Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
Oncotarget ; 7(14): 17726-36, 2016 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-26894857

RESUMEN

Gemcitabine is an important anticancer therapeutics approved for treatment of several human cancers including locally advanced or metastatic pancreatic ductal adenocarcinoma (PDAC). Its clinical effectiveness, however, is hindered by existence of intrinsic and development of acquired resistances. Previously, it was found that 14-3-3σ expression associates with poor clinical outcome of PDAC patients. It was also found that 14-3-3σ expression is up-regulated in gemcitabine resistant PDAC cells and contributes to the acquired gemcitabine resistance. In this study, we investigated the molecular mechanism of 14-3-3σ function in gemcitabine resistance and found that 14-3-3σ up-regulates YAP1 expression and then binds to YAP1 to inhibit gemcitabine-induced caspase 8 activation and apoptosis. 14-3-3σ association with YAP1 up-regulates the expression of ribonucleotide reductase M1 and M2, which may mediate 14-3-3σ/YAP1 function in the acquired gemcitabine resistance. These findings suggest a possible role of YAP1 signaling in gemcitabine resistance.


Asunto(s)
Proteínas 14-3-3/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Desoxicitidina/análogos & derivados , Neoplasias Pancreáticas/metabolismo , Fosfoproteínas/metabolismo , Ribonucleótido Reductasas/biosíntesis , Antimetabolitos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Desoxicitidina/farmacología , Resistencia a Antineoplásicos , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Ribonucleótido Reductasas/metabolismo , Factores de Transcripción , Transfección , Regulación hacia Arriba/efectos de los fármacos , Proteínas Señalizadoras YAP , Gemcitabina
2.
J Hepatol ; 63(4): 789-96, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26026873

RESUMEN

BACKGROUND & AIMS: Hepatitis B virus (HBV) infects and replicates in quiescent hepatocytes, which are deficient in dNTPs, the critical precursors of HBV replication. Most tumor viruses promote dNTP production in host cells by inducing cell proliferation. Although HBV is known as a major cause of hepatocellular carcinoma, it does not lead to cellular proliferation. Instead, HBV acquires dNTPs by activating the expression of the R2 subunit of the Ribonucleotide Reductase (RNR) holoenzyme, the cell cycle gene that is rate-limiting for generation of dNTPs, without inducing the cell cycle. We wished to elucidate the molecular basis of HBV-dependent R2 expression in quiescent cells. METHODS: Quiescent HepG2 cells were transduced with an HBV-containing lentiviral vector, and primary human hepatocytes were infected with HBV. DNA damage response and RNR-R2 gene expression were monitored under this condition. RESULTS: We report here that HBV-induced R2 expression is mediated by the E2F1 transcription factor, and that HBV induces E2F1 accumulation, modification and binding to the R2 promoter. We found that Chk1, a known E2F1 kinase that functions in response to DNA damage, was activated by HBV. In cells where Chk1 was pharmacologically inhibited, or depleted by shRNA-mediated knockdown, HBV-mediated R2 expression was severely attenuated. Furthermore, we found that HBV attenuates DNA repair, thus reducing cellular dNTP consumption. CONCLUSIONS: Our findings demonstrate that HBV exploits the Chk1-E2F1 axis of the DNA damage response pathway to induce R2 expression in a cell cycle-independent manner. This suggests that inhibition of this pathway may have a therapeutic value for HBV carriers.


Asunto(s)
Daño del ADN/genética , Regulación Viral de la Expresión Génica , Virus de la Hepatitis B/genética , Hepatitis C/virología , ARN Viral/genética , Ribonucleótido Reductasas/genética , Activación Viral/genética , Apoptosis , Southern Blotting , Western Blotting , Ciclo Celular , División Celular , Proliferación Celular , Electroforesis en Gel de Campo Pulsado , Virus de la Hepatitis B/metabolismo , Hepatitis C/metabolismo , Hepatitis C/patología , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Inmunoprecipitación , Reacción en Cadena de la Polimerasa , Ribonucleótido Reductasas/biosíntesis
5.
Proc Natl Acad Sci U S A ; 110(42): E3997-4006, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24082141

RESUMEN

Macrophages are a major target cell for HIV-1, and their infection contributes to HIV pathogenesis. We have previously shown that the cyclin-dependent kinase inhibitor p21 inhibits the replication of HIV-1 and other primate lentiviruses in human monocyte-derived macrophages by impairing reverse transcription of the viral genome. In the attempt to understand the p21-mediated restriction mechanisms, we found that p21 impairs HIV-1 and simian immunodeficiency virus (SIV)mac reverse transcription in macrophages by reducing the intracellular deoxyribonucleotide (dNTP) pool to levels below those required for viral cDNA synthesis by a SAM domain and HD domain-containing protein 1 (SAMHD1)-independent pathway. We found that p21 blocks dNTP biosynthesis by down-regulating the expression of the RNR2 subunit of ribonucleotide reductase, an enzyme essential for the reduction of ribonucleotides to dNTP. p21 inhibits RNR2 transcription by repressing E2F1 transcription factor, its transcriptional activator. Our findings unravel a cellular pathway that restricts HIV-1 and other primate lentiviruses by affecting dNTP synthesis, thereby pointing to new potential cellular targets for anti-HIV therapeutic strategies.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Desoxirribonucleótidos/biosíntesis , Regulación Enzimológica de la Expresión Génica , Infecciones por VIH/metabolismo , VIH-1/fisiología , Macrófagos/metabolismo , Ribonucleótido Reductasas/biosíntesis , Replicación Viral/fisiología , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , ADN Complementario/biosíntesis , ADN Complementario/genética , ADN Viral/biosíntesis , ADN Viral/genética , Desoxirribonucleótidos/genética , Regulación hacia Abajo/genética , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F1/metabolismo , Infecciones por VIH/terapia , Infecciones por VIH/virología , Macrófagos/virología , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Ribonucleótido Reductasas/genética , Proteína 1 que Contiene Dominios SAM y HD , Virus de la Inmunodeficiencia de los Simios/fisiología , Transcripción Genética/genética
6.
Int J Gynecol Cancer ; 23(4): 615-21, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23552804

RESUMEN

OBJECTIVE: To evaluate pretherapy ribonucleotide reductase (RNR) expression and its effect on radiochemotherapeutic outcome in women with cervical cancer. METHODS/MATERIALS: Pretherapy RNR M1, M2, and M2b immunohistochemistry was done on cervical cancer specimens retrieved from women treated on Radiation Therapy Oncology Group (RTOG) 0116 and 0128 clinical trials. Enrollees of RTOG 0116 (node-positive stages IA-IVA) received weekly cisplatin (40 mg/m(2)) with amifostine (500 mg) and extended-field radiation then brachytherapy (85 Gy). Enrollees of RTOG 0128 (node-positive or bulky ≥5 cm, stages IB-IIA or stages IIB-IVA) received cisplatin (75 mg/m(2)) on days 1, 23, and 43 and 5-FU (1 g/m(2) for 4 days) during pelvic radiation then brachytherapy (85 Gy), plus celecoxib (400 mg twice daily, day 1 through 1 year). Disease-free survival (DFS) was estimated univariately by the Kaplan-Meier method. Cox proportional hazards models evaluated the impact of RNR immunoreactivity on DFS. RESULTS: Fifty-one tissue samples were analyzed: 13 from RTOG 0116 and 38 from RTOG 0128. M1, M2, and M2b overexpression (3+) frequencies were 2%, 80%, and 47%, respectively. Low-level (0-1+, n = 44/51) expression of the regulatory subunit M1 did not associate with DFS (P = 0.38). High (3+) M2 expression occurred in most (n = 41/51) but without impact alone on DFS (hazard ratio, 0.54; 95% confidence interval, 0.2-1.4; P = 0.20). After adjusting for M2b status, pelvic node-positive women had increased hazard for relapse or death (hazard ratio, 5.5; 95% confidence interval, 2.2-13.8; P = 0.0003). CONCLUSIONS: These results suggest that RNR subunit expression may discriminate cervical cancer phenotype and radiochemotherapy outcome. Future RNR biomarker studies are warranted.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Ribonucleótido Reductasas/biosíntesis , Neoplasias del Cuello Uterino/enzimología , Adulto , Anciano , Biomarcadores de Tumor/genética , Ensayos Clínicos Fase II como Asunto , Supervivencia sin Enfermedad , Femenino , Humanos , Persona de Mediana Edad , Fenotipo , Estudios Retrospectivos , Ribonucleótido Reductasas/genética , Investigación Biomédica Traslacional , Resultado del Tratamiento , Estados Unidos/epidemiología , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/terapia
7.
Am J Pathol ; 182(1): 142-51, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23245831

RESUMEN

In normal human cells, oncogene-induced senescence (OIS) depends on induction of DNA damage response. Oxidative stress and hyperreplication of genomic DNA have been proposed as major causes of DNA damage in OIS cells. Here, we report that down-regulation of deoxyribonucleoside pools is another endogenous source of DNA damage in normal human fibroblasts (NHFs) undergoing HRAS(G12V)-induced senescence. NHF-HRAS(G12V) cells underexpressed thymidylate synthase (TS) and ribonucleotide reductase (RR), two enzymes required for the entire de novo deoxyribonucleotide biosynthesis, and possessed low dNTP levels. Chromatin at the promoters of the genes encoding TS and RR was enriched with retinoblastoma tumor suppressor protein and histone H3 tri-methylated at lysine 9. Importantly, ectopic coexpression of TS and RR or addition of deoxyribonucleosides substantially suppressed DNA damage, senescence-associated phenotypes, and proliferation arrest in two types of NHF-expressing HRAS(G12V). Reciprocally, short hairpin RNA-mediated suppression of TS and RR caused DNA damage and senescence in NHFs, although less efficiently than HRAS(G12V). However, overexpression of TS and RR in quiescent NHFs did not overcome proliferation arrest, suggesting that unlike quiescence, OIS requires depletion of dNTP pools and activated DNA replication. Our data identify a previously unknown role of deoxyribonucleotides in regulation of OIS.


Asunto(s)
Senescencia Celular/genética , Daño del ADN/genética , Desoxirribonucleótidos/metabolismo , Oncogenes/fisiología , Proliferación Celular , Células Cultivadas , Senescencia Celular/fisiología , Replicación del ADN/genética , Desoxirribonucleótidos/genética , Fibroblastos/metabolismo , Fibroblastos/fisiología , Humanos , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Ribonucleótido Reductasas/biosíntesis , Ribonucleótido Reductasas/fisiología , Timidilato Sintasa/biosíntesis , Timidilato Sintasa/fisiología
8.
Anticancer Res ; 32(11): 4685-90, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23155230

RESUMEN

BACKGROUND: Ribonucleotide reductase composed of the hRRM1 and hRRM2 subunits catalyzes the conversion of ribonucleotides to their corresponding deoxy forms for DNA replication. Anti-hRRM2 siRNA degrades hRRM2's mRNA and suppresses tumorigenesis. A Phase I clinical trial demonstrated its therapy potential. HN-1 represents a tumor-specifically internalizing peptide for targeted-drug delivery into human head and neck squamous cell carcinoma. MATERIALS AND METHODS: Internalization of peptide was monitored by fluorescence microscopy. The peptide-siRNA conjugate was chemically synthesized. The hRRM2 expression was monitored by western blot analysis. RESULTS: HN-1(TYR) (HN-1 with two N-terminally added tyrosines) was internalized by human head and neck or breast cancer cells. Anti-hRRM2 siRNA(R) (resistant to RNase degradation) was conjugated to HN-1(TYR) without compromising their properties. The treatment with HN-1(TYR)-anti-hRRM2 siRNA(R) partly suppressed the endogenously expressed hRRM2 in human breast cancer cells. CONCLUSION: Our results establish the utility of tumor-specifically internalizing peptides for targeted siRNA delivery into human cancer cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Terapia Genética/métodos , Neoplasias de Cabeza y Cuello/metabolismo , Oligopéptidos/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/terapia , Humanos , Microscopía Fluorescente , Oligopéptidos/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ribonucleótido Reductasas/biosíntesis , Ribonucleótido Reductasas/deficiencia , Ribonucleótido Reductasas/genética
9.
Osteoarthritis Cartilage ; 20(12): 1603-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22954457

RESUMEN

OBJECTIVE: The p53 tumor-suppressor protein p53R2 is activated in response to various stressors that act on cell signaling. When DNA is damaged, phosphorylation of p53 at its Ser 15 residue induces p53R2 production. The role of p53R2 in chondrocytes remains poorly understood. In this study, we evaluated in chondrocytes, p53R2 expression and its regulation in response to mechanical stress. Furthermore, we investigated the function of p53R2 in relation to mechanotransduction. METHODS: Osteoarthritis (OA) cartilage obtained from total knee replacements and normal cartilage obtained from femoral neck fractures was used to measure p53R2 expression by using immunohistochemistry, western blotting, and real-time polymerase chain reaction (PCR). The OA chondrocytes were subjected to a high magnitude of cyclical tensile strain by using an FX-2000 Flexercell system. Next, sulfated glycosaminoglycan (sGAG) production was quantified in these cells. Protein expression of p53R2, and phosphorylation of Akt, p38MAPK, ERK1/2, and JNK was also detected using western blotting. Moreover, Akt phosphorylation was detected after transfecting the cells with p53R2-specific small interfering RNA (siRNA). RESULTS: Expression of p53R2 was significantly increased in OA chondrocytes and in chondrocytes after applying 5% tensile strain to the cells. However, Akt phosphorylation was down-regulated in OA chondrocytes after the strain, and was up-regulated after transfection of p53R2. sGAG protein as well as collagen type II and aggrecan mRNA was increased following transfection of p53R2-specific siRNA after 5% tensile strain. CONCLUSIONS: p53R2 could regulate matrix synthesis via Akt phosphorylation during chondrocyte mechanotransduction. Down-regulation of p53R2 may be a new therapeutic approach in OA therapy.


Asunto(s)
Cartílago Articular/metabolismo , Proteínas de Ciclo Celular/genética , Condrocitos/metabolismo , Regulación de la Expresión Génica , Osteoartritis de la Rodilla/genética , Proteínas Proto-Oncogénicas c-akt/genética , ARN Mensajero/genética , Ribonucleótido Reductasas/genética , Western Blotting , Cartílago Articular/patología , Proteínas de Ciclo Celular/biosíntesis , Células Cultivadas , Condrocitos/patología , Reparación del ADN , Humanos , Inmunohistoquímica , Osteoartritis de la Rodilla/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Ribonucleótido Reductasas/biosíntesis , Transducción de Señal , Estrés Mecánico
10.
PLoS Pathog ; 8(4): e1002635, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22496656

RESUMEN

Leishmaniasis is a parasitic disease that is widely prevalent in many tropical and sub-tropical regions of the world. Infection with Leishmania has been recognized to induce a striking acceleration of Human Immunodeficiency Virus Type 1 (HIV-1) infection in coinfected individuals through as yet incompletely understood mechanisms. Cells of the monocyte/macrophage lineage are the predominant cell types coinfected by both pathogens. Monocytes and macrophages contain extremely low levels of deoxynucleoside triphosphates (dNTPs) due to their lack of cell cycling and S phase, where dNTP biosynthesis is specifically activated. Lentiviruses, such as HIV-1, are unique among retroviruses in their ability to replicate in these non-dividing cells due, at least in part, to their highly efficient reverse transcriptase (RT). Nonetheless, viral replication progresses more efficiently in the setting of higher intracellular dNTP concentrations related to enhanced enzyme kinetics of the viral RT. In the present study, in vitro infection of CD14+ peripheral blood-derived human monocytes with Leishmania major was found to induce differentiation, marked elevation of cellular p53R2 ribonucleotide reductase subunit and R2 subunit expression. The R2 subunit is restricted to the S phase of the cell cycle. Our dNTP assay demonstrated significant elevation of intracellular monocyte-derived macrophages (MDMs) dNTP concentrations in Leishmania-infected cell populations as compared to control cells. Infection of Leishmania-maturated MDMs with a pseudotyped GFP expressing HIV-1 resulted in increased numbers of GFP+ cells in the Leishmania-maturated MDMs as compared to control cells. Interestingly, a sub-population of Leishmania-maturated MDMs was found to have re-entered the cell cycle, as demonstrated by BrdU labeling. In conclusion, Leishmania infection of primary human monocytes promotes the induction of an S phase environment and elevated dNTP levels with notable elevation of HIV-1 expression in the setting of coinfection.


Asunto(s)
Desoxirribonucleótidos/metabolismo , Infecciones por VIH , VIH-1/metabolismo , Leishmania major/metabolismo , Leishmaniasis Cutánea , Macrófagos , Proteínas de Ciclo Celular/biosíntesis , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Viral de la Expresión Génica , Infecciones por VIH/complicaciones , Infecciones por VIH/metabolismo , Transcriptasa Inversa del VIH/metabolismo , Humanos , Leishmaniasis Cutánea/complicaciones , Leishmaniasis Cutánea/metabolismo , Macrófagos/metabolismo , Macrófagos/parasitología , Macrófagos/virología , Masculino , Monocitos , Ribonucleótido Reductasas/biosíntesis , Fase S
11.
J Bacteriol ; 194(6): 1515-22, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22247510

RESUMEN

Production of deoxyribonucleotides for DNA synthesis is an essential and tightly regulated process. The class Ia ribonucleotide reductase (RNR), the product of the nrdAB genes, is required for aerobic growth of Escherichia coli. In catalyzing the reduction of ribonucleotides, two of the cysteines of RNR become oxidized, forming a disulfide bond. To regenerate active RNR, the cell uses thioredoxins and glutaredoxins to reduce the disulfide bond. Strains that lack thioredoxins 1 and 2 and glutaredoxin 1 do not grow because RNR remains in its oxidized, inactive form. However, suppressor mutations that lead to RNR overproduction allow glutaredoxin 3 to reduce sufficient RNR for growth of these mutant strains. We previously described suppressor mutations in the dnaA and dnaN genes that had such effects. Here we report the isolation of new mutations that lead to increased levels of RNR. These include mutations that were not known to influence production of RNR previously, such as a mutation in the hda gene and insertions in the nrdAB promoter region of insertion elements IS1 and IS5. Bioinformatic analysis raises the possibility that IS element insertion in this region represents an adaptive mechanism in nrdAB regulation in E. coli and closely related species. We also characterize mutations altering different amino acids in DnaA and DnaN from those isolated before.


Asunto(s)
Escherichia coli/enzimología , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Ribonucleótido Reductasas/biosíntesis , Ribonucleótido Reductasas/genética , Supresión Genética , Proteínas Bacterianas/genética , ADN Polimerasa III/genética , Proteínas de Unión al ADN/genética , Proteínas de Escherichia coli/genética , Genes Bacterianos , Mutagénesis Insercional , Regiones Promotoras Genéticas , Ribonucleósido Difosfato Reductasa/genética
12.
J Hepatobiliary Pancreat Sci ; 18(5): 700-11, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21451941

RESUMEN

BACKGROUND: Gemcitabine is a promising drug for cholangiocarcinoma treatment. However, the kinetics and metabolism of this drug in cholangiocarcinoma treatment are not well defined. We aimed to investigate the potential clinical role of gemcitabine metabolism-related genes in the gemcitabine sensitivity of cholangiocarcinoma and identify and characterize novel gemcitabine resistance-related genes. METHODS: Expressions of genes related to gemcitabine sensitivity and gemcitabine metabolism were measured in 10 cholangiocarcinoma cell lines, and the association between gene expression and gemcitabine sensitivity was evaluated. Furthermore, gemcitabine-resistant cell lines were established from YSCCC cells and subjected to genome-wide microarray analysis. The 2-fold upregulated and downregulated genes were then subjected to pathway analysis. RESULTS: p53R2 mRNA expression was significantly higher in gemcitabine-resistant cell lines (IC(50) > 1000 nM), and all subunits of ribonucleotide reductase were upregulated in the established gemcitabine-resistant cell lines. Microarray analysis revealed that the upregulated genes in the resistant cells belonged to the glutathione and pyrimidine metabolism pathways, and that the downregulated genes belonged to the N-glycan biosynthesis pathway. CONCLUSIONS: Increased expression of p53R2 may predict gemcitabine resistance, and upregulated RNR activity may influence gemcitabine resistance in cholangiocarcinoma cells. Glutathione pathway-related genes were induced by continuous exposure to gemcitabine and may contribute to gemcitabine resistance.


Asunto(s)
Neoplasias de los Conductos Biliares/tratamiento farmacológico , Conductos Biliares Intrahepáticos , Proteínas de Ciclo Celular/genética , Colangiocarcinoma/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Ribonucleótido Reductasas/genética , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/metabolismo , Proteínas de Ciclo Celular/biosíntesis , Línea Celular Tumoral , Colangiocarcinoma/genética , Colangiocarcinoma/metabolismo , Reparación del ADN , Desoxicitidina/farmacología , Humanos , Inmunosupresores/farmacología , ARN Neoplásico/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Ribonucleótido Reductasas/biosíntesis , Gemcitabina
13.
Cancer Res ; 71(9): 3202-13, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21415168

RESUMEN

Ribonucleotide reductase subunit RRM2B (p53R2) has been reported to suppress invasion and metastasis in colorectal cancer (CRC). Here, we report that high levels of RRM2B expression are correlated with markedly better survival in CRC patients. In a fluorescence-labeled orthotopic mouse xenograft model, we confirmed that overexpression of RRM2B in nonmetastatic CRC cells prevented lung and/or liver metastasis, relative to control cells that did metastasize. Clinical outcome studies were conducted on a training set with 103 CRCs and a validation set with 220 CRCs. All participants underwent surgery with periodic follow-up to determine survivability. A newly developed specific RRM2B antibody was employed to carry out immunohistochemistry for determining RRM2B expression levels on tissue arrays. In the training set, the Kaplan-Meier and multivariate Cox analysis revealed that RRM2B is associated with better survival of CRCs, especially in stage IV patients (HR = 0.40; 95% CI = 0.18-0.86, P = 0.016). In the validation set, RRM2B was negatively related to tumor invasion (OR = 0.45, 95% CI = 0.19-0.99, P = 0.040) and lymph node involvement (OR = 0.48, 95% CI = 0.25-0.92, P = 0.026). Furthermore, elevated expression of RRM2B was associated with better prognosis in this set as determined by multivariate analyses (HR = 0.48, 95% CI = 0.26-0.91, P = 0.030). Further investigations revealed that RRM2B was correlated with better survival of CRCs with advanced stage III and IV tumors rather than earlier stage I and II tumors. Taken together, our findings establish that RRM2B suppresses invasiveness of cancer cells and that its expression is associated with a better survival prognosis for CRC patients.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Proteínas de Ciclo Celular/biosíntesis , Neoplasias Colorrectales/enzimología , Ribonucleótido Reductasas/biosíntesis , Secuencia de Aminoácidos , Animales , Anticuerpos/química , Anticuerpos/inmunología , Especificidad de Anticuerpos , Biomarcadores de Tumor/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/inmunología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Femenino , Células HCT116 , Humanos , Inmunohistoquímica/métodos , Ratones , Ratones Endogámicos NOD , Datos de Secuencia Molecular , Invasividad Neoplásica , Estadificación de Neoplasias , Pronóstico , Ribonucleótido Reductasas/genética , Ribonucleótido Reductasas/inmunología , Tasa de Supervivencia , Transfección , Trasplante Heterólogo
14.
Eksp Klin Farmakol ; 73(4): 31-4, 2010 Apr.
Artículo en Ruso | MEDLINE | ID: mdl-20486557

RESUMEN

Results of a comparative study of the influence of doxorubicine (DOX) and dehydroepiandrosterone (DHEA) on cell proliferation and oxidative stress in Saccharomyces cerevisiae cells are presented. Three treatment schedules were assessed--DOX only, DHEA only, and DOX simultaneously with DHEA--in examining cell proliferation, measuring the content of glutathione, and evaluating the expression of ribonucleotide reductase in the test cells. The results indicate that the separate treatment with DOX or DHEA stimulates the expression of ribonucleotide reductase and leads to a decrease in the rate of cell proliferation. DHEA produces a dose-dependent decrease in the content of a reduced form of glutathione in cells, whereas the concentration of the oxidized form remains unchanged. In contrast, the treatment with DOX increased the concentrations of both forms of glutathione. The simultaneous treatment of cells by DOX and DHEA increased the accumulation of intracellular glutathione and decreased the total antiproliferative effect.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Antibióticos Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Deshidroepiandrosterona/farmacología , Doxorrubicina/farmacología , Estrés Oxidativo/efectos de los fármacos , Saccharomyces cerevisiae/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Fúngica de la Expresión Génica/efectos de los fármacos , Glutatión/metabolismo , Ribonucleótido Reductasas/biosíntesis , Proteínas de Saccharomyces cerevisiae/biosíntesis
15.
Int J Oncol ; 36(2): 341-9, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20043067

RESUMEN

Gemcitabine (Gem) is a dFdC analogue with activity against several solid tumors. Gem is intracellularly phosphorylated by dCK, leading to the production of the metabolite dFdCDP. dFdCDP exhibits the cytotoxic effect by inactivating ribonucleotide reductase larger subunit 1 (RRM1), which is a rate limiting enzyme for de novo DNA synthesis. To date, RRM1 expression is believed to determine sensitivity to Gem in pancreatic and non-small cell lung cancer. In the present study, we found that an anti-allergic drug, tranilast strongly enhanced the sensitivity of pancreatic cancer cell line KP4 to Gem. In growth inhibition assay, 100 microM of tranilast plus 1 microM of Gem more strongly suppressed the growth of KP4 at 12.7-fold in IC50 than single Gem treatment, while this compound no longer affected the sensitivity to other drugs such as 5-fluorouracil, irinotecan or paclitaxel. FACS and TUNEL analysis demonstrated the increased apoptotic population in KP4 cells under tranilast plus Gem, compared with single Gem treatment. In Western blot analysis, tranilast treatment decreased RRM1 expression at protein level with dose-dependency in KP4 cells. Proteasome inhibitor MG132 disturbed the reduction of RRM1 expression in tranilast treated KP4 cells, indicating protein degradation by the activated proteasome. Transfection using siRNA against RRM1 increased the sensitivity of KP4 to Gem, suggesting that RRM1 suppression is an important step in increasing Gem efficacy. Finally, we demonstrated that tranilast reduced RRM1 protein and increased Gem efficacy in 4 other pancreatic cell lines. In a future, a novel chemotherapeutic strategy by Gem along with tranilast might improve Gem efficacy against pancreatic cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Desoxicitidina/análogos & derivados , Neoplasias Pancreáticas/enzimología , Ribonucleótido Reductasas/efectos de los fármacos , ortoaminobenzoatos/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Separación Celular , Desoxicitidina/administración & dosificación , Sinergismo Farmacológico , Citometría de Flujo , Humanos , Etiquetado Corte-Fin in Situ , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleótido Reductasas/biosíntesis , Gemcitabina
16.
Methods Enzymol ; 462: 45-76, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19632469

RESUMEN

Since the discovery of the essential tyrosyl radical (Y*) in E. coli ribonucleotide reductase (RNR), a number of enzymes involved in primary metabolism have been found that use transient or stable tyrosyl (Y) or tryptophanyl (W) radicals in catalysis. These enzymes engage in a myriad of charge transfer reactions that occur with exquisite control and specificity. The unavailability of natural amino acids that can perturb the reduction potential and/or protonation states of redox-active Y or W residues has limited the usefulness of site-directed mutagenesis methods to probe the attendant mechanism of charge transport at these residues. However, recent technologies designed to site-specifically incorporate unnatural amino acids into proteins have now made viable the study of these mechanisms. The class Ia RNR from E. coli serves as a paradigm for enzymes that use amino acid radicals in catalysis. It catalyzes the conversion of nucleotides to deoxynucleotides and utilizes both stable and transient protein radicals. This reaction requires radical transfer from a stable tyrosyl radical (Y(122)*) in the beta subunit to an active-site cysteine (C(439)) in the alpha subunit, where nucleotide reduction occurs. The distance between the sites is proposed to be >35 A. A pathway between these sites has been proposed in which transient aromatic amino acid radicals mediate radical transport. To examine the pathway for radical propagation as well as requirements for coupled electron and proton transfers, a suppressor tRNA/aminoacyl-tRNA synthetase (RS) pair has been evolved that allows for site-specific incorporation of 3-aminotyrosine (NH(2)Y). NH(2)Y was chosen because it is structurally similar to Y with a similar phenolic pK(a). However, at pH 7, it is more easily oxidized than Y by 190 mV (approximately 4.4 kcal/mol), thus allowing it to act as a radical trap. Here we present the detailed procedures involved in evolving an NH(2)Y-specific RS, assessing its efficiency in NH(2)Y insertion, generating RNR mutants with NH(2)Y at selected sites, and determining the spectroscopic properties of NH(2)Y* and the kinetics of its formation.


Asunto(s)
Sustitución de Aminoácidos , Evolución Molecular Dirigida , ARN de Transferencia de Tirosina/metabolismo , Ribonucleótido Reductasas/biosíntesis , Tirosina/análogos & derivados , Tirosina/metabolismo , Secuencia de Aminoácidos , Biocatálisis , Dominio Catalítico , Escherichia coli , Proteínas de Escherichia coli/antagonistas & inhibidores , Proteínas de Escherichia coli/biosíntesis , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Proteínas Mutantes/biosíntesis , Oxidación-Reducción , Ribonucleótido Reductasas/antagonistas & inhibidores , Ribonucleótido Reductasas/química , Ribonucleótido Reductasas/metabolismo , Tirosina-ARNt Ligasa
17.
Cancer Chemother Pharmacol ; 63(2): 321-30, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18575868

RESUMEN

PURPOSE: Oncolytic viral therapy is a newly developed modality for treating tumors. Many clinical trials using oncolytic virus have been performed worldwide, but most of them have used local injection in the tumor. Determination of the effect and safety of intravascular virus injection instead of local injection is necessary for clinical use against multiple liver metastases and systemic metastases. METHODS: To evaluate the efficacy and safety of intravascular virus therapy, mice bearing multiple liver metastases were treated by intraportal or intravenous administration of the herpes simplex virus type 1 (HSV-1) mutant, hrR3. Mice treated with hrR3 were killed and organs were harvested for lacZ staining and PCR analysis. Inactivation of oncolytic virus in bloodstream was assessed by neutralization assay in vitro. Infectious activity of hrR3 with vascular endothelial cells was evaluated by replication and cytotoxicity assay. RESULTS: The survival rate of animals treated by hrR3 was significantly improved compared with the untreated group. lacZ staining and PCR analysis demonstrated detectable virus in the tumor but not in normal tissue or other organs except for the adrenal glands. We also showed that vascular endothelial cells allowed virus replication, while normal hepatocytes did not, and human anti-HSV antibody revealed attenuation of the infectious activity of hrR3. CONCLUSIONS: Intravascular delivery of hrR3 is effective in treating multiple liver metastases, however, several points must be kept in mind at the time of human clinical trials using intravascular virus administration in order to avoid critical side effects.


Asunto(s)
Herpesvirus Humano 1/genética , Neoplasias Hepáticas/secundario , Neoplasias Hepáticas/terapia , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Células Endoteliales/enzimología , Células Endoteliales/virología , Vectores Genéticos , Hepatocitos/enzimología , Hepatocitos/virología , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/fisiología , Humanos , Inmunoglobulina G/sangre , Inyecciones Intravenosas , Operón Lac , Neoplasias Hepáticas/enzimología , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa , Ribonucleótido Reductasas/biosíntesis , Células Vero , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Genetics ; 180(1): 681-6, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18716329

RESUMEN

Here we show that inactivation of the ATR-related kinase ATL-1 results in a significant reduction in mitochondrial DNA (mtDNA) copy numbers in Caenorhabditis elegans. Although ribonucleotide reductase (RNR) expression and the ATP/dATP ratio remained unaltered in atl-1 deletion mutants, inhibition of RNR by RNAi or hydroxyurea treatment caused further reductions in mtDNA copy number. These results suggest that ATL-1 functions to maintain mtDNA independently of RNR.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , ADN Mitocondrial , Eliminación de Gen , Mutación , Fosfotransferasas/genética , Adenosina Trifosfato/química , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Caenorhabditis elegans , Cromosomas , ADN Mitocondrial/genética , Homocigoto , Hidroxiurea/farmacología , Modelos Biológicos , Modelos Genéticos , Interferencia de ARN , Ribonucleótido Reductasas/biosíntesis , Ribonucleótido Reductasas/genética
19.
Biochem Biophys Res Commun ; 361(1): 249-55, 2007 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-17658469

RESUMEN

Homeodomain Interacting Protein Kinase-2 (HIPK2) is a protein with many functions and a modulator of p53 oncosuppressor functions. TP53 is the "guardian of the genome" thus, is the most critical tumor suppressor gene product that inhibits malignant transformation. P53R2 gene is directly induced by p53 in response to DNA damage and is involved in the p53 checkpoint for repairing damaged DNA to block genome instability. Here we wanted to explore the involvement of HIPK2 in damaged-DNA repair by regulating p53-induced p53R2 gene. We show that, induction of p53R2 expression, p53 recruitment onto p53R2 promoter, and its transcriptional activation was strongly impaired by HIPK2 knock-down, in response to drug. The failure of p53-induced p53R2 activation markedly compromised damaged-DNA repair efficiency. Finally, overexpression of exogenous p53 overcame the inability of endogenous p53 to activate p53R2-luc promoter in HIPK2 depleted cells. These data suggest that HIPK2 is involved in damaged-DNA repair taking part in restraining tumor progression, at least in part depending on p53 regulation.


Asunto(s)
Proteínas Portadoras/fisiología , Daño del ADN , Reparación del ADN , Neoplasias/enzimología , Proteínas Serina-Treonina Quinasas/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Humanos , Neoplasias/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Ribonucleótido Reductasas/biosíntesis , Ribonucleótido Reductasas/genética , Transcripción Genética , Proteína p53 Supresora de Tumor/antagonistas & inhibidores
20.
Anticancer Drugs ; 18(4): 377-88, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17351390

RESUMEN

RNA interference, a posttranscriptional gene-silencing mechanism, has received considerable attention for its potential as a new therapeutic strategy to treat human diseases and conditions including cancer. Various studies have supported a role for the R2 subunit of ribonucleotide reductase in cancer progression and metastasis. Short interfering siRNA 1284 was designed to target R2. In vitro studies, in which three different human tumor cell lines (A498, HT-29 and A2058) were transfected with short interfering siRNA 1284, demonstrate sequence-specific down-regulation of R2, which coincides with a decrease in cell proliferation, and cell cycle inhibition. In vivo studies with xenograft mouse models, generated from the same tumor cell lines, indicate that treatment with short interfering siRNA 1284 leads to inhibition of tumor growth and this effect was found to be dose dependent. Taken together, these results suggest that short interfering siRNA 1284, targeting R2, has great potential to serve as a therapeutic agent towards the treatment of human cancers.


Asunto(s)
Neoplasias/terapia , ARN Interferente Pequeño/uso terapéutico , Ribonucleótido Reductasas/biosíntesis , Animales , Northern Blotting , Western Blotting , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Densitometría , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Células HT29 , Humanos , Ratones , Ratones SCID , Trasplante de Neoplasias , Neoplasias/genética , Ribonucleótido Reductasas/genética , Fase S/efectos de los fármacos , Transfección , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA