Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 51(9): 4363-4384, 2023 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-36942481

RESUMO

Crossing over between homologs is critical for the stable segregation of chromosomes during the first meiotic division. Saccharomyces cerevisiae Mer3 (HFM1 in mammals) is a SF2 helicase and member of the ZMM group of proteins, that facilitates the formation of the majority of crossovers during meiosis. Here, we describe the structural organisation of Mer3 and using AlphaFold modelling and XL-MS we further characterise the previously described interaction with Mlh1-Mlh2. We find that Mer3 also forms a previously undescribed complex with the recombination regulating factors Top3 and Rmi1 and that this interaction is competitive with Sgs1BLM helicase. Using in vitro reconstituted D-loop assays we show that Mer3 inhibits the anti-recombination activity of Sgs1 helicase, but only in the presence of Dmc1. Thus we provide a mechanism whereby Mer3 interacts with a network of proteins to protect Dmc1 derived D-loops from dissolution.


Assuntos
DNA Helicases , Recombinação Homóloga , Meiose , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Proteínas de Ciclo Celular/genética , Troca Genética , DNA Helicases/química , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Meiose/genética , Ligação Proteica , Dobramento de Proteína , RecQ Helicases/antagonistas & inibidores , RecQ Helicases/química , RecQ Helicases/metabolismo , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Ligação Competitiva
2.
Cytogenet Genome Res ; 161(6-7): 305-327, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34474412

RESUMO

Human RecQ helicases play diverse roles in the maintenance of genomic stability. Inactivating mutations in 3 of the 5 human RecQ helicases are responsible for the pathogenesis of Werner syndrome (WS), Bloom syndrome (BS), Rothmund-Thomson syndrome (RTS), RAPADILINO, and Baller-Gerold syndrome (BGS). WS, BS, and RTS patients are at increased risk for developing many age-associated diseases including cancer. Mutations in RecQL1 and RecQL5 have not yet been associated with any human diseases so far. In terms of disease outcome, RecQL4 deserves special attention because mutations in RecQL4 result in 3 autosomal recessive syndromes (RTS type II, RAPADILINO, and BGS). RecQL4, like other human RecQ helicases, has been demonstrated to play a crucial role in the maintenance of genomic stability through participation in diverse DNA metabolic activities. Increased incidence of osteosarcoma in RecQL4-mutated RTS patients and elevated expression of RecQL4 in sporadic cancers including osteosarcoma suggest that loss or gain of RecQL4 expression is linked with cancer susceptibility. In this review, current and future perspectives are discussed on the potential use of RecQL4 as a novel cancer therapeutic target.


Assuntos
Síndrome de Bloom/genética , Predisposição Genética para Doença/genética , Mutação , RecQ Helicases/genética , Síndrome de Rothmund-Thomson/genética , Síndrome de Werner/genética , Humanos , Terapia de Alvo Molecular , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/terapia , RecQ Helicases/antagonistas & inibidores , RecQ Helicases/metabolismo , Fatores de Risco
3.
Mol Ther ; 29(3): 1016-1027, 2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33678249

RESUMO

Recombinant adeno-associated virus (rAAV) vectors have the unique property of being able to perform genomic targeted integration (TI) without inducing a double-strand break (DSB). In order to improve our understanding of the mechanism behind TI mediated by AAV and improve its efficiency, we performed an unbiased genetic screen in human cells using a promoterless AAV-homologous recombination (AAV-HR) vector system. We identified that the inhibition of the Fanconi anemia complementation group M (FANCM) protein enhanced AAV-HR-mediated TI efficiencies in different cultured human cells by ∼6- to 9-fold. The combined knockdown of the FANCM and two proteins also associated with the FANCM complex, RecQ-mediated genome instability 1 (RMI1) and Bloom DNA helicase (BLM) from the BLM-topoisomerase IIIα (TOP3A)-RMI (BTR) dissolvase complex (RMI1, having also been identified in our screen), led to the enhancement of AAV-HR-mediated TI up to ∼17 times. AAV-HR-mediated TI in the presence of a nuclease (CRISPR-Cas9) was also increased by ∼1.5- to 2-fold in FANCM and RMI1 knockout cells, respectively. Furthermore, knockdown of FANCM in human CD34+ hematopoietic stem and progenitor cells (HSPCs) increased AAV-HR-mediated TI by ∼3.5-fold. This study expands our knowledge on the mechanisms related to AAV-mediated TI, and it highlights new pathways that might be manipulated for future improvements in AAV-HR-mediated TI.


Assuntos
Sistemas CRISPR-Cas , DNA Helicases/antagonistas & inibidores , Proteínas de Ligação a DNA/antagonistas & inibidores , Dependovirus/genética , Edição de Genes , Células-Tronco Hematopoéticas/metabolismo , RecQ Helicases/antagonistas & inibidores , DNA Helicases/genética , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Vetores Genéticos , Células HeLa , Células-Tronco Hematopoéticas/citologia , Recombinação Homóloga , Humanos , RecQ Helicases/genética , RecQ Helicases/metabolismo
4.
Elife ; 102021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33647232

RESUMO

BLM (Bloom syndrome protein) is a RECQ-family helicase involved in the dissolution of complex DNA structures and repair intermediates. Synthetic lethality analysis implicates BLM as a promising target in a range of cancers with defects in the DNA damage response; however, selective small molecule inhibitors of defined mechanism are currently lacking. Here, we identify and characterise a specific inhibitor of BLM's ATPase-coupled DNA helicase activity, by allosteric trapping of a DNA-bound translocation intermediate. Crystallographic structures of BLM-DNA-ADP-inhibitor complexes identify a hitherto unknown interdomain interface, whose opening and closing are integral to translocation of ssDNA, and which provides a highly selective pocket for drug discovery. Comparison with structures of other RECQ helicases provides a model for branch migration of Holliday junctions by BLM.


Assuntos
RecQ Helicases/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , DNA/metabolismo , DNA Cruciforme , DNA de Cadeia Simples , Descoberta de Drogas , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Escherichia coli , Ensaios de Triagem em Larga Escala , Humanos , RecQ Helicases/metabolismo
5.
Trends Cancer ; 7(2): 146-161, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33041245

RESUMO

DNA helicases have risen to the forefront as genome caretakers. Their prominent roles in chromosomal stability are demonstrated by the linkage of mutations in helicase genes to hereditary disorders with defects in DNA repair, the replication stress response, and/or transcriptional activation. Conversely, accumulating evidence suggests that DNA helicases in cancer cells have a network of pathway interactions such that codeficiency of some helicases and their genetically interacting proteins results in synthetic lethality (SL). Such genetic interactions may potentially be exploited for cancer therapies. We discuss the roles of RECQ DNA helicases in cancer, emphasizing some of the more recent developments in SL.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/genética , RecQ Helicases/metabolismo , Mutações Sintéticas Letais/efeitos dos fármacos , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Reparo do DNA/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Instabilidade Genômica/efeitos dos fármacos , Humanos , Oncologia/métodos , Oncologia/tendências , Camundongos , Mutação , Neoplasias/tratamento farmacológico , Medicina de Precisão/métodos , Medicina de Precisão/tendências , RecQ Helicases/antagonistas & inibidores , RecQ Helicases/genética , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Med Chem ; 63(17): 9752-9772, 2020 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-32697083

RESUMO

DNA damage response (DDR) pathways are crucial for the survival of cancer cells and are attractive targets for cancer therapy. Bloom syndrome protein (BLM) is a DNA helicase that performs important roles in DDR pathways. Our previous study discovered an effective new BLM inhibitor with a quinazolinone scaffold by a screening assay. Herein, to better understand the structure-activity relationship (SAR) and biological roles of the BLM inhibitor, a series of new derivatives were designed, synthesized, and evaluated based on this scaffold. Among them, compound 9h exhibited nanomolar inhibitory activity and binding affinity for BLM. 9h could effectively disrupt BLM recruitment to DNA in cells. Furthermore, 9h inhibited the proliferation of the colorectal cell line HCT116 by significantly triggering DNA damage in the telomere region and inducing apoptosis, especially in combination with a poly (ADP-ribose) polymerase (PARP) inhibitor. This result suggested a synthetic lethal effect between the BLM and PARP inhibitors in DDR pathways.


Assuntos
Dano ao DNA , Desenho de Fármacos , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Quinazolinonas/síntese química , Quinazolinonas/farmacologia , RecQ Helicases/antagonistas & inibidores , Telômero/genética , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Técnicas de Química Sintética , Sinergismo Farmacológico , Células HCT116 , Humanos , Modelos Moleculares , Conformação Proteica , Quinazolinonas/química , RecQ Helicases/química , Relação Estrutura-Atividade
7.
Anticancer Agents Med Chem ; 20(11): 1311-1326, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32418530

RESUMO

Replication fork reversal and restart has gained immense interest as a central response mechanism to replication stress following DNA damage. Although the exact mechanism of fork reversal has not been elucidated precisely, the involvement of diverse pathways and different factors has been demonstrated, which are central to this phenomenon. RecQ helicases known for their vital role in DNA repair and maintaining genome stability has recently been implicated in the restart of regressed replication forks. Through interaction with vital proteins like Poly (ADP) ribose polymerase 1 (PARP1), these helicases participate in the replication fork reversal and restart phenomenon. Most therapeutic agents used for cancer chemotherapy act by causing DNA damage in replicating cells and subsequent cell death. These DNA damages can be repaired by mechanisms involving fork reversal as the key phenomenon eventually reducing the efficacy of the therapeutic agent. Hence the factors contributing to this repair process can be good selective targets for developing more efficient chemotherapeutic agents. In this review, we have discussed in detail the role of various proteins in replication fork reversal and restart with special emphasis on RecQ helicases. Involvement of other proteins like PARP1, recombinase rad51, SWI/SNF complex has also been discussed. Since RecQ helicases play a central role in the DNA damage response following chemotherapeutic treatment, we propose that targeting these helicases can emerge as an alternative to available intervention strategies. We have also summarized the current research status of available RecQ inhibitors and siRNA based therapeutic approaches that targets RecQ helicases. In summary, our review gives an overview of the DNA damage responses involving replication fork reversal and provides new directions for the development of more efficient and sustainable chemotherapeutic approaches.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Neoplasias/tratamento farmacológico , RecQ Helicases/antagonistas & inibidores , Antineoplásicos/química , Reparo do DNA , Replicação do DNA , Inibidores Enzimáticos/química , Humanos , Neoplasias/metabolismo , RecQ Helicases/metabolismo
8.
J Med Chem ; 62(6): 3147-3162, 2019 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-30827110

RESUMO

Homologous recombination repair (HRR), a crucial approach in DNA damage repair, is an attractive target in cancer therapy and drug design. The Bloom syndrome protein (BLM) is a 3'-5' DNA helicase that performs an important role in HRR regulation. However, limited studies about BLM inhibitors and their biological effects have been reported. Here, we identified a class of isaindigotone derivatives as novel BLM inhibitors by synthesis, screening, and evaluating. Among them, compound 29 was found as an effective BLM inhibitor with a high binding affinity and good inhibitory effect on BLM. Cellular evaluation indicated that 29 effectively disrupted the recruitment of BLM at DNA double-strand break sites, promoted an accumulation of RAD51, and regulated the HRR process. Meanwhile, 29 significantly induced DNA damage responses, as well as apoptosis and proliferation arrest in cancer cells. Our finding provides a potential anticancer strategy based on interfering with BLM via small molecules.


Assuntos
Alcaloides/farmacologia , DNA/metabolismo , Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Quinazolinas/farmacologia , RecQ Helicases/antagonistas & inibidores , Reparo de DNA por Recombinação , Alcaloides/química , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Inibidores Enzimáticos/química , Células HCT116 , Humanos , Quinazolinas/química , Rad51 Recombinase/metabolismo , RecQ Helicases/metabolismo
9.
Proc Natl Acad Sci U S A ; 115(14): 3680-3685, 2018 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-29563231

RESUMO

Discovering the genetic changes underlying species differences is a central goal in evolutionary genetics. However, hybrid crosses between species in mammals often suffer from hybrid sterility, greatly complicating genetic mapping of trait variation across species. Here, we describe a simple, robust, and transgene-free technique to generate "in vitro crosses" in hybrid mouse embryonic stem (ES) cells by inducing random mitotic cross-overs with the drug ML216, which inhibits the DNA helicase Bloom syndrome (BLM). Starting with an interspecific F1 hybrid ES cell line between the Mus musculus laboratory mouse and Mus spretus (∼1.5 million years of divergence), we mapped the genetic basis of drug resistance to the antimetabolite tioguanine to a single region containing hypoxanthine-guanine phosphoribosyltransferase (Hprt) in as few as 21 d through "flow mapping" by coupling in vitro crosses with fluorescence-activated cell sorting (FACS). We also show how our platform can enable direct study of developmental variation by rederiving embryos with contribution from the recombinant ES cell lines. We demonstrate how in vitro crosses can overcome major bottlenecks in mouse complex trait genetics and address fundamental questions in evolutionary biology that are otherwise intractable through traditional breeding due to high cost, small litter sizes, and/or hybrid sterility. In doing so, we describe an experimental platform toward studying evolutionary systems biology in mouse and potentially in human and other mammals, including cross-species hybrids.


Assuntos
Cruzamentos Genéticos , Células-Tronco Embrionárias Murinas/citologia , Locos de Características Quantitativas , Animais , Antimetabólitos Antineoplásicos/farmacologia , Evolução Biológica , Células Cultivadas , Mapeamento Cromossômico , Resistência a Medicamentos/genética , Feminino , Hibridização Genética , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/metabolismo , Fenótipo , Gravidez , RecQ Helicases/antagonistas & inibidores , Especificidade da Espécie , Tioguanina/farmacologia
10.
Cancer Lett ; 413: 1-10, 2018 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-29080750

RESUMO

Human RecQ helicases that share homology with E. coli RecQ helicase play critical roles in diverse biological activities such as DNA replication, transcription, recombination and repair. Mutations in three of the five human RecQ helicases (RecQ1, WRN, BLM, RecQL4 and RecQ5) result in autosomal recessive syndromes characterized by accelerated aging symptoms and cancer incidence. Mutational inactivation of Werner (WRN) and Bloom (BLM) genes results in Werner syndrome (WS) and Bloom syndrome (BS) respectively. However, mutations in RecQL4 result in three human disorders: (I) Rothmund-Thomson syndrome (RTS), (II) RAPADILINO and (III) Baller-Gerold syndrome (BGS). Cells from WS, BS and RTS are characterized by a unique chromosomal anomaly indicating that each of the RecQ helicases performs specialized function(s) in a non-redundant manner. Elucidating the biological functions of RecQ helicases will enable us to understand not only the aging process but also to determine the cause for age-associated human diseases. Recent biochemical and molecular studies have given new insights into the multifaceted roles of RecQL4 that range from genomic stability to carcinogenesis and beyond. This review summarizes some of the existing and emerging knowledge on diverse biological functions of RecQL4 and its significance as a potential molecular target for cancer therapy.


Assuntos
Canal Anal/anormalidades , Biomarcadores Tumorais/metabolismo , Transformação Celular Neoplásica/metabolismo , Craniossinostoses/enzimologia , Nanismo/enzimologia , Instabilidade Genômica , Comunicação Interatrial/enzimologia , Deformidades Congênitas dos Membros/enzimologia , Neoplasias/enzimologia , Patela/anormalidades , Rádio (Anatomia)/anormalidades , RecQ Helicases/metabolismo , Síndrome de Rothmund-Thomson/enzimologia , Canal Anal/enzimologia , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/antagonistas & inibidores , Biomarcadores Tumorais/genética , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Craniossinostoses/genética , Reparo do DNA , Replicação do DNA , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Nanismo/genética , Inibidores Enzimáticos/uso terapêutico , Predisposição Genética para Doença , Comunicação Interatrial/genética , Humanos , Deformidades Congênitas dos Membros/genética , Mutação , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/patologia , Patela/enzimologia , Fenótipo , Rádio (Anatomia)/enzimologia , RecQ Helicases/antagonistas & inibidores , RecQ Helicases/genética , Síndrome de Rothmund-Thomson/genética
11.
Sci Rep ; 7: 44358, 2017 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-28287182

RESUMO

Helicase are essential enzymes which are widespread in all life-forms. Due to their central role in nucleic acid metabolism, they are emerging as important targets for anti-viral, antibacterial and anti-cancer drugs. The development of easy, cheap, fast and robust biochemical assays to measure helicase activity, overcoming the limitations of the current methods, is a pre-requisite for the discovery of helicase inhibitors through high-throughput screenings. We have developed a method which exploits the optical properties of DNA-conjugated gold nanoparticles (AuNP) and meets the required criteria. The method was tested with the catalytic domain of the human RecQ4 helicase and compared with a conventional FRET-based assay. The AuNP-based assay produced similar results but is simpler, more robust and cheaper than FRET. Therefore, our nanotechnology-based platform shows the potential to provide a useful alternative to the existing conventional methods for following helicase activity and to screen small-molecule libraries as potential helicase inhibitors.


Assuntos
Colorimetria/métodos , DNA/química , Ouro/química , Nanopartículas Metálicas/química , RecQ Helicases/metabolismo , Sequência de Bases , Domínio Catalítico , DNA/genética , Ensaios Enzimáticos/métodos , Transferência Ressonante de Energia de Fluorescência , Ensaios de Triagem em Larga Escala , Humanos , RecQ Helicases/antagonistas & inibidores , RecQ Helicases/química , Bibliotecas de Moléculas Pequenas/isolamento & purificação , Bibliotecas de Moléculas Pequenas/farmacologia
12.
Leukemia ; 31(10): 2104-2113, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28186131

RESUMO

Multiple myeloma (MM) is a plasma cell cancer with poor survival, characterized by the expansion of multiple myeloma cells (MMCs) in the bone marrow. Using a microarray-based genome-wide screen for genes responding to DNA methyltransferases (DNMT) inhibition in MM cells, we identified RECQ1 among the most downregulated genes. RecQ helicases are DNA unwinding enzymes involved in the maintenance of chromosome stability. Here we show that RECQ1 is significantly overexpressed in MMCs compared to normal plasma cells and that increased RECQ1 expression is associated with poor prognosis in three independent cohorts of patients. Interestingly, RECQ1 knockdown inhibits cells growth and induces apoptosis in MMCs. Moreover, RECQ1 depletion promotes the development of DNA double-strand breaks, as evidenced by the formation of 53BP1 foci and the phosphorylation of ataxia-telangiectasia mutated (ATM) and histone variant H2A.X (H2AX). In contrast, RECQ1 overexpression protects MMCs from melphalan and bortezomib cytotoxicity. RECQ1 interacts with PARP1 in MMCs exposed to treatment and RECQ1 depletion sensitizes MMCs to poly(ADP-ribose) polymerase (PARP) inhibitor. DNMT inhibitor treatment results in RECQ1 downregulation through miR-203 deregulation in MMC. Altogether, these data suggest that association of DNA damaging agents and/or PARP inhibitors with DNMT inhibitors may represent a therapeutic approach in patients with high RECQ1 expression associated with a poor prognosis.


Assuntos
DNA de Neoplasias/genética , Resistencia a Medicamentos Antineoplásicos/fisiologia , Mieloma Múltiplo/enzimologia , Proteínas de Neoplasias/fisiologia , RecQ Helicases/fisiologia , Bortezomib/farmacologia , Ciclo Celular/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla , Dano ao DNA , Metilação de DNA/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , DNA de Neoplasias/metabolismo , DNA-Citosina Metilases/antagonistas & inibidores , Indução Enzimática , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Regulação Neoplásica da Expressão Gênica , Humanos , Melfalan/farmacologia , MicroRNAs/genética , Terapia de Alvo Molecular , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/enzimologia , Plasmócitos/efeitos dos fármacos , Plasmócitos/enzimologia , Poli(ADP-Ribose) Polimerase-1/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , RecQ Helicases/antagonistas & inibidores , RecQ Helicases/genética , Células Tumorais Cultivadas
13.
Biochim Biophys Acta Rev Cancer ; 1867(1): 42-48, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27902925

RESUMO

WRN helicase has several roles in genome maintenance, such as replication, base excision repair, recombination, DNA damage response and transcription. These processes are often found upregulated in human cancers, many of which display increased levels of WRN. Therefore, directed inhibition of this RecQ helicase could be beneficial to selective cancer therapy. Inhibition of WRN is feasible by the use of small-molecule inhibitors or application of RNA interference and EGS/RNase P targeting systems. Remarkably, helicase depletion leads to a severe reduction in cell viability due to mitotic catastrophe, which is triggered by replication stress induced by DNA repair failure and fork progression arrest. Moreover, we present new evidence that WRN depletion results in early changes of RNA polymerase III and RNase P activities, thereby implicating chromatin-associated tRNA enzymes in WRN-related stress response. Combined with the recently discovered roles of RecQ helicases in cancer, current data support the targeting prospect of these genome guardians, as a means of developing clinical phases aimed at diminishing adaptive resistance to present targeted therapies.


Assuntos
Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Replicação do DNA/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Helicase da Síndrome de Werner/antagonistas & inibidores , Animais , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/genética , Replicação do DNA/genética , Humanos , RecQ Helicases/antagonistas & inibidores
14.
Biochem Cell Biol ; 94(6): 551-559, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27813658

RESUMO

RecQL4, one of the 5 human RecQ helicases, is a key mediator of genomic stability and its deficiency can cause premature aging phenotypes. Here, by using CRISPR/Cas and RNAi technology, we demonstrated that autophagy level was elevated in both RecQL4 knockdown and knockout cells compared with those of the control cells. Surprisingly, mitochondrial content was increased and LC3 co-localization with mitochondria was partially lost in RecQL4 knockout cells compared with the control cells, suggesting that RecQL4 deficiency impaired mitophagic processes in U2OS cells. Furthermore, we found that knockout of RecQL4 destabilized PINK1. In addition, RecQL4 knockout cells were more susceptible to apoptosis under mitochondrial stress than the control cells. In conclusion, our findings indicated a novel role of RecQL4 in the regulation of autophagy/mitophagy in U2OS cells.


Assuntos
Apoptose , Autofagia , Neoplasias Ósseas/patologia , Mitocôndrias/patologia , Osteossarcoma/patologia , RecQ Helicases/metabolismo , Sequência de Bases , Western Blotting , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Sistemas CRISPR-Cas , Dano ao DNA , Imunofluorescência , Instabilidade Genômica , Humanos , Mitocôndrias/metabolismo , Osteossarcoma/genética , Osteossarcoma/metabolismo , Fenótipo , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , RecQ Helicases/antagonistas & inibidores , RecQ Helicases/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
15.
Oncotarget ; 7(46): 76140-76150, 2016 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-27764811

RESUMO

RECQ helicases are a family of enzymes with both over lapping and unique functions. Functional autosomal recessive loss of three members of the family BLM, WRN and RECQL4, results in hereditary human syndromes characterized by cancer predisposition and premature aging, but despite the finding that RECQL5 deficient mice are cancer prone, no such link has been made to human RECQL5. Here we demonstrate that human urothelial carcinoma of the bladder (UCC) has increased expression of RECQL5 compared to normal bladder tissue and that increasing RECQL5 expression can drive proliferation of normal bladder cells and is associated with poor prognosis. Further, by expressing a helicase dead RECQL5 and by depleting bladder cancer cells of RECQL5 we show that inhibition of RECQL5 activity has potential as a new target for treatment of UCC.


Assuntos
Carcinoma de Células de Transição/genética , Carcinoma de Células de Transição/metabolismo , Regulação Neoplásica da Expressão Gênica , RecQ Helicases/genética , RecQ Helicases/metabolismo , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/metabolismo , Antineoplásicos/farmacologia , Carcinoma de Células de Transição/tratamento farmacológico , Carcinoma de Células de Transição/patologia , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular/genética , Progressão da Doença , Ativação Enzimática , Humanos , Prognóstico , RecQ Helicases/antagonistas & inibidores , Estresse Fisiológico , Análise de Sobrevida , Neoplasias da Bexiga Urinária/tratamento farmacológico , Neoplasias da Bexiga Urinária/patologia
16.
Sci Rep ; 6: 26225, 2016 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-27194376

RESUMO

Cadmium is a toxic metal that inactivates DNA-repair proteins via multiple mechanisms, including zinc substitution. In this study, we investigated the effect of Cd(2+) on the Bloom protein (BLM), a DNA-repair helicase carrying a zinc-binding domain (ZBD) and playing a critical role to ensure genomic stability. One characteristics of BLM-deficient cells is the elevated rate of sister chromatid exchanges, a phenomenon that is also induced by Cd(2+). Here, we show that Cd(2+) strongly inhibits both ATPase and helicase activities of BLM. Cd(2+) primarily prevents BLM-DNA interaction via its binding to sulfhydryl groups of solvent-exposed cysteine residues and, concomitantly, promotes the formation of large BLM multimers/aggregates. In contrast to previously described Cd(2+) effects on other zinc-containing DNA-repair proteins, the ZBD appears to play a minor role in the Cd(2+)-mediated inhibition. While the Cd(2+)-dependent formation of inactive multimers and the defect of DNA-binding were fully reversible upon addition of EDTA, the inhibition of the DNA unwinding activity was not counteracted by EDTA, indicating another mechanism of inhibition by Cd(2+) relative to the targeting of a catalytic residue. Altogether, our results provide new clues for understanding the mechanism behind the ZBD-independent inactivation of BLM by Cd(2+) leading to accumulation of DNA double-strand breaks.


Assuntos
Cádmio/toxicidade , Inibidores Enzimáticos/toxicidade , RecQ Helicases/antagonistas & inibidores , Adenosina Trifosfatases/antagonistas & inibidores , DNA/metabolismo , DNA Helicases/antagonistas & inibidores , Ácido Edético/metabolismo , Ligação Proteica
17.
J Biomol Screen ; 21(6): 626-33, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26962873

RESUMO

Induction of the Fanconi anemia (FA) DNA repair pathway is a common mechanism by which tumors evolve resistance to DNA crosslinking chemotherapies. Proper execution of the FA pathway requires interaction between the FA complementation group M protein (FANCM) and the RecQ-mediated genome instability protein (RMI) complex, and mutations that disrupt FANCM/RMI interactions sensitize cells to DNA crosslinking agents. Inhibitors that block FANCM/RMI complex formation could be useful therapeutics for resensitizing tumors that have acquired chemotherapeutic resistance. To identify such inhibitors, we have developed and validated high-throughput fluorescence polarization and proximity assays that are sensitive to inhibitors that disrupt interactions between the RMI complex and its binding site on FANCM (a peptide referred to as MM2). A pilot screen of 74,807 small molecules was performed using the fluorescence polarization assay. Hits from the primary screen were further tested using the proximity assay, and an orthogonal proximity assay was used to assess inhibitor selectivity. Direct physical interaction between the RMI complex and the most selective inhibitor identified through the screening process was measured by surface plasmon resonance and isothermal titration calorimetry. Observation of direct binding by this small molecule validates the screening protocol.


Assuntos
Antineoplásicos/química , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Anemia de Fanconi/tratamento farmacológico , Ensaios de Triagem em Larga Escala/métodos , Complexos Multiproteicos/antagonistas & inibidores , Antineoplásicos/uso terapêutico , Dano ao DNA/efeitos dos fármacos , DNA Helicases/antagonistas & inibidores , DNA Helicases/genética , Reparo do DNA/efeitos dos fármacos , Anemia de Fanconi/genética , Anemia de Fanconi/patologia , Humanos , Mapas de Interação de Proteínas/efeitos dos fármacos , RecQ Helicases/antagonistas & inibidores , RecQ Helicases/genética
18.
Biochim Biophys Acta ; 1859(4): 572-80, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26808708

RESUMO

Human WRN, a RecQ helicase encoded by the Werner syndrome gene, is implicated in genome maintenance, including replication, recombination, excision repair and DNA damage response. These genetic processes and expression of WRN are concomitantly upregulated in many types of cancers. Therefore, targeted destruction of this helicase could be useful for elimination of cancer cells. Here, we provide a proof of concept for applying the external guide sequence (EGS) approach in directing an RNase P RNA to efficiently cleave the WRN mRNA in cultured human cell lines, thus abolishing translation and activity of this distinctive 3'-5' DNA helicase-nuclease. Remarkably, EGS-directed knockdown of WRN leads to severe inhibition of cell viability. Hence, further assessment of this targeting system could be beneficial for selective cancer therapies, particularly in the light of the recent improvements introduced into EGSs.


Assuntos
Exodesoxirribonucleases/genética , Biossíntese de Proteínas , RecQ Helicases/genética , Ribonuclease P/genética , Síndrome de Werner/genética , Linhagem Celular , Dano ao DNA , Reparo do DNA/genética , Replicação do DNA/genética , Exodesoxirribonucleases/antagonistas & inibidores , Genoma Humano , Instabilidade Genômica/genética , Humanos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RecQ Helicases/antagonistas & inibidores , Síndrome de Werner/patologia , Helicase da Síndrome de Werner
19.
Nucleic Acids Res ; 43(20): 9788-803, 2015 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-26275776

RESUMO

The WRN helicase/exonuclease protein is required for proper replication fork recovery and maintenance of genome stability. However, whether the different catalytic activities of WRN cooperate to recover replication forks in vivo is unknown. Here, we show that, in response to replication perturbation induced by low doses of the TOP1 inhibitor camptothecin, loss of the WRN exonuclease resulted in enhanced degradation and ssDNA formation at nascent strands by the combined action of MRE11 and EXO1, as opposed to the limited processing of nascent strands performed by DNA2 in wild-type cells. Nascent strand degradation by MRE11/EXO1 took place downstream of RAD51 and affected the ability to resume replication, which correlated with slow replication rates in WRN exonuclease-deficient cells. In contrast, loss of the WRN helicase reduced exonucleolytic processing at nascent strands and led to severe genome instability. Our findings identify a novel role of the WRN exonuclease at perturbed forks, thus providing the first in vivo evidence for a distinct action of the two WRN enzymatic activities upon fork stalling and providing insights into the pathological mechanisms underlying the processing of perturbed forks.


Assuntos
Replicação do DNA , Exodesoxirribonucleases/metabolismo , RecQ Helicases/metabolismo , Camptotecina/farmacologia , Linhagem Celular Transformada , DNA Helicases/metabolismo , Replicação do DNA/efeitos dos fármacos , DNA de Cadeia Simples/metabolismo , Proteínas de Ligação a DNA/metabolismo , Exodesoxirribonucleases/química , Exodesoxirribonucleases/genética , Instabilidade Genômica , Humanos , Mutação , Poli(ADP-Ribose) Polimerases/metabolismo , Estrutura Terciária de Proteína , RecQ Helicases/antagonistas & inibidores , RecQ Helicases/genética , Inibidores da Topoisomerase I/farmacologia
20.
Nucleic Acids Res ; 42(17): 11119-35, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25170083

RESUMO

4-Hydroxy-2-nonenal (HNE) is a reactive α,ß-unsaturated aldehyde generated during oxidative stress and subsequent peroxidation of polyunsaturated fatty acids. Here, Werner protein (WRN) was identified as a novel target for modification by HNE. Werner syndrome arises through mutations in the WRN gene that encodes the RecQ DNA helicase which is critical for maintaining genomic stability. This hereditary disease is associated with chromosomal instability, premature aging and cancer predisposition. WRN appears to participate in the cellular response to oxidative stress and cells devoid of WRN display elevated levels of oxidative DNA damage. We demonstrated that helicase/ATPase and exonuclease activities of HNE-modified WRN protein were inhibited both in vitro and in immunocomplexes purified from the cell extracts. Sites of HNE adduction in human WRN were identified at Lys577, Cys727, His1290, Cys1367, Lys1371 and Lys1389. We applied in silico modeling of the helicase and RQC domains of WRN protein with HNE adducted to Lys577 and Cys727 and provided a potential mechanism of the observed deregulation of the protein catalytic activities. In light of the obtained results, we postulate that HNE adduction to WRN is a post-translational modification, which may affect WRN conformational stability and function, contributing to features and diseases associated with premature senescence.


Assuntos
Aldeídos/química , Exodesoxirribonucleases/química , Exodesoxirribonucleases/metabolismo , RecQ Helicases/química , RecQ Helicases/metabolismo , Adenosina Trifosfatases/antagonistas & inibidores , Aldeídos/análise , Animais , Biocatálise , Células Cultivadas , DNA/metabolismo , Exodesoxirribonucleases/antagonistas & inibidores , Humanos , Modelos Moleculares , RecQ Helicases/antagonistas & inibidores , Helicase da Síndrome de Werner
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...