Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
iScience ; 26(11): 108262, 2023 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-38026177

RESUMO

Hunner-type interstitial cystitis (HIC) is a rare, enigmatic inflammatory disease of the urinary bladder with no curative treatments. In this study, we aimed to characterize the unique cellular and immunological factors specifically involved in HIC by comparing with cystitis induced by Mycobacterium bovis bacillus Calmette-Guérin, which presents similar clinicopathological features to HIC. Here, we show that T helper 1/17 +polarized immune responses accompanied by prominent overexpression of interferon (IFN)-γ, enhanced cGAS-STING cytosolic DNA sensing pathway, and increased plasma cell infiltration are the characteristic inflammatory features in HIC bladder. Further, we developed a mouse anti-IFN-γ DNA aptamer and observed that the intravesical instillation of the aptamer significantly ameliorated bladder inflammation, pelvic pain and voiding dysfunction in a recently developed murine HIC model with little migration into the blood. Our study provides the plausible basis for the clinical translation of the anti-IFN-γ DNA aptamer in the treatment of human HIC.

2.
Mol Ther Nucleic Acids ; 14: 158-170, 2019 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-30594072

RESUMO

The potential of genetic alphabet expansion technologies using artificial extra base pairs (unnatural base pairs) has been rapidly expanding and increasing. We present that the hydrophobic unnatural base, 7-(2-thienyl)imidazo[4,5-b]pyridine (Ds), which acts as a fifth letter in a DNA library, provides a series of high-affinity DNA aptamers with versatile binding specificities and activities to cancer cells. These Ds-containing DNA aptamers were generated by a method called cell-ExSELEX to target three breast cancer cell lines: MCF7, MDA-MB-231, and T-47D. Aptamer 14A-MCF7, which targets MCF7 cells, specifically binds to MCF7 cells, but not other cancer cell lines. Aptamer 07-MB231, which targets MDA-MB-231 cells, binds to a series of metastatic bone and lung cancer cell lines. Aptamer 05-MB231 targets MDA-MB-231 cells, but it also binds to all of the cancer and leukemia cell lines that we examined. None of these aptamers bind to normal cell lines, such as MCF10A and HUVEC. In addition, aptamers 14A-MCF7 and 05-MB231 are internalized within the cancer cells, and aptamer 05-MB231 possesses anti-proliferative properties against most cancer cell lines that we examined. These aptamers and the generation method are broadly applicable to cancer cell imaging, biomarker discovery, cancer cell profiling, anti-cancer therapies, and drug delivery systems.

3.
Front Genet ; 5: 441, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25620975

RESUMO

RECQL1 and WRN helicases in the human RecQ helicase family participate in maintaining genome stability, DNA repair, replication, and recombination pathways in the cell cycle. They are expressed highly in rapidly proliferating cells and tumor cells, suggesting that they have important roles in the replication of a genome. Although mice deficient in these helicases are indistinguishable from wild-type mice, their embryonic fibroblasts are sensitive to DNA damage. In tumor cells, silencing the expression of RECQL1 or WRN helicase by RNA interference induces mitotic catastrophe that eventually kills tumor cells at the mitosis stage of the cell cycle. By contrast, the same gene silencing by cognate small RNA (siRNA) never kills normal cells, although cell growth is slightly delayed. These findings indicate that RECQL1 and WRN helicases are ideal molecular targets for cancer therapy. The molecular mechanisms underlying these events has been studied extensively, which may help development of anticancer drugs free from adverse effects by targeting DNA repair helicases RECQL1 and WRN. As expected, the anticancer activity of conventional genotoxic drugs is significantly augmented by combined treatment with RECQL1- or WRN-siRNAs that prevents DNA repair in cancer cells. In this review, we focus on studies that clarified the mechanisms that lead to the specific killing of cancer cells and introduce efforts to develop anticancer RecQ-siRNA drugs free from adverse effects.

4.
PLoS One ; 8(8): e72820, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23951333

RESUMO

OBJECTIVE: This study analyzed the clinicopathological correlation between ovarian cancer (OC) and RECQL1 DNA helicase to assess its therapeutic potential. METHODS: Surgically resected OC from 118 retrospective cases, for which paraffin blocks and all clinical data were complete, were used in this study. RECQL1 and Ki-67 immunostaining were performed on sections to correlate RECQL1 staining with subtype and patient survival. Ten OC and two normal cell lines were then examined for RECQL1 expression and were treated with siRNA against RECQL1 to assess its effect on cell proliferation. RESULTS: Of the 118 cases of adenocarcinoma (50, serous; 26, endometrioid; 21, clear cell; 15, mucinous; 6, other histology), 104 (90%) showed varying levels of RECQL1 expression in the nuclei of OC cells. The Cox hazards model confirmed that diffuse and strong staining of RECQL1 was correlated with histological type. However, RECQL1 expression did not correlate with overall patient survival or FIGO stage. In vitro, RECQL1 expression was exceptionally high in rapidly growing OC cell lines, as compared with normal cells. Using a time-course analysis of RECQL1-siRNA transfection, we observed a significant inhibition in cell proliferation. CONCLUSIONS: RECQL1 DNA helicase is a marker of highly proliferative cells. RECQL1-siRNA may offer a new therapeutic strategy against various subtypes of OC, including platinum-resistant cancers, or in recurrent cancers that gain platinum resistance.


Assuntos
Adenocarcinoma de Células Claras/genética , Adenocarcinoma Mucinoso/genética , Biomarcadores Tumorais/genética , Cistadenocarcinoma Seroso/genética , Reparo do DNA , Recidiva Local de Neoplasia/genética , Neoplasias Ovarianas/genética , RecQ Helicases/genética , Adenocarcinoma de Células Claras/enzimologia , Adenocarcinoma de Células Claras/mortalidade , Adenocarcinoma de Células Claras/patologia , Adenocarcinoma Mucinoso/enzimologia , Adenocarcinoma Mucinoso/mortalidade , Adenocarcinoma Mucinoso/patologia , Biomarcadores Tumorais/antagonistas & inibidores , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Cistadenocarcinoma Seroso/enzimologia , Cistadenocarcinoma Seroso/mortalidade , Cistadenocarcinoma Seroso/patologia , Feminino , Expressão Gênica , Humanos , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/enzimologia , Recidiva Local de Neoplasia/mortalidade , Recidiva Local de Neoplasia/patologia , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/mortalidade , Neoplasias Ovarianas/patologia , Prognóstico , Modelos de Riscos Proporcionais , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , RecQ Helicases/antagonistas & inibidores , RecQ Helicases/metabolismo , Estudos Retrospectivos , Análise de Sobrevida
5.
J Cell Sci ; 125(Pt 10): 2509-22, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22357944

RESUMO

Mutations in RECQL4 helicase are associated with Rothmund-Thomson syndrome (RTS). A subset of RTS patients is predisposed to cancer and is sensitive to DNA damaging agents. The enhanced sensitivity of cells from RTS patients correlates with the accumulation of transcriptionally active nuclear p53. We found that in untreated normal human cells these two nuclear proteins, p53 and RECQL4, instead colocalize in the mitochondrial nucleoids. RECQL4 accumulates in mitochondria in all phases of the cell cycle except S phase and physically interacts with p53 only in the absence of DNA damage. p53-RECQL4 binding leads to the masking of the nuclear localization signal of p53. The N-terminal 84 amino acids of RECQL4 contain a mitochondrial localization signal, which causes the localization of RECQL4-p53 complex to the mitochondria. RECQL4-p53 interaction is disrupted after stress, allowing p53 translocation to the nucleus. In untreated normal cells RECQL4 optimizes de novo replication of mtDNA, which is consequently decreased in fibroblasts from RTS patients. Wild-type RECQL4-complemented RTS cells show relocalization of both RECQL4 and p53 to the mitochondria, loss of p53 activation, restoration of de novo mtDNA replication and resistance to different types of DNA damage. In cells expressing Δ84 RECQL4, which cannot translocate to mitochondria, all the above functions are compromised. The recruitment of p53 to the sites of de novo mtDNA replication is also regulated by RECQL4. Thus these findings elucidate the mechanism by which p53 is regulated by RECQL4 in unstressed normal cells and also delineates the mitochondrial functions of the helicase.


Assuntos
Mitocôndrias/metabolismo , RecQ Helicases/metabolismo , Síndrome de Rothmund-Thomson/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular , DNA Helicases/análise , Humanos , Mitocôndrias/enzimologia , Transporte Proteico , RecQ Helicases/genética , Síndrome de Rothmund-Thomson/enzimologia , Síndrome de Rothmund-Thomson/genética , Estresse Fisiológico , Proteína Supressora de Tumor p53/genética
6.
Geriatr Gerontol Int ; 12(1): 140-6, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22188495

RESUMO

Werner syndrome (WS) is an autosomal recessive progeroid disorder caused by mutations in the WRN DNA helicase. It is characterized by the graying and loss of hair, juvenile cataracts, sclerosis and ulceration of skin, insulin-resistant diabetes mellitus, dyslipidemia, abdominal adiposity, osteoporosis, atherosclerosis, and malignant neoplasm. Patients are usually diagnosed in their 30s or 40s, but the early pathophysiology of the syndrome is still not fully understood. Here we report a 29-year-old female patient who displayed cataracts, hair graying, and tendinous calcinosis. Her parents were first cousins. Interestingly, the patient lacked the metabolic signs typical for WS, including glucose intolerance, dyslipidemia, and visceral fat accumulation. A hyperinsulinemic response at 30 min was observed in an oral glucose tolerance test. Mutational analysis for the WRN gene revealed a homozygous nucleotide substitution 3190C>T in exon 24, resulting in a protein product with replacement of an arginine residue at position 573 by termination codon (Arg987Ter). The mutated WRN protein was unable to translocate into the nucleus in an in vitro cell assay. A WS patient with an Arg987Ter mutation has been previously reported in Switzerland, the present case is the first to be identified in Asia. This case demonstrates the early clinical features of WS and suggests that metabolic abnormality, including insulin resistance, is not an essential component of WS at disease onset. Moreover, a follow-up study of such case would be useful to understand how the various clinical symptoms in WS develop and progress over the years.


Assuntos
Resistência à Insulina/fisiologia , Síndrome de Werner/fisiopatologia , Adulto , Análise Mutacional de DNA , Exodesoxirribonucleases/genética , Exodesoxirribonucleases/metabolismo , Feminino , Seguimentos , Humanos , Mutação , RecQ Helicases/genética , RecQ Helicases/metabolismo , Síndrome de Werner/genética , Síndrome de Werner/metabolismo , Helicase da Síndrome de Werner
7.
Cancer Res ; 71(13): 4598-607, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21571861

RESUMO

RECQL1 and WRN proteins are RecQ DNA helicases that participate in suppression of DNA hyper-recombination and repair. In this study, we report evidence supporting their candidacy as cancer therapeutic targets. In hypopharyngeal carcinomas, which have the worst prognosis among head and neck squamous cell carcinomas (HNSCC) that are rapidly rising in incidence, we found that RECQL1 and WRN proteins are highly expressed and that siRNA-mediated silencing of either gene suppressed carcinoma cell growth in vitro. Similarly, siRNA administration in a murine xenograft model of hypopharyngeal carcinoma markedly inhibited tumor growth. Moreover, combining either siRNA with cis-platinum (II) diammine dichloride significantly augmented the in vivo anticancer effects of this drug that is used commonly in HNSCC treatment. Notably, we observed no recurrence of some tumors following siRNA treatment in this model. Our findings offer a preclinical proof of concept for RECQL1 and WRN proteins as novel therapeutic targets to treat aggressive HNSCC and perhaps other cancers.


Assuntos
Carcinoma/enzimologia , Carcinoma/terapia , Exodesoxirribonucleases/antagonistas & inibidores , Neoplasias de Cabeça e Pescoço/enzimologia , Neoplasias de Cabeça e Pescoço/terapia , Neoplasias Hipofaríngeas/enzimologia , Neoplasias Hipofaríngeas/terapia , Terapia de Alvo Molecular/métodos , Neoplasias de Células Escamosas/enzimologia , Neoplasias de Células Escamosas/terapia , RecQ Helicases/antagonistas & inibidores , Animais , Carcinoma/tratamento farmacológico , Carcinoma/genética , Carcinoma de Células Escamosas , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Linhagem Celular Tumoral , Cisplatino/farmacologia , Terapia Combinada , Exodesoxirribonucleases/biossíntese , Exodesoxirribonucleases/genética , Inativação Gênica , Células HeLa , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/genética , Humanos , Neoplasias Hipofaríngeas/tratamento farmacológico , Neoplasias Hipofaríngeas/genética , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias de Células Escamosas/tratamento farmacológico , Neoplasias de Células Escamosas/genética , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Distribuição Aleatória , RecQ Helicases/biossíntese , RecQ Helicases/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço , Helicase da Síndrome de Werner , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Int J Mol Med ; 25(4): 537-45, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20198302

RESUMO

RecQL1 in the human RecQ DNA helicase family participates in DNA repair and recombination pathways in cell cycle replication. Immunohistochemical analysis of human hepatocellular carcinoma (HCC) tissues showed that RecQL1 expression is strongly correlated with histological grade and MIB-1 indices of HCC, and that the expression was greater in simple HCCs inducing extranodular growth or portal vein invasion than in HCCs not inducing extranodular growth or portal vein invasion. These histological data reveal the potential of RecQL1 as a biological marker predicting the malignancy and progression of liver cancer. High expression profiles were also produced by various HCC cells, including HCC cell lines established by us. When RecQL1 expression was silenced by siRNA in vitro, most HCC cells died of mitotic catastrophe. In a mouse orthotopic xenograft model of liver cancer with transplanted human HCC, RecQL1-siRNA mixed with cationic liposomes exhibited a strong anticancer effect that prevented the growth of the cancer. RecQL1-siRNA inhibited the growth of human HCC in the mouse liver, confirming that RecQL1 is an excellent molecular agent against liver cancer and suggests that RecQL1-siRNA formulated with liver-prone liposomes has excellent potential as a therapeutic drug against liver cancers.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/enzimologia , Carcinoma Hepatocelular/terapia , Reparo do DNA , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/terapia , RecQ Helicases/metabolismo , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Lipossomos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Camundongos , Mitose , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Nucleic Acids Res ; 37(7): e56, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19282452

RESUMO

Small interfering RNA (siRNA) has excellent pharmacological features and is expected to be used for therapeutic drug development. To this end, however, new RNA technology needs to be established so that extremely small amounts (less than 1 pmol) of siRNA can be detected in organs of experimental animals and in human blood to facilitate pharmacokinetics studies. An important feature is that this new technology is not dependent on radioisotopes and can detect siRNA molecules identical to those used for drug development in preclinical tests with experimental animals or in clinical tests with humans. We report a convenient method that can detect small amounts of siRNA. The method uses high-power confocal microscopic analysis of fluorescence polarization in DNA probes that are bound to one of the strands of siRNA and directly quantitates the copy number of siRNA molecule after extraction from specimens. A pharmacokinetic study to examine the blood retention time of siRNA/cationic liposomes in mice showed that this straightforward method is consistent with the other reverse transcriptase polymerase chain reaction amplification-based method. We believe that the entire process is simple and applicable for a high-throughput analysis, which provides excellent technical support for fundamental research on RNA interference and development of siRNA drugs.


Assuntos
Polarização de Fluorescência/métodos , RNA Interferente Pequeno/análise , Animais , Sequência de Bases , Linhagem Celular Tumoral , Sondas de DNA/química , Humanos , Injeções Intravenosas , Camundongos , Microscopia Confocal , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/sangue , Distribuição Tecidual , Transfecção
10.
Cancer Sci ; 99(6): 1227-36, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18422747

RESUMO

Small interfering RNAs (siRNAs) are expected to have a medical application in human therapy as drugs with a high specificity for their molecular target mRNAs. RecQL1 DNA helicase in the human RecQ helicase family participates in DNA repair and recombination pathways in the cell cycle of replication. Silencing the RecQL1 expression by RecQL1-siRNA induces mitotic death in vitro specifically in growing cancer cells. By contrast, the same RecQL1 silencing does not affect the growth of normal cells, emphasizing that RecQL1 helicase is an ideal molecular target for cancer therapy. In this study, we show that local and systemic administration of RecQL1-siRNA mixed with polyethyleneimine polymer or cationic liposomes prevented cancer cell proliferation in vivo in mouse models of cancer without noticeable adverse effects. The results indicate that RecQL1-siRNA in a complex with a cationic polymer is a very promising anticancer drug candidate, and that in particular, RecQL1-siRNA formulated with a cationic liposome has an enormous potential to be used by intravenous injection for therapy specific for liver cancers, including metastasized cancers from the colon and pancreas.


Assuntos
Neoplasias Experimentais/tratamento farmacológico , RNA Interferente Pequeno/farmacologia , RecQ Helicases/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Carcinoma Hepatocelular/enzimologia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/terapia , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/terapia , Citometria de Fluxo , Humanos , Técnicas Imunoenzimáticas , Lipossomos , Neoplasias Hepáticas Experimentais/genética , Neoplasias Hepáticas Experimentais/secundário , Neoplasias Hepáticas Experimentais/terapia , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Experimentais/enzimologia , Neoplasias Experimentais/genética , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Polietilenoimina/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RecQ Helicases/antagonistas & inibidores , RecQ Helicases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Cancer Sci ; 99(5): 843-8, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18312465

RESUMO

Werner syndrome (WS) is an autosomal recessive genetic disorder causing premature aging, and WRN has been identified as the causative gene of WS. The product of the WRN gene (WRN) acts as a DNA helicase with exonuclease activity, and data have accumulated showing that the WRN gene strongly participates in carcinogenesis: (1) the normal WRN gene likely participates in the immortalization of B-lymphoblastoid cell lines through telomeric crisis caused by telomere shortening, (2) a much higher incidence of rare cancers occurs in WS patients than in other kinds of patients, and (3) levels of WRN expressed in virus-transformed cells and cancer cells are usually markedly up-regulated and are inversely correlated with the sensitivity of these cells against various genotoxins, including camptothecin. In this paper, we review the events that show a close correlation of the WRN gene and WRN with carcinogenesis and their underlying molecular mechanisms.


Assuntos
DNA Helicases/metabolismo , Exodesoxirribonucleases/metabolismo , Mutagênicos/toxicidade , Neoplasias/genética , RecQ Helicases/metabolismo , Transformação Celular Neoplásica , Instabilidade Cromossômica , DNA Helicases/genética , Reparo do DNA , Exodesoxirribonucleases/genética , Humanos , Modelos Biológicos , Mutação , Neoplasias/enzimologia , RecQ Helicases/genética , Telômero/metabolismo , Síndrome de Werner/genética , Síndrome de Werner/metabolismo , Helicase da Síndrome de Werner
12.
Cancer Sci ; 99(1): 71-80, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17953710

RESUMO

RecQL1 DNA helicase of the human RecQ helicase family participates in DNA repair and recombination pathways during cell-cycle replication. When we examined the effect of RecQL1 suppression on cell growth, we found that RecQL1 silencing by small interference RNA efficiently prevented proliferation of a wide range of cancer cells by inducing mitotic catastrophe and mitotic cell death. In contrast, such mitotic cell death was not seen in the growing normal fibroblasts used as controls, even if RecQL1 expression was fully downregulated. Our results support the hypothesis that endogenous DNA damage that occurs during DNA replication and remains unrepaired in cancer cells due to RecQL1 silencing induces cancer cell-specific mitotic catastrophe through a less-strict checkpoint in cancer cells than in normal cells. We speculate that normal cells are exempt from such mitotic cell death, despite slow growth, because cell-cycle progression is controlled strictly by a strong checkpoint system that detects DNA damage and arrests progression of the cell cycle until DNA damage is repaired completely. These results suggest that RecQL1 helicase is an excellent molecular target for cancer chemotherapy.


Assuntos
Neoplasias/enzimologia , Neoplasias/genética , RNA Interferente Pequeno/genética , RecQ Helicases/genética , Morte Celular/genética , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Dano ao DNA , Reparo do DNA , Regulação para Baixo , Inativação Gênica , Células HCT116 , Células HeLa , Humanos , Microscopia de Fluorescência , Mitose/genética , Neoplasias/patologia , RecQ Helicases/antagonistas & inibidores , RecQ Helicases/biossíntese
13.
Biol Pharm Bull ; 30(10): 1958-61, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17917271

RESUMO

Werner syndrome helicase (WRN) participates in a wide range of DNA activities, including replication, double-strand DNA break repair, telomere and retrovirus long terminal repeat maintenance. Mutations of the WRN gene cause Werner syndrome (WS), an autosomal recessive premature ageing disorder associated with various symptoms related to ageing. In this study, we investigated the siRNA that specifically down-regulates WRN expression. WRN silencing increased markedly the chemotherapeutic activity of camptothecin (CPT) on cancer cells in terms of the extent of efficacy and lowering effective drug dosage, accompanied by suppressing recovery from DNA damage caused by CPT. Here, we propose a potential combination therapy of WRN-siRNA and CPT, looking forward to minimizing the inevitable adverse effects associated with cancer chemotherapy.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Camptotecina/uso terapêutico , Inativação Gênica/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , RecQ Helicases/antagonistas & inibidores , RecQ Helicases/genética , Sobrevivência Celular/efeitos dos fármacos , Dano ao DNA , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Sinergismo Farmacológico , Exodesoxirribonucleases , Células HeLa , Humanos , Immunoblotting , Microscopia Confocal , Helicase da Síndrome de Werner
14.
Biol Pharm Bull ; 30(9): 1685-92, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17827721

RESUMO

Yeast Pif1 DNA helicase is the prototype member of a helicase subfamily participating in the maintenance of telomere, ribosome, and mitochondria DNAs. The Pif1 DNA helicase family is highly conserved from yeast to human, but the biochemical nature of human homologues remains to be clarified. To this end, we investigated the transcriptional unit of human Pif1 gene and its encoded protein hPif1. The results showed that the hPif1 gene product has at least two isoforms consisting of the conserved helicase motif and differential C-terminal regions derived from alternative splicing of the gene transcript. Deletion mutant analysis showed that Pif1 helicase has nuclear localization signal and mitochondria targeting signal at the N-terminal and C-terminal regions, respectively. In HeLa cells, hPif1 helicase expression was induced by the release of cells from serum starvation, suggesting that hPif1 has roles in the S phase. Consistently, the down regulation of the hPif1 helicase by RNA interference with siRNA caused a cell cycle delay at the S phase. These findings suggest that hPif1 in the nucleus may be involved in chromosome maintenance in association with DNA replication, while the function of hPif1 remains to be clarified.


Assuntos
Núcleo Celular/enzimologia , DNA Helicases/metabolismo , Mitocôndrias/enzimologia , Adenosina Trifosfatases/metabolismo , Western Blotting , Ciclo Celular/efeitos dos fármacos , DNA/biossíntese , DNA/genética , DNA Helicases/genética , DNA Helicases/isolamento & purificação , Citometria de Fluxo , Células HeLa , Humanos , Microscopia Confocal , RNA Interferente Pequeno/farmacologia , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Fase S/efeitos dos fármacos , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo
15.
Biol Pharm Bull ; 30(2): 266-71, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17268063

RESUMO

We report that the expression of Bloom helicase (BLM) was up-regulated by 17beta-estradiol (E2) in estrogen receptor (ER)-positive mammary tumor MCF-7 cells, but was hardly modulated in ER-negative mammary tumor MDA-MB-231 cells. ER antagonist ICI182780 blocked the E2 effect on BLM expression in MCF-7 cells. From these results we conclude that ER participates in up-regulation of BLM expression in MCF-7 cells by means of E2. Similar results were obtained when MCF-7 cells were treated with bisphenol A (BPA), an endocrine-disrupting chemical having a weak estrogenic activity. The ER binding ability of BPA is estimated at 1/1000 of E2 ability, and in this study about 1000-times more BPA was needed for the same levels of estrogenic effect of E2. The expression of cell-cycle associated genes, cdc6, MCM5, MCM2, Myt1, PCNA and AuroraA were up-regulated by E2 and BPA treatment in MCF-7 cells accompanied by up-regulation of BLM. In this BLM promoter study, Sp1 elements in the upper region of BLM modulated transcription, but were not indispensable for E2 response. Our results suggested that up-regulation of BLM expression by E2 and BPA is ER-dependent and may be responsible for repair of DNA damage caused by the genotoxicity of these estrogenic agents.


Assuntos
Adenosina Trifosfatases/biossíntese , DNA Helicases/biossíntese , Estradiol/farmacologia , Estrogênios/farmacologia , Fenóis/farmacologia , Adenosina Trifosfatases/genética , Compostos Benzidrílicos , Linhagem Celular Tumoral , DNA Helicases/genética , Humanos , Luciferases/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/biossíntese , RecQ Helicases , Receptores de Estrogênio/metabolismo , Regulação para Cima
16.
Exp Cell Res ; 312(17): 3443-57, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16949575

RESUMO

Mutations in the RECQL4 helicase gene have been linked to Rothmund-Thomson syndrome (RTS), which is characterized by poikiloderma, growth deficiency, and a predisposition to cancer. Examination of RECQL4 subcellular localization in live cells demonstrated a nucleoplasmic pattern and, to a lesser degree, staining in nucleoli. Analysis of RECQL4-GFP deletion mutants revealed two nuclear localization regions in the N-terminal region of RECQL4 and a nucleolar localization signal at amino acids 376-386. RECQL4 localization did not change after treatment with the DNA-damaging agents bleomycin, etoposide, UV irradiation and gamma irradiation, in contrast to the Bloom and Werner syndrome helicases that relocate to distinct nuclear foci after damage. However, in a significant number of cells exposed to hydrogen peroxide or streptonigrin, RECQL4 accumulated in nucleoli. Using a T7 phage display screen, we determined that RECQL4 interacts with poly(ADP-ribose) polymerase-1 (PARP-1), a nuclear enzyme that promotes genomic integrity through its involvement in DNA repair and signaling pathways. The RECQL4 nucleolar localization was inhibited by pretreatment with a PARP-1 inhibitor. The C-terminal portion of RECQL4 was found to be an in vitro substrate for PARP-1. These results demonstrate changes in the intracellular localization of RECQL4 in response to oxidative stress and identify an interaction between RECQL4 and PARP-1.


Assuntos
Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Nucléolo Celular/metabolismo , DNA Helicases/genética , DNA Helicases/metabolismo , Estresse Oxidativo , Adenosina Trifosfatases/química , Linhagem Celular , Núcleo Celular/química , DNA Helicases/química , Proteínas de Fluorescência Verde/genética , Células HeLa , Humanos , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/metabolismo , RecQ Helicases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA