Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 16(5): e0251261, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33970942

RESUMO

Chl1 is a member of the XPD family of 5'-3' DNA helicases, which perform a variety of roles in genome maintenance and transmission. They possess a variety of unique structural features, including the presence of a highly variable, partially-ordered insertion in the helicase domain 1. Chl1 has been shown to be required for chromosome segregation in yeast due to its role in the formation of persistent chromosome cohesion during S-phase. Here we present structural and biochemical data to show that Chl1 has the same overall domain organisation as other members of the XPD family, but with some conformational alterations. We also present data suggesting the insert domain in Chl1 regulates its DNA binding.


Assuntos
Chaetomium/enzimologia , DNA Helicases/química , Proteína Grupo D do Xeroderma Pigmentoso/química , Chaetomium/química , Chaetomium/genética , Cristalografia por Raios X , DNA Helicases/genética , DNA Helicases/metabolismo , Conformação Proteica , Fase S/fisiologia , Troca de Cromátide Irmã , Proteína Grupo D do Xeroderma Pigmentoso/genética , Proteína Grupo D do Xeroderma Pigmentoso/metabolismo
2.
Nucleic Acids Res ; 49(9): e53, 2021 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-33660771

RESUMO

Molecular machines within cells dynamically assemble, disassemble and reorganize. Molecular interactions between their components can be observed at the single-molecule level and quantified using colocalization single-molecule spectroscopy, in which individual labeled molecules are seen transiently associating with a surface-tethered partner, or other total internal reflection fluorescence microscopy approaches in which the interactions elicit changes in fluorescence in the labeled surface-tethered partner. When multiple interacting partners can form ternary, quaternary and higher order complexes, the types of spatial and temporal organization of these complexes can be deduced from the order of appearance and reorganization of the components. Time evolution of complex architectures can be followed by changes in the fluorescence behavior in multiple channels. Here, we describe the kinetic event resolving algorithm (KERA), a software tool for organizing and sorting the discretized fluorescent trajectories from a range of single-molecule experiments. KERA organizes the data in groups by transition patterns, and displays exhaustive dwell time data for each interaction sequence. Enumerating and quantifying sequences of molecular interactions provides important information regarding the underlying mechanism of the assembly, dynamics and architecture of the macromolecular complexes. We demonstrate KERA's utility by analyzing conformational dynamics of two DNA binding proteins: replication protein A and xeroderma pigmentosum complementation group D helicase.


Assuntos
Software , Algoritmos , DNA/química , Fluorescência , Cinética , Conformação Proteica , Domínios Proteicos , Proteína de Replicação A/química , Proteína Grupo D do Xeroderma Pigmentoso/química
3.
Mol Biol Rep ; 47(7): 5081-5090, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32519309

RESUMO

Identification of modifier genes predisposing to breast cancer (BC) phenotype remains a significant challenge and varies with ethnicity. The genetic variability observed in DNA repair genes may modulate the cell's ability to repair the damaged DNA and hence, evaluation of genetic variants in crucial DNA damage repair genes is of clinical importance. We performed the present study to evaluate the role of ERCC2-Lys751Gln, hOGG1-Ser326Cys, and XRCC1-Arg399Gln gene polymorphisms on the risk of BC development and its molecular profile in Indian women. Three non-synonymous variants (rs13181, rs1052133, and rs25487) were genotyped in 464 BC patients and 450 healthy controls. Logistic regression was employed to evaluate the association of genotypes with BC risk. Also, in silico analysis was carried out to map the Arg399Gln variant on the BRCT1 domain of XRCC1 protein. XRCC1 Gln/Gln genotype frequency was significantly elevated in BC patients [odd ratio (OR) = 1.73; 95% confidence interval (CI) = 1.13-2.65]. No significant association was observed between hOGG1-Ser326Cys and ERCC2-Lys751Gln variants and BC risk. Subgroup analysis revealed that ERCC2-Lys751Gln and XRCC1-Arg399Gln variants contributed towards tumor progression. A positive interaction between the investigated SNPs and BC was revealed by MDR analysis. Arg399Gln variant resulted in a change in the surface charge of XRCC1 protein. The rs25487 variant of XRCC1 might be associated with an elevated risk of BC. Furthermore, we demonstrated that high order gene-gene interaction plays a significant role in BC etiology. Hence, understanding the impact of low penetrant gene polymorphisms might enable a better understanding of the genetic background of breast cancer.


Assuntos
Neoplasias da Mama/genética , DNA Glicosilases/genética , Polimorfismo de Nucleotídeo Único , Proteína 1 Complementadora Cruzada de Reparo de Raio-X/genética , Proteína Grupo D do Xeroderma Pigmentoso/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , DNA Glicosilases/química , Feminino , Humanos , Índia , Pessoa de Meia-Idade , Simulação de Dinâmica Molecular , Mutação de Sentido Incorreto , Domínios Proteicos , Proteína 1 Complementadora Cruzada de Reparo de Raio-X/química , Proteína Grupo D do Xeroderma Pigmentoso/química
4.
Elife ; 82019 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-30860024

RESUMO

Transcription factor IIH (TFIIH) is a heterodecameric protein complex critical for transcription initiation by RNA polymerase II and nucleotide excision DNA repair. The TFIIH core complex is sufficient for its repair functions and harbors the XPB and XPD DNA-dependent ATPase/helicase subunits, which are affected by human disease mutations. Transcription initiation additionally requires the CdK activating kinase subcomplex. Previous structural work has provided only partial insight into the architecture of TFIIH and its interactions within transcription pre-initiation complexes. Here, we present the complete structure of the human TFIIH core complex, determined by phase-plate cryo-electron microscopy at 3.7 Å resolution. The structure uncovers the molecular basis of TFIIH assembly, revealing how the recruitment of XPB by p52 depends on a pseudo-symmetric dimer of homologous domains in these two proteins. The structure also suggests a function for p62 in the regulation of XPD, and allows the mapping of previously unresolved human disease mutations.


Assuntos
DNA Helicases/química , Proteínas de Ligação a DNA/química , Subunidade p52 de NF-kappa B/química , Fator de Transcrição TFIIH/química , Fator de Transcrição TFIIH/fisiologia , Proteínas de Ciclo Celular/química , Microscopia Crioeletrônica , Dano ao DNA , DNA Helicases/metabolismo , Reparo do DNA , Células HeLa , Humanos , Mutação , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Proteínas de Ligação a RNA/química , Fator de Transcrição TFIIH/genética , Fatores de Transcrição/química , Transcrição Gênica , Proteína Grupo D do Xeroderma Pigmentoso/química
5.
Nucleic Acids Res ; 46(D1): D465-D470, 2018 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-29140456

RESUMO

Minimotif Miner (MnM) is a database and web system for analyzing short functional peptide motifs, termed minimotifs. We present an update to MnM growing the database from ∼300 000 to >1 000 000 minimotif consensus sequences and instances. This growth comes largely from updating data from existing databases and annotation of articles with high-throughput approaches analyzing different types of post-translational modifications. Another update is mapping human proteins and their minimotifs to know human variants from the dbSNP, build 150. Now MnM 4 can be used to generate mechanistic hypotheses about how human genetic variation affect minimotifs and outcomes. One example of the utility of the combined minimotif/SNP tool identifies a loss of function missense SNP in a ubiquitylation minimotif encoded in the excision repair cross-complementing 2 (ERCC2) nucleotide excision repair gene. This SNP reaches genome wide significance for many types of cancer and the variant identified with MnM 4 reveals a more detailed mechanistic hypothesis concerning the role of ERCC2 in cancer. Other updates to the web system include a new architecture with migration of the web system and database to Docker containers for better performance and management. Weblinks:minimotifminer.org and mnm.engr.uconn.edu.


Assuntos
Bases de Dados de Proteínas , Peptídeos/química , Processamento de Proteína Pós-Traducional , Receptores Acoplados a Proteínas G/química , Software , Proteína Grupo D do Xeroderma Pigmentoso/química , Sequência de Aminoácidos , Sítios de Ligação , Sequência Consenso , Ontologia Genética , Genoma Humano , Humanos , Internet , Modelos Moleculares , Anotação de Sequência Molecular , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Peptídeos/genética , Peptídeos/metabolismo , Polimorfismo de Nucleotídeo Único , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Alinhamento de Sequência , Proteína Grupo D do Xeroderma Pigmentoso/genética , Proteína Grupo D do Xeroderma Pigmentoso/metabolismo
6.
J Vis Exp ; (123)2017 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-28570512

RESUMO

AFM imaging is a powerful technique for the study of protein-DNA interactions. This single molecule method allows the simultaneous resolution of different molecules and molecular assemblies in a heterogeneous sample. In the particular context of DNA interacting protein systems, different protein complex forms and their corresponding binding positions on target sites containing DNA fragments can thus be distinguished. Here, an application of AFM to the study of DNA lesion recognition in the prokaryotic and eukaryotic nucleotide excision DNA repair (NER) systems is presented. The procedures of DNA and protein sample preparations are described and experimental as well as analytical details of the experiments are provided. The data allow important conclusions on the strategies by which target site verification may be achieved by the NER proteins. Interestingly, they indicate different approaches of lesion recognition and identification for the eukaryotic NER system, depending on the type of lesion. Furthermore, distinct structural properties of the two different helicases involved in prokaryotic and eukaryotic NER result in and explain the different strategies observed for these two systems. Importantly, these experimental and analytical approaches can be applied not only to the study of DNA repair but also very similarly to other DNA interacting protein systems such as those involved in replication or transcription processes.


Assuntos
Dano ao DNA , Reparo do DNA , Microscopia de Força Atômica/métodos , DNA/química , Humanos , Conformação de Ácido Nucleico , Ligação Proteica , Proteína Grupo D do Xeroderma Pigmentoso/química
7.
PLoS Genet ; 12(8): e1006248, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27504877

RESUMO

The increasing application of gene panels for familial cancer susceptibility disorders will probably lead to an increased proposal of susceptibility gene candidates. Using ERCC2 DNA repair gene as an example, we show that proof of a possible role in cancer susceptibility requires a detailed dissection and characterization of the underlying mutations for genes with diverse cellular functions (in this case mainly DNA repair and basic cellular transcription). In case of ERCC2, panel sequencing of 1345 index cases from 587 German, 405 Lithuanian and 353 Czech families with breast and ovarian cancer (BC/OC) predisposition revealed 25 mutations (3 frameshift, 2 splice-affecting, 20 missense), all absent or very rare in the ExAC database. While 16 mutations were unique, 9 mutations showed up repeatedly with population-specific appearance. Ten out of eleven mutations that were tested exemplarily in cell-based functional assays exert diminished excision repair efficiency and/or decreased transcriptional activation capability. In order to provide evidence for BC/OC predisposition, we performed familial segregation analyses and screened ethnically matching controls. However, unlike the recently published RECQL example, none of our recurrent ERCC2 mutations showed convincing co-segregation with BC/OC or significant overrepresentation in the BC/OC cohort. Interestingly, we detected that some deleterious founder mutations had an unexpectedly high frequency of > 1% in the corresponding populations, suggesting that either homozygous carriers are not clinically recognized or homozygosity for these mutations is embryonically lethal. In conclusion, we provide a useful resource on the mutational landscape of ERCC2 mutations in hereditary BC/OC patients and, as our key finding, we demonstrate the complexity of correct interpretation for the discovery of "bonafide" breast cancer susceptibility genes.


Assuntos
Neoplasias da Mama/genética , Predisposição Genética para Doença , Neoplasias Ovarianas/genética , Proteína Grupo D do Xeroderma Pigmentoso/genética , Neoplasias da Mama/patologia , Reparo do DNA/genética , Feminino , Mutação em Linhagem Germinativa , Heterozigoto , Humanos , Mutação de Sentido Incorreto , Neoplasias Ovarianas/patologia , Proteína Grupo D do Xeroderma Pigmentoso/química
8.
DNA Repair (Amst) ; 44: 136-142, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27262611

RESUMO

XPD, as part of the TFIIH complex, has classically been linked to the damage verification step of nucleotide excision repair (NER). However, recent data indicate that XPD, due to its iron-sulfur center interacts with the iron sulfur cluster assembly proteins, and may interact with other proteins in the cell to mediate a diverse set of biological functions including cell cycle regulation, mitosis, and mitochondrial function. In this perspective, after first reviewing the function and some of the key disease causing variants that affect XPD's interaction with TFIIH and the CDK-activating kinase complex (CAK), we investigate these intriguing cellular roles of XPD and highlight important unanswered questions that provide a fertile ground for further scientific exploration.


Assuntos
Reparo do DNA , DNA/metabolismo , Fator de Transcrição TFIIH/metabolismo , Proteína Grupo D do Xeroderma Pigmentoso/metabolismo , Xeroderma Pigmentoso/genética , Ciclo Celular , Núcleo Celular/metabolismo , Quinases Ciclina-Dependentes/genética , Quinases Ciclina-Dependentes/metabolismo , DNA/química , Dano ao DNA , Humanos , Proteínas Ferro-Enxofre/genética , Proteínas Ferro-Enxofre/metabolismo , Mitocôndrias/metabolismo , Modelos Moleculares , Mutação , Ligação Proteica , Estrutura Secundária de Proteína , Fator de Transcrição TFIIH/química , Fator de Transcrição TFIIH/genética , Xeroderma Pigmentoso/metabolismo , Xeroderma Pigmentoso/patologia , Proteína Grupo D do Xeroderma Pigmentoso/química , Proteína Grupo D do Xeroderma Pigmentoso/genética , Quinase Ativadora de Quinase Dependente de Ciclina
9.
Nucleic Acids Res ; 44(6): 2806-15, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-26896802

RESUMO

The xeroderma pigmentosum group D (XPD) helicase is a component of the transcription factor IIH complex in eukaryotes and plays an essential role in DNA repair in the nucleotide excision repair pathway. XPD is a 5' to 3' helicase with an essential iron-sulfur cluster. Structural and biochemical studies of the monomeric archaeal XPD homologues have aided a mechanistic understanding of this important class of helicase, but several important questions remain open. In particular, the mechanism for DNA loading, which is assumed to require large protein conformational change, is not fully understood. Here, DNA binding by the archaeal XPD helicase from Thermoplasma acidophilum has been investigated using a combination of crystallography, cross-linking, modified substrates and biochemical assays. The data are consistent with an initial tight binding of ssDNA to helicase domain 2, followed by transient opening of the interface between the Arch and 4FeS domains, allowing access to a second binding site on helicase domain 1 that directs DNA through the pore. A crystal structure of XPD from Sulfolobus acidocaldiarius that lacks helicase domain 2 has an otherwise unperturbed structure, emphasizing the stability of the interface between the Arch and 4FeS domains in XPD.


Assuntos
Proteínas Arqueais/química , Reparo do DNA , DNA Arqueal/química , DNA de Cadeia Simples/química , Thermoplasma/química , Proteína Grupo D do Xeroderma Pigmentoso/química , Motivos de Aminoácidos , Proteínas Arqueais/antagonistas & inibidores , Proteínas Arqueais/genética , Proteínas Arqueais/metabolismo , Sítios de Ligação , Clonagem Molecular , Cristalografia por Raios X , Dano ao DNA , DNA Arqueal/genética , DNA Arqueal/metabolismo , DNA de Cadeia Simples/genética , DNA de Cadeia Simples/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Estabilidade Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sulfolobus/química , Sulfolobus/enzimologia , Thermoplasma/enzimologia , Proteína Grupo D do Xeroderma Pigmentoso/genética , Proteína Grupo D do Xeroderma Pigmentoso/metabolismo
10.
Nucleic Acids Res ; 44(2): 954-68, 2016 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-26657627

RESUMO

The complex molecular motions central to the functions of helicases have long attracted attention. Protein crystallography has provided transformative insights into these dynamic conformational changes, however important questions about the true nature of helicase configurations during the catalytic cycle remain. Using pulsed EPR (PELDOR or DEER) to measure interdomain distances in solution, we have examined two representative helicases: PcrA from superfamily 1 and XPD from superfamily 2. The data show that PcrA is a dynamic structure with domain movements that correlate with particular functional states, confirming and extending the information gleaned from crystal structures and other techniques. XPD in contrast is shown to be a rigid protein with almost no conformational changes resulting from nucleotide or DNA binding, which is well described by static crystal structures. Our results highlight the complimentary nature of PELDOR to crystallography and the power of its precision in understanding the conformational changes relevant to helicase function.


Assuntos
Proteínas de Bactérias/química , DNA Helicases/química , Espectroscopia de Ressonância de Spin Eletrônica/métodos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , DNA/química , DNA/metabolismo , DNA Helicases/genética , DNA Helicases/metabolismo , Geobacillus stearothermophilus/enzimologia , Modelos Moleculares , Mutação , Nucleotídeos/metabolismo , Estrutura Terciária de Proteína , Marcadores de Spin , Proteína Grupo D do Xeroderma Pigmentoso/química , Proteína Grupo D do Xeroderma Pigmentoso/genética , Proteína Grupo D do Xeroderma Pigmentoso/metabolismo
11.
Mol Cell ; 59(5): 794-806, 2015 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-26340423

RESUMO

TFIIH is essential for both RNA polymerase II transcription and DNA repair, and mutations in TFIIH can result in human disease. Here, we determine the molecular architecture of human and yeast TFIIH by an integrative approach using chemical crosslinking/mass spectrometry (CXMS) data, biochemical analyses, and previously published electron microscopy maps. We identified four new conserved "topological regions" that function as hubs for TFIIH assembly and more than 35 conserved topological features within TFIIH, illuminating a network of interactions involved in TFIIH assembly and regulation of its activities. We show that one of these conserved regions, the p62/Tfb1 Anchor region, directly interacts with the DNA helicase subunit XPD/Rad3 in native TFIIH and is required for the integrity and function of TFIIH. We also reveal the structural basis for defects in patients with xeroderma pigmentosum and trichothiodystrophy, with mutations found at the interface between the p62 Anchor region and the XPD subunit.


Assuntos
Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Fator de Transcrição TFIIH/química , Fator de Transcrição TFIIH/metabolismo , Reagentes de Ligações Cruzadas , DNA Helicases/química , DNA Helicases/genética , DNA Helicases/metabolismo , Reparo do DNA , Humanos , Espectrometria de Massas , Modelos Moleculares , Mutação , Domínios e Motivos de Interação entre Proteínas , Subunidades Proteicas , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Fator de Transcrição TFIIH/genética , Fatores de Transcrição TFII/química , Fatores de Transcrição TFII/genética , Fatores de Transcrição TFII/metabolismo , Transcrição Gênica , Xeroderma Pigmentoso/genética , Xeroderma Pigmentoso/metabolismo , Proteína Grupo D do Xeroderma Pigmentoso/química , Proteína Grupo D do Xeroderma Pigmentoso/genética , Proteína Grupo D do Xeroderma Pigmentoso/metabolismo
12.
Protoplasma ; 252(6): 1487-504, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25708921

RESUMO

XPD helicase, a TFIIH subunit, is essential for several processes including transcription, NER, cell cycle regulation, and apoptosis in eukaryotes. Another component of TFIIH, namely p44, is among the well-known interacting partners of XPD and is vital in regulating the helicase activities of latter. However, none of the above mentioned proteins have been functionally characterized in Plasmodium falciparum. Consequently, in this study, we performed detailed studies on XPD and its interacting partner, p44, from P. falciparum 3D7 strain. Accordingly, we expressed and purified recombinant PfXPD and its fragments and Pfp44 proteins and characterized the enzymatic activities of PfXPD and its fragments. The in vivo stage-specific expression and subcellular localizations of PfXPD and Pfp44 proteins were studied using the specific antibodies in the intraerythrocytic developmental stages of P. falciparum 3D7 strain. Our results suggest that PfXPD displays the characteristic ssDNA-dependent ATPase and 5'-3' DNA helicase activities. We also report the existence of two high molecular weight forms of p44 in P. falciparum 3D7 strain. Both PfXPD and Pfp44 colocalize in the nucleus and interact with each other, which suggest that they are most likely components of the same complex apparently, TFIIH. Furthermore, during trophozoite and schizont stages, both proteins exhibit a distinct cytoplasmic distribution pattern which implies that PfXPD and Pfp44 might also be involved in other functions. These studies will aid in understanding the basic biology of malaria parasite.


Assuntos
DNA/metabolismo , Plasmodium falciparum/enzimologia , Proteínas de Protozoários/metabolismo , Fator de Transcrição TFIIH/metabolismo , Proteína Grupo D do Xeroderma Pigmentoso/metabolismo , Transporte Ativo do Núcleo Celular , Trifosfato de Adenosina/metabolismo , Núcleo Celular/enzimologia , Clonagem Molecular , Citoplasma/enzimologia , DNA/química , DNA/genética , Regulação da Expressão Gênica no Desenvolvimento , Hidrólise , Cinética , Estágios do Ciclo de Vida , Peso Molecular , Conformação de Ácido Nucleico , Plasmodium falciparum/genética , Plasmodium falciparum/crescimento & desenvolvimento , Ligação Proteica , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Proteínas Recombinantes/metabolismo , Análise de Sequência de DNA , Fator de Transcrição TFIIH/química , Fator de Transcrição TFIIH/genética , Transcrição Gênica , Proteína Grupo D do Xeroderma Pigmentoso/química , Proteína Grupo D do Xeroderma Pigmentoso/genética
13.
Cancer Discov ; 4(10): 1140-53, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25096233

RESUMO

UNLABELLED: Cisplatin-based chemotherapy is the standard of care for patients with muscle-invasive urothelial carcinoma. Pathologic downstaging to pT0/pTis after neoadjuvant cisplatin-based chemotherapy is associated with improved survival, although molecular determinants of cisplatin response are incompletely understood. We performed whole-exome sequencing on pretreatment tumor and germline DNA from 50 patients with muscle-invasive urothelial carcinoma who received neoadjuvant cisplatin-based chemotherapy followed by cystectomy (25 pT0/pTis "responders," 25 pT2+ "nonresponders") to identify somatic mutations that occurred preferentially in responders. ERCC2, a nucleotide excision repair gene, was the only significantly mutated gene enriched in the cisplatin responders compared with nonresponders (q < 0.01). Expression of representative ERCC2 mutants in an ERCC2-deficient cell line failed to rescue cisplatin and UV sensitivity compared with wild-type ERCC2. The lack of normal ERCC2 function may contribute to cisplatin sensitivity in urothelial cancer, and somatic ERCC2 mutation status may inform cisplatin-containing regimen usage in muscle-invasive urothelial carcinoma. SIGNIFICANCE: Somatic ERCC2 mutations correlate with complete response to cisplatin-based chemosensitivity in muscle-invasive urothelial carcinoma, and clinically identified mutations lead to cisplatin sensitivity in vitro. Nucleotide excision repair pathway defects may drive exceptional response to conventional chemotherapy.


Assuntos
Cisplatino/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Mutação , Neoplasias Urológicas/tratamento farmacológico , Neoplasias Urológicas/genética , Urotélio/patologia , Proteína Grupo D do Xeroderma Pigmentoso/genética , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Cisplatino/administração & dosagem , Sequência Conservada , Reparo do DNA , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Terapia Neoadjuvante , Invasividade Neoplásica , Metástase Neoplásica , Estadiamento de Neoplasias , Conformação Proteica , Fatores de Risco , Resultado do Tratamento , Neoplasias Urológicas/patologia , Proteína Grupo D do Xeroderma Pigmentoso/química
14.
Artigo em Inglês | MEDLINE | ID: mdl-24731924

RESUMO

Transcriptional factor IIH (TFIIH) is involved in cell cycle regulation, nucleotide excision repair, and gene transcription. Mutations in three of its subunits, XPB, XPD, and TTDA, lead to human recessive genetic disorders such as trichothiodystrophy and xeroderma pigmentosum, the latter of which is sometimes associated with Cockayne's syndrome. In the present study, we investigate the sequence conservation of TFIIH subunits among several teleost fish species and compare their characteristics and putative regulation by transcription factors to those of human and zebrafish. We report the following findings: (i) comparisons among protein sequences revealed a high sequence identity for each TFIIH subunit analysed; (ii) among transcription factors identified as putative regulators, OCT1 and AP1 have the highest binding-site frequencies in the promoters of TFIIH genes, and (iii) TFIIH genes have alternatively spliced isoforms. Finally, we compared the protein primary structure in human and zebrafish of XPD and XPB - two important ATP-dependent helicases that catalyse the unwinding of the DNA duplex at promoters during transcription - highlighting the conservation of domain regions such as the helicase domains. Our study suggests that zebrafish, a widely used model for many human diseases, could also act as an important model to study the function of TFIIH complex in repair and transcription regulation in humans.


Assuntos
Evolução Molecular , Fator de Transcrição TFIIH/fisiologia , Processamento Alternativo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Mapeamento Cromossômico , Sequência Conservada , Peixes/genética , Humanos , Dados de Sequência Molecular , Fator 1 de Transcrição de Octâmero/genética , Fator 1 de Transcrição de Octâmero/metabolismo , Regiões Promotoras Genéticas , Conformação Proteica , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Fator de Transcrição TFIIH/química , Proteína Grupo D do Xeroderma Pigmentoso/química , Proteína Grupo D do Xeroderma Pigmentoso/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
15.
DNA Repair (Amst) ; 20: 58-70, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24560558

RESUMO

XPD-like helicases constitute a prominent DNA helicase family critical for many aspects of genome maintenance. These enzymes share a unique structural feature, an auxiliary domain stabilized by an iron-sulphur (FeS) cluster, and a 5'-3' polarity of DNA translocation and duplex unwinding. Biochemical analyses alongside two single-molecule approaches, total internal reflection fluorescence microscopy and high-resolution optical tweezers, have shown how the unique structural features of XPD helicase and its specific patterns of substrate interactions tune the helicase for its specific cellular function and shape its molecular mechanism. The FeS domain forms a duplex separation wedge and contributes to an extended DNA binding site. Interactions within this site position the helicase in an orientation to unwind the duplex, control the helicase rate, and verify the integrity of the translocating strand. Consistent with its cellular role, processivity of XPD is limited and is defined by an idiosyncratic stepping kinetics. DNA duplex separation occurs in single base pair steps punctuated by frequent backward steps and conformational rearrangements of the protein-DNA complex. As such, the helicase in isolation mainly stabilizes spontaneous base pair opening and exhibits a limited ability to unwind stable DNA duplexes. The presence of a cognate ssDNA binding protein converts XPD into a vigorous helicase by destabilizing the upstream dsDNA as well as by trapping the unwound strands. Remarkably, the two proteins can co-exist on the same DNA strand without competing for binding. The current model of the XPD unwinding mechanism will be discussed along with possible modifications to this mechanism by the helicase interacting partners and unique features of such bio-medically important XPD-like helicases as FANCJ (BACH1), RTEL1 and CHLR1 (DDX11).


Assuntos
DNA de Cadeia Simples/química , Pinças Ópticas , Proteína Grupo D do Xeroderma Pigmentoso/química , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , DNA de Cadeia Simples/metabolismo , Humanos , Microscopia de Fluorescência , Dados de Sequência Molecular , Ligação Proteica , Proteína Grupo D do Xeroderma Pigmentoso/metabolismo
16.
J Biol Chem ; 289(6): 3613-24, 2014 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-24338567

RESUMO

Recognition and removal of DNA damages is essential for cellular and organismal viability. Nucleotide excision repair (NER) is the sole mechanism in humans for the repair of carcinogenic UV irradiation-induced photoproducts in the DNA, such as cyclobutane pyrimidine dimers. The broad substrate versatility of NER further includes, among others, various bulky DNA adducts. It has been proposed that the 5'-3' helicase XPD (xeroderma pigmentosum group D) protein plays a decisive role in damage verification. However, despite recent advances such as the identification of a DNA-binding channel and central pore in the protein, through which the DNA is threaded, as well as a dedicated lesion recognition pocket near the pore, the exact process of target site recognition and verification in eukaryotic NER still remained elusive. Our single molecule analysis by atomic force microscopy reveals for the first time that XPD utilizes different recognition strategies to verify structurally diverse lesions. Bulky fluorescein damage is preferentially detected on the translocated strand, whereas the opposite strand preference is observed for a cyclobutane pyrimidine dimer lesion. Both states, however, lead to similar conformational changes in the resulting specific complexes, indicating a merge to a "final" verification state, which may then trigger the recruitment of further NER proteins.


Assuntos
Proteínas Arqueais/metabolismo , Dano ao DNA , Reparo do DNA/fisiologia , DNA Arqueal/metabolismo , Thermoplasma/enzimologia , Proteína Grupo D do Xeroderma Pigmentoso/metabolismo , Proteínas Arqueais/química , Proteínas Arqueais/genética , DNA Arqueal/química , DNA Arqueal/genética , Humanos , Dímeros de Pirimidina/química , Dímeros de Pirimidina/genética , Dímeros de Pirimidina/metabolismo , Thermoplasma/genética , Proteína Grupo D do Xeroderma Pigmentoso/química , Proteína Grupo D do Xeroderma Pigmentoso/genética
17.
Elife ; 2: e00334, 2013 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-23741615

RESUMO

Helicases couple the chemical energy of ATP hydrolysis to directional translocation along nucleic acids and transient duplex separation. Understanding helicase mechanism requires that the basic physicochemical process of base pair separation be understood. This necessitates monitoring helicase activity directly, at high spatio-temporal resolution. Using optical tweezers with single base pair (bp) resolution, we analyzed DNA unwinding by XPD helicase, a Superfamily 2 (SF2) DNA helicase involved in DNA repair and transcription initiation. We show that monomeric XPD unwinds duplex DNA in 1-bp steps, yet exhibits frequent backsteps and undergoes conformational transitions manifested in 5-bp backward and forward steps. Quantifying the sequence dependence of XPD stepping dynamics with near base pair resolution, we provide the strongest and most direct evidence thus far that forward, single-base pair stepping of a helicase utilizes the spontaneous opening of the duplex. The proposed unwinding mechanism may be a universal feature of DNA helicases that move along DNA phosphodiester backbones. DOI:http://dx.doi.org/10.7554/eLife.00334.001.


Assuntos
Proteínas Arqueais/metabolismo , Replicação do DNA , DNA/metabolismo , Proteína Grupo D do Xeroderma Pigmentoso/metabolismo , Trifosfato de Adenosina/metabolismo , Proteínas Arqueais/química , Sequência de Bases , Simulação por Computador , DNA/química , Reparo do DNA , Hidrólise , Cinética , Modelos Biológicos , Conformação de Ácido Nucleico , Pinças Ópticas , RNA/biossíntese , Processos Estocásticos , Iniciação da Transcrição Genética , Proteína Grupo D do Xeroderma Pigmentoso/química
18.
Int J Cancer ; 133(4): 1016-22, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23390054

RESUMO

The use of platinated agents in combination chemotherapy regimens for advanced pancreatic cancer is controversial owing to the lack of an outstanding impact on the outcome and a substantial increase in hematologic and extra-hematologic toxicities. Pharmacogenetic studies to identify patients who could benefit most from such therapies are urgently needed. The Xeroderma-Pigmentosum group-D polymorphism at codon-751 (XPD-Lys751Gln) emerged as the most significant independent predictor for death- and progression-risk in our previous study on functional polymorphisms in 122 advanced pancreatic cancer patients treated with cisplatin-docetaxel-capecitabine-gemcitabine and cisplatin-epirubicin-capecitabine-gemcitabine (or EC-GemCap). To confirm the prognostic role of this variable, we further evaluated the correlation of XPD-Lys751Gln with outcome in another 125 patients treated with the same regimens, and 90 treated with gemcitabine monotherapy. Genotyping was successfully carried out in the vast majority of DNA samples. Genotype frequencies followed Hardy-Weinberg equilibrium, and XPD-Lys751Gln was associated with differential progression-free and overall-survival. Multivariate analysis confirmed its prognostic significance in platinum-based regimens. In particular, XPD-Gln751Gln was significantly associated with risk of death (hazard ratio, HR = 1.7, 95% confidence interval [CI], 1.1-2.6, p = 0.011) and risk of progression (HR = 1.7, 95% CI, 1.1-2.5, p = 0.013). No correlation was observed in gemcitabine monotherapy-treated patients. The analysis of DNA damage using extra-long-PCR in lymphocytes supported the association of XPD-Gln751Gln with greater resistance to cisplatin-induced damage. The increasing evidence of XPD-Lys751Gln impact on the outcome of gemcitabine-cisplatin-based polychemotherapy leads to plan prospective studies to validate the role of this polymorphism as a new tool for optimization of the currently available treatments in pancreatic cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Glicina/genética , Lisina/genética , Neoplasias Pancreáticas/tratamento farmacológico , Polimorfismo Genético , Proteína Grupo D do Xeroderma Pigmentoso/genética , Idoso , Cisplatino/administração & dosagem , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/genética , Reação em Cadeia da Polimerase , Prognóstico , Proteína Grupo D do Xeroderma Pigmentoso/química , Gencitabina
19.
Proc Natl Acad Sci U S A ; 110(8): E633-42, 2013 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-23382212

RESUMO

The xeroderma pigmentosum group D (XPD) helicase is a subunit of transcription/DNA repair factor, transcription factor II H (TFIIH) that catalyzes the unwinding of a damaged DNA duplex during nucleotide excision repair. Apart from two canonical helicase domains, XPD is composed of a 4Fe-S cluster domain involved in DNA damage recognition and a module of uncharacterized function termed the "ARCH domain." By investigating the consequences of a mutation found in a patient with trichothiodystrophy, we show that the ARCH domain is critical for the recruitment of the cyclin-dependent kinase (CDK)-activating kinase (CAK) complex. Indeed, this mutation not only affects the interaction with the MAT1 CAK subunit, thereby decreasing the in vitro basal transcription activity of TFIIH itself and impeding the efficient recruitment of the transcription machinery on the promoter of an activated gene, but also impairs the DNA unwinding activity of XPD and the nucleotide excision repair activity of TFIIH. We further demonstrate the role of CAK in downregulating the XPD helicase activity within TFIIH. Taken together, our results identify the ARCH domain of XPD as a platform for the recruitment of CAK and as a potential molecular switch that might control TFIIH composition and play a key role in the conversion of TFIIH from a factor active in transcription to a factor involved in DNA repair.


Assuntos
Reparo do DNA , Mutação , Receptores Proteína Tirosina Quinases/metabolismo , Fator de Transcrição TFIIH/fisiologia , Transcrição Gênica , Proteína Grupo D do Xeroderma Pigmentoso/genética , Linhagem Celular , Imunoprecipitação da Cromatina , Receptor com Domínio Discoidina 1 , Humanos , Proteínas Ferro-Enxofre/metabolismo , Modelos Moleculares , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Síndromes de Tricotiodistrofia/genética , Proteína Grupo D do Xeroderma Pigmentoso/química , Proteína Grupo D do Xeroderma Pigmentoso/metabolismo
20.
Proc Natl Acad Sci U S A ; 110(3): E212-20, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23267107

RESUMO

Specific mutations in the XPD subunit of transcription factor IIH result in combined xeroderma pigmentosum (XP)/Cockayne syndrome (CS), a severe DNA repair disorder characterized at the cellular level by a transcriptional arrest following UV irradiation. This transcriptional arrest has always been thought to be the result of faulty transcription-coupled repair. In the present study, we showed that, following UV irradiation, XP-D/CS cells displayed a gross transcriptional dysregulation compared with "pure" XP-D cells or WT cells. Furthermore, global RNA-sequencing analysis showed that XP-D/CS cells repressed the majority of genes after UV, whereas pure XP-D cells did not. By using housekeeping genes as a model, we demonstrated that XP-D/CS cells were unable to reassemble these gene promoters and thus to restart transcription after UV irradiation. Furthermore, we found that the repression of these promoters in XP-D/CS cells was not a simple consequence of deficient repair but rather an active heterochromatinization process mediated by the histone deacetylase Sirt1. Indeed, RNA-sequencing analysis showed that inhibition of and/or silencing of Sirt1 changed the chromatin environment at these promoters and restored the transcription of a large portion of the repressed genes in XP-D/CS cells after UV irradiation. Our work demonstrates that a significant part of the transcriptional arrest displayed by XP-D/CS cells arises as a result of an active repression process and not simply as a result of a DNA repair deficiency. This dysregulation of Sirt1 function that results in transcriptional repression may be the cause of various severe clinical features in patients with XP-D/CS that cannot be explained by a DNA repair defect.


Assuntos
Síndrome de Cockayne/genética , Síndrome de Cockayne/metabolismo , RNA/biossíntese , Sirtuína 1/metabolismo , Proteína Grupo D do Xeroderma Pigmentoso/genética , Proteína Grupo D do Xeroderma Pigmentoso/metabolismo , Xeroderma Pigmentoso/genética , Xeroderma Pigmentoso/metabolismo , Células Cultivadas , Síndrome de Cockayne/complicações , Reparo do DNA , Heterocromatina/genética , Heterocromatina/metabolismo , Heterocromatina/efeitos da radiação , Humanos , Mutação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/genética , Fator de Transcrição TFIIH/química , Fator de Transcrição TFIIH/genética , Fator de Transcrição TFIIH/metabolismo , Transcrição Gênica/efeitos da radiação , Raios Ultravioleta/efeitos adversos , Xeroderma Pigmentoso/complicações , Proteína Grupo D do Xeroderma Pigmentoso/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...