Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33619096

RESUMEN

The pathogenic consequences of 369 unique human HsMLH1 missense variants has been hampered by the lack of a detailed function in mismatch repair (MMR). Here single-molecule images show that HsMSH2-HsMSH6 provides a platform for HsMLH1-HsPMS2 to form a stable sliding clamp on mismatched DNA. The mechanics of sliding clamp progression solves a significant operational puzzle in MMR and provides explicit predictions for the distribution of clinically relevant HsMLH1 missense mutations.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Reparación de la Incompatibilidad de ADN , Proteínas de Unión al ADN/genética , ADN/genética , Homólogo 1 de la Proteína MutL/genética , Proteína 2 Homóloga a MutS/genética , Mutación Missense , Sitios de Unión , Neoplasias Colorrectales Hereditarias sin Poliposis/metabolismo , Neoplasias Colorrectales Hereditarias sin Poliposis/patología , ADN/química , ADN/metabolismo , Daño del ADN , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Humanos , Modelos Moleculares , Homólogo 1 de la Proteína MutL/química , Homólogo 1 de la Proteína MutL/metabolismo , Proteína 2 Homóloga a MutS/química , Proteína 2 Homóloga a MutS/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas
2.
J Biol Chem ; 298(11): 102505, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36126773

RESUMEN

MutS homologs (MSHs) are highly conserved core components of DNA mismatch repair. Mismatch recognition provokes ATP-binding by MSH proteins that drives a conformational transition from a short-lived lesion-searching clamp to an extremely stable sliding clamp on the DNA. Here, we have expanded on previous bulk biochemical studies to examine the stability, lifetime, and kinetics of bacterial and human MSH sliding clamps on mismatched DNA using surface plasmon resonance and single-molecule analysis of fluorescently labeled proteins. We found that ATP-bound MSH complexes bound to blocked-end or very long mismatched DNAs were extremely stable over a range of ionic conditions. These observations underpinned the development of a high-throughput Förster resonance energy transfer system that specifically detects the formation of MSH sliding clamps on mismatched DNA. The Förster resonance energy transfer system is capable of distinguishing between HsMSH2-HsMSH3 and HsMSH2-HsMSH6 and appears suitable for chemical inhibitor screens. Taken together, our results provide additional insight into MSH sliding clamps as well as methods to distinguish their functions in mismatch repair.


Asunto(s)
Proteínas de Escherichia coli , Proteína MutS de Unión a los Apareamientos Incorrectos del ADN , Humanos , Adenosina Trifosfato/metabolismo , Disparidad de Par Base , ADN/metabolismo , Reparación de la Incompatibilidad de ADN , Proteínas de Escherichia coli/metabolismo , Proteína MutS de Unión a los Apareamientos Incorrectos del ADN/genética , Proteína MutS de Unión a los Apareamientos Incorrectos del ADN/metabolismo , Proteína 2 Homóloga a MutS/genética , Proteína 2 Homóloga a MutS/metabolismo , Proteínas MutS/genética , Unión Proteica
3.
J Biol Chem ; 296: 100550, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33744295

RESUMEN

Retroviral integrases must navigate host DNA packaged as chromatin during integration of the viral genome. Prototype foamy virus (PFV) integrase (IN) forms a tetramer bound to two viral DNA (vDNA) ends in a complex termed an intasome. PFV IN consists of four domains: the amino terminal extension domain (NED), amino terminal domain (NTD), catalytic core domain (CCD), and carboxyl terminal domain (CTD). The domains of the two inner IN protomers have been visualized, as well as the CCDs of the two outer IN protomers. However, the roles of the amino and carboxyl terminal domains of the PFV intasome outer subunits during integration to a nucleosome target substrate are not clear. We used the well-characterized 601 nucleosome to assay integration activity as well as intasome binding. PFV intasome integration to 601 nucleosomes occurs in clusters at four independent sites. We find that the outer protomer NED and NTD domains have no significant effects on integration efficiency, site selection, or binding. The CTDs of the outer PFV intasome subunits dramatically affect nucleosome binding but have little effect on total integration efficiency. The outer PFV IN CTDs did significantly alter the integration efficiency at one site. Histone tails also significantly affect intasome binding, but have little impact on PFV integration efficiency or site selection. These results indicate that binding to nucleosomes does not correlate with integration efficiency and suggests most intasome-binding events are unproductive.


Asunto(s)
Histonas/metabolismo , Integrasas/metabolismo , Nucleosomas/metabolismo , Spumavirus/metabolismo , Proteínas Virales/metabolismo , Integración Viral , Dominio Catalítico , Cromatina/genética , Cromatina/metabolismo , ADN Viral/genética , ADN Viral/metabolismo , Genoma Viral , Humanos , Integrasas/genética , Multimerización de Proteína , Spumavirus/genética , Spumavirus/crecimiento & desarrollo , Proteínas Virales/química , Proteínas Virales/genética
4.
Nature ; 539(7630): 583-587, 2016 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-27851738

RESUMEN

Mismatched nucleotides arise from polymerase misincorporation errors, recombination between heteroallelic parents and chemical or physical DNA damage. Highly conserved MutS (MSH) and MutL (MLH/PMS) homologues initiate mismatch repair and, in higher eukaryotes, act as DNA damage sensors that can trigger apoptosis. Defects in human mismatch repair genes cause Lynch syndrome or hereditary non-polyposis colorectal cancer and 10-40% of related sporadic tumours. However, the collaborative mechanics of MSH and MLH/PMS proteins have not been resolved in any organism. We visualized Escherichia coli (Ec) ensemble mismatch repair and confirmed that EcMutS mismatch recognition results in the formation of stable ATP-bound sliding clamps that randomly diffuse along the DNA with intermittent backbone contact. The EcMutS sliding clamps act as a platform to recruit EcMutL onto the mismatched DNA, forming an EcMutS-EcMutL search complex that then closely follows the DNA backbone. ATP binding by EcMutL establishes a second long-lived DNA clamp that oscillates between the principal EcMutS-EcMutL search complex and unrestricted EcMutS and EcMutL sliding clamps. The EcMutH endonuclease that targets mismatch repair excision only binds clamped EcMutL, increasing its DNA association kinetics by more than 1,000-fold. The assembly of an EcMutS-EcMutL-EcMutH search complex illustrates how sequential stable sliding clamps can modulate one-dimensional diffusion mechanics along the DNA to direct mismatch repair.


Asunto(s)
Reparación de la Incompatibilidad de ADN , ADN/metabolismo , Difusión , Proteínas de Escherichia coli/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas MutL/metabolismo , Proteína MutS de Unión a los Apareamientos Incorrectos del ADN/metabolismo , Adenosina Trifosfato/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Endodesoxirribonucleasas/metabolismo , Endonucleasas/química , Endonucleasas/metabolismo , Escherichia coli/enzimología , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Cinética , Complejos Multiproteicos/química , Proteínas MutL/química , Proteína MutS de Unión a los Apareamientos Incorrectos del ADN/química , Transporte de Proteínas , Imagen Individual de Molécula
5.
J Biol Chem ; 293(24): 9473-9485, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29674319

RESUMEN

Conformations adopted by long stretches of single-stranded DNA (ssDNA) are of central interest in understanding the architecture of replication forks, R loops, and other structures generated during DNA metabolism in vivo This is particularly so if the ssDNA consists of short nucleotide repeats. Such studies have been hampered by the lack of defined substrates greater than ∼150 nt and the absence of high-resolution biophysical approaches. Here we describe the generation of very long ssDNA consisting of the mammalian telomeric repeat (5'-TTAGGG-3') n , as well as the interrogation of its structure by EM and single-molecule magnetic tweezers (smMT). This repeat is of particular interest because it contains a run of three contiguous guanine residues capable of forming G quartets as ssDNA. Fluorescent-dye exclusion assays confirmed that this G-strand ssDNA forms ubiquitous G-quadruplex folds. EM revealed thick bead-like filaments that condensed the DNA ∼12-fold. The bead-like structures were 5 and 8 nm in diameter and linked by thin filaments. The G-strand ssDNA displayed initial stability to smMT force extension that ultimately released in steps that were multiples ∼28 nm at forces between 6 and 12 pN, well below the >20 pN required to unravel G-quadruplexes. Most smMT steps were consistent with the disruption of the beads seen by EM. Binding by RAD51 distinctively altered the force extension properties of the G-strand ssDNA, suggesting a stochastic G-quadruplex-dependent condensation model that is discussed.


Asunto(s)
ADN de Cadena Simple/química , ADN de Cadena Simple/ultraestructura , G-Cuádruplex , Secuencia de Bases , ADN de Cadena Simple/metabolismo , Humanos , Unión Proteica , Recombinasa Rad51/metabolismo , Telómero/química , Telómero/metabolismo , Telómero/ultraestructura
6.
J Biol Chem ; 293(37): 14285-14294, 2018 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-30072380

RESUMEN

Sliding clamps on DNA consist of evolutionarily conserved enzymes that coordinate DNA replication, repair, and the cellular DNA damage response. MutS homolog (MSH) proteins initiate mismatch repair (MMR) by recognizing mispaired nucleotides and in the presence of ATP form stable sliding clamps that randomly diffuse along the DNA. The MSH sliding clamps subsequently load MutL homolog (MLH/PMS) proteins that form a second extremely stable sliding clamp, which together coordinate downstream MMR components with the excision-initiation site that may be hundreds to thousands of nucleotides distant from the mismatch. Specific or nonspecific binding of other proteins to the DNA between the mismatch and the distant excision-initiation site could conceivably obstruct the free diffusion of these MMR sliding clamps, inhibiting their ability to initiate repair. Here, we employed bulk biochemical analysis, single-molecule fluorescence imaging, and mathematical modeling to determine how sliding clamps might overcome such hindrances along the DNA. Using both bacterial and human MSH proteins, we found that increasing the number of MSH sliding clamps on a DNA decreased the association of the Escherichia coli transcriptional repressor LacI to its cognate promoter LacO. Our results suggest a simple mechanism whereby thermal diffusion of MSH sliding clamps along the DNA alters the association kinetics of other DNA-binding proteins over extended distances. These observations appear generally applicable to any stable sliding clamp that forms on DNA.


Asunto(s)
ADN Bacteriano/metabolismo , Proteína MutS de Unión a los Apareamientos Incorrectos del ADN/metabolismo , Thermus/metabolismo , Adenosina Trifosfato/metabolismo , Disparidad de Par Base , Modelos Teóricos , Unión Proteica , Resonancia por Plasmón de Superficie
7.
Nucleic Acids Res ; 45(2): 685-698, 2017 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-27738136

RESUMEN

Wrapping of genomic DNA into nucleosomes poses thermodynamic and kinetic barriers to biological processes such as replication, transcription, repair and recombination. Previous biochemical studies have demonstrated that in the presence of adenosine triphosphate (ATP) the human RAD51 (HsRAD51) recombinase can form a nucleoprotein filament (NPF) on double-stranded DNA (dsDNA) that is capable of unwrapping the nucleosomal DNA from the histone octamer (HO). Here, we have used single molecule Förster Resonance Energy Transfer (smFRET) to examine the real time nucleosome dynamics in the presence of the HsRAD51 NPF. We show that oligomerization of HsRAD51 leads to stepwise, but stochastic unwrapping of the DNA from the HO in the presence of ATP. The highly reversible dynamics observed in single-molecule trajectories suggests an antagonistic mechanism between HsRAD51 binding and rewrapping of the DNA around the HO. These stochastic dynamics were independent of the nucleosomal DNA sequence or the asymmetry created by the presence of a linker DNA. We also observed sliding and rotational oscillations of the HO with respect to the nucleosomal DNA. These studies underline the dynamic nature of even tightly associated protein-DNA complexes such as nucleosomes.


Asunto(s)
Histonas/metabolismo , Nucleosomas/metabolismo , Recombinasa Rad51/metabolismo , Adenosina Trifosfato/metabolismo , ADN/genética , ADN/metabolismo , Replicación del ADN , Histonas/química , Humanos , Hidrólisis , Modelos Biológicos , Nucleoproteínas/metabolismo , Unión Proteica , Multimerización de Proteína
8.
Proc Natl Acad Sci U S A ; 113(12): 3281-6, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-26951673

RESUMEN

Mismatch repair (MMR) is activated by evolutionarily conserved MutS homologs (MSH) and MutL homologs (MLH/PMS). MSH recognizes mismatched nucleotides and form extremely stable sliding clamps that may be bound by MLH/PMS to ultimately authorize strand-specific excision starting at a distant 3'- or 5'-DNA scission. The mechanical processes associated with a complete MMR reaction remain enigmatic. The purified human (Homo sapien or Hs) 5'-MMR excision reaction requires the HsMSH2-HsMSH6 heterodimer, the 5' → 3' exonuclease HsEXOI, and the single-stranded binding heterotrimer HsRPA. The HsMLH1-HsPMS2 heterodimer substantially influences 5'-MMR excision in cell extracts but is not required in the purified system. Using real-time single-molecule imaging, we show that HsRPA or Escherichia coli EcSSB restricts HsEXOI excision activity on nicked or gapped DNA. HsMSH2-HsMSH6 activates HsEXOI by overcoming HsRPA/EcSSB inhibition and exploits multiple dynamic sliding clamps to increase tract length. Conversely, HsMLH1-HsPMS2 regulates tract length by controlling the number of excision complexes, providing a link to 5' MMR.


Asunto(s)
Disparidad de Par Base , Reparación del ADN , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenosina Trifosfatasas/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Dimerización , Humanos , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto , Homólogo 1 de la Proteína MutL , Proteínas Nucleares/metabolismo
9.
Anal Biochem ; 556: 78-84, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29932890

RESUMEN

Single-molecule (SM) microscopy is a powerful tool capable of visualizing individual molecules and events in real time. SM imaging may rely on proteins or nucleic acids labelled with a fluorophore. Unfortunately photobleaching of fluorophores leads to irreversible loss of signal, impacting the collection of data from SM experiments. Trace amounts of dissolved oxygen (O2) are the main cause of photobleaching. Oxygen scavenging systems (OSS) have been developed that decrease dissolved O2. Commercial OSS enzyme preparations are frequently contaminated with nucleases that damage nucleic acid substrates. In this protocol, we purify highly active Pseudomonas putida protocatechuate 3,4-dioxygenase (PCD) without nuclease contaminations. Quantitation of Cy3 photostability revealed that PCD with its substrate protocatechuic acid (PCA) increased the fluorophore half-life 100-fold. This low cost purification method of recombinant PCD yields an enzyme superior to commercially available OSS that is effectively free of nuclease activity.


Asunto(s)
Proteínas Bacterianas , Expresión Génica , Hidroxibenzoatos/química , Imagen Óptica , Protocatecuato-3,4-Dioxigenasa , Pseudomonas putida , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Desoxirribonucleasas , Estabilidad de Enzimas , Oxígeno/química , Protocatecuato-3,4-Dioxigenasa/biosíntesis , Protocatecuato-3,4-Dioxigenasa/química , Protocatecuato-3,4-Dioxigenasa/genética , Protocatecuato-3,4-Dioxigenasa/aislamiento & purificación , Pseudomonas putida/enzimología , Pseudomonas putida/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación
10.
Mol Cell ; 36(6): 1086-94, 2009 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-20064472

RESUMEN

DNA nucleotide mismatches and lesions arise on chromosomes that are a complex assortment of protein and DNA (chromatin). The fundamental unit of chromatin is a nucleosome that contains approximately 146 bp DNA wrapped around an H2A, H2B, H3, and H4 histone octamer. We demonstrate that the mismatch recognition heterodimer hMSH2-hMSH6 disassembles a nucleosome. Disassembly requires a mismatch that provokes the formation of hMSH2-hMSH6 hydrolysis-independent sliding clamps, which translocate along the DNA to the nucleosome. The rate of disassembly is enhanced by actual or mimicked acetylation of histone H3 within the nucleosome entry-exit and dyad axis that occurs during replication and repair in vivo and reduces DNA-octamer affinity in vitro. Our results support a passive mechanism for chromatin remodeling whereby hMSH2-hMSH6 sliding clamps trap localized fluctuations in nucleosome positioning and/or wrapping that ultimately leads to disassembly, and highlight unanticipated strengths of the Molecular Switch Model for mismatch repair (MMR).


Asunto(s)
Ensamble y Desensamble de Cromatina , Proteínas de Unión al ADN/metabolismo , Proteína 2 Homóloga a MutS/metabolismo , Nucleosomas/metabolismo , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Disparidad de Par Base , ADN/metabolismo , Reparación de la Incompatibilidad de ADN , Proteínas de Unión al ADN/genética , Histonas/metabolismo , Humanos , Proteína 2 Homóloga a MutS/genética , Xenopus laevis
11.
Proc Natl Acad Sci U S A ; 111(3): E316-25, 2014 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-24395779

RESUMEN

High fidelity homologous DNA recombination depends on mismatch repair (MMR), which antagonizes recombination between divergent sequences by rejecting heteroduplex DNA containing excessive nucleotide mismatches. The hMSH2-hMSH6 heterodimer is the first responder in postreplicative MMR and also plays a prominent role in heteroduplex rejection. Whether a similar molecular mechanism underlies its function in these two processes remains enigmatic. We have determined that hMSH2-hMSH6 efficiently recognizes mismatches within a D-loop recombination initiation intermediate. Mismatch recognition by hMSH2-hMSH6 is not abrogated by human replication protein A (HsRPA) bound to the displaced single-stranded DNA (ssDNA) or by HsRAD51. In addition, ATP-bound hMSH2-hMSH6 sliding clamps that are essential for downstream MMR processes are formed and constrained within the heteroduplex region of the D-loop. Moreover, the hMSH2-hMSH6 sliding clamps are stabilized on the D-loop by HsRPA bound to the displaced ssDNA. Our findings reveal similarities and differences in hMSH2-hMSH6 mismatch recognition and sliding-clamp formation between a D-loop recombination intermediate and linear duplex DNA.


Asunto(s)
Reparación de la Incompatibilidad de ADN , Proteínas de Unión al ADN/química , ADN/química , Proteína 2 Homóloga a MutS/química , Recombinación Genética , Adenosina Difosfato/química , Adenosina Trifosfato/química , Disparidad de Par Base , Biotinilación , Humanos , Hidrólisis , Cinética , Unión Proteica , Estructura Terciaria de Proteína , Recombinasa Rad51/química , Proteína de Replicación A/química
12.
J Bacteriol ; 198(19): 2673-81, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27185826

RESUMEN

During late stages of cystic fibrosis pulmonary infections, Pseudomonas aeruginosa often overproduces the exopolysaccharide alginate, protecting the bacterial community from host immunity and antimicrobials. The transcription of the alginate biosynthesis operon is under tight control by a number of factors, including AmrZ, the focus of this study. Interestingly, multiple transcription factors interact with the far-upstream region of this promoter (PalgD), in which one AmrZ binding site has been identified previously. The mechanisms of AmrZ binding and subsequent activation remain unclear and require more-detailed investigation. In this study, in-depth examinations elucidated four AmrZ binding sites, and their disruption eliminated AmrZ binding and promoter activation. Furthermore, our in vitro fluorescence resonance energy transfer experiments suggest that AmrZ holds together multiple binding sites in PalgD and thereafter induces the formation of higher-order DNA-AmrZ complexes. To determine the importance of interactions between those AmrZ oligomers in the cell, a DNA phasing experiment was performed. PalgD transcription was significantly impaired when the relative phase between AmrZ binding sites was reversed (5 bp), while a full-DNA-turn insertion (10 bp) restored promoter activity. Taken together, the investigations presented here provide a deeper mechanistic understanding of AmrZ-mediated binding to PalgD IMPORTANCE: Overproduction of the exopolysaccharide alginate provides protection to Pseudomonas aeruginosa against antimicrobial treatments and is associated with chronic P. aeruginosa infections in the lungs of cystic fibrosis patients. In this study, we combined a variety of microbiological, genetic, biochemical, and biophysical approaches to investigate the activation of the alginate biosynthesis operon promoter by a key transcription factor named AmrZ. This study has provided important new information on the mechanism of activation of this extremely complex promoter.


Asunto(s)
Proteínas Bacterianas/metabolismo , ADN Bacteriano/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Regiones Promotoras Genéticas , Pseudomonas aeruginosa/metabolismo , Alginatos , Proteínas Bacterianas/genética , Sitios de Unión , Ácido Glucurónico/biosíntesis , Ácidos Hexurónicos , Mutación , Unión Proteica , Pseudomonas aeruginosa/genética
13.
J Biol Chem ; 290(44): 26395-403, 2015 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-26354434

RESUMEN

Highly conserved MutS homologs (MSH) and MutL homologs (MLH/PMS) are the fundamental components of mismatch repair (MMR). After decades of debate, it appears clear that the MSH proteins initiate MMR by recognizing a mismatch and forming multiple extremely stable ATP-bound sliding clamps that diffuse without hydrolysis along the adjacent DNA. The function(s) of MLH/PMS proteins is less clear, although they too bind ATP and are targeted to MMR by MSH sliding clamps. Structural analysis combined with recent real-time single molecule and cellular imaging technologies are providing new and detailed insight into the thermal-driven motions that animate the complete MMR mechanism.


Asunto(s)
Adenosina Trifosfato/metabolismo , Reparación de la Incompatibilidad de ADN/fisiología , Enzimas Reparadoras del ADN/metabolismo , ADN/metabolismo , Proteínas Nucleares/metabolismo , Adenosina Trifosfato/genética , Animales , ADN/genética , Enzimas Reparadoras del ADN/genética , Humanos , Proteínas Nucleares/genética
14.
Nucleic Acids Res ; 41(15): 7302-12, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23757189

RESUMEN

Double-strand breaks (DSB) occur in chromatin following replication fork collapse and chemical or physical damage [Symington and Gautier (Double-strand break end resection and repair pathway choice. Annu. Rev. Genet. 2011;45:247-271.)] and may be repaired by homologous recombination (HR) and non-homologous end-joining. Nucleosomes are the fundamental units of chromatin and must be remodeled during DSB repair by HR [Andrews and Luger (Nucleosome structure(s) and stability: variations on a theme. Annu. Rev. Biophys. 2011;40:99-117.)]. Physical initiation of HR requires RAD51, which forms a nucleoprotein filament (NPF) that catalyzes homologous pairing and strand exchange (recombinase) between DNAs that ultimately bridges the DSB gap [San Filippo, Sung and Klein. (Mechanism of eukaryotic HR. Annu. Rev. Biochem. 2008;77:229-257.)]. RAD51 forms an NPF on single-stranded DNA and double-stranded DNA (dsDNA). Although the single-stranded DNA NPF is essential for recombinase initiation, the role of the dsDNA NPF is less clear. Here, we demonstrate that the human RAD51 (HsRAD51) dsDNA NPF disassembles nucleosomes by unwrapping the DNA from the core histones. HsRAD51 that has been constitutively or biochemically activated for recombinase functions displays significantly reduced nucleosome disassembly activity. These results suggest that HsRAD51 can perform ATP hydrolysis-dependent nucleosome disassembly in addition to its recombinase functions.


Asunto(s)
Adenosina Trifosfato/metabolismo , Replicación del ADN , Nucleosomas/metabolismo , Recombinasa Rad51/metabolismo , Adenosina Trifosfato/genética , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Cromatina/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , ADN/genética , ADN/metabolismo , Roturas del ADN de Doble Cadena , ADN de Cadena Simple/metabolismo , Genoma Humano , Inestabilidad Genómica , Histonas/genética , Histonas/metabolismo , Humanos , Hidrólisis , Nucleosomas/genética , Recombinasa Rad51/genética , Rec A Recombinasas/genética , Rec A Recombinasas/metabolismo , Reparación del ADN por Recombinación , Origen de Réplica
15.
Nucleic Acids Res ; 40(20): 10215-27, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22965129

RESUMEN

Eukaryotic genomes are repetitively wrapped into nucleosomes that then regulate access of transcription and DNA repair complexes to DNA. The mechanisms that regulate extrinsic protein interactions within nucleosomes are unresolved. We demonstrate that modulation of the nucleosome unwrapping rate regulates protein binding within nucleosomes. Histone H3 acetyl-lysine 56 [H3(K56ac)] and DNA sequence within the nucleosome entry-exit region additively influence nucleosomal DNA accessibility by increasing the unwrapping rate without impacting rewrapping. These combined epigenetic and genetic factors influence transcription factor (TF) occupancy within the nucleosome by at least one order of magnitude and enhance nucleosome disassembly by the DNA mismatch repair complex, hMSH2-hMSH6. Our results combined with the observation that ∼30% of Saccharomyces cerevisiae TF-binding sites reside in the nucleosome entry-exit region suggest that modulation of nucleosome unwrapping is a mechanism for regulating transcription and DNA repair.


Asunto(s)
Ensamble y Desensamble de Cromatina , ADN/química , Nucleosomas/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Histonas , Proteína 2 Homóloga a MutS/metabolismo , Nucleosomas/química , Saccharomyces cerevisiae/genética , Factores de Transcripción/metabolismo , Xenopus laevis
17.
Proc Natl Acad Sci U S A ; 108(31): 12711-6, 2011 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-21768347

RESUMEN

Nucleosomes are stable DNA-histone protein complexes that must be unwrapped and disassembled for genome expression, replication, and repair. Histone posttranslational modifications (PTMs) are major regulatory factors of these nucleosome structural changes, but the molecular mechanisms associated with PTM function remains poorly understood. Here we demonstrate that histone PTMs within distinct structured regions of the nucleosome directly regulate the inherent dynamic properties of the nucleosome. Precise PTMs were introduced into nucleosomes by chemical ligation. Single molecule magnetic tweezers measurements determined that only PTMs near the nucleosome dyad increase the rate of histone release in unwrapped nucleosomes. In contrast, FRET and restriction enzyme analysis reveal that only PTMs throughout the DNA entry-exit region increase unwrapping and enhance transcription factor binding to nucleosomal DNA. These results demonstrate that PTMs in separate structural regions of the nucleosome control distinct dynamic events, where the dyad regulates disassembly while the DNA entry-exit region regulates unwrapping. These studies are consistent with the conclusion that histone PTMs may independently influence nucleosome dynamics and associated chromatin functions.


Asunto(s)
ADN/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Nucleosomas/metabolismo , Acetilación , Algoritmos , Animales , ADN/química , ADN/genética , Ensayo de Cambio de Movilidad Electroforética , Transferencia Resonante de Energía de Fluorescencia , Histonas/química , Histonas/genética , Cinética , Lisina/química , Lisina/genética , Microscopía de Fuerza Atómica , Modelos Moleculares , Mutación , Conformación de Ácido Nucleico , Nucleosomas/genética , Unión Proteica , Multimerización de Proteína , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
18.
J Biol Chem ; 287(12): 8724-36, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22275364

RESUMEN

RAD51 mediates homologous recombination by forming an active DNA nucleoprotein filament (NPF). A conserved aspartate that forms a salt bridge with the ATP γ-phosphate is found at the nucleotide-binding interface between RAD51 subunits of the NPF known as the ATP cap. The salt bridge accounts for the nonphysiological cation(s) required to fully activate the RAD51 NPF. In contrast, RecA homologs and most RAD51 paralogs contain a conserved lysine at the analogous structural position. We demonstrate that substitution of human RAD51(Asp-316) with lysine (HsRAD51(D316K)) decreases NPF turnover and facilitates considerably improved recombinase functions. Structural analysis shows that archaebacterial Methanococcus voltae RadA(D302K) (MvRAD51(D302K)) and HsRAD51(D316K) form extended active NPFs without salt. These studies suggest that the HsRAD51(Asp-316) salt bridge may function as a conformational sensor that enhances turnover at the expense of recombinase activity.


Asunto(s)
Adenosina Trifosfato/metabolismo , Proteínas Arqueales/química , Proteínas Arqueales/metabolismo , Methanococcus/enzimología , Nucleoproteínas/química , Recombinasa Rad51/química , Recombinasa Rad51/metabolismo , Secuencia de Aminoácidos , Proteínas Arqueales/genética , Cristalografía por Rayos X , Humanos , Methanococcus/química , Methanococcus/genética , Datos de Secuencia Molecular , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , Estabilidad Proteica , Recombinasa Rad51/genética , Alineación de Secuencia
20.
Nucleic Acids Res ; 39(19): 8306-13, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21764779

RESUMEN

The expression, replication and repair of eukaryotic genomes require the fundamental organizing unit of chromatin, the nucleosome, to be unwrapped and disassembled. We have developed a quantitative model of nucleosome dynamics which provides a fundamental understanding of these DNA processes. We calibrated this model using results from high precision single molecule nucleosome unzipping experiments, and then tested its predictions for experiments in which nucleosomes are disassembled by the DNA mismatch recognition complex hMSH2-hMSH6. We found that this calibrated model quantitatively describes hMSH2-hMSH6 induced disassembly rates of nucleosomes with two separate DNA sequences and four distinct histone modification states. In addition, this model provides mechanistic insight into nucleosome disassembly by hMSH2-hMSH6 and the influence of histone modifications on this disassembly reaction. This model's precise agreement with current experiments suggests that it can be applied more generally to provide important mechanistic understanding of the numerous nucleosome alterations that occur during DNA processing.


Asunto(s)
Ensamble y Desensamble de Cromatina , Modelos Genéticos , Nucleosomas/química , Proteínas de Unión al ADN/metabolismo , Histonas/metabolismo , Proteína 2 Homóloga a MutS/metabolismo , Nucleosomas/metabolismo , Procesamiento Proteico-Postraduccional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA