Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nucleic Acids Res ; 45(3): 1566, 2017 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-28180303

RESUMO

Humans have five RecQ helicases, whereas simpler organisms have only one. Little is known about whether and how these RecQ helicases co-operate and/or complement each other in response to cellular stress. Here we show that RECQL5 associates longer at laser-induced DNA double-strand breaks in the absence of Werner syndrome (WRN) protein, and that it interacts physically and functionally with WRN both in vivo and in vitro. RECQL5 co-operates with WRN on synthetic stalled replication fork-like structures and stimulates its helicase activity on DNA fork duplexes. Both RECQL5 and WRN re-localize from the nucleolus into the nucleus after replicative stress and significantly associate with each other during S-phase. Further, we show that RECQL5 is essential for cell survival in the absence of WRN. Loss of both RECQL5 and WRN severely compromises DNA replication, accumulates genomic instability and ultimately leads to cell death. Collectively, our results indicate that RECQL5 plays both co-operative and complementary roles with WRN. This is an early demonstration of a significant functional interplay and a novel synthetic lethal interaction among the human RecQ helicases.

2.
Nucleic Acids Res ; 41(2): 881-99, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23180761

RESUMO

Humans have five RecQ helicases, whereas simpler organisms have only one. Little is known about whether and how these RecQ helicases co-operate and/or complement each other in response to cellular stress. Here we show that RECQL5 associates longer at laser-induced DNA double-strand breaks in the absence of Werner syndrome (WRN) protein, and that it interacts physically and functionally with WRN both in vivo and in vitro. RECQL5 co-operates with WRN on synthetic stalled replication fork-like structures and stimulates its helicase activity on DNA fork duplexes. Both RECQL5 and WRN re-localize from the nucleolus into the nucleus after replicative stress and significantly associate with each other during S-phase. Further, we show that RECQL5 is essential for cell survival in the absence of WRN. Loss of both RECQL5 and WRN severely compromises DNA replication, accumulates genomic instability and ultimately leads to cell death. Collectively, our results indicate that RECQL5 plays both co-operative and complementary roles with WRN. This is an early demonstration of a significant functional interplay and a novel synthetic lethal interaction among the human RecQ helicases.


Assuntos
Quebras de DNA de Cadeia Dupla , Exodesoxirribonucleases/metabolismo , RecQ Helicases/metabolismo , Linhagem Celular , Sobrevivência Celular , Replicação do DNA , Exodesoxirribonucleases/fisiologia , Instabilidade Genômica , RecQ Helicases/antagonistas & inibidores , RecQ Helicases/fisiologia , Síndrome de Werner/genética , Helicase da Síndrome de Werner
3.
Nucleic Acids Res ; 40(4): 1621-35, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22013166

RESUMO

DNA decatenation mediated by Topoisomerase II is required to separate the interlinked sister chromatids post-replication. SGS1, a yeast homolog of the human RecQ family of helicases interacts with Topoisomerase II and plays a role in chromosome segregation, but this functional interaction has yet to be identified in higher organisms. Here, we report a physical and functional interaction of Topoisomerase IIα with RECQL5, one of five mammalian RecQ helicases, during DNA replication. Direct interaction of RECQL5 with Topoisomerase IIα stimulates the decatenation activity of Topoisomerase IIα. Consistent with these observations, RECQL5 co-localizes with Topoisomerase IIα during S-phase of the cell cycle. Moreover, cells with stable depletions of RECQL5 display a slow proliferation rate, a G2/M cell cycle arrest and late S-phase cycling defects. Metaphase spreads generated from RECQL5-depleted cells exhibit undercondensed and entangled chromosomes. Further, RECQL5-depleted cells activate a G2/M checkpoint and undergo apoptosis. These phenotypes are similar to those observed when Topoisomerase II catalytic activity is inhibited. These results reveal an important role for RECQL5 in the maintenance of genomic stability and a new insight into the decatenation process.


Assuntos
Antígenos de Neoplasias/metabolismo , Ciclo Celular , DNA Topoisomerases Tipo II/metabolismo , DNA Catenado/metabolismo , Proteínas de Ligação a DNA/metabolismo , RecQ Helicases/metabolismo , Apoptose , Pontos de Checagem do Ciclo Celular , Linhagem Celular , Proliferação de Células , Aberrações Cromossômicas , Exodesoxirribonucleases/metabolismo , Humanos , Metáfase/genética , RecQ Helicases/antagonistas & inibidores , Helicase da Síndrome de Werner
4.
Nucleic Acids Res ; 40(14): 6632-48, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22544709

RESUMO

Bacteria and yeast possess one RecQ helicase homolog whereas humans contain five RecQ helicases, all of which are important in preserving genome stability. Three of these, BLM, WRN and RECQL4, are mutated in human diseases manifesting in premature aging and cancer. We are interested in determining to which extent these RecQ helicases function cooperatively. Here, we report a novel physical and functional interaction between BLM and RECQL4. Both BLM and RECQL4 interact in vivo and in vitro. We have mapped the BLM interacting site to the N-terminus of RECQL4, comprising amino acids 361-478, and the region of BLM encompassing amino acids 1-902 interacts with RECQL4. RECQL4 specifically stimulates BLM helicase activity on DNA fork substrates in vitro. The in vivo interaction between RECQL4 and BLM is enhanced during the S-phase of the cell cycle, and after treatment with ionizing radiation. The retention of RECQL4 at DNA double-strand breaks is shortened in BLM-deficient cells. Further, depletion of RECQL4 in BLM-deficient cells leads to reduced proliferative capacity and an increased frequency of sister chromatid exchanges. Together, our results suggest that BLM and RECQL4 have coordinated activities that promote genome stability.


Assuntos
Instabilidade Genômica , RecQ Helicases/metabolismo , Linhagem Celular , DNA/metabolismo , Dano ao DNA , Guanina/análogos & derivados , Guanina/metabolismo , Células HeLa , Humanos , Domínios e Motivos de Interação entre Proteínas , RecQ Helicases/química , Fase S , Troca de Cromátide Irmã , Timina/análogos & derivados , Timina/metabolismo
5.
NAR Genom Bioinform ; 6(1): lqae019, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38344273

RESUMO

The correlation between messenger RNA (mRNA) and protein abundances has long been debated. RNA sequencing (RNA-seq), a high-throughput, commonly used method for analyzing transcriptional dynamics, leaves questions about whether we can translate RNA-seq-identified gene signatures directly to protein changes. In this study, we utilized a set of 17 widely assessed immune and wound healing mediators in the context of canine volumetric muscle loss to investigate the correlation of mRNA and protein abundances. Our data reveal an overall agreement between mRNA and protein levels on these 17 mediators when examining samples from the same experimental condition (e.g. the same biopsy). However, we observed a lack of correlation between mRNA and protein levels for individual genes under different conditions, underscoring the challenges in converting transcriptional changes into protein changes. To address this discrepancy, we developed a machine learning model to predict protein abundances from RNA-seq data, achieving high accuracy. Our approach also effectively corrected multiple extreme outliers measured by antibody-based protein assays. Additionally, this model has the potential to detect post-translational modification events, as shown by accurately estimating activated transforming growth factor ß1 levels. This study presents a promising approach for converting RNA-seq data into protein abundance and its biological significance.

6.
Carcinogenesis ; 34(10): 2218-30, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23715498

RESUMO

Interstrand cross-links (ICLs) are very severe lesions as they are absolute blocks of replication and transcription. This property of interstrand cross-linking agents has been exploited clinically for the treatment of cancers and other diseases. ICLs are repaired in human cells by specialized DNA repair pathways including components of the nucleotide excision repair pathway, double-strand break repair pathway and the Fanconi anemia pathway. In this report, we identify the role of RECQL5, a member of the RecQ family of helicases, in the repair of ICLs. Using laser-directed confocal microscopy, we demonstrate that RECQL5 is recruited to ICLs formed by trioxalen (a psoralen-derived compound) and ultraviolet irradiation A. Using single-cell gel electrophoresis and proliferation assays, we identify the role of RECQL5 in the repair of ICL lesions. The domain of RECQL5 that recruits to the site of ICL was mapped to the KIX region between amino acids 500 and 650. Inhibition of transcription and of topoisomerases did not affect recruitment, which was inhibited by DNA-intercalating agents, suggesting that the DNA structure itself may be responsible for the recruitment of RECQL5 to the sites of ICLs.


Assuntos
Reagentes de Ligações Cruzadas/toxicidade , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/fisiologia , Ficusina/toxicidade , RecQ Helicases/metabolismo , Linhagem Celular , DNA Topoisomerases/metabolismo , Exodesoxirribonucleases/metabolismo , Humanos , Cinética , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , RecQ Helicases/química , Inibidores da Topoisomerase/farmacologia , Transcrição Gênica , Helicase da Síndrome de Werner
7.
J Biol Chem ; 287(1): 196-209, 2012 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-22039056

RESUMO

Telomeres are structures at the ends of chromosomes and are composed of long tracks of short tandem repeat DNA sequences bound by a unique set of proteins (shelterin). Telomeric DNA is believed to form G-quadruplex and D-loop structures, which presents a challenge to the DNA replication and repair machinery. Although the RecQ helicases WRN and BLM are implicated in the resolution of telomeric secondary structures, very little is known about RECQL4, the RecQ helicase mutated in Rothmund-Thomson syndrome (RTS). Here, we report that RTS patient cells have elevated levels of fragile telomeric ends and that RECQL4-depleted human cells accumulate fragile sites, sister chromosome exchanges, and double strand breaks at telomeric sites. Further, RECQL4 localizes to telomeres and associates with shelterin proteins TRF1 and TRF2. Using recombinant proteins we showed that RECQL4 resolves telomeric D-loop structures with the help of shelterin proteins TRF1, TRF2, and POT1. We also found a novel functional synergistic interaction of this protein with WRN during D-loop unwinding. These data implicate RECQL4 in telomere maintenance.


Assuntos
Proteínas Mutantes/metabolismo , Mutação , RecQ Helicases/metabolismo , Síndrome de Rothmund-Thomson/genética , Telômero/metabolismo , Afidicolina/farmacologia , Sequência de Bases , DNA/biossíntese , DNA/química , DNA/metabolismo , Replicação do DNA/efeitos dos fármacos , Exodesoxirribonucleases/metabolismo , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Dados de Sequência Molecular , Proteínas Mutantes/genética , Conformação de Ácido Nucleico/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , RNA Interferente Pequeno/genética , RecQ Helicases/deficiência , RecQ Helicases/genética , Síndrome de Rothmund-Thomson/metabolismo , Síndrome de Rothmund-Thomson/patologia , Telômero/efeitos dos fármacos , Telômero/genética , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53 , Helicase da Síndrome de Werner
8.
Cell Rep ; 42(5): 112505, 2023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-37182209

RESUMO

Genes that are key to cell identity are generally regulated by cell-type-specific enhancer elements bound by transcription factors, some of which facilitate looping to distant gene promoters. In contrast, genes that encode housekeeping functions, whose regulation is essential for normal cell metabolism and growth, generally lack interactions with distal enhancers. We find that Ronin (Thap11) assembles multiple promoters of housekeeping and metabolic genes to regulate gene expression. This behavior is analogous to how enhancers are brought together with promoters to regulate cell identity genes. Thus, Ronin-dependent promoter assemblies provide a mechanism to explain why housekeeping genes can forgo distal enhancer elements and why Ronin is important for cellular metabolism and growth control. We propose that clustering of regulatory elements is a mechanism common to cell identity and housekeeping genes but is accomplished by different factors binding distinct control elements to establish enhancer-promoter or promoter-promoter interactions, respectively.


Assuntos
Elementos Facilitadores Genéticos , Genes Essenciais , Genes Essenciais/genética , Elementos Facilitadores Genéticos/genética , Fatores de Transcrição/metabolismo , Regiões Promotoras Genéticas/genética
9.
Carcinogenesis ; 33(2): 442-51, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22114072

RESUMO

The role of dominant transforming p53 in carcinogenesis is poorly understood. Our previous data suggested that aberrant p53 proteins can enhance tumorigenesis and metastasis. Here, we examined potential mechanisms through which gain-of-function (GOF) p53 proteins can induce motility. Cells expressing GOF p53 -R175H, -R273H and -D281G showed enhanced migration, which was reversed by RNA interference (RNAi) or transactivation-deficient mutants. In cells with engineered or endogenous p53 mutants, enhanced migration was reduced by downregulation of nuclear factor-kappaB2, a GOF p53 target. We found that GOF p53 proteins upregulate CXC-chemokine expression, the inflammatory mediators that contribute to multiple aspects of tumorigenesis. Elevated expression of CXCL5, CXCL8 and CXCL12 was found in cells expressing oncogenic p53. Transcription was elevated as CXCL5 and CXCL8 promoter activity was higher in cells expressing GOF p53, whereas wild-type p53 repressed promoter activity. Chromatin immunoprecipitation assays revealed enhanced presence of acetylated histone H3 on the CXCL5 promoter in H1299/R273H cells, in agreement with increased transcriptional activity of the promoter, whereas RNAi-mediated repression of CXCL5 inhibited cell migration. Consistent with this, knockdown of the endogenous mutant p53 in lung cancer or melanoma cells reduced CXCL5 expression and cell migration. Furthermore, short hairpin RNA knockdown of mutant p53 in MDA-MB-231 cells reduced expression of a number of key targets, including several chemokines and other inflammatory mediators. Finally, CXCL5 expression was also elevated in lung tumor samples containing GOF p53, indicating relevance to human cancer. The data suggest a mechanistic link between GOF p53 proteins and chemokines in enhanced cell motility.


Assuntos
Movimento Celular/genética , Quimiocinas CXC/genética , Mutação , Proteína Supressora de Tumor p53/genética , Acetilação , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Quimiocinas CXC/metabolismo , Imunoprecipitação da Cromatina/métodos , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes/métodos , Histonas/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Melanoma/genética , Melanoma/metabolismo , Subunidade p52 de NF-kappa B/genética , Subunidade p52 de NF-kappa B/metabolismo , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno/genética , Ativação Transcricional , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima
10.
Blood Adv ; 6(23): 6016-6022, 2022 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-35667093

RESUMO

The fetal-to-adult hemoglobin switching at about the time of birth involves a shift in expression from γ-globin to ß-globin in erythroid cells. Effective re-expression of fetal γ-globin can ameliorate sickle cell anemia and ß-thalassemia. Despite the physiological and clinical relevance of this switch, its posttranscriptional regulation is poorly understood. Here, we identify Pumilo 1 (PUM1), an RNA-binding protein with no previously reported functions in erythropoiesis, as a direct posttranscriptional regulator of ß-globin switching. PUM1, whose expression is regulated by the erythroid master transcription factor erythroid Krüppel-like factor (EKLF/KLF1), peaks during erythroid differentiation, binds γ-globin messenger RNA (mRNA), and reduces γ-globin (HBG1) mRNA stability and translational efficiency, which culminates in reduced γ-globin protein levels. Knockdown of PUM1 leads to a robust increase in fetal hemoglobin (∼22% HbF) without affecting ß-globin levels in human erythroid cells. Importantly, targeting PUM1 does not limit the progression of erythropoiesis, which provides a potentially safe and effective treatment strategy for sickle cell anemia and ß-thalassemia. In support of this idea, we report elevated levels of HbF in the absence of anemia in an individual with a novel heterozygous PUM1 mutation in the RNA-binding domain (p.(His1090Profs∗16); c.3267_3270delTCAC), which suggests that PUM1-mediated posttranscriptional regulation is a critical player during human hemoglobin switching.


Assuntos
Anemia Falciforme , Talassemia beta , Adulto , Humanos , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , gama-Globinas/genética , gama-Globinas/metabolismo , Talassemia beta/genética , Globinas beta/genética , Proteínas de Transporte , Anemia Falciforme/genética , Proteínas de Ligação a RNA/genética
11.
Stem Cell Res ; 41: 101617, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31731178

RESUMO

Parkinson's disease is associated with the loss of dopaminergic neurons in the midbrain. Clinical studies investigating replacement of these neurons with in vitro-generated neurons are currently underway. However, this approach has been limited by difficulties in scaling up on-demand production of midbrain dopaminergic (mDA) neurons from pluripotent stem cells. Cryo-preservation may offer a solution, as it allows for banking of quality controlled mDA neurons. In this study, we tested different freezing conditions and found that optimal cryopreservation of immature human mDA neurons at an early differentiation time point was achieved in STEM-CELLBANKER medium using a controlled freezing program.


Assuntos
Diferenciação Celular , Criopreservação , Neurônios Dopaminérgicos , Células-Tronco Pluripotentes Induzidas , Mesencéfalo , Doença de Parkinson Secundária , Animais , Linhagem Celular , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Neurônios Dopaminérgicos/transplante , Xenoenxertos , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Mesencéfalo/metabolismo , Mesencéfalo/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Oxidopamina/farmacologia , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/metabolismo , Doença de Parkinson Secundária/patologia , Doença de Parkinson Secundária/terapia
12.
Cancer Cell ; 28(3): 357-69, 2015 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-26373281

RESUMO

The chromatin-remodeler ATRX is frequently lost in cancer cells that use ALT (alternative lengthening of telomeres) for telomere maintenance, but its function in telomere recombination is unknown. Here we show that loss of ATRX suppresses the timely resolution of sister telomere cohesion that normally occurs prior to mitosis. In the absence of ATRX, the histone variant macroH2A1.1 binds to the poly(ADP-ribose) polymerase tankyrase 1, preventing it from localizing to telomeres and resolving cohesion. The resulting persistent telomere cohesion promotes recombination between sister telomeres, while it suppresses inappropriate recombination between non-sisters. Forced resolution of sister telomere cohesion induces excessive recombination between non-homologs, genomic instability, and impaired cell growth, indicating the ATRX-macroH2A1.1-tankyrase axis as a potential therapeutic target in ALT tumors.


Assuntos
DNA Helicases/genética , Proteínas Nucleares/genética , Recombinação Genética/genética , Telômero/genética , Linhagem Celular , Linhagem Celular Tumoral , Montagem e Desmontagem da Cromatina/genética , Instabilidade Genômica/genética , Células HEK293 , Células HeLa , Histonas/genética , Humanos , Poli(ADP-Ribose) Polimerases/genética , Telomerase/genética , Homeostase do Telômero/genética , Proteína Nuclear Ligada ao X
13.
Methods Mol Biol ; 962: 127-33, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23150442

RESUMO

Chemoresistance and increased growth rate are two gain-of-function functions that mutant p53 is thought to possess. Here, we describe two methods for measuring the sensitiveness of cells to chemotherapeutic drugs and the rate of cell growth. Both of which can be used with a wide range of cell types. The clonogenic assay can be used with many different chemotoxic drugs and the growth assay described here presents an alternative to the MTT assay and allows for a long-term measurement of cell growth. These protocols are both easy, flexible, require relatively little effort, and are inexpensive to carry out.


Assuntos
Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Proteína Supressora de Tumor p53/metabolismo , Antineoplásicos/farmacologia , Apoptose , Ciclo Celular/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias/métodos , Humanos , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética
14.
Free Radic Biol Med ; 53(6): 1371-80, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22885031

RESUMO

Alzheimer disease (AD) is a major health problem in the United States, affecting one in eight Americans over the age of 65. The number of elderly suffering from AD is expected to continue to increase over the next decade, as the average age of the U.S. population increases. The risk factors for and etiology of AD are not well understood; however, recent studies suggest that exposure to oxidative stress may be a contributing factor. Here, microarray gene expression signatures were compared in AD-patient-derived fibroblasts and normal fibroblasts exposed to hydrogen peroxide or menadione (to simulate conditions of oxidative stress). Using the 23K Illumina cDNA microarray to screen expression of >14,000 human genes, we identified a total of 1017 genes that are chronically up- or downregulated in AD fibroblasts, 215 of which were also differentially expressed in normal human fibroblasts within 12h after exposure to hydrogen peroxide or menadione. Pathway analysis of these 215 genes and their associated pathways revealed cellular functions that may be critically dysregulated by oxidative stress and play a critical role in the etiology and/or pathology of AD. We then examined the AD fibroblasts for the presence of oxidative DNA damage and found increased accumulation of 8-oxo-guanine. These results indicate the possible role of oxidative stress in the gene expression profile seen in AD.


Assuntos
Doença de Alzheimer/patologia , Fibroblastos/metabolismo , Estresse Oxidativo , Fenótipo , Transcriptoma , Doença de Alzheimer/metabolismo , Estudos de Casos e Controles , Células Cultivadas , Dano ao DNA , Fibroblastos/efeitos dos fármacos , Regulação da Expressão Gênica , Humanos , Peróxido de Hidrogênio/farmacologia , Redes e Vias Metabólicas/genética , Análise de Sequência com Séries de Oligonucleotídeos , Oxidantes/farmacologia , Transcrição Gênica , Vitamina K 3/farmacologia
15.
DNA Repair (Amst) ; 11(7): 624-35, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22633600

RESUMO

RECQL5 is one of the five human RecQ helicases, involved in the maintenance of genomic integrity. While much insight has been gained into the function of the Werner (WRN) and Bloom syndrome proteins (BLM), little is known about RECQL5. We have analyzed the recruitment and retention dynamics of RECQL5 at laser-induced DNA double strand breaks (DSBs) relative to other human RecQ helicases. RECQL5-depleted cells accumulate persistent 53BP1 foci followed by γ-irradiation, indicating a potential role of RECQL5 in the processing of DSBs. Real time imaging of live cells using confocal laser microscopy shows that RECQL5 is recruited early to laser-induced DSBs and remains for a shorter duration than BLM and WRN, but persist longer than RECQL4. These studies illustrate the differential involvement of RecQ helicases in the DSB repair process. Mapping of domains within RECQL5 that are necessary for recruitment to DSBs revealed that both the helicase and KIX domains are required for DNA damage recognition and stable association of RECQL5 to the DSB sites. Previous studies have shown that MRE11 is essential for the recruitment of RECQL5 to the DSB sites. Here we show that the recruitment of RECQL5 does not depend on the exonuclease activity of MRE11 or on active transcription by RNA polymerase II, one of the prominent interacting partners of RECQL5. Also, the recruitment of RECQL5 to laser-induced damage sites is independent of the presence of other DNA damage signaling and repair proteins BLM, WRN and ATM.


Assuntos
Quebras de DNA de Cadeia Dupla , RecQ Helicases/metabolismo , Linhagem Celular , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática , Exodesoxirribonucleases/metabolismo , Raios gama/efeitos adversos , Inativação Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Cinética , Lasers/efeitos adversos , Proteína Homóloga a MRE11 , Domínios e Motivos de Interação entre Proteínas/fisiologia , RNA Polimerase II/metabolismo , RecQ Helicases/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53 , Helicase da Síndrome de Werner
16.
Mol Biol Cell ; 23(21): 4273-85, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22973052

RESUMO

Human RECQL5 is a member of the RecQ helicase family, which maintains genome stability via participation in many DNA metabolic processes, including DNA repair. Human cells lacking RECQL5 display chromosomal instability. We find that cells depleted of RECQL5 are sensitive to oxidative stress, accumulate endogenous DNA damage, and increase the cellular poly(ADP-ribosyl)ate response. In contrast to the RECQ helicase family members WRN, BLM, and RECQL4, RECQL5 accumulates at laser-induced single-strand breaks in normal human cells. RECQL5 depletion affects the levels of PARP-1 and XRCC1, and our collective results suggest that RECQL5 modulates and/or directly participates in base excision repair of endogenous DNA damage, thereby promoting chromosome stability in normal human cells.


Assuntos
Dano ao DNA , RecQ Helicases/metabolismo , Dano ao DNA/genética , Reparo do DNA/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo/genética , Técnicas de Silenciamento de Genes , Proteínas de Fluorescência Verde/metabolismo , Células HCT116 , Células HeLa , Humanos , Lasers , Modelos Biológicos , Oxirredução , Estresse Oxidativo/genética , Poli Adenosina Difosfato Ribose/metabolismo , RecQ Helicases/deficiência , Proteínas Recombinantes de Fusão/metabolismo , Proteína 1 Complementadora Cruzada de Reparo de Raio-X
17.
Aging Cell ; 11(3): 456-66, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22296597

RESUMO

RECQL4 is associated with Rothmund-Thomson Syndrome (RTS), a rare autosomal recessive disorder characterized by premature aging, genomic instability, and cancer predisposition. RECQL4 is a member of the RecQ helicase family, and has many similarities to WRN protein, which is also implicated in premature aging. There is no information about whether any of the RecQ helicases play roles in mitochondrial biogenesis, which is strongly implicated in the aging process. Here, we used microscopy to visualize RECQL4 in mitochondria. Fractionation of human and mouse cells also showed that RECQL4 was present in mitochondria. Q-PCR amplification of mitochondrial DNA demonstrated that mtDNA damage accumulated in RECQL4-deficient cells. Microarray analysis suggested that mitochondrial bioenergetic pathways might be affected in RTS. Measurements of mitochondrial bioenergetics showed a reduction in the mitochondrial reserve capacity after lentiviral knockdown of RECQL4 in two different primary cell lines. Additionally, biochemical assays with RECQL4, mitochondrial transcription factor A, and mitochondrial DNA polymerase γ showed that the polymerase inhibited RECQL4's helicase activity. RECQL4 is the first 3'-5' RecQ helicase to be found in both human and mouse mitochondria, and the loss of RECQL4 alters mitochondrial integrity.


Assuntos
DNA Mitocondrial/genética , Mitocôndrias/genética , Mitocôndrias/metabolismo , RecQ Helicases/genética , RecQ Helicases/metabolismo , Fatores Etários , Idoso de 80 Anos ou mais , Animais , Fracionamento Celular/métodos , Linhagem Celular Tumoral , Dano ao DNA , Instabilidade Genômica , Células HeLa , Humanos , Camundongos
18.
J Exp Med ; 209(4): 855-69, 2012 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-22473955

RESUMO

Cockayne syndrome (CS) is a devastating autosomal recessive disease characterized by neurodegeneration, cachexia, and accelerated aging. 80% of the cases are caused by mutations in the CS complementation group B (CSB) gene known to be involved in DNA repair and transcription. Recent evidence indicates that CSB is present in mitochondria, where it associates with mitochondrial DNA (mtDNA). We report an increase in metabolism in the CSB(m/m) mouse model and CSB-deficient cells. Mitochondrial content is increased in CSB-deficient cells, whereas autophagy is down-regulated, presumably as a result of defects in the recruitment of P62 and mitochondrial ubiquitination. CSB-deficient cells show increased free radical production and an accumulation of damaged mitochondria. Accordingly, treatment with the autophagic stimulators lithium chloride or rapamycin reverses the bioenergetic phenotype of CSB-deficient cells. Our data imply that CSB acts as an mtDNA damage sensor, inducing mitochondrial autophagy in response to stress, and that pharmacological modulators of autophagy are potential treatment options for this accelerated aging phenotype.


Assuntos
Autofagia , DNA Helicases/fisiologia , Enzimas Reparadoras do DNA/fisiologia , Mitocôndrias/metabolismo , Animais , Cálcio/metabolismo , Células Cultivadas , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Transporte da Membrana Mitocondrial , Poro de Transição de Permeabilidade Mitocondrial , Proteínas de Ligação a Poli-ADP-Ribose , Espécies Reativas de Oxigênio/metabolismo
19.
Genes Cancer ; 2(10): 943-55, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22701761

RESUMO

The current model predicts that MDM2 is primarily overexpressed in cancers with wild-type (WT) p53 and contributes to oncogenesis by degrading p53. Following a correlated expression of MDM2 and NF-κB2 transcripts in human lung tumors, we have identified a novel transactivation function of MDM2. Here, we report that in human lung tumors, overexpression of MDM2 was found in approximately 30% of cases irrespective of their p53 status, and expression of MDM2 and NF-κB2 transcripts showed a highly significant statistical correlation in tumors with WT p53. We investigated the significance of this correlated expression in terms of mechanism and biological function. Increase in MDM2 expression from its own promoter in transgenic mice remarkably enhanced expression of NF-κB2 compared with its non-transgenic littermates. Knockdown or elimination of endogenous MDM2 expression in cultured non-transformed or lung tumor cells drastically reduced expression of NF-κB2 transcripts, suggesting a normal physiological role of MDM2 in regulating NF-κB2 transcription. MDM2 could up-regulate expression of NF-κB2 transcripts when its p53-interaction domain was blocked with Nutlin-3, indicating that the MDM2-p53 interaction is dispensable for up-regulation of NF-κB2 expression. Consistently, analysis of functional domains of MDM2 indicated that although the p53-interaction domain of MDM2 contributes to the up-regulation of the NFκB2 promoter, MDM2 does not require direct interactions with p53 for this function. Accordingly, MDM2 overexpression in non-transformed or lung cancer cells devoid of p53 also generated a significant increase in the expression of NF-κB2 transcript and its targets CXCL-1 and CXCL-10, whereas elimination of MDM2 expression had the opposite effects. MDM2-mediated increase in p100/NF-κB2 expression reduced cell death mediated by paclitaxel. Furthermore, knockdown of NF-κB2 expression retarded cell proliferation. Based on these data, we propose that MDM2-mediated NF-κB2 up-regulation is a combined effect of p53-dependent and independent mechanisms and that it confers a survival advantage to lung cancer cells.

20.
Mol Cancer Res ; 7(8): 1253-67, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19671680

RESUMO

Overexpression of MDM2 has been related to oncogenesis. In this communication, we present evidence to show that MDM2 controls the cell cycle-dependent expression of cyclin A by using a pathway that ensures its timely expression. MDM2 does not inhibit cyclin D or E expression. Silencing of endogenous MDM2 expression elevates cyclin A expression. The p53-binding domain of MDM2 harbors a SWIB region homologous to a conserved domain of a chromosome remodeling factor BRG1-associated protein. The SWIB domain of MDM2 inhibits cyclin A expression in a p53- and BRG1-dependent fashion, suggesting that MDM2 interferes with p53 binding of the BRG1 complex freeing it to repress cyclin A expression. Silencing of cyclin-dependent kinase (cdk) inhibitor p16 prevents MDM2-mediated inhibition of cyclin A expression, implicating its role in the process. MDM2-mediated repression of cyclin A expression induces G(1)-S arrest, which can be rescued by ectopic expression of cyclin A. Cancer cells lacking p53, p16, or BRG1 escape MDM2-mediated repression of cyclin A expression and growth arrest. Our data propose a novel mechanism by which MDM2 controls the cell cycle in normal cells and how cancer cells may escape this important safety barrier.


Assuntos
Ciclo Celular , Ciclina A/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Linhagem Celular Transformada , Linhagem Celular Tumoral , Ciclina E/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , DNA Helicases/metabolismo , Regulação para Baixo , Citometria de Fluxo , Fase G1 , Humanos , Microscopia Confocal , Modelos Biológicos , Proteínas Nucleares/metabolismo , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-mdm2/química , Fatores de Tempo , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA