Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 827
Filtrar
Mais filtros

País/Região como assunto
Intervalo de ano de publicação
1.
Cell ; 176(1-2): 167-181.e21, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30595447

RESUMO

Covalent DNA-protein cross-links (DPCs) impede replication fork progression and threaten genome integrity. Using Xenopus egg extracts, we previously showed that replication fork collision with DPCs causes their proteolysis, followed by translesion DNA synthesis. We show here that when DPC proteolysis is blocked, the replicative DNA helicase CMG (CDC45, MCM2-7, GINS), which travels on the leading strand template, bypasses an intact leading strand DPC. Single-molecule imaging reveals that GINS does not dissociate from CMG during bypass and that CMG slows dramatically after bypass, likely due to uncoupling from the stalled leading strand. The DNA helicase RTEL1 facilitates bypass, apparently by generating single-stranded DNA beyond the DPC. The absence of RTEL1 impairs DPC proteolysis, suggesting that CMG must bypass the DPC to enable proteolysis. Our results suggest a mechanism that prevents inadvertent CMG destruction by DPC proteases, and they reveal CMG's remarkable capacity to overcome obstacles on its translocation strand.


Assuntos
DNA Helicases/metabolismo , DNA Helicases/fisiologia , Reparo do DNA/fisiologia , Animais , Proteínas de Ciclo Celular/metabolismo , DNA/metabolismo , Replicação do DNA , DNA de Cadeia Simples , Proteínas de Ligação a DNA/fisiologia , Feminino , Masculino , Proteólise , Imagem Individual de Molécula/métodos , Xenopus laevis/metabolismo
2.
Mol Cell ; 72(3): 568-582.e6, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30344097

RESUMO

Protecting stalled DNA replication forks from degradation by promiscuous nucleases is essential to prevent genomic instability, a major driving force of tumorigenesis. Several proteins commonly associated with the repair of DNA double-strand breaks (DSBs) by homologous recombination (HR) have been implicated in the stabilization of stalled forks. Human CtIP, in conjunction with the MRE11 nuclease complex, plays an important role in HR by promoting DSB resection. Here, we report an unanticipated function for CtIP in protecting reversed forks from degradation. Unlike BRCA proteins, which defend nascent DNA strands from nucleolytic attack by MRE11, we find that CtIP protects perturbed forks from erroneous over-resection by DNA2. Finally, we uncover functionally synergistic effects between CtIP and BRCA1 in mitigating replication-stress-induced genomic instability. Collectively, our findings reveal a DSB-resection- and MRE11-independent role for CtIP in preserving fork integrity that contributes to the survival of BRCA1-deficient cells.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Transporte/fisiologia , Replicação do DNA/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiologia , Proteína BRCA1 , Proteína BRCA2 , Linhagem Celular , Quebras de DNA de Cadeia Dupla , DNA Helicases/fisiologia , Reparo do DNA , Proteínas de Ligação a DNA , Desoxirribonucleases , Endodesoxirribonucleases , Instabilidade Genômica/fisiologia , Recombinação Homóloga/genética , Humanos , Proteína Homóloga a MRE11/metabolismo , Ligação Proteica
3.
Genes Dev ; 32(2): 165-180, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29440260

RESUMO

Multiple congenital disorders often present complex phenotypes, but how the mutation of individual genetic factors can lead to multiple defects remains poorly understood. In the present study, we used human neuroepithelial (NE) cells and CHARGE patient-derived cells as an in vitro model system to identify the function of chromodomain helicase DNA-binding 7 (CHD7) in NE-neural crest bifurcation, thus revealing an etiological link between the central nervous system (CNS) and craniofacial anomalies observed in CHARGE syndrome. We found that CHD7 is required for epigenetic activation of superenhancers and CNS-specific enhancers, which support the maintenance of the NE and CNS lineage identities. Furthermore, we found that BRN2 and SOX21 are downstream effectors of CHD7, which shapes cellular identities by enhancing a CNS-specific cellular program and indirectly repressing non-CNS-specific cellular programs. Based on our results, CHD7, through its interactions with superenhancer elements, acts as a regulatory hub in the orchestration of the spatiotemporal dynamics of transcription factors to regulate NE and CNS lineage identities.


Assuntos
DNA Helicases/fisiologia , Proteínas de Ligação a DNA/fisiologia , Epigênese Genética , Células-Tronco Neurais/metabolismo , Células Neuroepiteliais/metabolismo , Síndrome CHARGE/genética , Linhagem Celular , Linhagem da Célula/genética , DNA Helicases/genética , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Elementos Facilitadores Genéticos , Haploinsuficiência , Humanos , Crista Neural/metabolismo , Transcrição Gênica
4.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35042797

RESUMO

Srs2 is a superfamily 1 (SF1) helicase that participates in several pathways necessary for the repair of damaged DNA. Srs2 regulates formation of early homologous recombination (HR) intermediates by actively removing the recombinase Rad51 from single-stranded DNA (ssDNA). It is not known whether and how Srs2 itself is down-regulated to allow for timely HR progression. Rad54 and Rdh54 are two closely related superfamily 2 (SF2) motor proteins that promote the formation of Rad51-dependent recombination intermediates. Rad54 and Rdh54 bind tightly to Rad51-ssDNA and act downstream of Srs2, suggesting that they may affect the ability of Srs2 to dismantle Rad51 filaments. Here, we used DNA curtains to determine whether Rad54 and Rdh54 alter the ability of Srs2 to disrupt Rad51 filaments. We show that Rad54 and Rdh54 act synergistically to greatly restrict the antirecombinase activity of Srs2. Our findings suggest that Srs2 may be accorded only a limited time window to act and that Rad54 and Rdh54 fulfill a role of prorecombinogenic licensing factors.


Assuntos
DNA Helicases/metabolismo , Enzimas Reparadoras do DNA/metabolismo , DNA Topoisomerases/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Ciclo Celular/metabolismo , Dano ao DNA/fisiologia , DNA Helicases/fisiologia , Reparo do DNA/genética , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/fisiologia , DNA Topoisomerases/fisiologia , DNA de Cadeia Simples/metabolismo , Proteínas de Ligação a DNA/metabolismo , Recombinação Homóloga/genética , Ligação Proteica/genética , Rad51 Recombinase/metabolismo , Rad51 Recombinase/fisiologia , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/fisiologia
5.
Genes Dev ; 31(14): 1469-1482, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28860160

RESUMO

Protection of the stalled replication fork is crucial for responding to replication stress and minimizing its impact on chromosome instability, thus preventing diseases, including cancer. We found a new component, Abro1, in the protection of stalled replication fork integrity. Abro1 deficiency results in increased chromosome instability, and Abro1-null mice are tumor-prone. We show that Abro1 protects stalled replication fork stability by inhibiting DNA2 nuclease/WRN helicase-mediated degradation of stalled forks. Depletion of RAD51 prevents the DNA2/WRN-dependent degradation of stalled forks in Abro1-deficient cells. This mechanism is distinct from the BRCA2-dependent fork protection pathway, in which stable RAD51 filament formation prevents MRE11-dependent degradation of the newly synthesized DNA at stalled forks. Thus, our data reveal a new aspect of regulated protection of stalled replication forks that involves Abro1.


Assuntos
Replicação do DNA , Instabilidade Genômica , Proteínas Associadas à Matriz Nuclear/fisiologia , Proteases Específicas de Ubiquitina/fisiologia , Animais , Proteína BRCA2/genética , Linhagem Celular , Células Cultivadas , DNA/biossíntese , DNA Helicases/fisiologia , Endodesoxirribonucleases/fisiologia , Proteína Homóloga a MRE11/fisiologia , Camundongos Knockout , Enzimas Multifuncionais/fisiologia , Neoplasias Experimentais/genética , Proteínas Associadas à Matriz Nuclear/genética , Rad51 Recombinase/genética , Estresse Fisiológico , Proteases Específicas de Ubiquitina/genética
6.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-33846247

RESUMO

The P-loop Walker A motif underlies hundreds of essential enzyme families that bind nucleotide triphosphates (NTPs) and mediate phosphoryl transfer (P-loop NTPases), including the earliest DNA/RNA helicases, translocases, and recombinases. What were the primordial precursors of these enzymes? Could these large and complex proteins emerge from simple polypeptides? Previously, we showed that P-loops embedded in simple ßα repeat proteins bind NTPs but also, unexpectedly so, ssDNA and RNA. Here, we extend beyond the purely biophysical function of ligand binding to demonstrate rudimentary helicase-like activities. We further constructed simple 40-residue polypeptides comprising just one ß-(P-loop)-α element. Despite their simplicity, these P-loop prototypes confer functions such as strand separation and exchange. Foremost, these polypeptides unwind dsDNA, and upon addition of NTPs, or inorganic polyphosphates, release the bound ssDNA strands to allow reformation of dsDNA. Binding kinetics and low-resolution structural analyses indicate that activity is mediated by oligomeric forms spanning from dimers to high-order assemblies. The latter are reminiscent of extant P-loop recombinases such as RecA. Overall, these P-loop prototypes compose a plausible description of the sequence, structure, and function of the earliest P-loop NTPases. They also indicate that multifunctionality and dynamic assembly were key in endowing short polypeptides with elaborate, evolutionarily relevant functions.


Assuntos
Domínio AAA/genética , Domínio AAA/fisiologia , Motivos de Aminoácidos/fisiologia , Sequência de Aminoácidos/genética , DNA Helicases/metabolismo , DNA Helicases/fisiologia , DNA de Cadeia Simples/química , DNA de Cadeia Simples/metabolismo , Modelos Moleculares , Nucleosídeo-Trifosfatase/química , Peptídeos/química , Fosfatos/química , Conformação Proteica em alfa-Hélice/fisiologia , Conformação Proteica em Folha beta/fisiologia , Proteínas/química , RNA/química , Recombinases Rec A/metabolismo
7.
Nat Immunol ; 12(10): 929-30, 2011 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-21934672

RESUMO

The sensing of pathogen-associated DNA in the cytoplasm is an important trigger of host-defense responses that include the production of type I interferon. A new study suggests that the DExDc helicase DDX41 may function in dendritic cells as a DNA sensor to activate STING-dependent innate immune responses.


Assuntos
RNA Helicases DEAD-box/fisiologia , DNA Helicases/fisiologia , DNA/metabolismo , Células Dendríticas/fisiologia , Proteínas de Membrana/fisiologia , Animais , Humanos
8.
Nat Immunol ; 12(10): 959-65, 2011 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-21892174

RESUMO

The recognition of pathogenic DNA is important to the initiation of antiviral responses. Here we report the identification of DDX41, a member of the DEXDc family of helicases, as an intracellular DNA sensor in myeloid dendritic cells (mDCs). Knockdown of DDX41 expression by short hairpin RNA blocked the ability of mDCs to mount type I interferon and cytokine responses to DNA and DNA viruses. Overexpression of both DDX41 and the membrane-associated adaptor STING together had a synergistic effect in promoting Ifnb promoter activity. DDX41 bound both DNA and STING and localized together with STING in the cytosol. Knockdown of DDX41 expression blocked activation of the mitogen-activated protein kinase TBK1 and the transcription factors NF-κB and IRF3 by B-form DNA. Our results suggest that DDX41 is an additional DNA sensor that depends on STING to sense pathogenic DNA.


Assuntos
RNA Helicases DEAD-box/fisiologia , DNA Helicases/fisiologia , DNA/metabolismo , Células Dendríticas/fisiologia , Proteínas de Membrana/fisiologia , Animais , Células da Medula Óssea/metabolismo , Células Cultivadas , Humanos , Interferons/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/virologia , Estrutura Terciária de Proteína , Transdução de Sinais
9.
Cell ; 135(4): 623-34, 2008 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-19013274

RESUMO

The loading of oligomeric helicases onto replication origins marks an essential step in replisome assembly. In cells, dedicated AAA+ ATPases regulate loading, however, the mechanism by which these factors recruit and deposit helicases has remained unclear. To better understand this process, we determined the structure of the ATPase region of the bacterial helicase loader DnaC from Aquifex aeolicus to 2.7 A resolution. The structure shows that DnaC is a close paralog of the bacterial replication initiator, DnaA, and unexpectedly shares an ability to form a helical assembly similar to that of ATP-bound DnaA. Complementation and ssDNA-binding assays validate the importance of homomeric DnaC interactions, while pull-down experiments show that the DnaC and DnaA AAA+ domains interact in a nucleotide-dependent manner. These findings implicate DnaC as a molecular adaptor that uses ATP-activated DnaA as a docking site for regulating the recruitment and correct spatial deposition of the DnaB helicase onto origins.


Assuntos
Proteínas de Bactérias/química , DNA Helicases/fisiologia , Replicação do DNA , Proteínas de Ligação a DNA/química , DnaB Helicases/química , Proteínas de Escherichia coli/química , Adenosina Trifosfatases/metabolismo , Sequência de Aminoácidos , Bactérias/enzimologia , Cristalografia por Raios X/métodos , DNA Helicases/metabolismo , DNA de Cadeia Simples/química , Modelos Biológicos , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Homologia de Sequência de Aminoácidos
10.
Mol Cell ; 57(4): 622-635, 2015 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-25620558

RESUMO

The helicase RTEL1 promotes t-loop unwinding and suppresses telomere fragility to maintain the integrity of vertebrate telomeres. An interaction between RTEL1 and PCNA is important to prevent telomere fragility, but how RTEL1 engages with the telomere to promote t-loop unwinding is unclear. Here, we establish that the shelterin protein TRF2 recruits RTEL1 to telomeres in S phase, which is required to prevent catastrophic t-loop processing by structure-specific nucleases. We show that the TRF2-RTEL1 interaction is mediated by a metal-coordinating C4C4 motif in RTEL1, which is compromised by the Hoyeraal-Hreidarsson syndrome (HHS) mutation, RTEL1(R1264H). Conversely, we define a TRF2(I124D) substitution mutation within the TRFH domain of TRF2, which eliminates RTEL1 binding and phenocopies the RTEL1(R1264H) mutation, giving rise to aberrant t-loop excision, telomere length heterogeneity, and loss of the telomere as a circle. These results implicate TRF2 in the recruitment of RTEL1 to facilitate t-loop disassembly at telomeres in S phase.


Assuntos
DNA Helicases/fisiologia , Modelos Genéticos , Fase S , Telômero/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/fisiologia , Animais , Células Cultivadas , DNA Helicases/química , DNA Helicases/metabolismo , Humanos , Metáfase , Camundongos , Estrutura Terciária de Proteína , Transporte Proteico , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo
11.
Mol Cell ; 57(4): 595-606, 2015 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-25699708

RESUMO

The displacement loop (D loop) is a DNA strand invasion product formed during homologous recombination. Disruption of nascent D loops prevents recombination, and during synthesis-dependent strand annealing (SDSA), disruption of D loops extended by DNA polymerase ensures a non-crossover outcome. The proteins implicated in D loop disruption are DNA motor proteins/helicases that act by moving DNA junctions. Here we report that D loops can also be disrupted by DNA topoisomerase 3 (Top3), and this disruption depends on Top3's catalytic activity. Yeast Top3 specifically disrupts D loops mediated by yeast Rad51/Rad54; protein-free D loops or D loop mediated by bacterial RecA protein or human RAD51/RAD54 resist dissolution. Also, the human Topoisomerase IIIa-RMI1-RMI2 complex is capable of dissolving D loops. Consistent with genetic data, we suggest that the extreme growth defect and hyper-recombination phenotype of Top3-deficient yeast cells is partially a result of unprocessed D loops.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Recombinação Homóloga/fisiologia , Modelos Genéticos , Rad51 Recombinase/fisiologia , RecQ Helicases/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia , DNA Helicases/genética , DNA Helicases/metabolismo , DNA Helicases/fisiologia , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Enzimas Reparadoras do DNA/fisiologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Rad51 Recombinase/metabolismo , RecQ Helicases/genética , RecQ Helicases/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Especificidade da Espécie
12.
Nucleic Acids Res ; 49(5): 2418-2434, 2021 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-33590097

RESUMO

Cockayne syndrome (CS) is a segmental premature aging syndrome caused primarily by defects in the CSA or CSB genes. In addition to premature aging, CS patients typically exhibit microcephaly, progressive mental and sensorial retardation and cutaneous photosensitivity. Defects in the CSB gene were initially thought to primarily impair transcription-coupled nucleotide excision repair (TC-NER), predicting a relatively consistent phenotype among CS patients. In contrast, the phenotypes of CS patients are pleiotropic and variable. The latter is consistent with recent work that implicates CSB in multiple cellular systems and pathways, including DNA base excision repair, interstrand cross-link repair, transcription, chromatin remodeling, RNAPII processing, nucleolin regulation, rDNA transcription, redox homeostasis, and mitochondrial function. The discovery of additional functions for CSB could potentially explain the many clinical phenotypes of CSB patients. This review focuses on the diverse roles played by CSB in cellular pathways that enhance genome stability, providing insight into the molecular features of this complex premature aging disease.


Assuntos
DNA Helicases/fisiologia , Enzimas Reparadoras do DNA/fisiologia , Proteínas de Ligação a Poli-ADP-Ribose/fisiologia , Montagem e Desmontagem da Cromatina , Quebras de DNA de Cadeia Dupla , DNA Helicases/química , Reparo do DNA , Enzimas Reparadoras do DNA/química , DNA Ribossômico/biossíntese , Regulação da Expressão Gênica , Humanos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose/química , RNA Polimerase II/metabolismo , Transcrição Gênica
13.
Int J Cancer ; 150(4): 551-561, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-34460104

RESUMO

Stress granules (SGs) contain mRNAs and proteins stalled in translation during stress; these are increasingly being implicated in diseases, including neurological disorders and cancer. The dysregulated assembly, persistence, disassembly and clearance of SGs contribute to the process of senescence. Senescence has long been a mysterious player in cellular physiology and associated diseases. The systemic process of aging has been pivotal in the development of various neurological disorders like age-related neuropathy, Alzheimer's disease and Parkinson's disease. Glioma is a cancer of neurological origin with a very poor prognosis and high rate of recurrence, SGs have only recently been implicated in its pathogenesis. Senescence has long been established to play an antitumorigenic role, however, relatively less studied is its protumorigenic importance. Here, we have evaluated the existing literature to assess the crosstalk of the two biological phenomena of senescence and SG formation in the context of tumorigenesis. In this review, we have attempted to analyze the contribution of senescence in regulating diverse cellular processes, like, senescence associated secretory phenotype (SASP), microtubular reorganization, telomeric alteration, autophagic clearance and how intricately these phenomena are tied with the formation of SGs. Finally, we propose that interplay between senescence, its contributing factors and the genesis of SGs can drive tumorigenicity of gliomas, which can potentially be utilized for therapeutic intervention.


Assuntos
Neoplasias Encefálicas/etiologia , Senescência Celular/fisiologia , Glioma/etiologia , Grânulos de Estresse/fisiologia , Autofagia , Neoplasias Encefálicas/patologia , DNA Helicases/fisiologia , Progressão da Doença , Glioma/patologia , Humanos , Microtúbulos/química , Proteínas de Ligação a Poli-ADP-Ribose/fisiologia , RNA Helicases/fisiologia , Proteínas com Motivo de Reconhecimento de RNA/fisiologia , Telômero , Quinases Associadas a rho/fisiologia
14.
Hum Mol Genet ; 29(20): 3412-3430, 2020 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-33075803

RESUMO

Although ARID1A mutations are a hallmark feature, mutations in other SWI/SNF (SWItch/Sucrose Non-Fermentable) chromatin remodeling subunits are also observed in endometrial neoplasms. Here, we interrogated the roles of Brahma/SWI2-related gene 1 (BRG1, SMARCA4), the SWI/SNF catalytic subunit, in the endometrial epithelium. BRG1 loss affects more than one-third of all active genes and highly overlaps with the ARID1A gene regulatory network. Chromatin immunoprecipitation studies revealed widespread subunit-specific differences in transcriptional regulation, as BRG1 promoter interactions are associated with gene activation, while ARID1A binding is associated with gene repression. However, we identified a physiologically relevant subset of BRG1 and ARID1A co-regulated epithelial identity genes. Mice were genetically engineered to inactivate BRG1 specifically in the endometrial epithelium. Endometrial glands were observed embedded in uterine myometrium, indicating adenomyosis-like phenotypes. Molecular similarities were observed between BRG1 and ARID1A mutant endometrial cells in vivo, including loss of epithelial cell adhesion and junction genes. Collectively, these studies illustrate overlapping contributions of multiple SWI/SNF subunit mutations in the translocation of endometrium to distal sites, with loss of cell integrity being a common feature in SWI/SNF mutant endometrial epithelia.


Assuntos
Montagem e Desmontagem da Cromatina , DNA Helicases/fisiologia , Proteínas de Ligação a DNA/fisiologia , Endométrio/patologia , Epitélio/patologia , Regulação da Expressão Gênica , Mutação , Proteínas Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Animais , Endométrio/metabolismo , Epitélio/metabolismo , Feminino , Camundongos , Camundongos Knockout
15.
Mol Cell ; 53(3): 420-32, 2014 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-24486020

RESUMO

The displacement loop (D loop) is the product of homology search and DNA strand invasion, constituting a central intermediate in homologous recombination (HR). In eukaryotes, the Rad51 DNA strand exchange protein is assisted in D loop formation by the Rad54 motor protein. Curiously, Rad54 also disrupts D loops. How these opposing activities are coordinated toward productive recombination is unknown. Moreover, a seemingly disparate function of Rad54 is removal of Rad51 from heteroduplex DNA (hDNA) to allow HR-associated DNA synthesis. Here, we uncover features of D loop formation/dissociation dynamics, employing Rad51 filaments formed on ssDNAs that mimic the physiological length and structure of in vivo substrates. The Rad54 motor is activated by Rad51 bound to synapsed DNAs and guided by a ssDNA-binding domain. We present a unified model wherein Rad54 acts as an hDNA pump that drives D loop formation while simultaneously removing Rad51 from hDNA, consolidating both ATP-dependent activities of Rad54 into a single mechanistic step.


Assuntos
DNA Helicases/fisiologia , Enzimas Reparadoras do DNA/fisiologia , Recombinação Homóloga/fisiologia , Ácidos Nucleicos Heteroduplexes/metabolismo , Rad51 Recombinase/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/genética , DNA Helicases/genética , DNA Helicases/metabolismo , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , DNA de Cadeia Simples/metabolismo , DNA Super-Helicoidal/metabolismo , Modelos Genéticos , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
16.
Nucleic Acids Res ; 48(19): 10998-11015, 2020 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-33045725

RESUMO

G-quadruplex (G4) structures are stable non-canonical DNA structures that are implicated in the regulation of many cellular pathways. We show here that the G4-stabilizing compound PhenDC3 causes growth defects in Schizosaccharomyces pombe cells, especially during S-phase in synchronized cultures. By visualizing individual DNA molecules, we observed shorter DNA fragments of newly replicated DNA in the PhenDC3-treated cells, suggesting that PhenDC3 impedes replication fork progression. Furthermore, a novel single DNA molecule damage assay revealed increased single-strand DNA lesions in the PhenDC3-treated cells. Moreover, chromatin immunoprecipitation showed enrichment of the leading-strand DNA polymerase at sites of predicted G4 structures, suggesting that these structures impede DNA replication. We tested a subset of these sites and showed that they form G4 structures, that they stall DNA synthesis in vitro and that they can be resolved by the breast cancer-associated Pif1 family helicases. Our results thus suggest that G4 structures occur in S. pombe and that stabilized/unresolved G4 structures are obstacles for the replication machinery. The increased levels of DNA damage might further highlight the association of the human Pif1 helicase with familial breast cancer and the onset of other human diseases connected to unresolved G4 structures.


Assuntos
Quebras de DNA de Cadeia Simples , Replicação do DNA , DNA Fúngico/química , Quadruplex G , Schizosaccharomyces/genética , DNA Helicases/fisiologia , Compostos de Anéis Fundidos/farmacologia , Fase S , Proteínas de Schizosaccharomyces pombe/fisiologia
17.
J Am Soc Nephrol ; 32(11): 2815-2833, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34716243

RESUMO

BACKGROUND: Eya1 is a critical regulator of nephron progenitor cell specification and interacts with Six2 to promote NPC self-renewal. Haploinsufficiency of these genes causes kidney hypoplasia. However, how the Eya1-centered network operates remains unknown. METHODS: We engineered a 2×HA-3×Flag-Eya1 knock-in mouse line and performed coimmunoprecipitation with anti-HA or -Flag to precipitate the multitagged-Eya1 and its associated proteins. Loss-of-function, transcriptome profiling, and genome-wide binding analyses for Eya1's interacting chromatin-remodeling ATPase Brg1 were carried out. We assayed the activity of the cis-regulatory elements co-occupied by Brg1/Six2 in vivo. RESULTS: Eya1 and Six2 interact with the Brg1-based SWI/SNF complex during kidney development. Knockout of Brg1 results in failure of metanephric mesenchyme formation and depletion of nephron progenitors, which has been linked to loss of Eya1 expression. Transcriptional profiling shows conspicuous downregulation of important regulators for nephrogenesis in Brg1-deficient cells, including Lin28, Pbx1, and Dchs1-Fat4 signaling, but upregulation of podocyte lineage, oncogenic, and cell death-inducing genes, many of which Brg1 targets. Genome-wide binding analysis identifies Brg1 occupancy to a distal enhancer of Eya1 that drives nephron progenitor-specific expression. We demonstrate that Brg1 enrichment to two distal intronic enhancers of Pbx1 and a proximal promoter region of Mycn requires Six2 activity and that these Brg1/Six2-bound enhancers govern nephron progenitor-specific expression in response to Six2 activity. CONCLUSIONS: Our results reveal an essential role for Brg1, its downstream pathways, and its interaction with Eya1-Six2 in mediating the fine balance among the self-renewal, differentiation, and survival of nephron progenitors.


Assuntos
Montagem e Desmontagem da Cromatina , DNA Helicases/fisiologia , Elementos Facilitadores Genéticos , Proteínas de Homeodomínio/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Néfrons/citologia , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiologia , Proteínas Tirosina Fosfatases/metabolismo , Células-Tronco/citologia , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , Animais , Sequência de Bases , Diferenciação Celular , Autorrenovação Celular , Imunoprecipitação da Cromatina , Técnicas de Introdução de Genes , Células HEK293 , Proteínas de Homeodomínio/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Rim/embriologia , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Complexos Multiproteicos , Proteínas Nucleares/genética , Mapeamento de Interação de Proteínas , Proteínas Tirosina Fosfatases/genética , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Transcriptoma
18.
Int J Mol Sci ; 23(3)2022 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-35163266

RESUMO

DNA helicase and polymerase work cooperatively at the replication fork to perform leading-strand DNA synthesis. It was believed that the helicase migrates to the forefront of the replication fork where it unwinds the duplex to provide templates for DNA polymerases. However, the molecular basis of the helicase-polymerase coupling is not fully understood. The recently elucidated T7 replisome structure suggests that the helicase and polymerase sandwich parental DNA and each enzyme pulls a daughter strand in opposite directions. Interestingly, the T7 polymerase, but not the helicase, carries the parental DNA with a positively charged cleft and stacks at the fork opening using a ß-hairpin loop. Here, we created and characterized T7 polymerases each with a perturbed ß-hairpin loop and positively charged cleft. Mutations on both structural elements significantly reduced the strand-displacement synthesis by T7 polymerase but had only a minor effect on DNA synthesis performed against a linear DNA substrate. Moreover, the aforementioned mutations eliminated synergistic helicase-polymerase binding and unwinding at the DNA fork and processive fork progressions. Thus, our data suggested that T7 polymerase plays a dominant role in helicase-polymerase coupling and replisome progression.


Assuntos
DNA Helicases/metabolismo , Replicação do DNA/genética , DNA Polimerase Dirigida por DNA/metabolismo , Bacteriófago T7/enzimologia , Bacteriófago T7/metabolismo , DNA Helicases/fisiologia , Replicação do DNA/fisiologia , DNA Viral/metabolismo , DNA Polimerase Dirigida por DNA/fisiologia , Proteínas Virais/metabolismo
19.
FASEB J ; 34(9): 12646-12662, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32748509

RESUMO

The integrity of the genetic material is crucial for every organism. One intrinsic attack to genome stability is stalling of the replication fork which can result in DNA breakage. Several factors, such as DNA lesions or the formation of stable secondary structures (eg, G-quadruplexes) can lead to replication fork stalling. G-quadruplexes (G4s) are well-characterized stable secondary DNA structures that can form within specific single-stranded DNA sequence motifs and have been shown to block/pause the replication machinery. In most genomes several helicases have been described to regulate G4 unfolding to preserve genome integrity, however, different experiments raise the hypothesis that processing of G4s during DNA replication is more complex and requires additional, so far unknown, proteins. Here, we show that the Saccharomyces cerevisiae Mgs1 protein robustly binds to G4 structures in vitro and preferentially acts at regions with a strong potential to form G4 structures in vivo. Our results suggest that Mgs1 binds to G4-forming sites and has a role in the maintenance of genome integrity.


Assuntos
DNA Helicases/fisiologia , Proteínas de Ligação a DNA/fisiologia , Quadruplex G , Instabilidade Genômica , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/genética , DNA Fúngico/química , Ligação Proteica
20.
J Immunol ; 203(5): 1252-1264, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31375545

RESUMO

Somatic hypermutation generates a myriad of Ab mutants in Ag-specific B cells, from which high-affinity mutants are selected. Chickens, sheep, and rabbits use nontemplated point mutations and templated mutations via gene conversion to diversify their expressed Ig loci, whereas mice and humans rely solely on untemplated somatic point mutations. In this study, we demonstrate that, in addition to untemplated point mutations, templated mutagenesis readily occurs at the murine and human Ig loci. We provide two distinct lines of evidence that are not explained by the Neuberger model of somatic hypermutation: 1) across multiple data sets there is significant linkage disequilibrium between individual mutations, especially among close mutations, and 2) among those mutations, those <8 bp apart are significantly more likely to match microhomologous regions in the IgHV repertoire than predicted by the mutation profiles of somatic hypermutation. Together, this supports the role of templated mutagenesis during somatic diversification of Ag-activated B cells.


Assuntos
Ligação Genética , Loci Gênicos , Cadeias Pesadas de Imunoglobulinas/genética , Mutagênese , Hipermutação Somática de Imunoglobulina , Animais , DNA Helicases/fisiologia , Proteínas de Ligação a DNA/fisiologia , Centro Germinativo/imunologia , Humanos , Região Variável de Imunoglobulina/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mutação , Plasmócitos/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA