Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Mol Cell ; 84(4): 640-658.e10, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38266639

RESUMEN

The Bloom syndrome helicase BLM interacts with topoisomerase IIIα (TOP3A), RMI1, and RMI2 to form the BTR complex, which dissolves double Holliday junctions and DNA replication intermediates to promote sister chromatid disjunction before cell division. In its absence, structure-specific nucleases like the SMX complex (comprising SLX1-SLX4, MUS81-EME1, and XPF-ERCC1) can cleave joint DNA molecules instead, but cells deficient in both BTR and SMX are not viable. Here, we identify a negative genetic interaction between BLM loss and deficiency in the BRCA1-BARD1 tumor suppressor complex. We show that this is due to a previously overlooked role for BARD1 in recruiting SLX4 to resolve DNA intermediates left unprocessed by BLM in the preceding interphase. Consequently, cells with defective BLM and BRCA1-BARD1 accumulate catastrophic levels of chromosome breakage and micronucleation, leading to cell death. Thus, we reveal mechanistic insights into SLX4 recruitment to DNA lesions, with potential clinical implications for treating BRCA1-deficient tumors.


Asunto(s)
Proteínas de Unión al ADN , Recombinasas , Humanos , ADN/genética , Reparación del ADN , Replicación del ADN , ADN Cruciforme , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Recombinasas/genética , RecQ Helicasas/genética , RecQ Helicasas/metabolismo
2.
Mol Cell ; 82(18): 3382-3397.e7, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36002001

RESUMEN

Aberrant replication causes cells lacking BRCA2 to enter mitosis with under-replicated DNA, which activates a repair mechanism known as mitotic DNA synthesis (MiDAS). Here, we identify genome-wide the sites where MiDAS reactions occur when BRCA2 is abrogated. High-resolution profiling revealed that these sites are different from MiDAS at aphidicolin-induced common fragile sites in that they map to genomic regions replicating in the early S-phase, which are close to early-firing replication origins, are highly transcribed, and display R-loop-forming potential. Both transcription inhibition in early S-phase and RNaseH1 overexpression reduced MiDAS in BRCA2-deficient cells, indicating that transcription-replication conflicts (TRCs) and R-loops are the source of MiDAS. Importantly, the MiDAS sites identified in BRCA2-deficient cells also represent hotspots for genomic rearrangements in BRCA2-mutated breast tumors. Thus, our work provides a mechanism for how tumor-predisposing BRCA2 inactivation links transcription-induced DNA damage with mitotic DNA repair to fuel the genomic instability characteristic of cancer cells.


Asunto(s)
Replicación del ADN , Mitosis , Afidicolina/farmacología , Proteína BRCA2/genética , Sitios Frágiles del Cromosoma/genética , ADN/genética , Daño del ADN , Inestabilidad Genómica , Humanos , Mitosis/genética
3.
Mol Cell ; 74(3): 571-583.e8, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30898438

RESUMEN

In mitosis, cells inactivate DNA double-strand break (DSB) repair pathways to preserve genome stability. However, some early signaling events still occur, such as recruitment of the scaffold protein MDC1 to phosphorylated histone H2AX at DSBs. Yet, it remains unclear whether these events are important for maintaining genome stability during mitosis. Here, we identify a highly conserved protein-interaction surface in MDC1 that is phosphorylated by CK2 and recognized by the DNA-damage response mediator protein TOPBP1. Disruption of MDC1-TOPBP1 binding causes a specific loss of TOPBP1 recruitment to DSBs in mitotic but not interphase cells, accompanied by mitotic radiosensitivity, increased micronuclei, and chromosomal instability. Mechanistically, we find that TOPBP1 forms filamentous structures capable of bridging MDC1 foci in mitosis, indicating that MDC1-TOPBP1 complexes tether DSBs until repair is reactivated in the following G1 phase. Thus, we reveal an important, hitherto-unnoticed cooperation between MDC1 and TOPBP1 in maintaining genome stability during cell division.


Asunto(s)
Proteínas Portadoras/genética , Inestabilidad Cromosómica/genética , Proteínas de Unión al ADN/genética , Mitosis/genética , Proteínas Nucleares/genética , Transactivadores/genética , Proteínas Adaptadoras Transductoras de Señales , Proteínas de Ciclo Celular , Roturas del ADN de Doble Cadena , Daño del ADN/genética , Reparación del ADN/genética , Fase G1/genética , Genoma Humano/genética , Inestabilidad Genómica/genética , Histonas , Humanos , Fosforilación , Transducción de Señal/genética
4.
Mol Cell ; 66(6): 801-817, 2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-28622525

RESUMEN

In vertebrate cells, the DNA damage response is controlled by three related kinases: ATM, ATR, and DNA-PK. It has been 20 years since the cloning of ATR, the last of the three to be identified. During this time, our understanding of how these kinases regulate DNA repair and associated events has grown profoundly, although major questions remain unanswered. Here, we provide a historical perspective of their discovery and discuss their established functions in sensing and responding to genotoxic stress. We also highlight what is known regarding their structural similarities and common mechanisms of regulation, as well as emerging non-canonical roles and how our knowledge of ATM, ATR, and DNA-PK is being translated to benefit human health.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Unión al Calcio/metabolismo , Núcleo Celular/enzimología , Daño del ADN , Reparación del ADN , Animales , Antineoplásicos/uso terapéutico , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Proteínas de la Ataxia Telangiectasia Mutada/química , Proteínas de la Ataxia Telangiectasia Mutada/historia , Proteínas de Unión al Calcio/antagonistas & inhibidores , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/historia , Reparación del ADN/efectos de los fármacos , Activación Enzimática , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Modelos Moleculares , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/genética , Neoplasias/patología , Fosforilación , Conformación Proteica , Inhibidores de Proteínas Quinasas/uso terapéutico , Transducción de Señal , Relación Estructura-Actividad , Proteína p53 Supresora de Tumor/metabolismo
5.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35115399

RESUMEN

The RecQ-like helicase BLM cooperates with topoisomerase IIIα, RMI1, and RMI2 in a heterotetrameric complex (the "Bloom syndrome complex") for dissolution of double Holliday junctions, key intermediates in homologous recombination. Mutations in any component of the Bloom syndrome complex can cause genome instability and a highly cancer-prone disorder called Bloom syndrome. Some heterozygous carriers are also predisposed to breast cancer. To understand how the activities of BLM helicase and topoisomerase IIIα are coupled, we purified the active four-subunit complex. Chemical cross-linking and mass spectrometry revealed a unique architecture that links the helicase and topoisomerase domains. Using biochemical experiments, we demonstrated dimerization mediated by the N terminus of BLM with a 2:2:2:2 stoichiometry within the Bloom syndrome complex. We identified mutations that independently abrogate dimerization or association of BLM with RMI1, and we show that both are dysfunctional for dissolution using in vitro assays and cause genome instability and synthetic lethal interactions with GEN1/MUS81 in cells. Truncated BLM can also inhibit the activity of full-length BLM in mixed dimers, suggesting a putative mechanism of dominant-negative action in carriers of BLM truncation alleles. Our results identify critical molecular determinants of Bloom syndrome complex assembly required for double Holliday junction dissolution and maintenance of genome stability.


Asunto(s)
Síndrome de Bloom/genética , ADN Cruciforme/genética , Inestabilidad Genómica/genética , Alelos , Proteínas Portadoras/genética , Línea Celular , ADN-Topoisomerasas de Tipo I/genética , Humanos , Mutación/genética , Unión Proteica/genética , RecQ Helicasas/genética , Recombinación Genética/genética , Solubilidad
6.
Trends Biochem Sci ; 45(4): 321-331, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32001093

RESUMEN

DNA double-strand breaks (DSBs) are highly toxic lesions that can lead to chromosomal instability if they are not repaired correctly. DSBs are especially dangerous in mitosis when cells go through the complex process of equal chromosome segregation into daughter cells. When cells encounter DSBs in interphase, they are able to arrest the cell cycle until the breaks are repaired before entering mitosis. However, when DSBs occur during mitosis, cells no longer arrest but prioritize completion of cell division over repair of DNA damage. This review focuses on recent progress in our understanding of the mechanisms that allow mitotic cells to postpone DSB repair without accumulating massive chromosomal instability. Additionally, we review possible physiological consequences of failed DSB responses in mitosis.


Asunto(s)
Células/metabolismo , Mitosis , Roturas del ADN de Doble Cadena , Daño del ADN , Humanos
7.
Genes Dev ; 30(19): 2152-2157, 2016 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-27798842

RESUMEN

PAXX was identified recently as a novel nonhomologous end-joining DNA repair factor in human cells. To characterize its physiological roles, we generated Paxx-deficient mice. Like Xlf-/- mice, Paxx-/- mice are viable, grow normally, and are fertile but show mild radiosensitivity. Strikingly, while Paxx loss is epistatic with Ku80, Lig4, and Atm deficiency, Paxx/Xlf double-knockout mice display embryonic lethality associated with genomic instability, cell death in the central nervous system, and an almost complete block in lymphogenesis, phenotypes that closely resemble those of Xrcc4-/- and Lig4-/- mice. Thus, combined loss of Paxx and Xlf is synthetic-lethal in mammals.


Asunto(s)
Proteínas de Unión al ADN/genética , Desarrollo Embrionario/genética , Mutaciones Letales Sintéticas/genética , Trisacáridos/genética , Animales , Apoptosis/genética , Proteínas de Unión al ADN/metabolismo , Epistasis Genética , Inestabilidad Genómica/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Tolerancia a Radiación/genética , Trisacáridos/metabolismo
8.
Mol Cell ; 60(3): 362-73, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26455393

RESUMEN

Repair of DNA double-strand breaks is crucial for maintaining genome integrity and is governed by post-translational modifications such as protein ubiquitylation. Here, we establish that the deubiquitylating enzyme USP4 promotes DNA-end resection and DNA repair by homologous recombination. We also report that USP4 interacts with CtIP and the MRE11-RAD50-NBS1 (MRN) complex and is required for CtIP recruitment to DNA damage sites. Furthermore, we show that USP4 is ubiquitylated on multiple sites including those on cysteine residues and that deubiquitylation of these sites requires USP4 catalytic activity and is required for USP4 to interact with CtIP/MRN and to promote CtIP recruitment and DNA repair. Lastly, we establish that regulation of interactor binding by ubiquitylation occurs more generally among USP-family enzymes. Our findings thus identify USP4 as a novel DNA repair regulator and invoke a model in which ubiquitin adducts regulate USP enzyme interactions and functions.


Asunto(s)
Roturas del ADN de Doble Cadena , Modelos Biológicos , Reparación del ADN por Recombinación , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinación/fisiología , Ácido Anhídrido Hidrolasas , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Endodesoxirribonucleasas , Humanos , Proteína Homóloga de MRE11 , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ubiquitina Tiolesterasa/genética , Proteasas Ubiquitina-Específicas
9.
Mol Cell ; 57(6): 1133-1141, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25794620

RESUMEN

The Bloom syndrome helicase BLM and topoisomerase-IIß-binding protein 1 (TopBP1) are key regulators of genome stability. It was recently proposed that BLM phosphorylation on Ser338 mediates its interaction with TopBP1, to protect BLM from ubiquitylation and degradation (Wang et al., 2013). Here, we show that the BLM-TopBP1 interaction does not involve Ser338 but instead requires BLM phosphorylation on Ser304. Furthermore, we establish that disrupting this interaction does not markedly affect BLM stability. However, BLM-TopBP1 binding is important for maintaining genome integrity, because in its absence cells display increased sister chromatid exchanges, replication origin firing and chromosomal aberrations. Therefore, the BLM-TopBP1 interaction maintains genome stability not by controlling BLM protein levels, but via another as-yet undetermined mechanism. Finally, we identify critical residues that mediate interactions between TopBP1 and MDC1, and between BLM and TOP3A/RMI1/RMI2. Taken together, our findings provide molecular insights into a key tumor suppressor and genome stability network.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Unión al ADN/metabolismo , Inestabilidad Genómica , Proteínas Nucleares/metabolismo , RecQ Helicasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Aminoácidos , Proteínas Portadoras/genética , Proteínas de Ciclo Celular , ADN-Topoisomerasas de Tipo I/genética , ADN-Topoisomerasas de Tipo I/metabolismo , Proteínas de Unión al ADN/genética , Células HeLa , Humanos , Datos de Secuencia Molecular , Mutación , Proteínas Nucleares/genética , Fosforilación , RecQ Helicasas/genética , Serina/metabolismo , Transactivadores/genética , Transactivadores/metabolismo
10.
Mol Cell ; 59(3): 462-77, 2015 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-26166705

RESUMEN

Recognition and repair of damaged replication forks are essential to maintain genome stability and are coordinated by the combined action of the Fanconi anemia and homologous recombination pathways. These pathways are vital to protect stalled replication forks from uncontrolled nucleolytic activity, which otherwise causes irreparable genomic damage. Here, we identify BOD1L as a component of this fork protection pathway, which safeguards genome stability after replication stress. Loss of BOD1L confers exquisite cellular sensitivity to replication stress and uncontrolled resection of damaged replication forks, due to a failure to stabilize RAD51 at these forks. Blocking DNA2-dependent resection, or downregulation of the helicases BLM and FBH1, suppresses both catastrophic fork processing and the accumulation of chromosomal damage in BOD1L-deficient cells. Thus, our work implicates BOD1L as a critical regulator of genome integrity that restrains nucleolytic degradation of damaged replication forks.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Replicación del ADN , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Línea Celular , Supervivencia Celular , Daño del ADN , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Genoma Humano , Inestabilidad Genómica , Células HeLa , Humanos , RecQ Helicasas/metabolismo
11.
Mol Cell ; 45(4): 505-16, 2012 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-22365830

RESUMEN

DNA double-strand break (DSB) signaling and repair are critical for cell viability, and rely on highly coordinated pathways whose molecular organization is still incompletely understood. Here, we show that heterogeneous nuclear ribonucleoprotein U-like (hnRNPUL) proteins 1 and 2 play key roles in cellular responses to DSBs. We identify human hnRNPUL1 and -2 as binding partners for the DSB sensor complex MRE11-RAD50-NBS1 (MRN) and demonstrate that hnRNPUL1 and -2 are recruited to DNA damage in an interdependent manner that requires MRN. Moreover, we show that hnRNPUL1 and -2 stimulate DNA-end resection and promote ATR-dependent signaling and DSB repair by homologous recombination, thereby contributing to cell survival upon exposure to DSB-inducing agents. Finally, we establish that hnRNPUL1 and -2 function downstream of MRN and CtBP-interacting protein (CtIP) to promote recruitment of the BLM helicase to DNA breaks. Collectively, these results provide insights into how mammalian cells respond to DSBs.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Ribonucleoproteínas Nucleares Heterogéneas/fisiología , Proteínas Nucleares/fisiología , Factores de Transcripción/fisiología , Ácido Anhídrido Hidrolasas , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas Portadoras/fisiología , Proteínas de Ciclo Celular/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Endodesoxirribonucleasas , Ribonucleoproteínas Nucleares Heterogéneas/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Proteína Homóloga de MRE11 , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
Hum Mol Genet ; 21(9): 2005-16, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22279085

RESUMEN

FANCM is the most highly conserved protein within the Fanconi anaemia (FA) tumour suppressor pathway. However, although FANCM contains a helicase domain with translocase activity, this is not required for its role in activating the FA pathway. Instead, we show here that FANCM translocaseactivity is essential for promoting replication fork stability. We demonstrate that cells expressing translocase-defective FANCM show altered global replication dynamics due to increased accumulation of stalled forks that subsequently degenerate into DNA double-strand breaks, leading to ATM activation, CTBP-interacting protein (CTIP)-dependent end resection and homologous recombination repair. Accordingly, abrogation of ATM or CTIP function in FANCM-deficient cells results in decreased cell survival. We also found that FANCM translocase activity protects cells from accumulating 53BP1-OPT domains, which mark lesions resulting from problems arising during replication. Taken together, these data show that FANCM plays an essential role in maintaining chromosomal integrity by promoting the recovery of stalled replication forks and hence preventing tumourigenesis.


Asunto(s)
ADN Helicasas/metabolismo , Replicación del ADN/fisiología , Proteínas de Transporte de Nucleótidos/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Roturas del ADN de Doble Cadena , ADN Helicasas/antagonistas & inhibidores , ADN Helicasas/genética , Reparación del ADN , Replicación del ADN/genética , Proteínas de Unión al ADN/metabolismo , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Técnicas de Inactivación de Genes , Células HEK293 , Células HeLa , Recombinación Homóloga , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Modelos Biológicos , Proteínas de Transporte de Nucleótidos/antagonistas & inhibidores , Proteínas de Transporte de Nucleótidos/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/genética , Proteínas Supresoras de Tumor/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53
13.
EMBO J ; 29(4): 806-18, 2010 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-20057355

RESUMEN

Fanconi anaemia is a chromosomal instability disorder associated with cancer predisposition and bone marrow failure. Among the 13 identified FA gene products only one, the DNA translocase FANCM, has homologues in lower organisms, suggesting a conserved function in DNA metabolism. However, a precise role for FANCM in DNA repair remains elusive. Here, we show a novel function for FANCM that is distinct from its role in the FA pathway: promoting replication fork restart and simultaneously limiting the accumulation of RPA-ssDNA. We show that in DT40 cells this process is controlled by ATR and PLK1, and that in the absence of FANCM, stalled replication forks are unable to resume DNA synthesis and genome duplication is ensured by excess origin firing. Unexpectedly, we also uncover an early role for FANCM in ATR-mediated checkpoint signalling by promoting chromatin retention of TopBP1. Failure to retain TopBP1 on chromatin impacts on the ability of ATR to phosphorylate downstream molecular targets, including Chk1 and SMC1. Our data therefore indicate a fundamental role for FANCM in the maintenance of genome integrity during S phase.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , ADN Helicasas/metabolismo , Replicación del ADN/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas Aviares/deficiencia , Proteínas Aviares/genética , Proteínas Aviares/metabolismo , Línea Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Pollos , Cromatina/metabolismo , ADN Helicasas/deficiencia , ADN Helicasas/genética , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Activación Enzimática , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Fase S , Transducción de Señal , Estrés Fisiológico , Proteínas Supresoras de Tumor/metabolismo , Quinasa Tipo Polo 1
14.
Proc Natl Acad Sci U S A ; 107(27): 12251-6, 2010 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-20566845

RESUMEN

Activation of the cellular DNA damage response is detrimental to adenovirus (Ad) infection. Ad has therefore evolved a number of strategies to inhibit ATM- and ATR-dependent signaling pathways during infection. Recent work suggests that the Ad5 E4orf3 protein prevents ATR activation through its ability to mislocalize the MRN complex. Here we provide evidence to indicate that Ad12 has evolved a different strategy from Ad5 to inhibit ATR. We show that Ad12 utilizes a CUL2/RBX1/elongin C-containing ubiquitin ligase to promote the proteasomal degradation of the ATR activator protein topoisomerase-IIbeta-binding protein 1 (TOPBP1). Ad12 also uses this complex to degrade p53 during infection, in contrast to Ad5, which requires a CUL5-based ubiquitin ligase. Although Ad12-mediated degradation of p53 is dependent upon both E1B-55K and E4orf6, Ad12-mediated degradation of TOPBP1 is solely dependent on E4orf6. We propose that Ad12 E4orf6 has two principal activities: to recruit the CUL2-based ubiquitin ligase and to act as substrate receptor for TOPBP1. In support of the idea that Ad12 E4orf6 specifically prevents ATR activation during infection by targeting TOPBP1 for degradation, we demonstrate that Ad12 E4orf6 can inhibit the ATR-dependent phosphorylation of CHK1 in response to replication stress. Taken together, these data provide insights into how Ad modulates ATR signaling pathways during infection.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Virales/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Adenoviridae/fisiología , Proteínas E1B de Adenovirus/genética , Proteínas E1B de Adenovirus/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada , Western Blotting , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Proteínas de Unión al ADN/genética , Elonguina , Técnica del Anticuerpo Fluorescente , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Microscopía Confocal , Mutación , Proteínas Nucleares/genética , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Virales/genética
15.
Nat Rev Cancer ; 23(2): 78-94, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36471053

RESUMEN

Cells have evolved a complex network of biochemical pathways, collectively known as the DNA damage response (DDR), to prevent detrimental mutations from being passed on to their progeny. The DDR coordinates DNA repair with cell-cycle checkpoint activation and other global cellular responses. Genes encoding DDR factors are frequently mutated in cancer, causing genomic instability, an intrinsic feature of many tumours that underlies their ability to grow, metastasize and respond to treatments that inflict DNA damage (such as radiotherapy). One instance where we have greater insight into how genetic DDR abrogation impacts on therapy responses is in tumours with mutated BRCA1 or BRCA2. Due to compromised homologous recombination DNA repair, these tumours rely on alternative repair mechanisms and are susceptible to chemical inhibitors of poly(ADP-ribose) polymerase (PARP), which specifically kill homologous recombination-deficient cancer cells, and have become a paradigm for targeted cancer therapy. It is now clear that many other synthetic-lethal relationships exist between DDR genes. Crucially, some of these interactions could be exploited in the clinic to target tumours that become resistant to PARP inhibition. In this Review, we discuss state-of-the-art strategies for DDR inactivation using small-molecule inhibitors and highlight those compounds currently being evaluated in the clinic.


Asunto(s)
Neoplasias , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Humanos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Reparación del ADN , Daño del ADN , Mutación , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Poli(ADP-Ribosa) Polimerasas/uso terapéutico
16.
J Virol ; 85(5): 2201-11, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21159879

RESUMEN

Adenovirus type 5 (Ad5) inactivates the host cell DNA damage response by facilitating the degradation of Mre11, DNA ligase IV, and p53. In the case of p53, this is achieved through polyubiquitylation by Ad5E1B55K and Ad5E4orf6, which recruit a Cul5-based E3 ubiquitin ligase. Recent evidence indicates that this paradigm does not apply to other adenovirus serotypes, since Ad12, but not Ad5, causes the degradation of TOPBP1 through the action of E4orf6 alone and a Cul2-based E3 ubiquitin ligase. We now have extended these studies to adenovirus groups A to E. While infection by Ad4, Ad5, and Ad12 (groups E, C, and A, respectively) cause the degradation of Mre11, DNA ligase IV, and p53, infection with Ad3, Ad7, Ad9, and Ad11 (groups B1, B1, D, and B2, respectively) only affects DNA ligase IV levels. Indeed, Ad3, Ad7, and Ad11 cause the marked accumulation of p53. Despite this, MDM2 levels were very low following infection with all of the viruses examined here, regardless of whether they increase p53 expression. In addition, we found that only Ad12 causes the degradation of TOPBP1, and, like Ad5, Ad4 recruits a Cul5-based E3 ubiquitin ligase to degrade p53. Surprisingly, Mre11 and DNA ligase IV degradation do not appear to be significantly affected in Ad4-, Ad5-, or Ad12-infected cells depleted of Cul2 or Cul5, indicating that E1B55K and E4orf6 recruit multiple ubiquitin ligases to target cellular proteins. Finally, although Mre11 is not degraded by Ad3, Ad7, Ad9, and Ad11, no viral DNA concatemers could be detected. We suggest that group B and D adenoviruses have evolved mechanisms based on the loss of DNA ligase IV and perhaps other unknown molecules to disable the host cell DNA damage response to promote viral replication.


Asunto(s)
Infecciones por Adenoviridae/genética , Infecciones por Adenoviridae/metabolismo , Adenoviridae/fisiología , Daño del ADN , Adenoviridae/clasificación , Adenoviridae/genética , Infecciones por Adenoviridae/enzimología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , ADN Ligasa (ATP) , ADN Ligasas/genética , ADN Ligasas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Especificidad de la Especie , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
17.
Nat Commun ; 13(1): 4143, 2022 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-35842428

RESUMEN

The accurate repair of DNA double-strand breaks (DSBs), highly toxic DNA lesions, is crucial for genome integrity and is tightly regulated during the cell cycle. In mitosis, cells inactivate DSB repair in favor of a tethering mechanism that stabilizes broken chromosomes until they are repaired in the subsequent cell cycle phases. How this is achieved mechanistically is not yet understood, but the adaptor protein TOPBP1 is critically implicated in this process. Here, we identify CIP2A as a TOPBP1-interacting protein that regulates TOPBP1 localization specifically in mitosis. Cells lacking CIP2A display increased radio-sensitivity, micronuclei formation and chromosomal instability. CIP2A is actively exported from the cell nucleus in interphase but, upon nuclear envelope breakdown at the onset of mitosis, gains access to chromatin where it forms a complex with MDC1 and TOPBP1 to promote TOPBP1 recruitment to sites of mitotic DSBs. Collectively, our data uncover CIP2A-TOPBP1 as a mitosis-specific genome maintenance complex.


Asunto(s)
Autoantígenos , Proteínas Portadoras , Reparación del ADN , Proteínas de Unión al ADN , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana , Proteínas Nucleares , Autoantígenos/genética , Autoantígenos/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Inestabilidad Cromosómica , ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mitosis/fisiología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo
18.
Nat Commun ; 12(1): 585, 2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33500419

RESUMEN

The Bloom syndrome helicase BLM interacts with topoisomerase IIIα (TOP3A), RMI1 and RMI2 to form the BTR complex, which dissolves double Holliday junctions to produce non-crossover homologous recombination (HR) products. BLM also promotes DNA-end resection, restart of stalled replication forks, and processing of ultra-fine DNA bridges in mitosis. How these activities of the BTR complex are regulated in cells is still unclear. Here, we identify multiple conserved motifs within the BTR complex that interact cooperatively with the single-stranded DNA (ssDNA)-binding protein RPA. Furthermore, we demonstrate that RPA-binding is required for stable BLM recruitment to sites of DNA replication stress and for fork restart, but not for its roles in HR or mitosis. Our findings suggest a model in which the BTR complex contains the intrinsic ability to sense levels of RPA-ssDNA at replication forks, which controls BLM recruitment and activation in response to replication stress.


Asunto(s)
Síndrome de Bloom/genética , Replicación del ADN , ADN de Cadena Simple/metabolismo , RecQ Helicasas/metabolismo , Proteína de Replicación A/metabolismo , Secuencias de Aminoácidos/genética , Sistemas CRISPR-Cas/genética , Daño del ADN , ADN-Topoisomerasas de Tipo I/metabolismo , ADN de Cadena Simple/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Mitosis/genética , Mutación , Unión Proteica/genética , Dominios Proteicos/genética , RecQ Helicasas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Reparación del ADN por Recombinación/genética
19.
J Biol Chem ; 284(49): 33939-48, 2009 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-19826003

RESUMEN

Human mediator of DNA damage checkpoint 1 (hMDC1) is an essential component of the cellular response to DNA double strand breaks. Recently, hMDC1 has been shown to associate with a subunit of the anaphase-promoting complex/cyclosome (APC/C) (Coster, G., Hayouka, Z., Argaman, L., Strauss, C., Friedler, A., Brandeis, M., and Goldberg, M. (2007) J. Biol. Chem. 282, 32053-32064), a key regulator of mitosis, suggesting a possible role for hMDC1 in controlling normal cell cycle progression. Here, we extend this work to show that hMDC1 regulates normal metaphase-to-anaphase transition through its ability to bind directly to the APC/C and modulate its E3 ubiquitin ligase activity. In support of a role for hMDC1 in controlling mitotic progression, depletion of hMDC1 by small interfering RNA results in a metaphase arrest that appears to be independent of both BubR1-dependent signaling pathways and ATM/ATR activation. Mitotic cells lacking hMDC1 exhibit markedly reduced levels of APC/C activity characterized by reduced levels of Cdc20, and a failure of Cdc20 to bind the APC/C and CREB-binding protein. We suggest therefore that hMDC1 functionally regulates the normal metaphase-to-anaphase transition by modulating the Cdc20-dependent activation of the APC/C.


Asunto(s)
Mitosis , Proteínas Nucleares/fisiología , Transactivadores/fisiología , Proteínas Adaptadoras Transductoras de Señales , Anafase , Ciclosoma-Complejo Promotor de la Anafase , Proteínas Cdc20 , Proteínas de Ciclo Celular/metabolismo , Células HeLa , Humanos , Immunoblotting/métodos , Metafase , Microscopía Fluorescente/métodos , Modelos Biológicos , Proteínas Nucleares/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/química , Ubiquitina-Proteína Ligasas/química
20.
Nat Commun ; 11(1): 123, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31913317

RESUMEN

Induction of DNA double-strand breaks (DSBs) in ribosomal DNA (rDNA) repeats is associated with ATM-dependent repression of ribosomal RNA synthesis and large-scale reorganization of nucleolar architecture, but the signaling events that regulate these responses are largely elusive. Here we show that the nucleolar response to rDNA breaks is dependent on both ATM and ATR activity. We further demonstrate that ATM- and NBS1-dependent recruitment of TOPBP1 in the nucleoli is required for inhibition of ribosomal RNA synthesis and nucleolar segregation in response to rDNA breaks. Mechanistically, TOPBP1 recruitment is mediated by phosphorylation-dependent interactions between three of its BRCT domains and conserved phosphorylated Ser/Thr residues at the C-terminus of the nucleolar phosphoprotein Treacle. Our data thus reveal an important cooperation between TOPBP1 and Treacle in the signaling cascade that triggers transcriptional inhibition and nucleolar segregation in response to rDNA breaks.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas Portadoras/metabolismo , Nucléolo Celular/genética , ADN Ribosómico/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Secuencias de Aminoácidos , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Nucléolo Celular/metabolismo , Roturas del ADN de Doble Cadena , ADN Ribosómico/metabolismo , Proteínas de Unión al ADN/genética , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/genética , Fosfoproteínas/química , Fosfoproteínas/genética , Unión Proteica , ARN Ribosómico/genética , ARN Ribosómico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA