Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.588
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 8511, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39353945

RESUMEN

Nucleotide excision repair (NER) is vital for genome integrity. Yet, our understanding of the complex NER protein machinery remains incomplete. Combining cryo-EM and XL-MS data with AlphaFold2 predictions, we build an integrative model of the NER pre-incision complex(PInC). Here TFIIH serves as a molecular ruler, defining the DNA bubble size and precisely positioning the XPG and XPF nucleases for incision. Using simulations and graph theoretical analyses, we unveil PInC's assembly, global motions, and partitioning into dynamic communities. Remarkably, XPG caps XPD's DNA-binding groove and bridges both junctions of the DNA bubble, suggesting a novel coordination mechanism of PInC's dual incision. XPA rigging interlaces XPF/ERCC1 with RPA, XPD, XPB, and 5' ssDNA, exposing XPA's crucial role in licensing the XPF/ERCC1 incision. Mapping disease mutations onto our models reveals clustering into distinct mechanistic classes, elucidating xeroderma pigmentosum and Cockayne syndrome disease etiology.


Asunto(s)
Reparación del ADN , Proteínas de Unión al ADN , Endonucleasas , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/química , Humanos , Endonucleasas/metabolismo , Endonucleasas/genética , Factor de Transcripción TFIIH/metabolismo , Factor de Transcripción TFIIH/química , Factor de Transcripción TFIIH/genética , Proteína de la Xerodermia Pigmentosa del Grupo D/metabolismo , Proteína de la Xerodermia Pigmentosa del Grupo D/genética , Proteína de la Xerodermia Pigmentosa del Grupo D/química , Microscopía por Crioelectrón , Proteína de la Xerodermia Pigmentosa del Grupo A/metabolismo , Proteína de la Xerodermia Pigmentosa del Grupo A/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Unión Proteica , ADN/metabolismo , ADN/química , ADN/genética , Proteína de Replicación A/metabolismo , Proteína de Replicación A/genética , Modelos Moleculares , ADN de Cadena Simple/metabolismo , ADN de Cadena Simple/genética , Reparación por Escisión , Proteínas Nucleares
2.
Methods Enzymol ; 705: 271-309, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39389667

RESUMEN

In humans, DNA polymerase δ (pol δ) holoenzymes, comprised of pol δ and the processivity sliding clamp, proliferating cell nuclear antigen (PCNA), carry out DNA synthesis during lagging strand replication, the initiation of leading strand DNA replication as well as most of the major DNA damage repair pathways. In each of these contexts, pol δ holoenzymes are assembled at primer/template (P/T) junctions and initiate DNA synthesis in a stepwise process that involves the PCNA clamp loader, replication factor C and, depending on the DNA synthesis pathway, the major single strand DNA-binding protein complex, replication protein A (RPA). In a recent report from our laboratory, we designed and utilized direct, ensemble Förster Resonance Energy Transfer approaches to monitor the transient state kinetics of pol δ holoenzyme assembly and initiation of DNA synthesis on P/T junctions engaged by RPA. In this chapter, we detail the original approaches and discuss adaptations that can be utilized to monitor fast kinetic reactions in the millisecond (ms) timescale. All approaches described in this chapter utilize a commercially-available fluorescence spectrophotometer, can be readily evolved for alternative DNA polymerases and P/T DNA substrates, and permit incorporation of protein posttranslational modifications, accessory factors, DNA covalent modifications, accessory factors, enzymes, etc. Hence, these approaches are widely accessible and broadly applicable for characterizing DNA polymerase holoenzyme assembly and initiation of DNA synthesis during any PCNA-dependent DNA synthesis pathway.


Asunto(s)
ADN Polimerasa III , Replicación del ADN , ADN , Transferencia Resonante de Energía de Fluorescencia , Antígeno Nuclear de Célula en Proliferación , Transferencia Resonante de Energía de Fluorescencia/métodos , ADN Polimerasa III/metabolismo , ADN Polimerasa III/química , Humanos , Cinética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Antígeno Nuclear de Célula en Proliferación/química , Antígeno Nuclear de Célula en Proliferación/genética , Holoenzimas/metabolismo , Holoenzimas/química , ADN/metabolismo , ADN/química , Proteína de Replicación A/metabolismo , Proteína de Replicación A/química , Proteína de Replicación C/metabolismo , Proteína de Replicación C/genética , Proteína de Replicación C/química
3.
New Phytol ; 244(3): 855-869, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39229867

RESUMEN

Leaf senescence is a developmental program regulated by both endogenous and environmental cues. Abiotic stresses such as nutrient deprivation can induce premature leaf senescence, which profoundly impacts plant growth and crop yield. However, the molecular mechanisms underlying stress-induced senescence are not fully understood. In this work, employing a carbon deprivation (C-deprivation)-induced senescence assay in Arabidopsis seedlings, we identified PLEIOTROPIC REGULATORY LOCUS 1 (PRL1), a component of the NineTeen Complex, as a negative regulator of C-deprivation-induced senescence. Furthermore, we demonstrated that PRL1 directly interacts with the RPA2A subunit of the single-stranded DNA-binding Replication Protein A (RPA) complex. Consistently, the loss of RPA2A leads to premature senescence, while increased expression of RPA2A inhibits senescence. Moreover, overexpression of RPA2A reverses the accelerated senescence in prl1 mutants, and the interaction with PRL1 stabilizes RPA2A under C-deprivation. In summary, our findings reveal the involvement of the PRL1-RPA2A functional module in C-deprivation-induced plant senescence.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , Carbono , Regulación de la Expresión Génica de las Plantas , Senescencia de la Planta , Unión Proteica , Proteína de Replicación A , Arabidopsis/genética , Arabidopsis/metabolismo , Arabidopsis/fisiología , Proteínas de Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Carbono/metabolismo , Mutación/genética , Hojas de la Planta/metabolismo , Estabilidad Proteica , Proteína de Replicación A/metabolismo , Plantones/genética , Plantones/metabolismo , Plantones/crecimiento & desarrollo
4.
Nat Commun ; 15(1): 7797, 2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39242676

RESUMEN

Ribosomal DNA (rDNA) encodes the ribosomal RNA genes and represents an intrinsically unstable genomic region. However, the underlying mechanisms and implications for genome integrity remain elusive. Here, we use Bloom syndrome (BS), a rare genetic disease characterized by DNA repair defects and hyper-unstable rDNA, as a model to investigate the mechanisms leading to rDNA instability. We find that in Bloom helicase (BLM) proficient cells, the homologous recombination (HR) pathway in rDNA resembles that in nuclear chromatin; it is initiated by resection, replication protein A (RPA) loading and BRCA2-dependent RAD51 filament formation. However, BLM deficiency compromises RPA-loading and BRCA1/2 recruitment to rDNA, but not RAD51 accumulation. RAD51 accumulates at rDNA despite depletion of long-range resection nucleases and rDNA damage results in micronuclei when BLM is absent. In summary, our findings indicate that rDNA is permissive to RAD51 accumulation in the absence of BLM, leading to micronucleation and potentially global genomic instability.


Asunto(s)
ADN Ribosómico , Inestabilidad Genómica , Recombinasa Rad51 , RecQ Helicasas , Recombinasa Rad51/metabolismo , Recombinasa Rad51/genética , ADN Ribosómico/genética , ADN Ribosómico/metabolismo , Humanos , RecQ Helicasas/metabolismo , RecQ Helicasas/genética , Proteína de Replicación A/metabolismo , Proteína de Replicación A/genética , Recombinación Homóloga , Síndrome de Bloom/genética , Síndrome de Bloom/metabolismo , Proteína BRCA2/metabolismo , Proteína BRCA2/genética , Proteína BRCA1/metabolismo , Proteína BRCA1/genética , Reparación del ADN
5.
Genes Dev ; 38(15-16): 755-771, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39231615

RESUMEN

Premature telomere shortening or telomere instability is associated with a group of rare and heterogeneous diseases collectively known as telomere biology disorders (TBDs). Here we identified two unrelated individuals with clinical manifestations of TBDs and short telomeres associated with the identical monoallelic variant c.767A>G; Y256C in RPA2 Although the replication protein A2 (RPA2) mutant did not affect ssDNA binding and G-quadruplex-unfolding properties of RPA, the mutation reduced the affinity of RPA2 with the ubiquitin ligase RFWD3 and reduced RPA ubiquitination. Using engineered knock-in cell lines, we found an accumulation of RPA at telomeres that did not trigger ATR activation but caused short and dysfunctional telomeres. Finally, both patients acquired, in a subset of blood cells, somatic genetic rescue events in either POT1 genes or TERT promoters known to counteract the accelerated telomere shortening. Collectively, our study indicates that variants in RPA2 represent a novel genetic cause of TBDs. Our results further support the fundamental role of the RPA complex in regulating telomere length and stability in humans.


Asunto(s)
Proteína de Replicación A , Proteínas de Unión a Telómeros , Telómero , Humanos , Proteína de Replicación A/genética , Proteína de Replicación A/metabolismo , Telómero/genética , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo , Heterocigoto , Masculino , Femenino , Complejo Shelterina , Acortamiento del Telómero/genética , Mutación , Telomerasa/genética , Telomerasa/metabolismo , Ubiquitinación/genética , Ubiquitina-Proteína Ligasas/genética
6.
Nat Commun ; 15(1): 7464, 2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39198528

RESUMEN

RNase H1 has been acknowledged as an endoribonuclease specializing in the internal degradation of the RNA moiety within RNA-DNA hybrids, and its ribonuclease activity is indispensable in multifaceted aspects of nucleic acid metabolism. However, the molecular mechanism underlying RNase H1-mediated hybrid cleavage remains inadequately elucidated. Herein, using single-molecule approaches, we probe the dynamics of the hybrid cleavage by Saccharomyces cerevisiae RNase H1. Remarkably, a single RNase H1 enzyme displays 3'-to-5' exoribonuclease activity. The directional RNA degradation proceeds processively and yet discretely, wherein unwinding approximately 6-bp hybrids as a prerequisite for two consecutive 3-nt RNA excisions limits the overall rate within each catalytic cycle. Moreover, Replication Protein A (RPA) reinforces RNase H1's 3'-to-5' nucleolytic rate and processivity and stimulates its 5'-to-3' exoribonuclease activity. This stimulation is primarily realized through the pre-separation of the hybrids and consequently transfers RNase H1 to a bidirectional exoribonuclease, further potentiating its cleavage efficiency. These findings unveil unprecedented characteristics of an RNase and provide a dynamic view of RPA-enhanced processive hybrid cleavage by RNase H1.


Asunto(s)
Exorribonucleasas , ARN , Proteína de Replicación A , Ribonucleasa H , Saccharomyces cerevisiae , Ribonucleasa H/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Exorribonucleasas/metabolismo , Exorribonucleasas/genética , ARN/metabolismo , ARN/genética , Proteína de Replicación A/metabolismo , ADN/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Estabilidad del ARN , Hibridación de Ácido Nucleico
7.
Int J Mol Sci ; 25(15)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39125953

RESUMEN

Targeting CDC20 can enhance the radiosensitivity of tumor cells, but the function and mechanism of CDC20 on DNA damage repair response remains vague. To examine that issue, tumor cell lines, including KYSE200, KYSE450, and HCT116, were utilized to detect the expression, function, and underlying mechanism of CDC20 in radio-chemoresistance. Western blot and immunofluorescence staining were employed to confirm CDC20 expression and location, and radiation could upregulate the expression of CDC20 in the cell nucleus. The homologous recombination (HR) and non-homologous end joining (NHEJ) reporter gene systems were utilized to explore the impact of CDC20 on DNA damage repair, indicating that CDC20 could promote HR repair and radio/chemo-resistance. In the early stages of DNA damage, CDC20 stabilizes the RPA1 protein through protein-protein interactions, activating the ATR-mediated signaling cascade, thereby aiding in genomic repair. In the later stages, CDC20 assists in the subsequent steps of damage repair by the ubiquitin-mediated degradation of RPA1. CCK-8 and colony formation assay were used to detect the function of CDC20 in cell vitality and proliferation, and targeting CDC20 can exacerbate the increase in DNA damage levels caused by cisplatin or etoposide. A tumor xenograft model was conducted in BALB/c-nu/nu mice to confirm the function of CDC20 in vivo, confirming the in vitro results. In conclusion, this study provides further validation of the potential clinical significance of CDC20 as a strategy to overcome radio-chemoresistance via uncovering a novel role of CDC20 in regulating RPA1 during DNA damage repair.


Asunto(s)
Proteínas Cdc20 , Daño del ADN , Resistencia a Antineoplásicos , Tolerancia a Radiación , Proteína de Replicación A , Humanos , Animales , Proteína de Replicación A/metabolismo , Proteína de Replicación A/genética , Ratones , Tolerancia a Radiación/efectos de los fármacos , Tolerancia a Radiación/genética , Resistencia a Antineoplásicos/genética , Proteínas Cdc20/metabolismo , Proteínas Cdc20/genética , Línea Celular Tumoral , Ratones Endogámicos BALB C , Ratones Desnudos , Reparación del ADN/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Células HCT116 , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos
8.
DNA Repair (Amst) ; 142: 103741, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39153403

RESUMEN

PrimPol is a human DNA primase involved in DNA damage tolerance pathways by restarting DNA replication downstream of DNA lesions and non-canonical DNA structures. Activity and affinity to DNA relays on the interaction of PrimPol with replication protein A (RPA). In this work, we report that PrimPol has an intrinsic ability to copy DNA hairpins with a stem length of 5-9 base pairs (bp) but shows pronounced pausing of DNA synthesis. RPA greatly stimulates DNA synthesis across inverted DNA repeats by PrimPol. Moreover, deletion of the C-terminal RPA binding motif (RBM) facilitates DNA hairpin bypass and makes it independent of RPA. This work supports the idea that RBM is a negative regulator of PrimPol and its interaction with RPA is required to achieve the fully active state.


Asunto(s)
ADN Primasa , Replicación del ADN , ADN , Humanos , ADN Primasa/metabolismo , ADN Primasa/química , ADN Primasa/genética , ADN/metabolismo , Enzimas Multifuncionales/metabolismo , Enzimas Multifuncionales/genética , Enzimas Multifuncionales/química , Proteína de Replicación A/metabolismo , Conformación de Ácido Nucleico , ADN Polimerasa Dirigida por ADN/metabolismo , Secuencias Invertidas Repetidas , Unión Proteica
9.
G3 (Bethesda) ; 14(10)2024 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-39150943

RESUMEN

Human APOBEC single-strand (ss) specific DNA and RNA cytidine deaminases change cytosines to uracils (U's) and function in antiviral innate immunity and RNA editing and can cause hypermutation in chromosomes. The resulting U's can be directly replicated, resulting in C to T mutations, or U-DNA glycosylase can convert the U's to abasic (AP) sites which are then fixed as C to T or C to G mutations by translesion DNA polymerases. We noticed that in yeast and in human cancers, contributions of C to T and C to G mutations depend on the origin of ssDNA mutagenized by APOBECs. Since ssDNA in eukaryotic genomes readily binds to replication protein A (RPA) we asked if RPA could affect APOBEC-induced mutation spectrum in yeast. For that purpose, we expressed human APOBECs in the wild-type (WT) yeast and in strains carrying a hypomorph mutation rfa1-t33 in the large RPA subunit. We confirmed that the rfa1-t33 allele can facilitate mutagenesis by APOBECs. We also found that the rfa1-t33 mutation changed the ratio of APOBEC3A-induced T to C and T to G mutations in replicating yeast to resemble a ratio observed in long persistent ssDNA in yeast and in cancers. We present the data suggesting that RPA may shield APOBEC formed U's in ssDNA from Ung1, thereby facilitating C to T mutagenesis through the accurate copying of U's by replicative DNA polymerases. Unexpectedly, we also found that for U's shielded from Ung1 by WT RPA, the mutagenic outcome is reduced in the presence of translesion DNA polymerase zeta.


Asunto(s)
Mutagénesis , Mutación , Proteína de Replicación A , Saccharomyces cerevisiae , Proteína de Replicación A/metabolismo , Proteína de Replicación A/genética , Humanos , Saccharomyces cerevisiae/genética , Desaminasas APOBEC/metabolismo , Desaminasas APOBEC/genética , Citidina Desaminasa/metabolismo , Citidina Desaminasa/genética , ADN de Cadena Simple/metabolismo , Desaminasas APOBEC-1/genética , Desaminasas APOBEC-1/metabolismo , Subunidades de Proteína/metabolismo , Subunidades de Proteína/genética
10.
Nat Commun ; 15(1): 7197, 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39169038

RESUMEN

Templated DNA repair that occurs during homologous recombination and replication stress relies on RAD51. RAD51 activity is positively regulated by BRCA2 and the RAD51 paralogs. The Shu complex is a RAD51 paralog-containing complex consisting of SWSAP1, SWS1, and SPIDR. We demonstrate that SWSAP1-SWS1 binds RAD51, maintains RAD51 filament stability, and enables strand exchange. Using single-molecule confocal fluorescence microscopy combined with optical tweezers, we show that SWSAP1-SWS1 decorates RAD51 filaments proficient for homologous recombination. We also find SWSAP1-SWS1 enhances RPA diffusion on ssDNA. Importantly, we show human sgSWSAP1 and sgSWS1 knockout cells are sensitive to pharmacological inhibition of PARP and APE1. Lastly, we identify cancer variants in SWSAP1 that alter Shu complex formation. Together, we show that SWSAP1-SWS1 stimulates RAD51-dependent high-fidelity repair and may be an important new cancer therapeutic target.


Asunto(s)
ADN de Cadena Simple , Recombinasa Rad51 , Proteína de Replicación A , Humanos , Reparación del ADN , ADN de Cadena Simple/metabolismo , ADN de Cadena Simple/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Recombinación Homóloga , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Unión Proteica , Recombinasa Rad51/metabolismo , Recombinasa Rad51/genética , Proteína de Replicación A/metabolismo , Proteína de Replicación A/genética , Imagen Individual de Molécula , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo
11.
Sci Rep ; 14(1): 19791, 2024 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-39187637

RESUMEN

The ATR kinase responds to elevated levels of single-stranded DNA (ssDNA) to activate the G2/M checkpoint, regulate origin utilization, preserve fork stability, and allow DNA repair to ensure genome integrity. The intrinsic replication stress in cancer cells makes this pathway an attractive therapeutic target. The ssDNA that drives ATR signaling is sensed by the ssDNA-binding protein replication protein A (RPA), which acts as a platform for ATRIP recruitment and subsequent ATR activation by TopBP1. We have developed chemical RPA inhibitors (RPAi) that block RPA-ssDNA interactions (RPA-DBi) and RPA protein-protein interactions (RPA-PPIi); both activities are required for ATR activation. Here, we biochemically reconstitute the ATR kinase signaling pathway and demonstrate that RPA-DBi and RPA-PPIi abrogate ATR-dependent phosphorylation of target proteins with selectivity advantages over active site ATR inhibitors. We demonstrate that RPA post-translational modifications (PTMs) impact ATR kinase activation but do not alter sensitivity to RPAi. Specifically, phosphorylation of RPA32 and TopBP1 stimulate, while RPA70 acetylation does not affect ATR phosphorylation of target proteins. Collectively, this work reveals the RPAi mechanism of action to inhibit ATR signaling that can be regulated by RPA PTMs and offers insight into the anti-cancer activity of ATR pathway-targeted cancer therapeutics.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada , ADN de Cadena Simple , Procesamiento Proteico-Postraduccional , Proteína de Replicación A , Transducción de Señal , Proteína de Replicación A/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Humanos , Fosforilación , ADN de Cadena Simple/metabolismo , Proteínas de Unión al ADN/metabolismo , Unión Proteica , Proteínas Portadoras , Proteínas Nucleares
12.
Proc Natl Acad Sci U S A ; 121(34): e2402262121, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39145931

RESUMEN

Homologous recombination (HR) is essential for the maintenance of genome stability. During HR, Replication Protein A (RPA) rapidly coats the 3'-tailed single-strand DNA (ssDNA) generated by end resection. Then, the ssDNA-bound RPA must be timely replaced by Rad51 recombinase to form Rad51 nucleoprotein filaments that drive homology search and HR repair. How cells regulate Rad51 assembly dynamics and coordinate RPA and Rad51 actions to ensure proper HR remains poorly understood. Here, we identified that Rtt105, a Ty1 transposon regulator, acts to stimulate Rad51 assembly and orchestrate RPA and Rad51 actions during HR. We found that Rtt105 interacts with Rad51 in vitro and in vivo and restrains the adenosine 5' triphosphate (ATP) hydrolysis activity of Rad51. We showed that Rtt105 directly stimulates dynamic Rad51-ssDNA assembly, strand exchange, and D-loop formation in vitro. Notably, we found that Rtt105 physically regulates the binding of Rad51 and RPA to ssDNA via different motifs and that both regulations are necessary and epistatic in promoting Rad51 nucleation, strand exchange, and HR repair. Consequently, disrupting either of the interactions impaired HR and conferred DNA damage sensitivity, underscoring the importance of Rtt105 in orchestrating the actions of Rad51 and RPA. Our work reveals additional layers of mechanisms regulating Rad51 filament dynamics and the coordination of HR.


Asunto(s)
ADN de Cadena Simple , Recombinasa Rad51 , Reparación del ADN por Recombinación , Proteína de Replicación A , Proteínas de Saccharomyces cerevisiae , Recombinasa Rad51/metabolismo , Proteína de Replicación A/metabolismo , Proteína de Replicación A/genética , ADN de Cadena Simple/metabolismo , ADN de Cadena Simple/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Unión Proteica
13.
J Struct Biol ; 216(3): 108115, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39117045

RESUMEN

Human RAD52 protein binds DNA and is involved in genomic stability maintenance and several forms of DNA repair, including homologous recombination and single-strand annealing. Despite its importance, there are very few structural details about the variability of the RAD52 ring size and the RAD52 C-terminal protein-protein interaction domains. Even recent attempts to employ cryogenic electron microscopy (cryoEM) methods on full-length yeast and human RAD52 do not reveal interpretable structures for the C-terminal half that contains the replication protein A (RPA) and RAD51 binding domains. In this study, we employed the monodisperse purification of two RAD52 deletion constructs and small angle X-ray scattering (SAXS) to construct a structural model that includes RAD52's RPA binding domain. This model is of interest to DNA repair specialists as well as for drug development against HR-deficient cancers.


Asunto(s)
Unión Proteica , Proteína Recombinante y Reparadora de ADN Rad52 , Proteína de Replicación A , Dispersión del Ángulo Pequeño , Humanos , Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , Proteína Recombinante y Reparadora de ADN Rad52/genética , Proteína Recombinante y Reparadora de ADN Rad52/química , Proteína de Replicación A/metabolismo , Proteína de Replicación A/química , Proteína de Replicación A/genética , Recombinasa Rad51/metabolismo , Recombinasa Rad51/química , Recombinasa Rad51/genética , Difracción de Rayos X/métodos , Reparación del ADN , Modelos Moleculares , Dominios Proteicos
14.
Oncogene ; 43(32): 2475-2489, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38961202

RESUMEN

The preferential response to PARP inhibitors (PARPis) in BRCA-deficient and Schlafen 11 (SLFN11)-expressing ovarian cancers has been documented, yet the underlying molecular mechanisms remain unclear. As the accumulation of single-strand DNA (ssDNA) gaps behind replication forks is key for the lethality effect of PARPis, we investigated the combined effects of SLFN11 expression and BRCA deficiency on PARPi sensitivity and ssDNA gap formation in human cancer cells. PARPis increased chromatin-bound RPA2 and ssDNA gaps in SLFN11-expressing cells and even more in cells with BRCA1 or BRCA2 deficiency. SLFN11 was co-localized with chromatin-bound RPA2 under PARPis treatment, with enhanced recruitment in BRCA2-deficient cells. Notably, the chromatin-bound SLFN11 under PARPis did not block replication, contrary to its function under replication stress. SLFN11 recruitment was attenuated by the inactivation of MRE11. Hence, under PARPi treatment, MRE11 expression and BRCA deficiency lead to ssDNA gaps behind replication forks, where SLFN11 binds and increases their accumulation. As ovarian cancer patients who responded (progression-free survival >2 years) to olaparib maintenance therapy had a significantly higher SLFN11-positivity than short-responders (<6 months), our findings provide a mechanistic understanding of the favorable responses to PARPis in SLFN11-expressing and BRCA-deficient tumors. It highlight the clinical implications of SLFN11.


Asunto(s)
Proteína BRCA1 , Proteína BRCA2 , Replicación del ADN , ADN de Cadena Simple , Proteína Homóloga de MRE11 , Neoplasias Ováricas , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Humanos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Replicación del ADN/efectos de los fármacos , ADN de Cadena Simple/genética , ADN de Cadena Simple/metabolismo , Femenino , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Neoplasias Ováricas/metabolismo , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Proteína Homóloga de MRE11/metabolismo , Proteína Homóloga de MRE11/genética , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Línea Celular Tumoral , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Proteína de Replicación A/metabolismo , Proteína de Replicación A/genética , Cromatina/metabolismo , Ftalazinas/farmacología
15.
J Assist Reprod Genet ; 41(9): 2419-2439, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39023827

RESUMEN

PURPOSE: Ovarian aging is closely related to a decrease in follicular reserve and oocyte quality. The precise molecular mechanisms underlying these reductions have yet to be fully elucidated. Herein, we examine spatiotemporal distribution of key proteins responsible for DNA double-strand break (DSB) repair in ovaries from early to older ages. Functional studies have shown that the γH2AX, RAD51, BRCA1, and RPA70 proteins play indispensable roles in HR-based repair pathway, while the KU80 and XRCC4 proteins are essential for successfully operating cNHEJ pathway. METHODS: Female Balb/C mice were divided into five groups as follows: Prepuberty (3 weeks old; n = 6), puberty (7 weeks old; n = 7), postpuberty (18 weeks old; n = 7), early aged (52 weeks old; n = 7), and late aged (60 weeks old; n = 7). The expression of DSB repair proteins, cellular senescence (ß-GAL) and apoptosis (cCASP3) markers was evaluated in the ovaries using immunohistochemistry. RESULT: ß-GAL and cCASP3 levels progressively increased from prepuberty to aged groups (P < 0.05). Notably, γH2AX levels varied in preantral and antral follicles among the groups (P < 0.05). In aged groups, RAD51, BRCA1, KU80, and XRCC4 levels increased (P < 0.05), while RPA70 levels decreased (P < 0.05) compared to the other groups. CONCLUSIONS: The observed alterations were primarily attributed to altered expression in oocytes and granulosa cells of the follicles and other ovarian cells. As a result, the findings indicate that these DSB repair proteins may play a role in the repair processes and even other related cellular events in ovarian cells from early to older ages.


Asunto(s)
Proteína BRCA1 , Roturas del ADN de Doble Cadena , Reparación del ADN , Proteínas de Unión al ADN , Histonas , Autoantígeno Ku , Folículo Ovárico , Ovario , Recombinasa Rad51 , Animales , Femenino , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Ratones , Autoantígeno Ku/metabolismo , Autoantígeno Ku/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Reparación del ADN/genética , Folículo Ovárico/metabolismo , Folículo Ovárico/crecimiento & desarrollo , Histonas/genética , Histonas/metabolismo , Ovario/metabolismo , Ovario/crecimiento & desarrollo , Oocitos/metabolismo , Oocitos/crecimiento & desarrollo , Envejecimiento/genética , Envejecimiento/metabolismo , Proteína de Replicación A/metabolismo , Proteína de Replicación A/genética , Ratones Endogámicos BALB C
16.
J Biol Chem ; 300(9): 107579, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39025455

RESUMEN

NEIL1 is a DNA glycosylase that recognizes and initiates base excision repair of oxidized bases. The ubiquitous ssDNA binding scaffolding protein, replication protein A (RPA), modulates NEIL1 activity in a manner that depends on DNA structure. Interaction between NEIL1 and RPA has been reported, but the molecular basis of this interaction has yet to be investigated. Using a combination of NMR spectroscopy and isothermal titration calorimetry (ITC), we show that NEIL1 interacts with RPA through two contact points. An interaction with the RPA32C protein recruitment domain was mapped to a motif in the common interaction domain (CID) of NEIL1 and a dissociation constant (Kd) of 200 nM was measured. A substantially weaker secondary interaction with the tandem RPA70AB ssDNA binding domains was also mapped to the CID. Together these two contact points reveal NEIL1 has a high overall affinity (Kd ∼ 20 nM) for RPA. A homology model of the complex of RPA32C with the NEIL1 RPA binding motif in the CID was generated and used to design a set of mutations in NEIL1 to disrupt the interaction, which was confirmed by ITC. The mutant NEIL1 remains catalytically active against a thymine glycol lesion in duplex DNA in vitro. Testing the functional effect of disrupting the NEIL1-RPA interaction in vivo using a Fluorescence Multiplex-Host Cell Reactivation (FM-HCR) reporter assay revealed an unexpected role for NEIL1 in nucleotide excision repair. These findings are discussed in the context of the role of NEIL1 in replication-associated repair.


Asunto(s)
ADN Glicosilasas , Reparación del ADN , Unión Proteica , Proteína de Replicación A , Proteína de Replicación A/metabolismo , Proteína de Replicación A/genética , Proteína de Replicación A/química , ADN Glicosilasas/metabolismo , ADN Glicosilasas/química , ADN Glicosilasas/genética , Humanos , Modelos Moleculares , Dominios Proteicos , Reparación por Escisión
17.
Nucleic Acids Res ; 52(15): 8880-8896, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-38967018

RESUMEN

The simian virus 40 (SV40) replisome only encodes for its helicase; large T-antigen (L-Tag), while relying on the host for the remaining proteins, making it an intriguing model system. Despite being one of the earliest reconstituted eukaryotic systems, the interactions coordinating its activities and the identification of new factors remain largely unexplored. Herein, we in vitro reconstituted the SV40 replisome activities at the single-molecule level, including DNA unwinding by L-Tag and the single-stranded DNA-binding protein Replication Protein A (RPA), primer extension by DNA polymerase δ, and their concerted leading-strand synthesis. We show that RPA stimulates the processivity of L-Tag without altering its rate and that DNA polymerase δ forms a stable complex with L-Tag during leading-strand synthesis. Furthermore, similar to human and budding yeast Cdc45-MCM-GINS helicase, L-Tag uses the fork protection complex (FPC) and the mini-chromosome maintenance protein 10 (Mcm10) during synthesis. Hereby, we demonstrate that FPC increases this rate, and both FPC and Mcm10 increase the processivity by stabilizing stalled replisomes and increasing their chances of restarting synthesis. The detailed kinetics and novel factors of the SV40 replisome establish it as a closer mimic of the host replisome and expand its application as a model replication system.


Asunto(s)
Replicación del ADN , Proteínas de Mantenimiento de Minicromosoma , Proteína de Replicación A , Virus 40 de los Simios , Virus 40 de los Simios/metabolismo , Virus 40 de los Simios/genética , Humanos , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Proteínas de Mantenimiento de Minicromosoma/genética , Proteína de Replicación A/metabolismo , ADN Polimerasa III/metabolismo , ADN Polimerasa III/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , ADN Helicasas/metabolismo , ADN Helicasas/genética , ADN Viral/metabolismo , ADN Viral/genética , Replicación Viral , Imagen Individual de Molécula , Antígenos Transformadores de Poliomavirus/metabolismo , Antígenos Transformadores de Poliomavirus/genética , ADN de Cadena Simple/metabolismo , ADN Polimerasa Dirigida por ADN , Complejos Multienzimáticos
18.
Anal Chem ; 96(25): 10443-10450, 2024 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-38864271

RESUMEN

Due to their ability to selectively target pathogen-specific nucleic acids, CRISPR-Cas systems are increasingly being employed as diagnostic tools. "One-pot" assays that combine nucleic acid amplification and CRISPR-Cas systems (NAAT-CRISPR-Cas) in a single step have emerged as one of the most popular CRISPR-Cas biosensing formats. However, operational simplicity comes at a cost, with one-pot assays typically being less sensitive than corresponding two-step NAAT-CRISPR-Cas assays and often failing to detect targets at low concentrations. It is thought that these performance reductions result from the competition between the two enzymatic processes driving the assay, namely, Cas-mediated cis-cleavage and polymerase-mediated amplification of the target DNA. Herein, we describe a novel one-pot RPA-Cas12a assay that circumvents this issue by leveraging in situ complexation of the target-specific sgRNA and Cas12a to purposefully limit the concentration of active Cas12a during the early stages of the assay. Using a clinically relevant assay against a DNA target for HPV-16, we show how this in situ format reduces competition between target cleavage and amplification and engenders significant improvements in detection limit when compared to the traditional one-pot assay format, even in patient-derived samples. Finally, to gain further insight into the assay, we use experimental data to formulate a mechanistic model describing the competition between the Cas enzyme and nucleic acid amplification. These findings suggest that purposefully limiting cis-cleavage rates of Cas proteins is a viable strategy for improving the performance of one-pot NAAT-CRISPR-Cas assays.


Asunto(s)
Proteínas Asociadas a CRISPR , Sistemas CRISPR-Cas , ARN Guía de Sistemas CRISPR-Cas , Sistemas CRISPR-Cas/genética , Proteínas Asociadas a CRISPR/metabolismo , ARN Guía de Sistemas CRISPR-Cas/metabolismo , Humanos , Endodesoxirribonucleasas/metabolismo , Endodesoxirribonucleasas/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Técnicas de Amplificación de Ácido Nucleico , Proteína de Replicación A/metabolismo , Técnicas Biosensibles/métodos
19.
J Biol Chem ; 300(7): 107461, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38876299

RESUMEN

Theta-mediated end joining (TMEJ) is critical for survival of cancer cells when other DNA double-stranded break repair pathways are impaired. Human DNA polymerase theta (Pol θ) can extend ssDNA oligonucleotides, but little is known about preferred substrates and mechanism. We show that Pol θ can extend both ssDNA and RNA substrates by unimolecular stem-loop synthesis initiated by only two 3' terminal base pairs. Given sufficient time, Pol θ uses alternative pairing configurations that greatly expand the repertoire of sequence outcomes. Further primer-template adjustments yield low-fidelity outcomes when the nucleotide pool is imbalanced. Unimolecular stem-loop synthesis competes with bimolecular end joining, even when a longer terminal microhomology for end joining is available. Both reactions are partially suppressed by the ssDNA-binding protein replication protein A. Protein-primer grasp residues that are specific to Pol θ are needed for rapid stem-loop synthesis. The ability to perform stem-loop synthesis from a minimally paired primer is rare among human DNA polymerases, but we show that human DNA polymerases Pol η and Pol λ can catalyze related reactions. Using purified human Pol θ, we reconstituted in vitro TMEJ incorporating an insertion arising from a stem-loop extension. These activities may help explain TMEJ repair events that include inverted repeat sequences.


Asunto(s)
ADN Polimerasa theta , ADN Polimerasa Dirigida por ADN , Humanos , Reparación del ADN por Unión de Extremidades , ADN Polimerasa beta/metabolismo , ADN Polimerasa beta/genética , ADN Polimerasa beta/química , Reparación del ADN , ADN de Cadena Simple/metabolismo , ADN de Cadena Simple/genética , ADN de Cadena Simple/química , ADN Polimerasa Dirigida por ADN/metabolismo , ADN Polimerasa Dirigida por ADN/genética , Proteína de Replicación A/metabolismo , Proteína de Replicación A/genética
20.
Nucleic Acids Res ; 52(13): 7650-7664, 2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-38842913

RESUMEN

DNA polymerase δ (pol δ) holoenzymes, comprised of pol δ and the processivity sliding clamp, PCNA, carry out DNA synthesis during lagging strand replication, initiation of leading strand replication, and the major DNA damage repair and tolerance pathways. Pol δ holoenzymes are assembled at primer/template (P/T) junctions and initiate DNA synthesis in a stepwise process involving the major single strand DNA (ssDNA)-binding protein complex, RPA, the processivity sliding clamp loader, RFC, PCNA and pol δ. During this process, the interactions of RPA, RFC and pol δ with a P/T junction all significantly overlap. A burning issue that has yet to be resolved is how these overlapping interactions are accommodated during this process. To address this, we design and utilize novel, ensemble FRET assays that continuously monitor the interactions of RPA, RFC, PCNA and pol δ with DNA as pol δ holoenzymes are assembled and initiate DNA synthesis. Results from the present study reveal that RPA remains engaged with P/T junctions throughout this process and the RPA•DNA complexes dynamically re-organize to allow successive binding of RFC and pol δ. These results have broad implications as they highlight and distinguish the functional consequences of dynamic RPA•DNA interactions in RPA-dependent DNA metabolic processes.


Asunto(s)
ADN Polimerasa III , Replicación del ADN , ADN , Antígeno Nuclear de Célula en Proliferación , Proteína de Replicación A , Proteína de Replicación C , Moldes Genéticos , Proteína de Replicación A/metabolismo , ADN Polimerasa III/metabolismo , ADN Polimerasa III/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Antígeno Nuclear de Célula en Proliferación/genética , Holoenzimas/metabolismo , ADN/metabolismo , ADN/biosíntesis , Proteína de Replicación C/metabolismo , Proteína de Replicación C/genética , Cartilla de ADN/genética , Transferencia Resonante de Energía de Fluorescencia , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA