Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 221
Filtrar
1.
Biosci Rep ; 43(2)2023 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-36601994

RESUMO

Central to the universal process of recombination, RecA family proteins form nucleoprotein filaments to catalyze production of heteroduplex DNA between substrate ssDNAs and template dsDNAs. ATP binding assists the filament in assuming the necessary conformation for forming heteroduplex DNA, but hydrolysis is not required. ATP hydrolysis has two identified roles which are not universally conserved: promotion of filament dissociation and enhancing flexibility of the filament. In this work, we examine ATP utilization of the RecA family recombinase SsoRadA from Saccharolobus solfataricus to determine its function in recombinase-mediated heteroduplex DNA formation. Wild-type SsoRadA protein and two ATPase mutant proteins were evaluated for the effects of three divalent metal cofactors. We found that unlike other archaeal RadA proteins, SsoRadA-mediated strand exchange is not enhanced by Ca2+. Instead, the S. solfataricus recombinase can utilize Mn2+ to stimulate strand invasion and reduce ADP-binding stability. Additionally, reduction of SsoRadA ATPase activity by Walker Box mutation or cofactor alteration resulted in a loss of large, complete strand exchange products. Depletion of ADP was found to improve initial strand invasion but also led to a similar loss of large strand exchange events. Our results indicate that overall, SsoRadA is distinct in its use of divalent cofactors but its activity with Mn2+ shows similarity to human RAD51 protein with Ca2+.


Assuntos
Cálcio , Sulfolobus solfataricus , Humanos , Cálcio/metabolismo , Ácidos Nucleicos Heteroduplexes/metabolismo , Recombinases Rec A/metabolismo , Sulfolobus solfataricus/genética , Sulfolobus solfataricus/metabolismo , Recombinases/metabolismo , Adenosina Trifosfatases/genética , Trifosfato de Adenosina/metabolismo
2.
Cell Rep ; 37(10): 110088, 2021 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-34879271

RESUMO

Alternative lengthening of telomeres (ALT) is a telomere-elongation mechanism observed in ∼15% of cancer subtypes. Current models indicate that ALT is mediated by homology-directed repair mechanisms. By disrupting MSH6 gene expression, we show that the deficiency of MutSα (MSH2/MSH6) DNA mismatch repair complex causes striking telomere hyperextension. Mechanistically, we show MutSα is specifically recruited to telomeres in ALT cells by associating with the proliferating-cell nuclear antigen (PCNA) subunit of the ALT telomere replisome. We also provide evidence that MutSα counteracts Bloom (BLM) helicase, which adopts a crucial role in stabilizing hyper-extended telomeres and maintaining the survival of MutSα-deficient ALT cancer cells. Lastly, we propose a model in which MutSα deficiency impairs heteroduplex rejection, leading to premature initiation of telomere DNA synthesis that coincides with an accumulation of telomere variant repeats (TVRs). These findings provide evidence that the MutSα DNA mismatch repair complex acts to restrain unwarranted ALT.


Assuntos
DNA de Neoplasias/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteína 2 Homóloga a MutS/metabolismo , Neoplasias/enzimologia , Ácidos Nucleicos Heteroduplexes/metabolismo , Homeostase do Telômero , Telômero/metabolismo , Linhagem Celular Tumoral , Reparo de Erro de Pareamento de DNA , DNA de Neoplasias/genética , Proteínas de Ligação a DNA/genética , Instabilidade Genômica , Células HeLa , Humanos , Modelos Genéticos , Proteína 2 Homóloga a MutS/genética , Neoplasias/genética , Neoplasias/patologia , Conformação de Ácido Nucleico , Ácidos Nucleicos Heteroduplexes/genética , RecQ Helicases/genética , RecQ Helicases/metabolismo , Telômero/genética
3.
Nat Commun ; 12(1): 7344, 2021 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-34937876

RESUMO

Manipulating lymphocyte functions with gene silencing approaches is promising for treating autoimmunity, inflammation, and cancer. Although oligonucleotide therapy has been proven to be successful in treating several conditions, efficient in vivo delivery of oligonucleotide to lymphocyte populations remains a challenge. Here, we demonstrate that intravenous injection of a heteroduplex oligonucleotide (HDO), comprised of an antisense oligonucleotide (ASO) and its complementary RNA conjugated to α-tocopherol, silences lymphocyte endogenous gene expression with higher potency, efficacy, and longer retention time than ASOs. Importantly, reduction of Itga4 by HDO ameliorates symptoms in both adoptive transfer and active experimental autoimmune encephalomyelitis models. Our findings reveal the advantages of HDO with enhanced gene knockdown effect and different delivery mechanisms compared with ASO. Thus, regulation of lymphocyte functions by HDO is a potential therapeutic option for immune-mediated diseases.


Assuntos
Linfócitos/metabolismo , Ácidos Nucleicos Heteroduplexes/metabolismo , Oligonucleotídeos/metabolismo , RNA/metabolismo , Administração Intravenosa , Transferência Adotiva , Animais , Doenças Desmielinizantes/genética , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/patologia , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Endocitose/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica , Inativação Gênica , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/imunologia , Humanos , Integrina alfa4/genética , Integrina alfa4/metabolismo , Células Jurkat , Masculino , Camundongos Endogâmicos C57BL , Ácidos Nucleicos Heteroduplexes/administração & dosagem , Ácidos Nucleicos Heteroduplexes/farmacocinética , Ácidos Nucleicos Heteroduplexes/farmacologia , Oligonucleotídeos/administração & dosagem , Oligonucleotídeos/farmacocinética , Oligonucleotídeos/farmacologia , RNA Longo não Codificante/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Medula Espinal/patologia , Distribuição Tecidual/efeitos dos fármacos
4.
Biochem J ; 477(18): 3567-3582, 2020 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-32886094

RESUMO

Recombination activating genes (RAGs), consisting of RAG1 and RAG2 have ability to perform spatially and temporally regulated DNA recombination in a sequence specific manner. Besides, RAGs also cleave at non-B DNA structures and are thought to contribute towards genomic rearrangements and cancer. The nonamer binding domain of RAG1 binds to the nonamer sequence of the signal sequence during V(D)J recombination. However, deletion of NBD did not affect RAG cleavage on non-B DNA structures. In the present study, we investigated the involvement of other RAG domains when RAGs act as a structure-specific nuclease. Studies using purified central domain (CD) and C-terminal domain (CTD) of the RAG1 showed that CD of RAG1 exhibited high affinity and specific binding to heteroduplex DNA, which was irrespective of the sequence of single-stranded DNA, unlike CTD which showed minimal binding. Furthermore, we show that ZnC2 of RAG1 is crucial for its binding to DNA structures as deletion and point mutations abrogated the binding of CD to heteroduplex DNA. Our results also provide evidence that unlike RAG cleavage on RSS, central domain of RAG1 is sufficient to cleave heteroduplex DNA harbouring pyrimidines, but not purines. Finally, we show that a point mutation in the DDE catalytic motif is sufficient to block the cleavage of CD on heteroduplex DNA. Therefore, in the present study we demonstrate that the while ZnC2 module in central domain of RAG1 is required for binding to non-B DNA structures, active site amino acids are important for RAGs to function as a structure-specific nuclease.


Assuntos
Proteínas de Homeodomínio/química , Ácidos Nucleicos Heteroduplexes/química , Motivos de Aminoácidos , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células HEK293 , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ácidos Nucleicos Heteroduplexes/genética , Ácidos Nucleicos Heteroduplexes/metabolismo , Domínios Proteicos , Relação Estrutura-Atividade , Recombinação V(D)J
5.
Nat Commun ; 11(1): 2950, 2020 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-32528002

RESUMO

During homologous recombination, Rad51 forms a nucleoprotein filament on single-stranded DNA to promote DNA strand exchange. This filament binds to double-stranded DNA (dsDNA), searches for homology, and promotes transfer of the complementary strand, producing a new heteroduplex. Strand exchange proceeds via two distinct three-strand intermediates, C1 and C2. C1 contains the intact donor dsDNA whereas C2 contains newly formed heteroduplex DNA. Here, we show that the conserved DNA binding motifs, loop 1 (L1) and loop 2 (L2) in site I of Rad51, play distinct roles in this process. L1 is involved in formation of the C1 complex whereas L2 mediates the C1-C2 transition, producing the heteroduplex. Another DNA binding motif, site II, serves as the DNA entry position for initial Rad51 filament formation, as well as for donor dsDNA incorporation. Our study provides a comprehensive molecular model for the catalytic process of strand exchange mediated by eukaryotic RecA-family recombinases.


Assuntos
DNA/metabolismo , Rad51 Recombinase/química , Rad51 Recombinase/metabolismo , Trifosfato de Adenosina/metabolismo , Sítios de Ligação/genética , DNA/genética , Dano ao DNA/genética , Dano ao DNA/fisiologia , Reparo do DNA/genética , Reparo do DNA/fisiologia , DNA de Cadeia Simples/genética , Recombinação Homóloga/genética , Recombinação Homóloga/fisiologia , Humanos , Mutação/genética , Ácidos Nucleicos Heteroduplexes/genética , Ácidos Nucleicos Heteroduplexes/metabolismo , Estrutura Secundária de Proteína , Rad51 Recombinase/genética , Saccharomyces cerevisiae/genética , Schizosaccharomyces/genética
6.
Nucleic Acids Res ; 47(15): 7798-7808, 2019 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-31372639

RESUMO

Homologous recombination is a fundamental process in all living organisms that allows the faithful repair of DNA double strand breaks, through the exchange of DNA strands between homologous regions of the genome. Results of three decades of investigation and recent fruitful observations have unveiled key elements of the reaction mechanism, which proceeds along nucleofilaments of recombinase proteins of the RecA family. Yet, one essential aspect of homologous recombination has largely been overlooked when deciphering the mechanism: while ATP is hydrolyzed in large quantity during the process, how exactly hydrolysis influences the DNA strand exchange reaction at the structural level remains to be elucidated. In this study, we build on a previous geometrical approach that studied the RecA filament variability without bound DNA to examine the putative implication of ATP hydrolysis on the structure, position, and interactions of up to three DNA strands within the RecA nucleofilament. Simulation results on modeled intermediates in the ATP cycle bring important clues about how local distortions in the DNA strand geometries resulting from ATP hydrolysis can aid sequence recognition by promoting local melting of already formed DNA heteroduplex and transient reverse strand exchange in a weaving type of mechanism.


Assuntos
Trifosfato de Adenosina/química , DNA de Cadeia Simples/química , DNA/química , Recombinação Homóloga , Ácidos Nucleicos Heteroduplexes/química , Recombinases Rec A/química , Trifosfato de Adenosina/metabolismo , Bactérias/genética , Bactérias/metabolismo , Sítios de Ligação , DNA/genética , DNA/metabolismo , Quebras de DNA de Cadeia Dupla , DNA de Cadeia Simples/genética , DNA de Cadeia Simples/metabolismo , Hidrólise , Simulação de Dinâmica Molecular , Conformação de Ácido Nucleico , Ácidos Nucleicos Heteroduplexes/genética , Ácidos Nucleicos Heteroduplexes/metabolismo , Ligação Proteica , Conformação Proteica , Recombinases Rec A/genética , Recombinases Rec A/metabolismo
7.
Spectrochim Acta A Mol Biomol Spectrosc ; 223: 117345, 2019 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-31310956

RESUMO

A novel luminescence energy transfer (LET) probe for detection of tumor related microRNAs using NaGdF4:Yb,Er@NaYF4 upconversion nanoparticles (UCNPs) as energy donors and gold nanoparticles (AuNPs) as energy acceptors was developed. Using the double modified complementary DNA sequences of microRNA155 (miRNA155) as a bridge, NaGdF4:Yb,Er@NaYF4 UCNPs and AuNPs were conjugated to form NaGdF4:Yb,Er@NaYF4 UCNPs-DNA-AuNPs nanocomplexes (UCNPs-DNA-AuNPs) probe. The energy transfer would occur when the distance between donor and acceptor gets closer. In the presence of target miRNA155, DNA-RNA heteroduplexes appeared as product, but the luminescence intensity was not changed obviously. In the existence of duplex-specific nuclease (DSN), DSN could hydrolyze the DNA strand of DNA-RNA heteroduplexes, the bridge linked NaGdF4:Yb,Er@NaYF4 UCNPs and AuNPs was destroyed, which induced that the quenched luminescence intensity was recovered and RNA was released. The released miRNA155 could react with another UCNPs-DNA-AuNPs probe to form DNA-RNA heteroduplexes again. This cyclic reaction generates an amplification of luminescence signal for quantitative detection of miRNA155. Under the illumination of 980 nm laser, the concentration ranges from 0.1 nM to 15 nM and the detection of limits was 0.045 nM for detection of miRNA155. Moreover, the UCNPs-DNA-AuNPs probe was used in quantify miRNA155 in cell lysates with satisfactory results.


Assuntos
Endonucleases/metabolismo , Transferência de Energia , Ouro/química , Luminescência , Nanopartículas Metálicas/química , MicroRNAs/metabolismo , Ácidos Nucleicos Heteroduplexes/metabolismo , Transferência Ressonante de Energia de Fluorescência , Humanos , Células K562 , Nanopartículas Metálicas/ultraestrutura , MicroRNAs/genética , Tamanho da Partícula
8.
Mol Cell ; 73(3): 398-411, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30735654

RESUMO

During transcription, the nascent RNA strand can base pair with its template DNA, displacing the non-template strand as ssDNA and forming a structure called an R-loop. R-loops are common across many domains of life and cause DNA damage in certain contexts. In this review, we summarize recent results implicating R-loops as important regulators of cellular processes such as transcription termination, gene regulation, and DNA repair. We also highlight recent work suggesting that R-loops can be problematic to cells as blocks to efficient transcription and replication that trigger the DNA damage response. Finally, we discuss how R-loops may contribute to cancer, neurodegeneration, and inflammatory diseases and compare the available next-generation sequencing-based approaches to map R-loops genome wide.


Assuntos
Núcleo Celular/fisiologia , DNA/genética , Genoma , Instabilidade Genômica , Ácidos Nucleicos Heteroduplexes/genética , RNA/genética , Animais , DNA/química , DNA/metabolismo , Dano ao DNA , Reparo do DNA , Regulação da Expressão Gênica , Humanos , Conformação de Ácido Nucleico , Ácidos Nucleicos Heteroduplexes/química , Ácidos Nucleicos Heteroduplexes/metabolismo , RNA/química , RNA/metabolismo , Relação Estrutura-Atividade , Transcrição Gênica
9.
Mol Cell ; 73(4): 670-683.e12, 2019 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-30639241

RESUMO

Cellular mechanisms that safeguard genome integrity are often subverted in cancer. To identify cancer-related genome caretakers, we employed a convergent multi-screening strategy coupled to quantitative image-based cytometry and ranked candidate genes according to multivariate readouts reflecting viability, proliferative capacity, replisome integrity, and DNA damage signaling. This unveiled regulators of replication stress resilience, including components of the pre-mRNA cleavage and polyadenylation complex. We show that deregulation of pre-mRNA cleavage impairs replication fork speed and leads to excessive origin activity, rendering cells highly dependent on ATR function. While excessive formation of RNA:DNA hybrids under these conditions was tightly associated with replication-stress-induced DNA damage, inhibition of transcription rescued fork speed, origin activation, and alleviated replication catastrophe. Uncoupling of pre-mRNA cleavage from co-transcriptional processing and export also protected cells from replication-stress-associated DNA damage, suggesting that pre-mRNA cleavage provides a mechanism to efficiently release nascent transcripts and thereby prevent gene gating-associated genomic instability.


Assuntos
Dano ao DNA , Replicação do DNA , Instabilidade Genômica , Neoplasias/genética , Clivagem do RNA , Precursores de RNA/genética , RNA Mensageiro/genética , RNA Neoplásico/genética , Transporte Ativo do Núcleo Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Proteínas de Ligação a DNA , Regulação Neoplásica da Expressão Gênica , Células HeLa , Humanos , Neoplasias/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ácidos Nucleicos Heteroduplexes/genética , Ácidos Nucleicos Heteroduplexes/metabolismo , Poliadenilação , Precursores de RNA/biossíntese , RNA Mensageiro/biossíntese , RNA Neoplásico/biossíntese , Proteínas de Ligação a RNA
10.
Biochemistry ; 58(4): 250-258, 2019 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-29870660

RESUMO

RNA helicase DHX33 has been shown to participate in a variety of cellular activities, including ribosome biogenesis, protein translation, and gene transcription. We and others further discovered that DHX33 is strongly expressed in several types of human cancers and plays important roles in promoting cancer cell proliferation. To better understand the molecular mechanism for DHX33 in exerting its biological functions, we purified recombinant DHX33 and performed biochemical studies in vitro. DHX33 protein was found to have ATPase activity that is dependent on DNA or RNA duplexes. The ATPase activity of DHX33 is coupled with its RNA/DNA unwinding activity. If a key residue in the ATP binding site were mutated, the mutant DHX33 could not unwind DNA/RNA duplexes. Furthermore, a deletion mutant of a RKK motif previously identified to be involved in ribosome DNA binding could still unwind DNA duplexes, albeit with reduced efficiency. In summary, our study reveals that purified DHX33 protein possesses unwinding activity toward DNA and RNA duplexes.


Assuntos
RNA Helicases DEAD-box/química , RNA Helicases DEAD-box/metabolismo , Ácidos Nucleicos Heteroduplexes/metabolismo , Trifosfato de Adenosina/metabolismo , Motivos de Aminoácidos , Dicroísmo Circular , RNA Helicases DEAD-box/genética , DNA Helicases/metabolismo , Hidrólise , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo
11.
Biochemistry ; 57(48): 6662-6668, 2018 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-30406989

RESUMO

RNA helicase A (RHA) as a member of the DExH/D-box subgroup of helicase superfamily II is involved in virtually all aspects of RNA metabolism. It exhibits robust RNA helicase activity in vitro. However, little is known about the molecular and physical determinants for RHA substrate recognition and RHA translocation along the nucleic acids. Here, our nondenaturing polyacrylamide gel electrophoresis (PAGE)-based unwinding assays of chemical and structural modified substrates indicate that RHA translocates efficiently along the 3' overhang of RNA, but not DNA, with a requirement of covalent continuity. Ribose-phosphate backbone lesions on both strands of the nucleic acids, especially on the 3' overhang of the loading strand, affect RHA unwinding significantly. Furthermore, RHA requires RNA on the 3' overhang which directly or indirectly connects with the duplex region to mediate productive unwinding. Collectively, these findings propose a basic mechanism of the substrate determinants for RHA backbone tracking during duplex unwinding.


Assuntos
RNA Helicases DEAD-box/metabolismo , Proteínas de Neoplasias/metabolismo , Conformação de Ácido Nucleico , RNA/química , RNA/metabolismo , Sequência de Bases , DNA/química , DNA/genética , DNA/metabolismo , Humanos , Cinética , Modelos Biológicos , Ácidos Nucleicos Heteroduplexes/química , Ácidos Nucleicos Heteroduplexes/genética , Ácidos Nucleicos Heteroduplexes/metabolismo , RNA/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato
12.
Mol Cell ; 72(6): 970-984.e7, 2018 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-30449723

RESUMO

Extensive tracts of the mammalian genome that lack protein-coding function are still transcribed into long noncoding RNA. While these lncRNAs are generally short lived, length restricted, and non-polyadenylated, how their expression is distinguished from protein-coding genes remains enigmatic. Surprisingly, depletion of the ubiquitous Pol-II-associated transcription elongation factor SPT6 promotes a redistribution of H3K36me3 histone marks from active protein coding to lncRNA genes, which correlates with increased lncRNA transcription. SPT6 knockdown also impairs the recruitment of the Integrator complex to chromatin, which results in a transcriptional termination defect for lncRNA genes. This leads to the formation of extended, polyadenylated lncRNAs that are both chromatin restricted and form increased levels of RNA:DNA hybrid (R-loops) that are associated with DNA damage. Additionally, these deregulated lncRNAs overlap with DNA replication origins leading to localized DNA replication stress and a cellular senescence phenotype. Overall, our results underline the importance of restricting lncRNA expression.


Assuntos
Proliferação de Células , Senescência Celular , Dano ao DNA , Replicação do DNA , DNA de Neoplasias/biossíntese , RNA Longo não Codificante/metabolismo , RNA Neoplásico/metabolismo , Fatores de Transcrição/metabolismo , Neoplasias Uterinas/metabolismo , Animais , Montagem e Desmontagem da Cromatina , DNA Polimerase II/genética , DNA Polimerase II/metabolismo , DNA de Neoplasias/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Células HeLa , Histonas/metabolismo , Humanos , Metilação , Conformação de Ácido Nucleico , Ácidos Nucleicos Heteroduplexes/genética , Ácidos Nucleicos Heteroduplexes/metabolismo , Estabilidade de RNA , RNA Longo não Codificante/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , Fatores de Transcrição/genética , Transcrição Gênica , Neoplasias Uterinas/genética
13.
Cell Rep ; 23(6): 1891-1905, 2018 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-29742442

RESUMO

R-loops comprise an RNA/DNA hybrid and displaced single-stranded DNA. They play important biological roles and are implicated in pathology. Even so, proteins recognizing these structures are largely undefined. Using affinity purification with the S9.6 antibody coupled to mass spectrometry, we defined the RNA/DNA hybrid interactome in HeLa cells. This consists of known R-loop-associated factors SRSF1, FACT, and Top1, and yet uncharacterized interactors, including helicases, RNA processing, DNA repair, and chromatin factors. We validate specific examples of these interactors and characterize their involvement in R-loop biology. A top candidate DHX9 helicase promotes R-loop suppression and transcriptional termination. DHX9 interacts with PARP1, and both proteins prevent R-loop-associated DNA damage. DHX9 and other interactome helicases are overexpressed in cancer, linking R-loop-mediated DNA damage and disease. Our RNA/DNA hybrid interactome provides a powerful resource to study R-loop biology in health and disease.


Assuntos
RNA Helicases DEAD-box/metabolismo , Dano ao DNA , DNA/metabolismo , Proteínas de Neoplasias/metabolismo , Conformação de Ácido Nucleico , Ácidos Nucleicos Heteroduplexes/metabolismo , RNA/metabolismo , Terminação da Transcrição Genética , Camptotecina/farmacologia , Células HEK293 , Células HeLa , Humanos , Imunoprecipitação , Reprodutibilidade dos Testes , Terminação da Transcrição Genética/efeitos dos fármacos
14.
Mol Cell ; 70(1): 9-20.e6, 2018 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-29625041

RESUMO

Meiotic recombination is essential for fertility and allelic shuffling. Canonical recombination models fail to capture the observed complexity of meiotic recombinants. Here, by combining genome-wide meiotic heteroduplex DNA patterns with meiotic DNA double-strand break (DSB) sites, we show that part of this complexity results from frequent template switching during synthesis-dependent strand annealing that yields noncrossovers and from branch migration of double Holliday junction (dHJ)-containing intermediates that mainly yield crossovers. This complexity also results from asymmetric positioning of crossover intermediates relative to the initiating DSB and Msh2-independent conversions promoted by the suspected dHJ resolvase Mlh1-3 as well as Exo1 and Sgs1. Finally, we show that dHJ resolution is biased toward cleavage of the pair of strands containing newly synthesized DNA near the junctions and that this bias can be decoupled from the crossover-biased dHJ resolution. These properties are likely conserved in eukaryotes containing ZMM proteins, which includes mammals.


Assuntos
Quebras de DNA de Cadeia Dupla , DNA Cruciforme , DNA Fúngico/genética , Meiose , Ácidos Nucleicos Heteroduplexes/genética , Recombinação Genética , Saccharomyces cerevisiae/genética , DNA Fúngico/metabolismo , Exodesoxirribonucleases/genética , Exodesoxirribonucleases/metabolismo , Proteína 1 Homóloga a MutL/genética , Proteína 1 Homóloga a MutL/metabolismo , Proteínas MutL/genética , Proteínas MutL/metabolismo , Conformação de Ácido Nucleico , Ácidos Nucleicos Heteroduplexes/metabolismo , RecQ Helicases/genética , RecQ Helicases/metabolismo , Saccharomyces cerevisiae/crescimento & desenvolvimento , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
15.
Protoplasma ; 255(2): 669-684, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29103092

RESUMO

RuvB, a member of AAA+ (ATPases Associated with diverse cellular Activities) superfamily of proteins, is essential, highly conserved and multifunctional in nature as it is involved in DNA damage repair, mitotic assembly, switching of histone variants and assembly of telomerase core complex. RuvB family is widely studied in various systems such as Escherichia coli, yeast, human, Drosophila, Plasmodium falciparum and mouse, but not well studied in plants. We have studied the transcript level of rice homologue of RuvB gene (OsRuvBL1a) under various abiotic stress conditions, and the results suggest that it is upregulated under salinity, cold and heat stress. Therefore, the OsRuvBL1a protein was characterized using in silico and biochemical approaches. In silico study confirmed the presence of all the four characteristic motifs of AAA+ superfamily-Walker A, Walker B, Sensor I and Sensor II. Structurally, OsRuvBL1a is similar to RuvB1 from Chaetomium thermophilum. The purified recombinant OsRuvBL1a protein shows unique DNA-independent ATPase activity. Using site-directed mutagenesis, the importance of two conserved motifs (Walker B and Sensor I) in ATPase activity has been also reported with mutants D302N and N332H. The OsRuvBL1a protein unwinds the duplex DNA in the 3' to 5' direction. The presence of unique DNA-independent ATPase and DNA unwinding activities of OsRuvBL1a protein and upregulation of its transcript under abiotic stress conditions suggest its involvement in multiple cellular pathways. The first detailed characterization of plant RuvBL1a in this study may provide important contribution in exploiting the role of RuvB for developing the stress tolerant plants of agricultural importance.


Assuntos
Adenosina Trifosfatases/metabolismo , DNA de Plantas/metabolismo , Ácidos Nucleicos Heteroduplexes/metabolismo , Oryza/enzimologia , Proteínas de Plantas/metabolismo , Estresse Fisiológico , Adenosina Trifosfatases/química , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , DNA Helicases/metabolismo , Regulação da Expressão Gênica de Plantas/efeitos dos fármacos , Cloreto de Magnésio/farmacologia , Modelos Moleculares , Mutagênese Sítio-Dirigida , Proteínas Mutantes/metabolismo , Oryza/efeitos dos fármacos , Oryza/genética , Proteínas de Plantas/química , Cloreto de Potássio/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes/metabolismo , Análise de Sequência de Proteína , Cloreto de Sódio/farmacologia , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/genética , Especificidade por Substrato/efeitos dos fármacos , Fatores de Tempo
16.
Cell Rep ; 21(11): 3166-3177, 2017 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-29241544

RESUMO

Srs2 is a superfamily 1 (SF1) helicase and antirecombinase that is required for genome integrity. However, the mechanisms that regulate Srs2 remain poorly understood. Here, we visualize Srs2 as it acts upon single-stranded DNA (ssDNA) bound by the Rad51 recombinase. We demonstrate that Srs2 is a processive translocase capable of stripping thousands of Rad51 molecules from ssDNA at a rate of ∼50 monomers/s. We show that Srs2 is recruited to RPA clusters embedded between Rad51 filaments and that multimeric arrays of Srs2 assemble during translocation on ssDNA through a mechanism involving iterative Srs2 loading events at sites cleared of Rad51. We also demonstrate that Srs2 acts on heteroduplex DNA joints through two alternative pathways, both of which result in rapid disruption of the heteroduplex intermediate. On the basis of these findings, we present a model describing the recruitment and regulation of Srs2 as it acts upon homologous recombination intermediates.


Assuntos
DNA Helicases/genética , Regulação Fúngica da Expressão Gênica , Recombinação Homóloga , Ácidos Nucleicos Heteroduplexes/genética , Rad51 Recombinase/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , DNA Helicases/metabolismo , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , DNA Fúngico/genética , DNA Fúngico/metabolismo , DNA de Cadeia Simples/genética , DNA de Cadeia Simples/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Mutação , Ácidos Nucleicos Heteroduplexes/metabolismo , Ligação Proteica , Rad51 Recombinase/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteína Vermelha Fluorescente
17.
Nature ; 544(7650): 377-380, 2017 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-28405019

RESUMO

The Rad51 (also known as RecA) family of recombinases executes the critical step in homologous recombination: the search for homologous DNA to serve as a template during the repair of DNA double-strand breaks (DSBs). Although budding yeast Rad51 has been extensively characterized in vitro, the stringency of its search and sensitivity to mismatched sequences in vivo remain poorly defined. Here, in Saccharomyces cerevisiae, we analysed Rad51-dependent break-induced replication in which the invading DSB end and its donor template share a 108-base-pair homology region and the donor carries different densities of single-base-pair mismatches. With every eighth base pair mismatched, repair was about 14% of that of completely homologous sequences. With every sixth base pair mismatched, repair was still more than 5%. Thus, completing break-induced replication in vivo overcomes the apparent requirement for at least 6-8 consecutive paired bases that has been inferred from in vitro studies. When recombination occurs without a protruding nonhomologous 3' tail, the mismatch repair protein Msh2 does not discourage homeologous recombination. However, when the DSB end contains a 3' protruding nonhomologous tail, Msh2 promotes the rejection of mismatched substrates. Mismatch correction of strand invasion heteroduplex DNA is strongly polar, favouring correction close to the DSB end. Nearly all mismatch correction depends on the proofreading activity of DNA polymerase-δ, although the repair proteins Msh2, Mlh1 and Exo1 influence the extent of correction.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo de Erro de Pareamento de DNA , Rad51 Recombinase/metabolismo , Reparo de DNA por Recombinação , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Pareamento de Bases , Sequência de Bases , DNA Polimerase III/metabolismo , Replicação do DNA , DNA Fúngico/genética , DNA Fúngico/metabolismo , Exodesoxirribonucleases/metabolismo , Proteína 1 Homóloga a MutL/metabolismo , Proteína 2 Homóloga a MutS/metabolismo , Ácidos Nucleicos Heteroduplexes/genética , Ácidos Nucleicos Heteroduplexes/metabolismo , Saccharomyces cerevisiae/metabolismo , Homologia de Sequência do Ácido Nucleico , Especificidade por Substrato , Moldes Genéticos
18.
Sci Rep ; 6: 38873, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27974823

RESUMO

DNA replication is essential for cellular proliferation. If improperly controlled it can constitute a major source of genome instability, frequently associated with cancer and aging. POLD1 is the catalytic subunit and POLD3 is an accessory subunit of the replicative Pol δ polymerase, which also functions in DNA repair, as well as the translesion synthesis polymerase Pol ζ, whose catalytic subunit is REV3L. In cells depleted of POLD1 or POLD3 we found a differential but general increase in genome instability as manifested by DNA breaks, S-phase progression impairment and chromosome abnormalities. Importantly, we showed that both proteins are needed to maintain the proper amount of active replication origins and that POLD3-depletion causes anaphase bridges accumulation. In addition, POLD3-associated DNA damage showed to be dependent on RNA-DNA hybrids pointing toward an additional and specific role of this subunit in genome stability. Interestingly, a similar increase in RNA-DNA hybrids-dependent genome instability was observed in REV3L-depleted cells. Our findings demonstrate a key role of POLD1 and POLD3 in genome stability and S-phase progression revealing RNA-DNA hybrids-dependent effects for POLD3 that might be partly due to its Pol ζ interaction.


Assuntos
DNA Polimerase III/metabolismo , Replicação do DNA , Instabilidade Genômica , Fase S , Quebras de DNA , DNA Polimerase III/genética , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Ácidos Nucleicos Heteroduplexes/genética , Ácidos Nucleicos Heteroduplexes/metabolismo
19.
J Biol Chem ; 291(19): 10006-20, 2016 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-26975377

RESUMO

The human primosome, a 340-kilodalton complex of primase and DNA polymerase α (Polα), synthesizes chimeric RNA-DNA primers to be extended by replicative DNA polymerases δ and ϵ. The intricate mechanism of concerted primer synthesis by two catalytic centers was an enigma for over three decades. Here we report the crystal structures of two key complexes, the human primosome and the C-terminal domain of the primase large subunit (p58C) with bound DNA/RNA duplex. These structures, along with analysis of primase/polymerase activities, provide a plausible mechanism for all transactions of the primosome including initiation, elongation, accurate counting of RNA primer length, primer transfer to Polα, and concerted autoregulation of alternate activation/inhibition of the catalytic centers. Our findings reveal a central role of p58C in the coordinated actions of two catalytic domains in the primosome and ultimately could impact the design of anticancer drugs.


Assuntos
DNA Polimerase I/química , DNA Primase/química , DNA/química , Complexos Multienzimáticos/química , Ácidos Nucleicos Heteroduplexes/química , DNA/biossíntese , DNA Polimerase I/metabolismo , DNA Primase/metabolismo , Humanos , Complexos Multienzimáticos/metabolismo , Ácidos Nucleicos Heteroduplexes/metabolismo
20.
Mol Cell ; 60(3): 351-61, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26593718

RESUMO

DNA replication stress can cause chromosomal instability and tumor progression. One key pathway that counteracts replication stress and promotes faithful DNA replication consists of the Fanconi anemia (FA) proteins. However, how these proteins limit replication stress remains largely elusive. Here we show that conflicts between replication and transcription activate the FA pathway. Inhibition of transcription or enzymatic degradation of transcription-associated R-loops (DNA:RNA hybrids) suppresses replication fork arrest and DNA damage occurring in the absence of a functional FA pathway. Furthermore, we show that simple aldehydes, known to cause leukemia in FA-deficient mice, induce DNA:RNA hybrids in FA-depleted cells. Finally, we demonstrate that the molecular mechanism by which the FA pathway limits R-loop accumulation requires FANCM translocase activity. Failure to activate a response to physiologically occurring DNA:RNA hybrids may critically contribute to the heightened cancer predisposition and bone marrow failure of individuals with mutated FA proteins.


Assuntos
Dano ao DNA , DNA Helicases/metabolismo , Replicação do DNA , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Instabilidade Genômica , Ácidos Nucleicos Heteroduplexes/metabolismo , Animais , DNA Helicases/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Células HeLa , Humanos , Leucemia/genética , Leucemia/metabolismo , Leucemia/patologia , Camundongos , Camundongos Knockout , Mutação , Ácidos Nucleicos Heteroduplexes/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA