step in the homologous recombination pathway of repairing DNA double-strand breaks (DSBs) that can be visualized in cells by detecting the generation of single-stranded DNA (ssDNA) intermediates formed during the resection of the DSBs. Here, we describe quantitative polymerase-chain-reaction-based procedures to quantitatively measure ssDNA intermediates formed during the DNA end resection. Using the ER-AsiSI system, we use differential digestion patterns by restriction endonucleases that digest unresected double-stranded DNA at DSB sites. For complete details on the use and execution of this protocol, please refer to Fitieh et al. (2022).1.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Reparación del ADN/genética , ADN , ADN de Cadena Simple/genética , Reacción en Cadena de la Polimerasa
9.
Int J Mol Sci ; 22(6)2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33804165

RESUMEN

The polycomb group (PcG) proteins are a class of transcriptional repressors that mediate gene silencing through histone post-translational modifications. They are involved in the maintenance of stem cell self-renewal and proliferation, processes that are often dysregulated in cancer. Apart from their canonical functions in epigenetic gene silencing, several studies have uncovered a function for PcG proteins in DNA damage signaling and repair. In particular, members of the poly-comb group complexes (PRC) 1 and 2 have been shown to recruit to sites of DNA damage and mediate DNA double-strand break repair. Here, we review current understanding of the PRCs and their roles in cancer development. We then focus on the PRC1 member BMI1, discussing the current state of knowledge of its role in DNA repair and genome integrity, and outline how it can be targeted pharmacologically.


Asunto(s)
Reparación del ADN/genética , Inestabilidad Genómica/genética , Complejo Represivo Polycomb 1/genética , Proteínas del Grupo Polycomb/genética , Animales , Roturas del ADN de Doble Cadena , Humanos , Complejo Represivo Polycomb 2/genética
10.
Nucleic Acids Res ; 49(2): 928-953, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33406258

RESUMEN

Double-strand breaks and stalled replication forks are a significant threat to genomic stability that can lead to chromosomal rearrangements or cell death. The protein CtIP promotes DNA end resection, an early step in homologous recombination repair, and has been found to protect perturbed forks from excessive nucleolytic degradation. However, it remains unknown how CtIP's function in fork protection is regulated. Here, we show that CtIP recruitment to sites of DNA damage and replication stress is impaired upon global inhibition of SUMOylation. We demonstrate that CtIP is a target for modification by SUMO-2 and that this occurs constitutively during S phase. The modification is dependent on the activities of cyclin-dependent kinases and the PI-3-kinase-related kinase ATR on CtIP's carboxyl-terminal region, an interaction with the replication factor PCNA, and the E3 SUMO ligase PIAS4. We also identify residue K578 as a key residue that contributes to CtIP SUMOylation. Functionally, a CtIP mutant where K578 is substituted with a non-SUMOylatable arginine residue is defective in promoting DNA end resection, homologous recombination, and in protecting stalled replication forks from excessive nucleolytic degradation. Our results shed further light on the tightly coordinated regulation of CtIP by SUMOylation in the maintenance of genome stability.


Asunto(s)
Reparación del ADN por Unión de Extremidades/fisiología , Replicación del ADN , Endodesoxirribonucleasas/fisiología , Procesamiento Proteico-Postraduccional , Sumoilación , Sustitución de Aminoácidos , Arginina/química , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular , Quinasas Ciclina-Dependientes/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades/genética , Endodesoxirribonucleasas/química , Endodesoxirribonucleasas/metabolismo , Genes Reporteros , Inestabilidad Genómica , Humanos , Lisina/química , Proteínas de Unión a Poli-ADP-Ribosa/fisiología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Inhibidoras de STAT Activados/fisiología , Mapeo de Interacción de Proteínas , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes de Fusión/metabolismo , Reparación del ADN por Recombinación/genética , Reparación del ADN por Recombinación/fisiología
11.
Cell Cycle ; 15(1): 84-94, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26771713

RESUMEN

Recent studies have found BCL10 can localize to the nucleus and that this is linked to tumor aggression and poorer prognosis. These studies suggest that BCL10 localization plays a novel role in the nucleus that may contribute to cellular transformation and carcinogenesis. In this study, we show that BCL10 functions as part of the DNA damage response (DDR). We found that BCL10 facilitates the rapid recruitment of RPA, BRCA1 and RAD51 to sites of DNA damage. Furthermore, we also found that ATM phosphorylates BCL10 in response to DNA damage. Functionally, BCL10 promoted DNA double-strand breaks repair, enhancing cell survival after DNA damage. Taken together our results suggest a novel role for BCL10 in the repair of DNA lesions.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Roturas del ADN de Doble Cadena , Daño del ADN/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteína 10 de la LLC-Linfoma de Células B , Sitios de Unión/fisiología , Línea Celular Tumoral , Supervivencia Celular/fisiología , Humanos
12.
Nat Cell Biol ; 17(11): 1446-57, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26502055

RESUMEN

DNA double-strand breaks (DSBs) are repaired mainly by non-homologous end joining or homologous recombination (HR). Cell cycle stage and DNA end resection are believed to regulate the commitment to HR repair. Here we identify RNF138 as a ubiquitin E3 ligase that regulates the HR pathway. RNF138 is recruited to DNA damage sites through zinc fingers that have a strong preference for DNA with 5'- or 3'-single-stranded overhangs. RNF138 stimulates DNA end resection and promotes ATR-dependent signalling and DSB repair by HR, thereby contributing to cell survival on exposure to DSB-inducing agents. Finally, we establish that RNF138-dependent Ku removal from DNA breaks is one mechanism whereby RNF138 can promote HR. These results establish RNF138 as an important regulator of DSB repair pathway choice.


Asunto(s)
Roturas del ADN de Doble Cadena , ADN Helicasas/metabolismo , Reparación del ADN , ADN de Neoplasias/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular Tumoral , Reparación del ADN por Unión de Extremidades , ADN Helicasas/genética , ADN de Neoplasias/genética , ADN de Cadena Simple/genética , ADN de Cadena Simple/metabolismo , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Autoantígeno Ku , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Células MCF-7 , Microscopía Confocal , Mutación , Unión Proteica , Interferencia de ARN , Reparación del ADN por Recombinación , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
13.
Cancer Res ; 74(16): 4282-94, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24894717

RESUMEN

The BRCA1-associated deubiquitylase BAP1 is mutated in several cancers, most notably mesothelioma and melanoma, where it is thought to promote oncogenesis. In this study, we present evidence that BAP1 functions as part of the DNA damage response (DDR). We found that BAP1 mediates rapid poly(ADP-ribose)-dependent recruitment of the polycomb deubiquitylase complex PR-DUB to sites of DNA damage. Furthermore, we identified BAP1 as a phosphorylation target for the DDR kinase ATM. Functionally, BAP1 promoted repair of DNA double-strand breaks, enhancing cell survival after DNA damage. Our results highlight the importance of ubiquitin turnover at sites of DNA damage, and they provide a mechanism to account for the tumor-suppressive function of BAP1.


Asunto(s)
Roturas del ADN de Doble Cadena , ADN de Neoplasias/genética , Mutación de Línea Germinal , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Línea Celular Tumoral , Reparación del ADN , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Osteosarcoma/genética , Osteosarcoma/metabolismo , Transfección , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo
14.
Cell Cycle ; 12(16): 2675-83, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23907130

RESUMEN

Polycomb protein histone methyltransferase, enhancer of Zeste homolog 2 (EZH2), is frequently overexpressed in human malignancy and is implicated in cancer cell proliferation and invasion. However, it is largely unknown whether EZH2 has a role in modulating the DNA damage response. Here, we show that polycomb repressive complex 2 (PRC2) is recruited to sites of DNA damage. This recruitment is independent of histone 2A variant X (H2AX) and the PI-3-related kinases ATM and DNA-PKcs. We establish that PARP activity is required for retaining PRC2 at sites of DNA damage. Furthermore, depletion of EZH2 in cells decreases the efficiency of DSB repair and increases sensitivity of cells to gamma-irradiation. These data unravel a crucial role of PRC2 in determining cancer cellular sensitivity following DNA damage and suggest that therapeutic targeting of EZH2 activity might serve as a strategy for improving conventional chemotherapy in a given malignancy.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN/fisiología , Complejo Represivo Polycomb 2/fisiología , Inmunoprecipitación de Cromatina , Ensayo de Unidades Formadoras de Colonias , Proteína Potenciadora del Homólogo Zeste 2 , Humanos , Microscopía Fluorescente , Complejo Represivo Polycomb 2/metabolismo
15.
J Biol Chem ; 288(37): 26944-54, 2013 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-23902761

RESUMEN

Polycomb-repressive complex 1 (PRC1)-mediated histone ubiquitylation plays an important role in aberrant gene silencing in human cancers and is a potential target for cancer therapy. Here we show that 2-pyridine-3-yl-methylene-indan-1,3-dione (PRT4165) is a potent inhibitor of PRC1-mediated H2A ubiquitylation in vivo and in vitro. The drug also inhibits the accumulation of all detectable ubiquitin at sites of DNA double-strand breaks (DSBs), the retention of several DNA damage response proteins in foci that form around DSBs, and the repair of the DSBs. In vitro E3 ubiquitin ligase activity assays revealed that PRT4165 inhibits both RNF2 and RING 1A, which are partially redundant paralogues that together account for the E3 ubiquitin ligase activity found in PRC1 complexes, but not RNF8 nor RNF168. Because ubiquitylation is completely inhibited despite the efficient recruitment of RNF8 to DSBs, our results suggest that PRC1-mediated monoubiquitylation is required for subsequent RNF8- and/or RNF168-mediated polyubiquitylation. Our results demonstrate the unique feature of PRT4165 as a novel chromatin-remodeling compound and provide a new tool for the inhibition of ubiquitylation signaling at DNA double-strand breaks.


Asunto(s)
Daño del ADN/efectos de los fármacos , Histonas/química , Indanos/química , Complejo Represivo Polycomb 1/antagonistas & inhibidores , Piridinas/química , Ubiquitina/metabolismo , Antineoplásicos/química , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Cromatina/metabolismo , ADN/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Microscopía Fluorescente , Transducción de Señal/efectos de los fármacos , Ubiquitina/química , Ubiquitina-Proteína Ligasas/metabolismo
16.
Nucleic Acids Res ; 40(12): 5497-510, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22402492

RESUMEN

Polycomb group (PcG) proteins are involved in epigenetic silencing where they function as major determinants of cell identity, stem cell pluripotency and the epigenetic gene silencing involved in cancer development. Recently numerous PcG proteins, including CBX4, have been shown to accumulate at sites of DNA damage. However, it remains unclear whether or not CBX4 or its E3 sumo ligase activity is directly involved in the DNA damage response (DDR). Here we define a novel role for CBX4 as an early DDR protein that mediates SUMO conjugation at sites of DNA lesions. DNA damage stimulates sumoylation of BMI1 by CBX4 at lysine 88, which is required for the accumulation of BMI1 at DNA damage sites. Moreover, we establish that CBX4 recruitment to the sites of laser micro-irradiation-induced DNA damage requires PARP activity but does not require H2AX, RNF8, BMI1 nor PI-3-related kinases. The importance of CBX4 in the DDR was confirmed by the depletion of CBX4, which resulted in decreased cellular resistance to ionizing radiation. Our results reveal a direct role for CBX4 in the DDR pathway.


Asunto(s)
Daño del ADN , Reparación del ADN , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Proteínas Represoras/fisiología , Sumoilación , Animales , Línea Celular , Roturas del ADN de Doble Cadena , Células HEK293 , Humanos , Ligasas , Lisina/metabolismo , Ratones , Proteínas Nucleares/química , Poli(ADP-Ribosa) Polimerasas/metabolismo , Complejo Represivo Polycomb 1 , Proteínas del Grupo Polycomb , Proteínas Inhibidoras de STAT Activados/metabolismo , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/química , Proteínas Represoras/química , Ubiquitina-Proteína Ligasas
17.
J Cell Biol ; 191(1): 45-60, 2010 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-20921134

RESUMEN

Polycomb group (PcG) proteins are major determinants of cell identity, stem cell pluripotency, and epigenetic gene silencing during development. The polycomb repressive complex 1, which contains BMI1, RING1, and RING2, functions as an E3-ubuiquitin ligase. We found that BMI1 and RING2 are recruited to sites of DNA double-strand breaks (DSBs) where they contribute to the ubiquitylation of γ-H2AX. In the absence of BMI1, several proteins dependent on ubiquitin signaling, including 53BP1, BRCA1, and RAP80, are impaired in recruitment to DSBs. Loss of BMI1 sensitizes cells to ionizing radiation to the same extent as loss of RNF8. The simultaneous depletion of both proteins revealed an additive increase in radiation sensitivity. These data uncover an unexpected link between the polycomb and the DNA damage response pathways, and suggest a novel function for BMI1 in maintaining genomic stability.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Histonas/metabolismo , Proteínas Nucleares/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Represoras/fisiología , Animales , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/fisiología , Línea Celular , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Humanos , Ratones , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Complejo Represivo Polycomb 1 , Señales de Clasificación de Proteína , Transporte de Proteínas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina-Proteína Ligasas , Ubiquitinación
18.
Nat Protoc ; 3(7): 1187-93, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18600224

RESUMEN

Phosphorylation of histone protein H2AX on serine 139 (gamma-H2AX) occurs at sites flanking DNA double-stranded breaks (DSBs) and can provide a measure of the number of DSBs within a cell. We describe a flow cytometry-based method optimized to measure gamma-H2AX in nonfixed mononuclear blood cells as well as in cultured cells, which is more sensitive and involves less steps compared with protocols involving fixed cells. This method can be used to monitor induction of gamma-H2AX in mononuclear cells from cancer patients undergoing radiotherapy and for detection of gamma-H2AX throughout the cell cycle in cultured cells. The method is based on the fact that H2AX like other histone proteins are retained in the nucleus when cells are lysed at physiological salt concentrations. Cells are therefore added without fixation to a solution containing detergent to lyse the cells along with a fluorescein isothiocyanate-labeled monoclonal gamma-H2AX antibody, DNA staining dye and blocking agents. The stained nuclei can be analyzed by flow cytometry to monitor the level of gamma-H2AX to determine the level of DSBs and DNA content and to determine the cell cycle stage. The omission of fixation simplifies staining and enhances the sensitivity. This protocol can be completed within 4-6 h.


Asunto(s)
Roturas del ADN de Doble Cadena , Citometría de Flujo/métodos , Histonas/análisis , Leucocitos Mononucleares/química , Núcleo Celular/química , Células Cultivadas , Humanos , Factores de Tiempo
19.
Environ Mol Mutagen ; 49(1): 73-82, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18095327

RESUMEN

In recent years, several histone modifications have been implicated in the cellular response to DNA double-strand breaks (DSBs). One of the best characterized histone modifications important in DSB repair is the phosphorylation of histone H2A variant, H2A.X. In response to DSBs, H2A.X is phosphorylated and this phosphorylation is required for DSB signaling and the retention of repair proteins at the break site. Despite the existing picture that the function of H2A.X is to promote DNA repair, very recent data suggest that the phosphorylation of histone H2A.X has additional functions. This is analogous to histone H3 phosphorylation on serine 10, which participates in seemingly incompatible functions--transcriptional activation and mitosis. In this review, we discuss the role of histone H2A.X in maintaining genomic stability and review emerging evidence that histone H2A.X is multifunctional.


Asunto(s)
Roturas del ADN de Doble Cadena , ADN/genética , Inestabilidad Genómica , Histonas/fisiología , Animales , Biomarcadores/análisis , Histonas/análisis , Humanos
20.
Nucleic Acids Res ; 35(5): e36, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17284459

RESUMEN

Phosphorylation of histone H2AX on serine 139 (gamma-H2AX, gammaH2AX) occurs at sites flanking DNA double-strand breaks (DSBs) and can provide a measure of the number of DSBs within a cell. Here we describe a rapid and simple flow-cytometry-based method, optimized to measure gamma-H2AX in non-fixed peripheral blood cells. No DSB induced signal was observed in H2AX-/- cells indicating that our FACS method specifically recognized gamma-H2AX accumulation. The gamma-H2AX assay was capable of detecting DNA damage at levels 100-fold below the detection limit of the alkaline comet assay. The gamma-H2AX signal was quantitative with a linear increase of the gamma-H2AX signal over two orders of magnitude. We found that all nucleated blood cell types examined, including the short-lived neutrophils induce gamma-H2AX in response to DSBs. Interindividual difference in the gamma-H2AX signal in response to ionizing radiation and the DSB-inducing drug calicheamicin was almost 2-fold in blood cells from patients, indicating that the amount of gamma-H2AX produced in response to a given dose of radiation varies significantly in the human population. This simple method could be used to monitor response to radiation or DNA-damaging drugs.


Asunto(s)
Roturas del ADN de Doble Cadena , Citometría de Flujo/métodos , Histonas/análisis , Leucocitos/química , Aminoglicósidos/toxicidad , Animales , Antibióticos Antineoplásicos/toxicidad , Células Cultivadas , Enediinos/toxicidad , Fluoresceína-5-Isotiocianato/análisis , Colorantes Fluorescentes/análisis , Humanos , Leucocitos/efectos de los fármacos , Leucocitos/efectos de la radiación , Ratones , Radiación Ionizante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...