Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Radiat Biol ; 100(6): 890-902, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38631047

RESUMEN

Purpose: Continuous exposure to ionizing radiation at a low dose rate poses significant health risks to humans on deep space missions, prompting the need for mechanistic studies to identify countermeasures against its deleterious effects. Mitochondria are a major subcellular locus of radiogenic injury, and may trigger secondary cellular responses through the production of reactive oxygen species (mtROS) with broader biological implications. Methods and Materials: To determine the contribution of mtROS to radiation-induced cellular responses, we investigated the impacts of protracted γ-ray exposures (IR; 1.1 Gy delivered at 0.16 mGy/min continuously over 5 days) on mitochondrial function, gene expression, and the protein secretome of human HCA2-hTERT fibroblasts in the presence and absence of a mitochondria-specific antioxidant mitoTEMPO (MT; 5 µM). Results: IR increased fibroblast mitochondrial oxygen consumption (JO2) and H2O2 release rates (JH2O2) under energized conditions, which corresponded to higher protein expression of NADPH Oxidase (NOX) 1, NOX4, and nuclear DNA-encoded subunits of respiratory chain Complexes I and III, but depleted mtDNA transcripts encoding subunits of the same complexes. This was associated with activation of gene programs related to DNA repair, oxidative stress, and protein ubiquination, all of which were attenuated by MT treatment along with radiation-induced increases in JO2 and JH2O2. IR also increased secreted levels of interleukin-8 and Type I collagens, while decreasing Type VI collagens and enzymes that coordinate assembly and remodeling of the extracellular matrix. MT treatment attenuated many of these effects while augmenting others, revealing complex effects of mtROS in fibroblast responses to IR. Conclusion: These results implicate mtROS production in fibroblast responses to protracted radiation exposure, and suggest potentially protective effects of mitochondrial-targeted antioxidants against radiogenic tissue injury in vivo.


Asunto(s)
Fibroblastos , Rayos gamma , Mitocondrias , Especies Reactivas de Oxígeno , Humanos , Fibroblastos/efectos de la radiación , Fibroblastos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Mitocondrias/efectos de la radiación , Mitocondrias/metabolismo , Rayos gamma/efectos adversos , Línea Celular , Exposición a la Radiación/efectos adversos , Compuestos Organofosforados , Piperidinas
2.
bioRxiv ; 2023 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-37546955

RESUMEN

RAD54L is a DNA motor protein with critical roles in homologous recombination DNA repair (HR). In vitro, RAD54L was also shown to catalyze the reversal and restoration of model replication forks. Little, however, is known about the role of RAD54L in regulating the dynamics of DNA replication in cells. Here, we show that RAD54L functions as a fork remodeler and restrains the progression of replication forks in human cells. Analogous to HLTF and FBH1, and consistent with a role in fork reversal, RAD54L catalyzes the slowing of fork progression in response to replication stress. In BRCA1/2-deficient cells, RAD54L activity leads to nascent strand DNA degradation, and loss of RAD54L reduces DNA double-strand break formation. Using a separation-of-function mutation, we show that RAD54L-mediated fork restraint depends on its ability to catalyze branch migration. Our results reveal a new role for RAD54L in regulating the dynamics of replication forks in cells and highlight the impact of RAD54L function on the treatment of patients with BRCA1/2-deficient tumors.

3.
Nucleic Acids Res ; 51(16): 8643-8662, 2023 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-37439356

RESUMEN

Environmental agents like ionizing radiation (IR) and chemotherapeutic drugs can cause severe damage to the DNA, often in the form of double-strand breaks (DSBs). Remaining unrepaired, DSBs can lead to chromosomal rearrangements, and cell death. One major error-free pathway to repair DSBs is homologous recombination repair (HRR). Tousled-like kinase 1 (TLK1), a Ser/Thr kinase that regulates the DNA damage checkpoint, has been found to interact with RAD54, a central DNA translocase in HRR. To determine how TLK1 regulates RAD54, we inhibited or depleted TLK1 and tested how this impacts HRR in human cells using a ISce-I-GR-DsRed fused reporter endonuclease. Our results show that TLK1 phosphorylates RAD54 at three threonines (T41, T59 and T700), two of which are located within its N-terminal domain (NTD) and one is located within its C-terminal domain (CTD). Phosphorylation at both T41 and T59 supports HRR and protects cells from DNA DSB damage. In contrast, phosphorylation of T700 leads to impaired HRR and engenders no protection to cells from cytotoxicity and rather results in repair delay. Further, our work enlightens the effect of RAD54-T700 (RAD54-CTD) phosphorylation by TLK1 in mammalian system and reveals a new site of interaction with RAD51.


Asunto(s)
Reparación del ADN , Reparación del ADN por Recombinación , Animales , Humanos , Fosforilación , Daño del ADN , ADN/metabolismo , Recombinasa Rad51/metabolismo , Recombinación Homóloga , Mamíferos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo
4.
Nat Commun ; 14(1): 432, 2023 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-36702902

RESUMEN

The tumor suppressor BRCA2 participates in DNA double-strand break repair by RAD51-dependent homologous recombination and protects stressed DNA replication forks from nucleolytic attack. We demonstrate that the C-terminal Recombinase Binding (CTRB) region of BRCA2, encoded by gene exon 27, harbors a DNA binding activity. CTRB alone stimulates the DNA strand exchange activity of RAD51 and permits the utilization of RPA-coated ssDNA by RAD51 for strand exchange. Moreover, CTRB functionally synergizes with the Oligonucleotide Binding fold containing DNA binding domain and BRC4 repeat of BRCA2 in RPA-RAD51 exchange on ssDNA. Importantly, we show that the DNA binding and RAD51 interaction attributes of the CTRB are crucial for homologous recombination and protection of replication forks against MRE11-mediated attrition. Our findings shed light on the role of the CTRB region in genome repair, reveal remarkable functional plasticity of BRCA2, and help explain why deletion of Brca2 exon 27 impacts upon embryonic lethality.


Asunto(s)
Replicación del ADN , Recombinasa Rad51 , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Reparación del ADN , Proteína BRCA2/metabolismo , ADN , Recombinación Homóloga
5.
Front Cell Dev Biol ; 10: 866601, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35652094

RESUMEN

Homologous recombination DNA repair (HR) is a complex DNA damage repair pathway and an attractive target of inhibition in anti-cancer therapy. To help guide the development of efficient HR inhibitors, it is critical to identify compensatory HR sub-pathways. In this study, we describe a novel synthetic interaction between RAD51AP1 and RAD54L, two structurally unrelated proteins that function downstream of the RAD51 recombinase in HR. We show that concomitant deletion of RAD51AP1 and RAD54L further sensitizes human cancer cell lines to treatment with olaparib, a Poly (adenosine 5'-diphosphate-ribose) polymerase inhibitor, to the DNA inter-strand crosslinking agent mitomycin C, and to hydroxyurea, which induces DNA replication stress. We also show that the RAD54L paralog RAD54B compensates for RAD54L deficiency, although, surprisingly, less extensively than RAD51AP1. These results, for the first time, delineate RAD51AP1- and RAD54L-dependent sub-pathways and will guide the development of inhibitors that target HR stimulators of strand invasion.

6.
Biochem J ; 479(11): 1205-1220, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35695515

RESUMEN

The Nuclear Casein and Cyclin-dependent Kinase Substrate 1 (NUCKS1) protein is highly conserved in vertebrates, predominantly localized to the nucleus and one of the most heavily modified proteins in the human proteome. NUCKS1 expression is high in stem cells and the brain, developmentally regulated in mice and associated with several diverse malignancies in humans, including cancer, metabolic syndrome and Parkinson's disease. NUCKS1 function has been linked to modulating chromatin architecture and transcription, DNA repair and cell cycle regulation. In this review, we summarize and discuss the published information on NUCKS1 and highlight the questions that remain to be addressed to better understand the complex biology of this multifaceted protein.


Asunto(s)
Proteínas Nucleares , Fosfoproteínas , Animales , Cromatina/genética , Reparación del ADN , Ratones , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo
7.
J Biol Chem ; 297(1): 100844, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34058198

RESUMEN

RAD51-associated protein 1 (RAD51AP1) is a key protein in the homologous recombination (HR) DNA repair pathway. Loss of RAD51AP1 leads to defective HR, genome instability, and telomere erosion. RAD51AP1 physically interacts with the RAD51 recombinase and promotes RAD51-mediated capture of donor DNA, synaptic complex assembly, and displacement-loop formation when tested with nucleosome-free DNA substrates. In cells, however, DNA is packaged into chromatin, posing an additional barrier to the complexities of the HR reaction. In this study, we show that RAD51AP1 binds to nucleosome core particles (NCPs), the minimum basic unit of chromatin in which approximately two superhelical turns of 147 bp double-stranded DNA are wrapped around one histone octamer with no free DNA ends remaining. We identified a C-terminal region in RAD51AP1, including its previously mapped DNA-binding domain, as critical for mediating the association between RAD51AP1 and both the NCP and the histone octamer. Using in vitro surrogate assays of HR activity, we show that RAD51AP1 is capable of promoting duplex DNA capture and initiating joint-molecule formation with the NCP and chromatinized template DNA, respectively. Together, our results suggest that RAD51AP1 directly assists in the RAD51-mediated search for donor DNA in chromatin. We present a model, in which RAD51AP1 anchors the DNA template through affinity for its nucleosomes to the RAD51-ssDNA nucleoprotein filament.


Asunto(s)
Cromatina/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ARN/genética , Recombinasa Rad51/genética , Reparación del ADN por Recombinación/genética , Cromatina/química , Emparejamiento Cromosómico/genética , ADN/química , ADN/genética , Proteínas de Unión al ADN/química , Inestabilidad Genómica/genética , Histonas/química , Histonas/genética , Humanos , Nucleosomas/genética , Dominios Proteicos/genética , Proteínas de Unión al ARN/química , Recombinasa Rad51/química , Telómero/genética
8.
J Cell Biol ; 219(10)2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32876692

RESUMEN

NUCKS1 (nuclear ubiquitous casein kinase and cyclin-dependent kinase substrate 1) is a chromatin-associated, vertebrate-specific, and multifunctional protein with a role in DNA damage signaling and repair. Previously, we have shown that NUCKS1 helps maintain homologous recombination (HR) DNA repair in human cells and functions as a tumor suppressor in mice. However, the mechanisms by which NUCKS1 positively impacts these processes had remained unclear. Here, we show that NUCKS1 physically and functionally interacts with the DNA motor protein RAD54. Upon exposure of human cells to DNA-damaging agents, NUCKS1 controls the resolution of RAD54 foci. In unperturbed cells, NUCKS1 prevents RAD54's inappropriate engagement with RAD51AP1. In vitro, NUCKS1 stimulates the ATPase activity of RAD54 and the RAD51-RAD54-mediated strand invasion step during displacement loop formation. Taken together, our data demonstrate that the NUCKS1 protein is an important new regulator of the spatiotemporal events in HR.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas Nucleares/genética , Fosfoproteínas/genética , Proteínas de Unión al ARN/genética , Reparación del ADN por Recombinación/genética , Proteína Nuclear Ligada al Cromosoma X/genética , Adenosina Trifosfatasas/genética , Línea Celular , Humanos , Unión Proteica/genética
9.
J Biol Chem ; 295(24): 8186-8194, 2020 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-32350107

RESUMEN

USP1-associated factor 1 (UAF1) is an integral component of the RAD51-associated protein 1 (RAD51AP1)-UAF1-ubiquitin-specific peptidase 1 (USP1) trimeric deubiquitinase complex. This complex acts on DNA-bound, monoubiquitinated Fanconi anemia complementation group D2 (FANCD2) protein in the Fanconi anemia pathway of the DNA damage response. Moreover, RAD51AP1 and UAF1 cooperate to enhance homologous DNA pairing mediated by the recombinase RAD51 in DNA repair via the homologous recombination (HR) pathway. However, whereas the DNA-binding activity of RAD51AP1 has been shown to be important for RAD51-mediated homologous DNA pairing and HR-mediated DNA repair, the role of DNA binding by UAF1 in these processes is unclear. We have isolated mutant UAF1 variants that are impaired in DNA binding and tested them together with RAD51AP1 in RAD51-mediated HR. This biochemical analysis revealed that the DNA-binding activity of UAF1 is indispensable for enhanced RAD51 recombinase activity within the context of the UAF1-RAD51AP1 complex. In cells, DNA-binding deficiency of UAF1 increased DNA damage sensitivity and impaired HR efficiency, suggesting that UAF1 and RAD51AP1 have coordinated roles in DNA binding during HR and DNA damage repair. Our findings show that even though UAF1's DNA-binding activity is redundant with that of RAD51AP1 in FANCD2 deubiquitination, it is required for efficient HR-mediated chromosome damage repair.


Asunto(s)
ADN/metabolismo , Proteínas Nucleares/metabolismo , Recombinasa Rad51/metabolismo , Reparación del ADN por Recombinación , Daño del ADN , Células HeLa , Humanos , Modelos Biológicos , Proteínas Nucleares/química , Unión Proteica , Estructura Secundaria de Proteína
10.
Nat Commun ; 10(1): 2849, 2019 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-31253762

RESUMEN

Fanconi anemia (FA) is a multigenic disease of bone marrow failure and cancer susceptibility stemming from a failure to remove DNA crosslinks and other chromosomal lesions. Within the FA DNA damage response pathway, DNA-dependent monoubiquitinaton of FANCD2 licenses downstream events, while timely FANCD2 deubiquitination serves to extinguish the response. Here, we show with reconstituted biochemical systems, which we developed, that efficient FANCD2 deubiquitination by the USP1-UAF1 complex is dependent on DNA and DNA binding by UAF1. Surprisingly, we find that the DNA binding activity of the UAF1-associated protein RAD51AP1 can substitute for that of UAF1 in FANCD2 deubiquitination in our biochemical system. We also reveal the importance of DNA binding by UAF1 and RAD51AP1 in FANCD2 deubiquitination in the cellular setting. Our results provide insights into a key step in the FA pathway and help define the multifaceted role of the USP1-UAF1-RAD51AP1 complex in DNA damage tolerance and genome repair.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Anemia de Fanconi/genética , Proteínas Nucleares/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Daño del ADN , Proteínas de Unión al ADN/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Regulación de la Expresión Génica/fisiología , Humanos , Mutación , Proteínas Nucleares/genética , Unión Proteica , Proteínas de Unión al ARN , Proteasas Ubiquitina-Específicas/genética , Ubiquitinación
11.
Annu Rev Biochem ; 88: 221-245, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-30917004

RESUMEN

Mutations in the BRCA1 and BRCA2 genes predispose afflicted individuals to breast, ovarian, and other cancers. The BRCA-encoded products form complexes with other tumor suppressor proteins and with the recombinase enzyme RAD51 to mediate chromosome damage repair by homologous recombination and also to protect stressed DNA replication forks against spurious nucleolytic attrition. Understanding how the BRCA tumor suppressor network executes its biological functions would provide the foundation for developing targeted cancer therapeutics, but progress in this area has been greatly hampered by the challenge of obtaining purified BRCA complexes for mechanistic studies. In this article, we review how recent effort begins to overcome this technical challenge, leading to functional and structural insights into the biochemical attributes of these complexes and the multifaceted roles that they fulfill in genome maintenance. We also highlight the major mechanistic questions that remain.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Neoplasias de la Mama/genética , Redes Reguladoras de Genes , Recombinasa Rad51/genética , Reparación del ADN por Recombinación , Proteínas Supresoras de Tumor/genética , Ubiquitina-Proteína Ligasas/genética , Proteína BRCA1/química , Proteína BRCA1/metabolismo , Proteína BRCA2/química , Proteína BRCA2/metabolismo , Sitios de Unión , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , ADN/química , ADN/genética , ADN/metabolismo , Roturas del ADN de Doble Cadena , Replicación del ADN , Femenino , Genoma Humano , Inestabilidad Genómica , Humanos , Modelos Moleculares , Unión Proteica , Estructura Secundaria de Proteína , Recombinasa Rad51/química , Recombinasa Rad51/metabolismo , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo
12.
J Clin Invest ; 128(12): 5307-5321, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30371505

RESUMEN

After the initial responsiveness of triple-negative breast cancers (TNBCs) to chemotherapy, they often recur as chemotherapy-resistant tumors, and this has been associated with upregulated homology-directed repair (HDR). Thus, inhibitors of HDR could be a useful adjunct to chemotherapy treatment of these cancers. We performed a high-throughput chemical screen for inhibitors of HDR from which we obtained a number of hits that disrupted microtubule dynamics. We postulated that high levels of the target molecules of our screen in tumors would correlate with poor chemotherapy response. We found that inhibition or knockdown of dynamin 2 (DNM2), known for its role in endocytic cell trafficking and microtubule dynamics, impaired HDR and improved response to chemotherapy of cells and of tumors in mice. In a retrospective analysis, levels of DNM2 at the time of treatment strongly predicted chemotherapy outcome for estrogen receptor-negative and especially for TNBC patients. We propose that DNM2-associated DNA repair enzyme trafficking is important for HDR efficiency and is a powerful predictor of sensitivity to breast cancer chemotherapy and an important target for therapy.


Asunto(s)
Antineoplásicos/farmacología , Dinaminas/metabolismo , Reparación del ADN por Recombinación , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/enzimología , Animales , Células CHO , Cricetulus , Dinamina II , Dinaminas/genética , Femenino , Humanos , Ratones , Ratones Desnudos , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Q J Exp Psychol (Hove) ; 71(9): 1988-2004, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30117383

RESUMEN

Using a novel eye-tracking paradigm, we assessed the development of 2- to 6-year-old children's intuitions about the physical support of symmetrical and asymmetrical objects in two experiments (Experiment 1: N = 98; Experiment 2: N = 288). Children were presented with video sequences demonstrating how two identical blocks were lowered onto a platform before being released simultaneously. In the critical test trials, both blocks remained in place although only one of them was sufficiently supported. As expected, children tended to look longer at the block, which should have fallen. Taken together, the results indicate that even 2-year-old children are sensitive to the amount of contact between symmetrical blocks and a supporting platform and even anticipate which block is going to fall. Nonetheless, we found a considerable improvement with age in this respect. Two-year-olds did not consider an object's weight distribution reliably when assessing its stability and even older preschoolers performed much more poorly with asymmetrical than symmetrical blocks. We conclude that intuitions about support are still weak and limited in toddlers and that they improve considerably during early childhood.


Asunto(s)
Desarrollo Infantil , Cognición/fisiología , Formación de Concepto/fisiología , Movimientos Oculares/fisiología , Intuición/fisiología , Psicología Infantil , Análisis de Varianza , Niño , Preescolar , Medidas del Movimiento Ocular , Femenino , Humanos , Masculino , Estimulación Luminosa , Factores de Tiempo
14.
Radiother Oncol ; 128(1): 68-75, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29910006

RESUMEN

In the clinic, proton beam therapy (PBT) is based on the use of a generic relative biological effectiveness (RBE) of 1.1 compared to photons in human cancers and normal tissues. However, the experimental basis for this RBE lacks any significant number of representative tumor models and clinically relevant endpoints for dose-limiting organs at risk. It is now increasingly appreciated that much of the variations of treatment responses in cancers are due to inter-tumoral genomic heterogeneity. Indeed, recently it has been shown that defects in certain DNA repair pathways, which are found in subsets of many cancers, are associated with a RBE increase in vitro. However, there currently exist little in vivo or clinical data that confirm the existence of similarly increased RBE values in human cancers. Furthermore, evidence for variable RBE values for normal tissue toxicity has been sparse and conflicting to date. If we could predict variable RBE values in patients, we would be able to optimally use and personalize PBT. For example, predictive tumor biomarkers may facilitate selection of patients with proton-sensitive cancers previously ineligible for PBT. Dose de-escalation may be possible to reduce normal tissue toxicity, especially in pediatric patients. Knowledge of increased tumor RBE may allow us to develop biologically optimized therapies to enhance local control while RBE biomarkers for normal tissues could lead to a better understanding and prevention of unusual PBT-associated toxicity. Here, we will review experimental data on the repair of proton damage to DNA that impact both RBE values and biophysical modeling to predict RBE variations. Experimental approaches for studying proton sensitivity in vitro and in vivo will be reviewed as well and recent clinical findings discussed. Ultimately, therapeutically exploiting the understudied biological advantages of protons and developing approaches to limit treatment toxicity should fundamentally impact the clinical use of PBT.


Asunto(s)
Neoplasias/radioterapia , Terapia de Protones , Efectividad Biológica Relativa , Reparación del ADN , Humanos
15.
Nature ; 550(7676): 360-365, 2017 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-28976962

RESUMEN

The tumour suppressor complex BRCA1-BARD1 functions in the repair of DNA double-stranded breaks by homologous recombination. During this process, BRCA1-BARD1 facilitates the nucleolytic resection of DNA ends to generate a single-stranded template for the recruitment of another tumour suppressor complex, BRCA2-PALB2, and the recombinase RAD51. Here, by examining purified wild-type and mutant BRCA1-BARD1, we show that both BRCA1 and BARD1 bind DNA and interact with RAD51, and that BRCA1-BARD1 enhances the recombinase activity of RAD51. Mechanistically, BRCA1-BARD1 promotes the assembly of the synaptic complex, an essential intermediate in RAD51-mediated DNA joint formation. We provide evidence that BRCA1 and BARD1 are indispensable for RAD51 stimulation. Notably, BRCA1-BARD1 mutants with weakened RAD51 interactions show compromised DNA joint formation and impaired mediation of homologous recombination and DNA repair in cells. Our results identify a late role of BRCA1-BARD1 in homologous recombination, an attribute of the tumour suppressor complex that could be targeted in cancer therapy.


Asunto(s)
Proteína BRCA1/metabolismo , Emparejamiento Base , Emparejamiento Cromosómico , Recombinasa Rad51/metabolismo , Reparación del ADN por Recombinación , Homología de Secuencia de Ácido Nucleico , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Proteína BRCA1/genética , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Proteína del Grupo de Complementación N de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Genes BRCA1 , Genes BRCA2 , Humanos , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Mutación , Unión Proteica , Recombinasa Rad51/genética , Reparación del ADN por Recombinación/genética , Moldes Genéticos , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/genética , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética
16.
DNA Repair (Amst) ; 59: 76-81, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28963981

RESUMEN

Homologous recombination (HR) serves to repair DNA double-strand breaks and damaged replication forks and is essential for maintaining genome stability and tumor suppression. HR capacity also determines the efficacy of anticancer therapy. Hence, there is an urgent need to better understand all HR proteins and sub-pathways. An emerging protein that is critical for RAD51-mediated HR is RAD51-associated protein 1 (RAD51AP1). Although much has been learned about its biochemical attributes, the precise molecular role of RAD51AP1 in the HR reaction is not yet fully understood. The available literature also suggests that RAD51AP1 expression may be relevant for cancer development and progression. Here, we review the efforts that led to the discovery of RAD51AP1 and elaborate on our current understanding of its biochemical profile and biological function. We also discuss how RAD51AP1 may help to promote cancer development and why it could potentially represent a promising new target for therapeutic intervention.


Asunto(s)
Carcinogénesis/metabolismo , Proteínas de Unión al ADN/metabolismo , Reparación del ADN por Recombinación , Animales , Proteínas Portadoras/metabolismo , Humanos , Ratones , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN
17.
Oncotarget ; 7(38): 61874-61889, 2016 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-27542204

RESUMEN

NUCKS1 is a 27 kD vertebrate-specific protein, with a role in the DNA damage response. Here, we show that after 4 Gy total-body X-irradiation, Trp53+/- Nucks1+/- mice more rapidly developed tumors, particularly thymic lymphoma (TL), than Trp53+/- mice. TLs in both cohorts showed loss of heterozygosity (LOH) of the Trp53+ allele in essentially all cases. In contrast, LOH of the Nucks1+ allele was rare. Nucks1 expression correlated well with Nucks1 gene dosage in normal thymi, but was increased in the majority of TLs from Trp53+/- Nucks1+/- mice, suggesting that elevated Nucks1 message may be associated with progression towards malignancy in vivo. Trp53+/- Nucks1+/- mice frequently succumbed to CD4- CD8- TLs harboring translocations involving Igh but not Tcra/d, indicating TLs in Trp53+/- Nucks1+/- mice mostly originated prior to the double positive stage and at earlier lineage than TLs in Trp53+/- mice. Monoclonal rearrangements at Tcrb were more prevalent in TLs from Trp53+/- Nucks1+/- mice, as was infiltration of primary TL cells to distant organs (liver, kidney and spleen). We propose that, in the context of Trp53 deficiency, wild type levels of Nucks1 are required to suppress radiation-induced TL, likely through the role of the NUCKS1 protein in the DNA damage response.


Asunto(s)
Linfoma/genética , Neoplasias Inducidas por Radiación/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Alelos , Animales , Anticuerpos Monoclonales/química , Hibridación Genómica Comparativa , Daño del ADN , Femenino , Dosificación de Gen , Genotipo , Haploinsuficiencia , Inmunofenotipificación , Riñón/metabolismo , Hígado/metabolismo , Pérdida de Heterocigocidad , Linfoma/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias Inducidas por Radiación/metabolismo , Bazo/metabolismo , Regulación hacia Arriba
18.
Cell Rep ; 15(10): 2118-2126, 2016 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-27239033

RESUMEN

The UAF1-USP1 complex deubiquitinates FANCD2 during execution of the Fanconi anemia DNA damage response pathway. As such, UAF1 depletion results in persistent FANCD2 ubiquitination and DNA damage hypersensitivity. UAF1-deficient cells are also impaired for DNA repair by homologous recombination. Herein, we show that UAF1 binds DNA and forms a dimeric complex with RAD51AP1, an accessory factor of the RAD51 recombinase, and a trimeric complex with RAD51 through RAD51AP1. Two small ubiquitin-like modifier (SUMO)-like domains in UAF1 and a SUMO-interacting motif in RAD51AP1 mediate complex formation. Importantly, UAF1 enhances RAD51-mediated homologous DNA pairing in a manner that is dependent on complex formation with RAD51AP1 but independent of USP1. Mechanistically, RAD51AP1-UAF1 co-operates with RAD51 to assemble the synaptic complex, a critical nucleoprotein intermediate in homologous recombination, and cellular studies reveal the biological significance of the RAD51AP1-UAF1 protein complex. Our findings provide insights into an apparently USP1-independent role of UAF1 in genome maintenance.


Asunto(s)
Emparejamiento Cromosómico , ADN/metabolismo , Recombinación Homóloga , Recombinasa Rad51/metabolismo , Secuencia de Aminoácidos , Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Células HeLa , Humanos , Modelos Biológicos , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Unión Proteica , Dominios Proteicos
19.
Mol Cell ; 61(4): 535-546, 2016 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26833090

RESUMEN

XPG is a structure-specific endonuclease required for nucleotide excision repair, and incision-defective XPG mutations cause the skin cancer-prone syndrome xeroderma pigmentosum. Truncating mutations instead cause the neurodevelopmental progeroid disorder Cockayne syndrome, but little is known about how XPG loss results in this devastating disease. We identify XPG as a partner of BRCA1 and BRCA2 in maintaining genomic stability through homologous recombination (HRR). XPG depletion causes DNA double-strand breaks, chromosomal abnormalities, cell-cycle delays, defective HRR, inability to overcome replication fork stalling, and replication stress. XPG directly interacts with BRCA2, RAD51, and PALB2, and XPG depletion reduces their chromatin binding and subsequent RAD51 foci formation. Upstream in HRR, XPG interacts directly with BRCA1. Its depletion causes BRCA1 hyper-phosphorylation and persistent chromatin binding. These unexpected findings establish XPG as an HRR protein with important roles in genome stability and suggest how XPG defects produce severe clinical consequences including cancer and accelerated aging.


Asunto(s)
Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , Síndrome de Cockayne/genética , Proteínas de Unión al ADN/genética , Endonucleasas/genética , Inestabilidad Genómica , Recombinación Homóloga , Proteínas Nucleares/genética , Factores de Transcripción/genética , Animales , Línea Celular Tumoral , Síndrome de Cockayne/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Endonucleasas/metabolismo , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Genoma Humano , Células HeLa , Humanos , Ratones , Proteínas Nucleares/metabolismo , Fosforilación , Recombinasa Rad51/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo
20.
Nucleic Acids Res ; 43(20): 9817-34, 2015 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-26323318

RESUMEN

NUCKS1 (nuclear casein kinase and cyclin-dependent kinase substrate 1) is a 27 kD chromosomal, vertebrate-specific protein, for which limited functional data exist. Here, we demonstrate that NUCKS1 shares extensive sequence homology with RAD51AP1 (RAD51 associated protein 1), suggesting that these two proteins are paralogs. Similar to the phenotypic effects of RAD51AP1 knockdown, we find that depletion of NUCKS1 in human cells impairs DNA repair by homologous recombination (HR) and chromosome stability. Depletion of NUCKS1 also results in greatly increased cellular sensitivity to mitomycin C (MMC), and in increased levels of spontaneous and MMC-induced chromatid breaks. NUCKS1 is critical to maintaining wild type HR capacity, and, as observed for a number of proteins involved in the HR pathway, functional loss of NUCKS1 leads to a slow down in DNA replication fork progression with a concomitant increase in the utilization of new replication origins. Interestingly, recombinant NUCKS1 shares the same DNA binding preference as RAD51AP1, but binds to DNA with reduced affinity when compared to RAD51AP1. Our results show that NUCKS1 is a chromatin-associated protein with a role in the DNA damage response and in HR, a DNA repair pathway critical for tumor suppression.


Asunto(s)
Inestabilidad Genómica , Proteínas Nucleares/fisiología , Fosfoproteínas/fisiología , Reparación del ADN por Recombinación , Línea Celular , Cromatina/metabolismo , Aberraciones Cromosómicas , ADN/metabolismo , Daño del ADN , Replicación del ADN , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/fisiología , Células HeLa/fisiología , Humanos , Mitomicina/farmacología , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/química , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación/efectos de la radiación , Proteínas de Unión al ARN , Recombinasa Rad51/metabolismo , Fase S/efectos de la radiación , Homología de Secuencia de Aminoácido , Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA