Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nucleic Acids Res ; 52(9): 5067-5087, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38416570

RESUMEN

CSB (Cockayne syndrome group B) and SMARCAL1 (SWI/SNF-related, matrix-associated, actin-dependent, regulator of chromatin, subfamily A-like 1) are DNA translocases that belong to the SNF2 helicase family. They both are enriched at stalled replication forks. While SMARCAL1 is recruited by RPA32 to stalled forks, little is known about whether RPA32 also regulates CSB's association with stalled forks. Here, we report that CSB directly interacts with RPA, at least in part via a RPA32C-interacting motif within the N-terminal region of CSB. Modeling of the CSB-RPA32C interaction suggests that CSB binds the RPA32C surface previously shown to be important for binding of UNG2 and SMARCAL1. We show that this interaction is necessary for promoting fork slowing and fork degradation in BRCA2-deficient cells but dispensable for mediating restart of stalled forks. CSB competes with SMARCAL1 for RPA32 at stalled forks and acts non-redundantly with SMARCAL1 to restrain fork progression in response to mild replication stress. In contrast to CSB stimulated restart of stalled forks, SMARCAL1 inhibits restart of stalled forks in BRCA2-deficient cells, likely by suppressing BIR-mediated repair of collapsed forks. Loss of CSB leads to re-sensitization of SMARCAL1-depleted BRCA2-deficient cells to chemodrugs, underscoring a role of CSB in targeted cancer therapy.


Asunto(s)
Proteína BRCA2 , ADN Helicasas , Enzimas Reparadoras del ADN , Replicación del ADN , Proteínas de Unión a Poli-ADP-Ribosa , Proteína de Replicación A , ADN Helicasas/metabolismo , ADN Helicasas/genética , Humanos , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteína BRCA2/metabolismo , Proteína BRCA2/genética , Enzimas Reparadoras del ADN/metabolismo , Enzimas Reparadoras del ADN/genética , Proteína de Replicación A/metabolismo , Proteína de Replicación A/genética , Unión Proteica , Línea Celular Tumoral , Reparación del ADN
2.
Int J Mol Sci ; 24(15)2023 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-37569794

RESUMEN

Topoisomerase inhibitor camptothecin (CPT) induces fork stalling and is highly toxic to proliferating cells. However, how cells respond to CPT-induced fork stalling has not been fully characterized. Here, we report that Cockayne syndrome group B (CSB) protein inhibits PRIMPOL-dependent fork repriming in response to a low dose of CPT. At a high concentration of CPT, CSB is required to promote the restart of DNA replication through MUS81-RAD52-POLD3-dependent break-induced replication (BIR). In the absence of CSB, resumption of DNA synthesis at a high concentration of CPT can occur through POLQ-LIG3-, LIG4-, or PRIMPOL-dependent pathways, which are inhibited, respectively, by RAD51, BRCA1, and BRCA2 proteins. POLQ and LIG3 are core components of alternative end joining (Alt-EJ), whereas LIG4 is a core component of nonhomologous end joining (NHEJ). These results suggest that CSB regulates fork restart pathway choice following high-dosage CPT-induced fork stalling, promoting BIR but inhibiting Alt-EJ, NHEJ, and fork repriming. We find that loss of CSB and BRCA2 is a toxic combination to genomic stability and cell survival at a high concentration of CPT, which is likely due to accumulation of ssDNA gaps, underscoring an important role of CSB in regulating the therapy response in cancers lacking functional BRCA2.


Asunto(s)
Reparación del ADN , Replicación del ADN , Reparación del ADN por Unión de Extremidades , Camptotecina/farmacología
3.
DNA Repair (Amst) ; 116: 103354, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35738143

RESUMEN

Mitotic DNA synthesis, also known as MiDAS, has been suggested to be a form of RAD52-dependent break-induced replication (BIR) that repairs under-replicated DNA regions of the genome in mitosis prior to chromosome segregation. Cockayne syndrome group B (CSB) protein, a chromatin remodeler of the SNF2 family, has been implicated in RAD52-dependent BIR repair of stalled replication forks. However, whether CSB plays a role in MiDAS has not been characterized. Here, we report that CSB functions epistatically with RAD52 to promote MiDAS at common fragile sites in response to replication stress, and prevents genomic instability associated with defects in MiDAS. We show that CSB is dependent upon the conserved phenylalanine at position 796 (F796), which lies in the recently-reported pulling pin that is required for CSB's translocase activity, to mediate MiDAS, suggesting that CSB uses its DNA translocase activity to promote MiDAS. Structural analysis reveals that CSB shares with a subset of SNF2 family proteins a translocase regulatory region (TRR), which is important for CSB's function in MiDAS. We further demonstrate that phosphorylation of S1013 in the TRR regulates the function of CSB in MiDAS and restart of stalled forks but not in fork degradation in BRCA2-deficient cells and UV repair. Taken together, these results suggest that the DNA translocase activity of CSB in vivo is likely to be highly regulated by post-translational modification in a context-specific manner.


Asunto(s)
Síndrome de Cockayne , Cromatina , Síndrome de Cockayne/genética , ADN/metabolismo , Reparación del ADN , Enzimas Reparadoras del ADN/metabolismo , Replicación del ADN , Humanos , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo
4.
Nucleic Acids Res ; 49(22): 12836-12854, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34871413

RESUMEN

Cockayne syndrome group B (CSB) protein has been implicated in the repair of a variety of DNA lesions that induce replication stress. However, little is known about its role at stalled replication forks. Here, we report that CSB is recruited to stalled forks in a manner dependent upon its T1031 phosphorylation by CDK. While dispensable for MRE11 association with stalled forks in wild-type cells, CSB is required for further accumulation of MRE11 at stalled forks in BRCA1/2-deficient cells. CSB promotes MRE11-mediated fork degradation in BRCA1/2-deficient cells. CSB possesses an intrinsic ATP-dependent fork reversal activity in vitro, which is activated upon removal of its N-terminal region that is known to autoinhibit CSB's ATPase domain. CSB functions similarly to fork reversal factors SMARCAL1, ZRANB3 and HLTF to regulate slowdown in fork progression upon exposure to replication stress, indicative of a role of CSB in fork reversal in vivo. Furthermore, CSB not only acts epistatically with MRE11 to facilitate fork restart but also promotes RAD52-mediated break-induced replication repair of double-strand breaks arising from cleavage of stalled forks by MUS81 in BRCA1/2-deficient cells. Loss of CSB exacerbates chemosensitivity in BRCA1/2-deficient cells, underscoring an important role of CSB in the treatment of cancer lacking functional BRCA1/2.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , ADN Helicasas/genética , Enzimas Reparadoras del ADN/genética , Reparación del ADN , ADN/genética , Proteína Homóloga de MRE11/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteína BRCA1/deficiencia , Proteína BRCA1/metabolismo , Proteína BRCA2/deficiencia , Proteína BRCA2/metabolismo , Línea Celular , Línea Celular Tumoral , ADN/química , ADN/metabolismo , Roturas del ADN de Doble Cadena , ADN Helicasas/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Replicación del ADN/genética , Células HCT116 , Células HEK293 , Humanos , Proteína Homóloga de MRE11/metabolismo , Mutación , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Interferencia de ARN
5.
Int J Mol Sci ; 22(7)2021 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-33806087

RESUMEN

Cockayne syndrome group B protein (CSB), a member of the SWI/SNF superfamily, resides in an elongating RNA polymerase II (RNAPII) complex and regulates transcription elongation. CSB contains a C-terminal winged helix domain (WHD) that binds to ubiquitin and plays an important role in DNA repair. However, little is known about the role of the CSB-WHD in transcription regulation. Here, we report that CSB is dependent upon its WHD to regulate RNAPII abundance at promoter proximal pause (PPP) sites of several actively transcribed genes, a key step in the regulation of transcription elongation. We show that two ubiquitin binding-defective mutations in the CSB-WHD, which impair CSB's ability to promote cell survival in response to treatment with cisplatin, have little impact on its ability to stimulate RNAPII occupancy at PPP sites. In addition, we demonstrate that two cancer-associated CSB mutations, which are located on the opposite side of the CSB-WHD away from its ubiquitin-binding pocket, impair CSB's ability to promote RNAPII occupancy at PPP sites. Taken together, these results suggest that CSB promotes RNAPII association with PPP sites in a manner requiring the CSB-WHD but independent of its ubiquitin-binding activity. These results further imply that CSB-mediated RNAPII occupancy at PPP sites is mechanistically separable from CSB-mediated repair of cisplatin-induced DNA damage.


Asunto(s)
ADN Helicasas/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Reparación del ADN , Regulación de la Expresión Génica , Mutación , Neoplasias/genética , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , ARN Polimerasa II/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Cisplatino/efectos adversos , Cisplatino/farmacología , Daño del ADN/efectos de los fármacos , ADN Helicasas/química , Enzimas Reparadoras del ADN/química , Humanos , Proteínas de Unión a Poli-ADP-Ribosa/química , Regiones Promotoras Genéticas , Unión Proteica , Factores de Transcripción/metabolismo , Ubiquitina/metabolismo
6.
J Cell Sci ; 133(4)2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-31974116

RESUMEN

Elevated replication stress is evident at telomeres of about 10-15% of cancer cells, which maintain their telomeres via a homologous recombination (HR)-based mechanism, referred to as alternative lengthening of telomeres (ALT). How ALT cells resolve replication stress to support their growth remains incompletely characterized. Here, we report that CSB (also known as ERCC6) promotes recruitment of HR repair proteins (MRN, BRCA1, BLM and RPA32) and POLD3 to ALT telomeres, a process that requires the ATPase activity of CSB and is controlled by ATM- and CDK2-dependent phosphorylation. Loss of CSB stimulates telomeric recruitment of MUS81 and SLX4, components of the structure-specific MUS81-EME1-SLX1-SLX4 (MUS-SLX) endonuclease complex, suggesting that CSB restricts MUS-SLX-mediated processing of stalled forks at ALT telomeres. Loss of CSB coupled with depletion of SMARCAL1, a chromatin remodeler implicated in catalyzing regression of stalled forks, synergistically promotes not only telomeric recruitment of MUS81 but also the formation of fragile telomeres, the latter of which is reported to arise from fork stalling. These results altogether suggest that CSB-mediated HR repair and SMARCAL1-mediated fork regression cooperate to prevent stalled forks from being processed into fragile telomeres in ALT cells.


Asunto(s)
Homeostasis del Telómero , Telómero , Reparación del ADN , Endonucleasas/metabolismo , Recombinación Homóloga , Telómero/genética , Telómero/metabolismo , Homeostasis del Telómero/genética
7.
Nucleic Acids Res ; 47(20): 10678-10692, 2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31501894

RESUMEN

CSB, a member of the SWI2/SNF2 superfamily, has been implicated in evicting histones to promote the DSB pathway choice towards homologous recombination (HR) repair. However, how CSB promotes HR repair remains poorly characterized. Here we demonstrate that CSB interacts with both MRE11/RAD50/NBS1 (MRN) and BRCA1 in a cell cycle regulated manner, with the former requiring its WHD and occurring predominantly in early S phase. CSB interacts with the BRCT domain of BRCA1 and this interaction is regulated by CDK-dependent phosphorylation of CSB on S1276. The CSB-BRCA1 interaction, which peaks in late S/G2 phase, is responsible for mediating the interaction of CSB with the BRCA1-C complex consisting of BRCA1, MRN and CtIP. While dispensable for histone eviction at DSBs, CSB phosphorylation on S1276 is necessary to promote efficient MRN- and CtIP-mediated DNA end resection, thereby restricting NHEJ and enforcing the DSB repair pathway choice to HR. CSB phosphorylation on S1276 is also necessary to support cell survival in response to DNA damage-inducing agents. These results altogether suggest that CSB interacts with BRCA1 to promote DNA end resection for HR repair and that although prerequisite, CSB-mediated histone eviction alone is insufficient to promote the pathway choice towards HR.


Asunto(s)
Proteína BRCA1/metabolismo , ADN Helicasas/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Reparación del ADN , Endodesoxirribonucleasas/metabolismo , Fase G2 , Complejos Multiproteicos/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Fase S , Proteína BRCA1/química , Camptotecina/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de los fármacos , ADN Helicasas/química , Reparación del ADN/efectos de los fármacos , Enzimas Reparadoras del ADN/química , Fase G2/efectos de los fármacos , Humanos , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Ftalazinas/farmacología , Piperazinas/farmacología , Proteínas de Unión a Poli-ADP-Ribosa/química , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Fase S/efectos de los fármacos , Proteínas de Unión a Telómeros/metabolismo
8.
DNA Repair (Amst) ; 68: 58-67, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29957539

RESUMEN

The ATP-dependent chromatin remodeler CSB is implicated in a variety of different DNA repair mechanisms, including transcription-coupled nucleotide excision repair (TC-NER), base excision repair and DNA double strand break (DSB) repair. However, how CSB is regulated in these various repair processes is not well understood. Here we report that the first 30 amino acids of CSB along with two phosphorylation events on S10 and S158, previously reported to be required for CSB function in homologous recombination (HR)-mediated repair, are dispensable for repairing UV-induced DNA damage, suggesting that the regulation of CSB in these two types of repair are carried out by distinct mechanisms. In addition, we show that although the central ATPase domain of CSB is engaged in interactions with both the N- and C-terminal regions, these interactions are disrupted following UV-induced DNA damage. The UV-induced disengagement of the C-terminal region of CSB from the ATPase domain requires two conserved amino acids W1486 and L1488, which are thought to contribute to the hydrophobic core formation of the winged helix domain (WHD) at its C-terminus. Failure to undergo UV-induced dissociation of the C-terminal region of CSB from the ATPase domain is associated with impairment in its UV-induced chromatin association, its UV-induced post-translational modification as well as cell survival. Collectively, these findings suggest that UV-induced dissociation of CSB domain interactions is a necessary step in repairing UV-induced DNA damage and that the WHD of CSB plays a key role in this dissociation.


Asunto(s)
Daño del ADN , ADN Helicasas/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Reparación del ADN , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Dominios Proteicos , Rayos Ultravioleta , Adenosina Trifosfatasas , Línea Celular , Síndrome de Cockayne , ADN/metabolismo , ADN/efectos de la radiación , ADN Helicasas/efectos de la radiación , Enzimas Reparadoras del ADN/efectos de la radiación , Humanos , Fosforilación , Proteínas de Unión a Poli-ADP-Ribosa/efectos de la radiación , Procesamiento Proteico-Postraduccional
9.
Nat Commun ; 8(1): 1921, 2017 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-29203878

RESUMEN

CSB, a member of the SWI2/SNF2 superfamily, is implicated in DNA double-strand break (DSB) repair. However, how it regulates this repair process is poorly understood. Here we uncover that CSB interacts via its newly identified winged helix domain with RIF1, an effector of 53BP1, and that this interaction mediates CSB recruitment to DSBs in S phase. At DSBs, CSB remodels chromatin by evicting histones, which limits RIF1 and its effector MAD2L2 but promotes BRCA1 accumulation. The chromatin remodeling activity of CSB requires not only damage-induced phosphorylation on S10 by ATM but also cell cycle-dependent phosphorylation on S158 by cyclin A-CDK2. Both modifications modulate the interaction of the CSB N-terminal region with its ATPase domain, the activity of which has been previously reported to be autorepressed by the N-terminal region. These results suggest that ATM and CDK2 control the chromatin remodeling activity of CSB in the regulation of DSB repair pathway choice.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Ensamble y Desensamble de Cromatina , Quinasa 2 Dependiente de la Ciclina/metabolismo , Roturas del ADN de Doble Cadena , ADN Helicasas/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Reparación del ADN , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Secuencias de Aminoácidos , Proteína BRCA1/metabolismo , Línea Celular Tumoral , Ciclina A/metabolismo , Células HCT116 , Humanos , Proteínas Mad2/metabolismo , Fosforilación , Fase S , Proteínas de Unión a Telómeros/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
10.
EMBO J ; 34(10): 1399-416, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25820262

RESUMEN

Mutations of CSB account for the majority of Cockayne syndrome (CS), a devastating hereditary disorder characterized by physical impairment, neurological degeneration and segmental premature aging. Here we report the generation of a human CSB-knockout cell line. We find that CSB facilitates HR and represses NHEJ. Loss of CSB or a CS-associated CSB mutation abrogating its ATPase activity impairs the recruitment of BRCA1, RPA and Rad51 proteins to damaged chromatin but promotes the formation of 53BP1-Rif1 damage foci in S and G2 cells. Depletion of 53BP1 rescues the formation of BRCA1 damage foci in CSB-knockout cells. In addition, knockout of CSB impairs the ATM- and Chk2-mediated DNA damage responses, promoting a premature entry into mitosis. Furthermore, we show that CSB accumulates at sites of DNA double-strand breaks (DSBs) in a transcription-dependent manner. The kinetics of DSB-induced chromatin association of CSB is distinct from that of its UV-induced chromatin association. These results reveal novel, important functions of CSB in regulating the DNA DSB repair pathway choice as well as G2/M checkpoint activation.


Asunto(s)
Roturas del ADN de Doble Cadena , ADN Helicasas/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Puntos de Control del Ciclo Celular/genética , Puntos de Control del Ciclo Celular/fisiología , Línea Celular , Células Cultivadas , Daño del ADN/genética , Humanos , Cinética , Modelos Biológicos , Proteínas de Unión a Poli-ADP-Ribosa , Ubiquitina-Proteína Ligasas/genética
11.
Nucleic Acids Res ; 40(19): 9661-74, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22904069

RESUMEN

The majority of Cockayne syndrome (CS) patients carry a mutation in Cockayne Syndrome group B (CSB), a large nuclear protein implicated in DNA repair, transcription and chromatin remodeling. However, whether CSB may play a role in telomere metabolism has not yet been characterized. Here, we report that CSB physically interacts with TRF2, a duplex telomeric DNA binding protein essential for telomere protection. We find that CSB localizes at a small subset of human telomeres and that it is required for preventing the formation of telomere dysfunction-induced foci (TIF) in CS cells. We find that CS cells or CSB knockdown cells accumulate telomere doublets, the suppression of which requires CSB. We find that overexpression of CSB in CS cells promotes telomerase-dependent telomere lengthening, a phenotype that is associated with a decrease in the amount of telomere-bound TRF1, a negative mediator of telomere length maintenance. Furthermore, we show that CS cells or CSB knockdown cells exhibit misregulation of TERRA, a large non-coding telomere repeat-containing RNA important for telomere maintenance. Taken together, these results suggest that CSB is required for maintaining the homeostatic level of TERRA, telomere length and integrity. These results further imply that CS patients carrying CSB mutations may be defective in telomere maintenance.


Asunto(s)
Síndrome de Cockayne/genética , ADN Helicasas/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Homeostasis del Telómero , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Línea Celular , ADN Helicasas/análisis , ADN Helicasas/genética , Enzimas Reparadoras del ADN/análisis , Enzimas Reparadoras del ADN/genética , Humanos , Mutación , Proteínas de Unión a Poli-ADP-Ribosa , ARN Largo no Codificante/metabolismo , Telomerasa/metabolismo , Telómero/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...