Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Cell ; 83(20): 3740-3753.e9, 2023 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-37832546

RESUMO

Mitochondrial DNA double-strand breaks (mtDSBs) lead to the degradation of circular genomes and a reduction in copy number; yet, the cellular response in human cells remains elusive. Here, using mitochondrial-targeted restriction enzymes, we show that a subset of cells with mtDSBs exhibited defective mitochondrial protein import, reduced respiratory complexes, and loss of membrane potential. Electron microscopy confirmed the altered mitochondrial membrane and cristae ultrastructure. Intriguingly, mtDSBs triggered the integrated stress response (ISR) via the phosphorylation of eukaryotic translation initiation factor 2α (eIF2α) by DELE1 and heme-regulated eIF2α kinase (HRI). When ISR was inhibited, the cells experienced intensified mitochondrial defects and slower mtDNA recovery post-breakage. Lastly, through proteomics, we identified ATAD3A-a membrane-bound protein interacting with nucleoids-as potentially pivotal in relaying signals from impaired genomes to the inner mitochondrial membrane. In summary, our study delineates the cascade connecting damaged mitochondrial genomes to the cytoplasm and highlights the significance of the ISR in maintaining mitochondrial homeostasis amid genome instability.


Assuntos
Mitocôndrias , eIF-2 Quinase , Humanos , Mitocôndrias/genética , Mitocôndrias/metabolismo , eIF-2 Quinase/metabolismo , Citoplasma/metabolismo , Fosforilação , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Fator de Iniciação 2 em Eucariotos/metabolismo , ATPases Associadas a Diversas Atividades Celulares/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo
2.
Science ; 381(6658): 653-660, 2023 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-37440612

RESUMO

Nonhomologous end-joining (NHEJ) and homologous recombination (HR) are the primary pathways for repairing DNA double-strand breaks (DSBs) during interphase, whereas microhomology-mediated end-joining (MMEJ) has been regarded as a backup mechanism. Through CRISPR-Cas9-based synthetic lethal screens in cancer cells, we identified subunits of the 9-1-1 complex (RAD9A-RAD1-HUS1) and its interacting partner, RHINO, as crucial MMEJ factors. We uncovered an unexpected function for RHINO in restricting MMEJ to mitosis. RHINO accumulates in M phase, undergoes Polo-like kinase 1 (PLK1) phosphorylation, and interacts with polymerase θ (Polθ), enabling its recruitment to DSBs for subsequent repair. Additionally, we provide evidence that MMEJ activity in mitosis repairs persistent DSBs that originate in S phase. Our findings offer insights into the synthetic lethal relationship between the genes POLQ and BRCA1 and BRAC2 and the synergistic effect of Polθ and poly(ADP-ribose) polymerase (PARP) inhibitors.


Assuntos
Proteínas de Ciclo Celular , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Exonucleases , Mitose , Humanos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Exonucleases/genética , Exonucleases/metabolismo , Células HEK293 , Mitose/genética , Poli(ADP-Ribose) Polimerases/metabolismo
3.
bioRxiv ; 2023 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-36993461

RESUMO

DNA double-strand breaks (DSBs) are toxic lesions that can lead to genome instability if not properly repaired. Breaks incurred in G1 phase of the cell cycle are predominantly fixed by non-homologous end-joining (NHEJ), while homologous recombination (HR) is the primary repair pathway in S and G2. Microhomology-mediated end-joining (MMEJ) is intrinsically error-prone and considered a backup DSB repair pathway that becomes essential when HR and NHEJ are compromised. In this study, we uncover MMEJ as the major DSB repair pathway in M phase. Using CRISPR/Cas9-based synthetic lethal screens, we identify subunits of the 9-1-1 complex (RAD9A-HUS1-RAD1) and its interacting partner, RHINO, as critical MMEJ factors. Mechanistically, we show that the function of 9-1-1 and RHINO in MMEJ is inconsistent with their well-established role in ATR signaling. Instead, RHINO plays an unexpected and essential role in directing mutagenic repair to M phase by directly binding to Polymerase theta (Polθ) and promoting its recruitment to DSBs in mitosis. In addition, we provide evidence that mitotic MMEJ repairs persistent DNA damage that originates in S phase but is not repaired by HR. The latter findings could explain the synthetic lethal relationship between POLQ and BRCA1/2 and the synergistic effect of Polθ and PARP inhibitors. In summary, our study identifies MMEJ as the primary pathway for repairing DSBs during mitosis and highlights an unanticipated role for RHINO in directing mutagenic repair to M phase.

4.
J Clin Invest ; 133(11)2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-36976649

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy that harbors mutations in homologous recombination-repair (HR-repair) proteins in 20%-25% of cases. Defects in HR impart a specific vulnerability to poly ADP ribose polymerase inhibitors and platinum-containing chemotherapy in tumor cells. However, not all patients who receive these therapies respond, and many who initially respond ultimately develop resistance. Inactivation of the HR pathway is associated with the overexpression of polymerase theta (Polθ, or POLQ). This key enzyme regulates the microhomology-mediated end-joining (MMEJ) pathway of double-strand break (DSB) repair. Using human and murine HR-deficient PDAC models, we found that POLQ knockdown is synthetically lethal in combination with mutations in HR genes such as BRCA1 and BRCA2 and the DNA damage repair gene ATM. Further, POLQ knockdown enhances cytosolic micronuclei formation and activates signaling of cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING), leading to enhanced infiltration of activated CD8+ T cells in BRCA2-deficient PDAC tumors in vivo. Overall, POLQ, a key mediator in the MMEJ pathway, is critical for DSB repair in BRCA2-deficient PDAC. Its inhibition represents a synthetic lethal approach to blocking tumor growth while concurrently activating the cGAS-STING signaling pathway to enhance tumor immune infiltration, highlighting what we believe to be a new role for POLQ in the tumor immune environment.


Assuntos
Adenocarcinoma , Neoplasias Pancreáticas , Humanos , Animais , Camundongos , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Quebras de DNA de Cadeia Dupla , Linhagem Celular Tumoral , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Recombinação Homóloga , Transdução de Sinais , Imunidade , Neoplasias Pancreáticas
5.
J Med Chem ; 65(19): 13198-13215, 2022 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-36126059

RESUMO

DNA polymerase theta (Polθ) is an attractive synthetic lethal target for drug discovery, predicted to be efficacious against breast and ovarian cancers harboring BRCA-mutant alleles. Here, we describe our hit-to-lead efforts in search of a selective inhibitor of human Polθ (encoded by POLQ). A high-throughput screening campaign of 350,000 compounds identified an 11 micromolar hit, giving rise to the N2-substituted fused pyrazolo series, which was validated by biophysical methods. Structure-based drug design efforts along with optimization of cellular potency and ADME ultimately led to the identification of RP-6685: a potent, selective, and orally bioavailable Polθ inhibitor that showed in vivo efficacy in an HCT116 BRCA2-/- mouse tumor xenograft model.


Assuntos
DNA Polimerase Dirigida por DNA , Neoplasias Ovarianas , Animais , Replicação do DNA , DNA Polimerase Dirigida por DNA/metabolismo , Desenho de Fármacos , Descoberta de Drogas , Feminino , Humanos , Camundongos
6.
Cell ; 185(5): 755-758, 2022 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-35245477

RESUMO

Support for basic science has been eclipsed by initiatives aimed at specific medical problems. The latest example is the dismantling of the Skirball Institute at NYU School of Medicine. Here, we reflect on the achievements and mission underlying the Skirball to gain insight into the dividends of maintaining a basic science vision within the academic enterprises.


Assuntos
Academias e Institutos , Pesquisa Biomédica , Faculdades de Medicina
7.
Genes Dev ; 36(5-6): 313-330, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35210222

RESUMO

In mammals, the conserved telomere binding protein Rap1 serves a diverse set of nontelomeric functions, including activation of the NF-kB signaling pathway, maintenance of metabolic function in vivo, and transcriptional regulation. Here, we uncover the mechanism by which Rap1 modulates gene expression. Using a separation-of-function allele, we show that Rap1 transcriptional regulation is largely independent of TRF2-mediated binding to telomeres and does not involve direct binding to genomic loci. Instead, Rap1 interacts with the TIP60/p400 complex and modulates its histone acetyltransferase activity. Notably, we show that deletion of Rap1 in mouse embryonic stem cells increases the fraction of two-cell-like cells. Specifically, Rap1 enhances the repressive activity of Tip60/p400 across a subset of two-cell-stage genes, including Zscan4 and the endogenous retrovirus MERVL. Preferential up-regulation of genes proximal to MERVL elements in Rap1-deficient settings implicates these endogenous retroviral elements in the derepression of proximal genes. Altogether, our study reveals an unprecedented link between Rap1 and the TIP60/p400 complex in the regulation of pluripotency.


Assuntos
Proteínas de Ligação a Telômeros , Telômero , Animais , Regulação da Expressão Gênica , Genoma , Mamíferos/genética , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Telômero/metabolismo , Proteínas de Ligação a Telômeros/genética , Proteínas de Ligação a Telômeros/metabolismo
8.
Trends Cell Biol ; 32(6): 527-536, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35063336

RESUMO

Telomeres are distinctive structures that protect the ends of linear chromosomes and ensure genome stability. They are composed of long tracks of repetitive and G-rich DNA that is bound by shelterin, a dedicated six-subunit protein complex. In somatic cells, shelterin protects telomeres from the DNA damage response and regulates telomere length. Telomere repeats are replenished by telomerase, a specialized ribonucleoprotein composed of telomerase reverse transcriptase and an integral RNA component. Telomere protection and telomerase regulation have been primarily studied in somatic cells. However, recent evidence points out striking differences in the context of embryonic stem cells (ESCs). In this review, we discuss insights into telomere protection in ESCs versus somatic cells and summarize findings on telomerase regulation as a function of pluripotency.


Assuntos
Telomerase , Células-Tronco Embrionárias , Instabilidade Genômica , Humanos , Complexo Shelterina , Telomerase/química , Telomerase/genética , Telomerase/metabolismo , Telômero/metabolismo
9.
Mol Cell ; 81(11): 2349-2360.e6, 2021 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-33852895

RESUMO

Telomere length control is critical for cellular lifespan and tumor suppression. Telomerase is transiently activated in the inner cell mass of the developing blastocyst to reset telomere reserves. Its silencing upon differentiation leads to gradual telomere shortening in somatic cells. Here, we report that transcriptional regulation through cis-regulatory elements only partially accounts for telomerase activation in pluripotent cells. Instead, developmental control of telomerase is primarily driven by an alternative splicing event, centered around hTERT exon 2. Skipping of exon 2 triggers hTERT mRNA decay in differentiated cells, and conversely, its retention promotes telomerase accumulation in pluripotent cells. We identify SON as a regulator of exon 2 alternative splicing and report a patient carrying a SON mutation and suffering from insufficient telomerase and short telomeres. In summary, our study highlights a critical role for hTERT alternative splicing in the developmental regulation of telomerase and implicates defective splicing in telomere biology disorders.


Assuntos
Processamento Alternativo , Proteínas de Ligação a DNA/genética , Elementos Facilitadores Genéticos , Antígenos de Histocompatibilidade Menor/genética , Telomerase/genética , Homeostase do Telômero , Telômero/metabolismo , Blastocisto/metabolismo , Blastocisto/patologia , Diferenciação Celular , Pré-Escolar , Proteínas de Ligação a DNA/deficiência , Feminino , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Embrionárias Humanas/patologia , Humanos , Linhagem , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/patologia , Cultura Primária de Células , Estabilidade de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Telomerase/deficiência , Telômero/patologia
10.
Nature ; 591(7850): 477-481, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33627873

RESUMO

Mitochondrial DNA double-strand breaks (mtDSBs) are toxic lesions that compromise the integrity of mitochondrial DNA (mtDNA) and alter mitochondrial function1. Communication between mitochondria and the nucleus is essential to maintain cellular homeostasis; however, the nuclear response to mtDSBs remains unknown2. Here, using mitochondrial-targeted transcription activator-like effector nucleases (TALENs)1,3,4, we show that mtDSBs activate a type-I interferon response that involves the phosphorylation of STAT1 and activation of interferon-stimulated genes. After the formation of breaks in the mtDNA, herniation5 mediated by BAX and BAK releases mitochondrial RNA into the cytoplasm and triggers a RIG-I-MAVS-dependent immune response. We further investigated the effect of mtDSBs on interferon signalling after treatment with ionizing radiation and found a reduction in the activation of interferon-stimulated genes when cells that lack mtDNA are exposed to gamma irradiation. We also show that mtDNA breaks synergize with nuclear DNA damage to mount a robust cellular immune response. Taken together, we conclude that cytoplasmic accumulation of mitochondrial RNA is an intrinsic immune surveillance mechanism for cells to cope with mtDSBs, including breaks produced by genotoxic agents.


Assuntos
Quebras de DNA de Cadeia Dupla , DNA Mitocondrial/imunologia , Imunidade Inata/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Linhagem Celular , Células Cultivadas , Quebras de DNA de Cadeia Dupla/efeitos da radiação , DNA Mitocondrial/efeitos da radiação , Humanos , Mitocôndrias/imunologia , Mitocôndrias/efeitos da radiação , Comunicação Parácrina , Radiação Ionizante , Transcrição Gênica , Ubiquitina-Proteína Ligases/metabolismo , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo
11.
Methods Mol Biol ; 2192: 21-34, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33230762

RESUMO

Single molecule analysis of replicating DNA (SMARD) is a powerful methodology that allows in vivo analysis of replicating DNA; identification of origins of replication, assessment of fork directionality, and measurement of replication fork speed. SMARD, which has been extensively used to study replication of nuclear DNA, involves incorporation of thymidine analogs to nascent DNA chains and their subsequent visualization through immune detection. Here, we adapt and fine-tune the SMARD technique to the specifics of human and mouse mitochondrial DNA. The mito-SMARD protocol allows researchers to gain in vivo insight into mitochondrial DNA (mtDNA) replication at the single molecule level and with high resolution.


Assuntos
Replicação do DNA/genética , DNA Mitocondrial/metabolismo , Imagem Individual de Molécula/métodos , Animais , Células Cultivadas , DNA Mitocondrial/genética , Genoma Mitocondrial , Humanos , Hibridização in Situ Fluorescente/métodos , Camundongos , Microscopia de Fluorescência/métodos , Mitocôndrias/metabolismo , Timidina/análogos & derivados , Timidina/metabolismo
12.
STAR Protoc ; 1(3): 100112, 2020 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-33377008

RESUMO

Advances in imaging technologies, gene editing, and fluorescent molecule development have made real-time imaging of nucleic acids practical. Here, we detail methods for imaging the human telomerase RNA template, hTR via the use of three inserted MS2 stem loops and cognate MS2 coat protein (MCP) tagged with superfolder GFP or photoactivatable GFP. These technologies enable tracking of the dynamics of RNA species through Cajal bodies and offer insight into their residence time in Cajal bodies through photobleaching and photoactivation experiments. For complete details on the use and execution of this protocol, please refer to Laprade et al. (2020).


Assuntos
Corpos Enovelados/metabolismo , RNA/análise , Imagem Individual de Molécula/métodos , Telomerase/análise , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Células HeLa , Humanos , Hibridização in Situ Fluorescente/métodos , Sequências Repetidas Invertidas/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Fotodegradação , RNA/genética , Telomerase/genética , Telômero/metabolismo
13.
Mol Cell Oncol ; 7(6): 1818537, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33241110

RESUMO

Telomerase plays a key role in the immortalization of cancer cells by maintaining telomeres length. Using single-molecule imaging of telomerase RNA molecules in cancer cells, we recently reported novel insights into the role of Cajal bodies in telomerase biogenesis and the regulation of telomerase recruitment to telomeres.

14.
STAR Protoc ; 1(2): 100104, 2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-33111129

RESUMO

Fluorescent in situ hybridization (FISH) on the RNA moiety of human telomerase (hTR) with 50-mer probes detects hTR RNA accumulated in Cajal bodies. Using both live-cell imaging and single-molecule inexpensive FISH, our published work revealed that only a fraction of hTR localizes to Cajal bodies, with the majority of hTR molecules distributed throughout the nucleoplasm. This protocol is an application guide to the smiFISH method for the dual detection of hTR RNA and telomeres or Cajal bodies by immunofluorescence. For complete details on the use and execution of this protocol, please refer to Laprade et al. (2020).


Assuntos
Hibridização in Situ Fluorescente/métodos , RNA/química , Imagem Individual de Molécula/métodos , Telomerase/química , Corpos Enovelados/metabolismo , Imunofluorescência/métodos , Células HeLa , Humanos , RNA/genética , Telomerase/genética , Telômero/metabolismo
15.
Genes Dev ; 34(23-24): 1619-1636, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33122293

RESUMO

Mutations in the telomere-binding protein POT1 are associated with solid tumors and leukemias. POT1 alterations cause rapid telomere elongation, ATR kinase activation, telomere fragility, and accelerated tumor development. Here, we define the impact of mutant POT1 alleles through complementary genetic and proteomic approaches based on CRISPR interference and biotin-based proximity labeling, respectively. These screens reveal that replication stress is a major vulnerability in cells expressing mutant POT1, which manifests as increased telomere mitotic DNA synthesis at telomeres. Our study also unveils a role for the nuclear pore complex in resolving replication defects at telomeres. Depletion of nuclear pore complex subunits in the context of POT1 dysfunction increases DNA damage signaling, telomere fragility and sister chromatid exchanges. Furthermore, we observed telomere repositioning to the nuclear periphery driven by nuclear F-actin polymerization in cells with POT1 mutations. In conclusion, our study establishes that relocalization of dysfunctional telomeres to the nuclear periphery is critical to preserve telomere repeat integrity.


Assuntos
Replicação do DNA/genética , Poro Nuclear/patologia , Proteínas de Ligação a Telômeros/genética , Telômero/genética , Linhagem Celular Tumoral , Dano ao DNA/genética , Humanos , Mitose/genética , Mutação , Neoplasias/genética , Neoplasias/fisiopatologia , Complexo Shelterina , Telômero/metabolismo , Proteínas de Ligação a Telômeros/metabolismo
16.
Nat Struct Mol Biol ; 27(8): 687-695, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32764737

RESUMO

Mitochondria respond to DNA damage and preserve their own genetic material in a manner distinct from that of the nucleus but that requires organized mito-nuclear communication. Failure to resolve mtDNA breaks leads to mitochondrial dysfunction and affects host cells and tissues. Here, we review the pathways that safeguard mitochondrial genomes and examine the insights gained from studies of cellular and tissue-wide responses to mtDNA damage and mito-nuclear genome incompatibility.


Assuntos
Dano ao DNA , Reparo do DNA , DNA Mitocondrial/genética , Genoma Mitocondrial , Animais , Eucariotos/genética , Humanos , Mitocôndrias/genética , Mitocôndrias/patologia
17.
Mol Cell ; 79(1): 115-126.e6, 2020 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-32497497

RESUMO

Extension of telomeres is a critical step in the immortalization of cancer cells. This complex reaction requires proper spatiotemporal coordination of telomerase and telomeres and remains poorly understood at the cellular level. To understand how cancer cells execute this process, we combine CRISPR genome editing and MS2 RNA tagging to image single molecules of telomerase RNA (hTR). Real-time dynamics and photoactivation experiments of hTR in Cajal bodies (CBs) reveal that hTERT controls the exit of hTR from CBs. Single-molecule tracking of hTR at telomeres shows that TPP1-mediated recruitment results in short telomere-telomerase scanning interactions, and then base pairing between hTR and telomere ssDNA promotes long interactions required for stable telomerase retention. Interestingly, POT1 OB-fold mutations that result in abnormally long telomeres in cancers act by enhancing this retention step. In summary, single-molecule imaging unveils the life cycle of telomerase RNA and provides a framework to reveal how cancer-associated mutations mechanistically drive defects in telomere homeostasis.


Assuntos
Corpos Enovelados/metabolismo , DNA de Cadeia Simples/metabolismo , RNA/metabolismo , Imagem Individual de Molécula/métodos , Telomerase/metabolismo , Homeostase do Telômero , Telômero/metabolismo , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , DNA de Cadeia Simples/genética , Edição de Genes , Células HeLa , Humanos , Mutação , RNA/genética , Complexo Shelterina , Telomerase/genética , Telômero/genética , Proteínas de Ligação a Telômeros/genética , Proteínas de Ligação a Telômeros/metabolismo
18.
Curr Opin Genet Dev ; 60: 119-126, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32302896

RESUMO

Mammalian cells have evolved multiple pathways to repair DNA double strand breaks (DSBs) and ensure genome stability. In addition to non-homologous end-joining (NHEJ) and homologous recombination (HR), cells evolved an error-prone repair pathway termed microhomology-mediated end joining (MMEJ). The mutagenic outcome of MMEJ derives from the activity of DNA polymerase theta (Polθ) - a multidomain enzyme that is minimally expressed in normal tissue but overexpressed in tumors. Polθ expression is particularly crucial for the proliferation of HR deficient cancer cells. As a result, this mutagenic repair emerged as an attractive target for cancer therapy, and inhibitors are currently in pre-clinical development. Here, we review the multifunctionality of this enigmatic polymerase, focusing on its role during DSB repair in mammalian cells and its impact on cancer genomes.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Replicação do DNA , DNA Polimerase Dirigida por DNA/metabolismo , Instabilidade Genômica , Neoplasias/genética , Neoplasias/patologia , DNA Polimerase Dirigida por DNA/genética , Recombinação Homóloga , Humanos , Neoplasias/enzimologia
19.
Methods Cell Biol ; 155: 401-414, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32183970

RESUMO

DNA combing technology is a powerful methodology for the study of DNA replication in vivo. This tool can be used to identify origins of replication, assess of directionality of forks, and measure fork speed. Over the years, the method has been used extensively to study nuclear DNA replication. The first step involves the incorporation of thymidine analogs (CldU and IdU) into nascent DNA chains and followed by their visualization with immunofluorescence using antibodies that can distinguish the two analogs. Recently, we adapted and fine-tuned DNA combing technology to the specifics of mitochondrial DNA (Phillips et al., 2017, p. 155). The protocol, which we termed mito-SMARD (mitochondrial single molecule analysis of replication DNA), provides in vivo insight into mitochondrial DNA (mtDNA) replication with high resolution.


Assuntos
Replicação do DNA/genética , DNA Mitocondrial/genética , Imagem Individual de Molécula/métodos , Linhagem Celular , DNA Mitocondrial/isolamento & purificação , Fluorescência , Humanos
20.
Nat Struct Mol Biol ; 24(12): 1116-1123, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29058711

RESUMO

Mammalian polymerase theta (Polθ) is a multifunctional enzyme that promotes error-prone DNA repair by alternative nonhomologous end joining (alt-NHEJ). Here we present structure-function analyses that reveal that, in addition to the polymerase domain, Polθ-helicase activity plays a central role during double-strand break (DSB) repair. Our results show that the helicase domain promotes chromosomal translocations by alt-NHEJ in mouse embryonic stem cells and also suppresses CRISPR-Cas9- mediated gene targeting by homologous recombination (HR). In vitro assays demonstrate that Polθ-helicase activity facilitates the removal of RPA from resected DSBs to allow their annealing and subsequent joining by alt-NHEJ. Consistent with an antagonistic role for RPA during alt-NHEJ, inhibition of RPA1 enhances end joining and suppresses recombination. Taken together, our results reveal that the balance between HR and alt-NHEJ is controlled by opposing activities of Polθ and RPA, providing further insight into the regulation of repair-pathway choice in mammalian cells.


Assuntos
Domínio Catalítico/genética , Reparo do DNA por Junção de Extremidades/genética , DNA Polimerase Dirigida por DNA/genética , Células-Tronco Embrionárias/citologia , Proteína de Replicação A/antagonistas & inibidores , Animais , Sistemas CRISPR-Cas/genética , Linhagem Celular , Quebras de DNA de Cadeia Dupla , Recombinação Homóloga/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína de Replicação A/genética , Relação Estrutura-Atividade , Translocação Genética/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...