Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neurotoxicology ; 59: 256-262, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27246648

RESUMO

Using a matched case-control design, we sought to determine whether the odds of konzo, a distinct spastic paraparesis associated with food (cassava) cyanogenic exposure in the tropics, were associated with lower cyanide detoxification rates (CDR) and malnutrition. Children with konzo (N=122, 5-17 years of age) were age- and sex-matched with presumably healthy controls (N=87) and assessed for motor and cognition performances, cyanogenic exposure, nutritional status, and cyanide detoxification rates (CDR). Cyanogenic exposure was ascertained by thiocyanate (SCN) concentrations in plasma (P-SCN) and urine (U-SCN). Children with a height-for-age z-score (HAZNCHS)<-2 were classified as nutritionally stunted. CDR was measured as time required to convert cyanide to SCN, and expressed as ms/µmol SCN/mg protein or as mmolSCN/ml plasma/min. Mean (SD) U-SCN in children with konzo was 521.9 (353.6) µmol/l and was, significantly higher than 384.6 (223.7) µmol/l in those without konzo. Conditional regression analysis of data for age- and sex- matched case-control pairs showed that konzo was associated with stunting (OR: 5.8; 95% CI: 2.7-12.8; p<0.01; N=83 paired groups) and higher U-SCN (OR: 1.1; 95% CI: 1.02-1.20 per 50-µmol increase in U-SCN; p=0.02; N=47 paired groups). After adjusting for stunting and U-SCN, the odds of developing konzo was reduced by 63% (95% CI: 11-85%, p=0.03; N=41 paired groups) for each 5mmol SCN/(ml plasma/min)-increase in CDR. Linear regression analysis indicated a significant association between BOT-2 or KABC-II scores and both the HAZNCHS z-score and the U-SCN concentration, but not the CDR. Our findings provide evidence in support of interventions to remove cyanogenic compounds from cassava prior to human consumption or, peharps, enhance the detoxification of cyanide in those relying on the cassava as the main source of food.


Assuntos
Cianetos/toxicidade , Paraparesia Espástica Tropical/induzido quimicamente , Sulfurtransferases/metabolismo , Adolescente , Análise de Variância , Estudos de Casos e Controles , Criança , Pré-Escolar , Transtornos Cognitivos/etiologia , Feminino , Humanos , Masculino , Transtornos Motores/etiologia , Nitrilas , Paraparesia Espástica Tropical/metabolismo , Estudos Retrospectivos , Estatísticas não Paramétricas
2.
Clin Toxicol (Phila) ; 52(5): 490-7, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24716792

RESUMO

BACKGROUND AND PURPOSE: Hydrogen sulfide (H2S) is a highly toxic gas for which no effective antidotes exist. It acts, at least in part, by binding to cytochrome c oxidase, causing cellular asphyxiation and anoxia. We investigated the effects of three different ligand forms of cobinamide, a vitamin B12 analog, to reverse sulfide (NaHS) toxicity. METHODS: New Zealand white rabbits received a continuous intravenous (IV) infusion of NaHS (3 mg/min) until expiration or a maximum 270 mg dose. Animals received six different treatments, administered at the time when they developed signs of severe toxicity: Group 1-saline (placebo group, N = 9); Group 2--IV hydroxocobalamin (N = 7); Group 3--IV aquohydroxocobinamide (N = 6); Group 4--IV sulfitocobinamide (N = 6); Group 5--intramuscular (IM) sulfitocobinamide (N = 6); and Group 6-IM dinitrocobinamide (N = 8). Blood was sampled intermittently, and systemic blood pressure and deoxygenated and oxygenated hemoglobin were measured continuously in peripheral muscle and over the brain region; the latter were measured by diffuse optical spectroscopy (DOS) and continuous wave near infrared spectroscopy (CWNIRS). RESULTS: Compared with the saline controls, all cobinamide derivatives significantly increased survival time and the amount of NaHS that was tolerated. Aquohydroxocobinamide was most effective (261.5 ± 2.4 mg NaHS tolerated vs. 93.8 ± 6.2 mg in controls, p < 0.0001). Dinitrocobinamide was more effective than sulfitocobinamide. Hydroxocobalamin was not significantly more effective than the saline control. CONCLUSIONS: Cobinamide is an effective agent for inhibiting lethal sulfide exposure in this rabbit model. Further studies are needed to determine the optimal dose and form of cobinamide and route of administration.


Assuntos
Antídotos/farmacologia , Cobamidas/farmacologia , Sulfeto de Hidrogênio/intoxicação , Hidroxocobalamina/farmacologia , Sulfetos/intoxicação , Animais , Antídotos/administração & dosagem , Antídotos/química , Cobamidas/administração & dosagem , Cobamidas/química , Modelos Animais de Doenças , Hemoglobinas/metabolismo , Sulfeto de Hidrogênio/administração & dosagem , Hidroxocobalamina/administração & dosagem , Infusões Intravenosas , Injeções Intramusculares , Injeções Intravenosas , Coelhos , Espectroscopia de Luz Próxima ao Infravermelho , Taxa de Sobrevida
3.
Ann Hematol ; 88(4): 319-24, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18784923

RESUMO

The 21-kD protein Ras of the low-molecular-weight GTP-binding (LMWG) family plays an important role in transduction of extracellular signals. Ras functions as a 'molecular switch' in transduction of signals from the membrane receptors of many growth factors, cytokines, and other second messengers to the cell nucleus. Numerous studies have shown that in multiple malignant tumors and hematopoietic malignancies, faulty signal transduction via the Ras pathway plays a key role in tumorigenesis. In this work, a non-radioactive assay was used to quantify Ras activity in hematologic malignancies. Ras activation was measured in six different cell lines and 24 patient samples, and sequence analysis of N- and K-ras was performed. The 24 patient samples comprised of seven acute myelogenous leukemia (AML) samples, five acute lymphocytic leukemia (ALL) samples, four myeloproliferative disease (MPD) samples, four lymphoma samples, four juvenile myelomonocytic leukemia (JMML) samples, and WBC from a healthy donor. The purpose of this study was to compare Ras activity determined by percentage of Ras-GTP with the mutational status of the Ras gene in the hematopoietic cells of the patients. Mutation analysis revealed ras mutations in two of the seven AML samples, one in codon 12 and one in codon 61; ras mutations were also found in two of the four JMML samples, and in one of the four lymphoma samples (codon 12). We found a mean Ras activation of 23.1% in cell lines with known constitutively activating ras mutations, which was significantly different from cell lines with ras wildtype sequence (Ras activation of 4.8%). Two of the five activating ras mutations in the patient samples correlated with increased Ras activation. In the other three samples, Ras was probably activated through "upstream" or "downstream" mechanisms.


Assuntos
Neoplasias Hematológicas/química , Neoplasias Hematológicas/genética , Proteínas ras/análise , Proteínas ras/genética , Análise Mutacional de DNA , Guanosina Difosfato/análise , Guanosina Trifosfato/análise , Neoplasias Hematológicas/etiologia , Humanos , Leucemia Mieloide Aguda , Leucemia Mielomonocítica Juvenil , Linfoma , Mutação , Transtornos Mieloproliferativos , Oncogenes , Leucemia-Linfoma Linfoblástico de Células Precursoras , Transdução de Sinais/genética , Células Tumorais Cultivadas
4.
J Biol Chem ; 276(29): 27296-303, 2001 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-11371572

RESUMO

Many of nitric oxide's biological effects are mediated via NO binding to the iron in heme-containing proteins. Cobalamin (vitamin B(12)) is structurally similar to heme and is a cofactor for methionine synthase, a key enzyme in folate metabolism. NO inhibits methionine synthase activity in vitro, but data concerning NO binding to cobalamin are controversial. We now show spectroscopically that NO reacts with all three valency states of cobalamin and that NO's inhibition of methionine synthase activity most likely involves its reaction with monovalent cobalamin. By following incorporation of the methyl moiety of [(14)C]methyltetrahydrofolic acid into protein, we show that NO inhibits methionine synthase activity in vivo, in cultured mammalian cells. The inhibition of methionine synthase activity disrupted carbon flow through the folate pathway as measured by decreased incorporation of [(14)C]formate into methionine, serine, and purine nucleotides. Homocysteine, but not cysteine, attenuated NO's inhibition of purine synthesis, providing further evidence that NO was acting through methionine synthase inhibition. NO's effect was observed both when NO donors were added to cells and when NO was produced physiologically in co-culture experiments. Treating cells with an NO synthase inhibitor increased formate incorporation into methionine, serine, and purines and methyl-tetrahydrofolate incorporation into protein. Thus, physiological concentrations of NO appear to regulate carbon flow through the folate pathway.


Assuntos
5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/antagonistas & inibidores , Carbono/metabolismo , Ácido Fólico/metabolismo , Óxido Nítrico/fisiologia , Animais , Linhagem Celular , Técnicas de Cocultura , Cricetinae , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Humanos , Metionina/biossíntese , NG-Nitroarginina Metil Éster/farmacologia , Doadores de Óxido Nítrico/farmacologia , Nucleotídeos de Purina/biossíntese , Ratos , Serina/biossíntese , Vitamina B 12/metabolismo
5.
Oncogene ; 20(16): 2044-9, 2001 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-11360188

RESUMO

We have previously demonstrated that amplification and overexpression of the Ki-ras gene is associated with mammary tumor progression in C3(1)/SV40Tag transgenic mice (Liu et al., 1998). To further evaluate the functional significance of the Ki-ras proto-oncogene in mammary cancer development, in vivo studies were conducted to examine the effect of Ki-ras gene dosage on tumor progression. The lack of one normal Ki-ras allele C3(1)/SV40Tag transgenic mice resulted in significantly delayed mammary intraepithelial neoplasia (MIN) formation as well as in a decreased number of mammary gland carcinomas. However, despite the retardation of tumor development by reduced Ki-ras gene dosage, overall survival was only modestly affected. This appears to be due to several factors including significant mammary tumor growth associated with Ki-ras gene amplification and over-expression that occurs during the advanced stage of oncogenesis in mice carrying either one or two normal Ki-ras alleles. The retardation of tumor progression due to the haploid loss of Ki-ras did not appear to be related to accelerated apoptosis, or a reduced rate of cell proliferation at the tumor stages examined. These data strongly suggest that the gene dosage of Ki-ras affects tumor promotion at an early stage of mammary tumor progression in this SV40 Tag-induced model of mammary oncogenesis.


Assuntos
Antígenos Transformantes de Poliomavirus/genética , Genes ras/genética , Neoplasias Mamárias Experimentais/genética , Animais , Apoptose/genética , Divisão Celular/genética , Progressão da Doença , Feminino , Haploidia , Masculino , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos
6.
J Biol Chem ; 276(15): 11804-11, 2001 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-11278453

RESUMO

Here we report the recurrent proviral activation of the Rap1-specific guanine nucleotide exchange factor CalDAG-GEF I (Kawasaki, H., Springett, G. M., Toki, S., Canales, J. J., Harlan, P., Blumenstiel, J. P., Chen, E. J., Bany, I. A., Mochizuki, N., Ashbacher, A., Matsuda, M., Housman, D. E., and Graybiel, A. M. (1998) Proc. Natl. Acad. Sci. U. S. A. 95, 13278-13283; Correction (1999) Proc. Natl. Acad. Sci. U. S. A. 96, 318) gene in BXH-2 acute myeloid leukemia. We also show that CalDAG-GEF I encodes two protein isoforms, a full-length isoform (CalDAG-GEF Ia) and a C-terminally truncated isoform (CalDAG-GEF Ib). Expression of the full-length CalDAG-GEF Ia isoform in Rat2 fibroblasts enhances growth in low serum, whereas expression in Swiss 3T3 cells causes morphological transformation and increased saturation density. In FDCP1 myeloid cells, CalDAG-GEF Ia expression increases growth and saturation density in the presence of the diacylglycerol analogs phorbol 12-myristate 13-acetate (PMA), which activates CalDAG-GEF Ia exchange activity. Likewise, in 32Dcl3 myeloblast cells, CalDAG-GEF Ia expression increases cell adherence to fibronectin in response to PMA and calcium ionophore and allows higher saturation densities and prolonged growth on fibronectin-coated plates. These effects were correlated with increased Rap1, but not Ras, protein activation following PMA and calcium ionophore treatment. Our results suggest that Rap1-GTP delivers signals that favor progression through the cell cycle and morphological transformation. The identification of CalDAG-GEF I as a proto-oncogene in BXH-2 acute myeloid leukemia is the first evidence implicating Rap1 signaling in myeloid leukemia.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/genética , Leucemia Mieloide/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , DNA , Fibroblastos/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Leucemia Mieloide/patologia , Camundongos , Dados de Sequência Molecular , Proto-Oncogene Mas , Ratos , Transdução de Sinais , Células Tumorais Cultivadas
7.
Breast Cancer Res Treat ; 62(1): 51-62, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10989985

RESUMO

Genetic ras mutations are infrequent in breast cancer but Ras may be pathologically activated in breast cancer by overexpression of growth factor receptors which signal through Ras. Using a highly sensitive, coupled enzymatic assay, we measured Ras activation in 20 breast cancers, two fibroadenomas, and seven normal breast samples. Ras was highly activated compared to benign tissue in 11 of the 20 cancers; 7 of these 11 cancers expressed both the epidermal growth factor (EGF) and ErbB-2/neu/HER-2 receptors with the remaining four cancers with high Ras activation expressing one of these two receptors. In the other nine cancers, Ras activation was similar to that observed in benign breast tissue with none of these cancers expressing the EGF receptor while one expressed the ErbB-2 receptor. None of the cancers tested had an activating K-ras mutation nor did any of the cancers express a truncated EGF receptor or the c-FMS receptor. The activity of mitogen-activated protein (MAP) kinase was high in the cancers, and reflected the degree of Ras activation. In cultured mammary tumor cell lines, we showed that Ras activation was ligand dependent in cells overexpressing the ErbB-2 receptor. Thus, Ras was abnormally activated in breast cancers overexpressing the EGF and/or ErbB-2 receptors indicating there are sufficient ligands in vivo to activate these receptors, and this work provides a basis for new target-based treatments of this disease.


Assuntos
Neoplasias da Mama/genética , Fibroadenoma/genética , Regulação Neoplásica da Expressão Gênica , Genes ras/genética , Proteínas ras/biossíntese , Feminino , Humanos , Imuno-Histoquímica , Ligantes , Neoplasias Mamárias Animais , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mutação Puntual , Células Tumorais Cultivadas
8.
Cancer Res ; 60(16): 4602-9, 2000 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-10969813

RESUMO

Azoxymethane (AOM)-induced colonic carcinogenesis involves a number of mutations, including those in the K-ras gene and CTNNB1, that codes for beta-catenin. Prior in vitro studies have also demonstrated that wild type p21(K-ras) can be activated by epigenetic events. We identified 15 K-ras mutations in 14 of 84 AOM-induced colonic tumors by three independent methods. By single strand conformational polymorphism, we also observed mutations in 22 of 68 tumors in exon 3 of CTNNB1. A highly sensitive method was then used to measure p21ras activation levels. All tumors assayed possessing K-ras mutations had significantly higher p21ras activation levels (8.8 +/- 1.5%; n = 13) compared with that of control colon (3.7 +/- 0.4; n = 6; P < 0.05) or tumors without such mutations (4.2 +/- 0.4%; n = 70; P < 0.05). Among tumors with wild-type K-ras, there was a subset of tumors (18 of 70) that had significantly higher p21ras activation levels (8.0 +/- 0.9%; n = 18) compared with control colons. In three of four tumors examined with activated wild-type p21ras, we observed increased c-erbB-2 receptor expression and decreased Ras-GAP expression. In contrast, only one of eight tumors examined with wild-type ras and nonactivated p21ras demonstrated these alterations. Mitogen-activated protein kinase (MAPK) activation and cyclooxygenase-2 (COX-2) expression were increased in tumors with mutated or activated wild-type p21ras, compared with their nonactivated counterparts. Although beta-catenin mutations did not alter COX-2 expression or MAPK activity, mutations in either K-ras or beta-catenin significantly increased cyclin D1 expression. In contrast, in tumors with wild-type but activated p21-ras, cyclin D1 expression was not enhanced. Thus, the spectrum of changes in MAPK, COX-2, and cyclin D1 is distinct among tumors with ras or beta-catenin mutations or nonmutational activation of p21ras.


Assuntos
Neoplasias do Colo/genética , Ciclina D1/biossíntese , Isoenzimas/biossíntese , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mutação/fisiologia , Prostaglandina-Endoperóxido Sintases/biossíntese , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Transativadores , Animais , Azoximetano , Carcinógenos , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/metabolismo , Ciclo-Oxigenase 2 , Proteínas do Citoesqueleto/genética , Ativação Enzimática , Genes ras/genética , Masculino , Polimorfismo de Fragmento de Restrição , Proteínas Proto-Oncogênicas p21(ras)/genética , Ratos , Ratos Endogâmicos F344 , beta Catenina
9.
Oncogene ; 19(35): 4029-34, 2000 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-10962559

RESUMO

We have previously isolated variant HL-60 cells that are resistant to cGMP-induced differentiation and showed that they are deficient in proteolytic cleavage and/or carboxyl methylation of Rap 1A (J. Biol. Chem. 269, 32155 - 32161, 1994 and Oncogene 17, 2211 - 2233, 1998). We have now developed an enzyme-based method for assessing Rap 1 activation which is quantitative and provides a measurement of the per cent of Rap molecules in the active GTP-bound state. Using this method, we show that cAMP and cGMP analogs activate Rap 1 in parental HL-60 cells but not in the variant cells and that H-89, a cAMP-dependent protein kinase inhibitor, has no effect on cAMP-induced Rap 1 activation in parental cells. Thus, cAMP activation of Rap 1 in HL-60 cells is likely through a cAMP-regulated guanine nucleotide exchange factor (cAMP-GEF) and since cAMP does not activate Rap 1 in the variant cells, the data suggest that full post-translational processing of Rap 1 is necessary for cAMP-GEF activation of Rap 1. Activation of Rap 1 by cGMP analogs has not been previously found and suggests possible cross-talk between the NO/cGMP signal transduction pathway and Rap 1 signaling. Oncogene (2000) 19, 4029 - 4034.


Assuntos
GMP Cíclico/farmacologia , Células HL-60/efeitos dos fármacos , Proteínas rap1 de Ligação ao GTP/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Cricetinae , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , GMP Cíclico/análogos & derivados , Resistência a Medicamentos , Ativação Enzimática , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Guanosina Trifosfato/metabolismo , Células HL-60/metabolismo , Humanos , Rim , Mesocricetus , Proteínas de Neoplasias/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Sistemas do Segundo Mensageiro , Tionucleotídeos/farmacologia
10.
J Biol Chem ; 275(41): 31921-9, 2000 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-10931830

RESUMO

Cyclic AMP can either activate or inhibit the mitogen-activated protein kinase (MAPK) pathway in different cell types; MAPK activation has been observed in B-Raf-expressing cells and has been attributed to Rap1 activation with subsequent B-Raf activation, whereas MAPK inhibition has been observed in cells lacking B-Raf and has been attributed to cAMP-dependent protein kinase (protein kinase A)-mediated phosphorylation and inhibition of Raf-1 kinase. We found that cAMP stimulated MAPK activity in CHO-K1 and PC12 cells but inhibited MAPK activity in C6 and NB2A cells. In all four cell types, cAMP activated Rap1, and the 95- and 68-kDa isoforms of B-Raf were expressed. cAMP activation or inhibition of MAPK correlated with activation or inhibition of endogenous and transfected B-Raf kinase. Although all cell types expressed similar amounts of 14-3-3 proteins, approximately 5-fold less 14-3-3 was associated with B-Raf in cells in which cAMP was inhibitory than in cells in which cAMP was stimulatory. We found that the cell type-specific inhibition of B-Raf could be completely prevented by overexpression of 14-3-3 isoforms, whereas expression of a dominant negative 14-3-3 mutant resulted in partial loss of B-Raf activity. Our data suggest that 14-3-3 bound to B-Raf protects the enzyme from protein kinase A-mediated inhibition; the amount of 14-3-3 associated with B-Raf may explain the tissue-specific effects of cAMP on B-Raf kinase activity.


Assuntos
AMP Cíclico/análogos & derivados , Proteínas Proto-Oncogênicas c-raf/metabolismo , Tionucleotídeos/farmacologia , Tirosina 3-Mono-Oxigenase/metabolismo , Proteínas 14-3-3 , Animais , Linhagem Celular , Cricetinae , AMP Cíclico/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ativação Enzimática/efeitos dos fármacos , Guanosina Trifosfato/metabolismo , Isoenzimas/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutação , Especificidade de Órgãos , Fosforilação/efeitos dos fármacos , Ligação Proteica , Proteínas Proto-Oncogênicas c-raf/antagonistas & inibidores , Ratos , Transfecção , Tirosina 3-Mono-Oxigenase/genética
11.
Clin Cancer Res ; 6(5): 1804-10, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10815901

RESUMO

Ras is an important cellular switch, relaying growth-promoting signals from the plasma membrane to the nucleus. In cultured cells, Ras is activated by various hematopoietic cytokines and growth factors, but the activation state of Ras in peripheral WBCs and bone marrow cells has not been studied nor has Ras activation been assessed in cells from patients with acute lymphoblastic leukemia (ALL). Using an enzyme-based method, we assessed Ras activation in peripheral WBCs, lymphocytes, and bone marrow cells from normal subjects and from children with T-cell ALL (T-ALL) and B-lineage ALL (B-ALL). In normal subjects, we found mean Ras activations of 14.3, 12.5, and 17.2% for peripheral blood WBCs, lymphocytes, and bone marrow cells, respectively. All three of these values are higher than we have found in other normal human cells, compatible with constitutive activation of Ras by cytokines and growth factors present in serum and bone marrow. In 9 of 18 children with T-ALL, Ras activation exceeded two SDs above the mean of the corresponding cells from normal subjects, whereas in none of 11 patients with B-ALL did Ras show increased activation; activating genetic mutations in ras occur in less than 10% of ALL patients. Thus, Ras is relatively activated in peripheral blood WBCs, lymphocytes, and bone marrow cells compared with other normal human cells, and Ras is activated frequently in T-ALL but not in B-ALL. Increased Ras activation in T-ALL compared with B-ALL may contribute to the more aggressive nature of the former disease.


Assuntos
Células Sanguíneas/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Proteínas ras/metabolismo , Adolescente , Células da Medula Óssea/metabolismo , Linfoma de Burkitt/sangue , Linfoma de Burkitt/metabolismo , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Leucemia-Linfoma de Células T do Adulto/sangue , Leucemia-Linfoma de Células T do Adulto/metabolismo , Leucócitos/metabolismo , Linfócitos/metabolismo , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/sangue
12.
Am J Physiol Cell Physiol ; 278(2): C372-80, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10666033

RESUMO

The small guanine nucleotide-binding protein Ras, activated by peptide mitogens and other stimuli, regulates downstream signaling events to influence transcription. The role of Ras in solute signaling to gene regulation was investigated in the murine inner medullary collecting duct (mIMCD3) cell line. Urea treatment (100-200 mM), but not sham treatment, increased Ras activation 124% at 2 min; the effect of NaCl did not achieve statistical significance. To determine the contribution of Ras activation to urea-inducible signal transduction, mIMCD3 cells were stably transfected with an expression plasmid encoding a dominant negative-acting N17Ras mutant driven by a dexamethasone-inducible (murine mammary tumor virus) promoter. After 24 h of induction, selected cell lines exhibited sufficient N17Ras overexpression to abolish epidermal growth factor- and hypotonicity-mediated signaling to extracellular signal-regulated kinase (ERK) phosphorylation, as determined by immunoblotting. Conditional N17Ras overexpression inhibited urea- and NaCl-inducible ERK phosphorylation by 40-50%, but only at 15 min, and not 5 min, of treatment. N17Ras induction, however, almost completely inhibited urea-inducible Egr-1 transcription, as quantitated by luciferase reporter gene assay, but failed to influence tonicity-inducible (TonE-mediated) transcription. N17Ras overexpression also blocked urea-inducible expression of the transcription factor Gadd153 but did not influence osmotic or urea-inducible apoptosis. In addition, urea treatment induced recruitment of the Ras activator Sos to the plasma membrane. Taken together, these observations suggest a role for Ras signaling in the IMCD cell response to urea stress.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT , Medula Renal/citologia , Sistema de Sinalização das MAP Quinases/fisiologia , Cloreto de Sódio/farmacologia , Ureia/farmacologia , Proteínas ras/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular , Tamanho Celular/fisiologia , Proteínas de Ligação a DNA/genética , Dexametasona/farmacologia , Regulação Enzimológica da Expressão Gênica , Soluções Hipertônicas/farmacologia , Medula Renal/enzimologia , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/enzimologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Vírus do Tumor Mamário do Camundongo , Fosforilação , Proteína Son Of Sevenless de Drosófila/metabolismo , Fator de Transcrição CHOP , Fatores de Transcrição/genética , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia , Transfecção , Proteínas ras/genética
13.
Oncogene ; 19(54): 6324-33, 2000 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-11175347

RESUMO

We have shown that nitric oxide (NO) regulates c-fos gene expression via cGMP-dependent protein kinase (G-kinase), but NO's precise mechanism of action is unclear. We now demonstrate that: (1) NO targets two transcriptional elements in the fos promoter, i.e., the fos AP-1 binding site and the cAMP-response element (CRE); (2) NO activation of these two enhancer elements requires the CRE binding protein CREB because a dominant negative CREB fully inhibits NO transactivation of reporter genes whereas dominant negative Fos or CCAAT enhancer binding proteins have no effect; (3) CREB is phosphorylated by G-kinase in vitro and its phosphorylation increases in vivo when G-kinase is activated either directly by cGMP or indirectly by NO via soluble guanylate cyclase; (4) NO activation of fos promoter elements requires nuclear translocation of G-kinase but not activation of mitogen-activated protein kinases.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Genes fos , Sistema de Sinalização das MAP Quinases , Óxido Nítrico/fisiologia , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Cricetinae , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteínas Quinases Dependentes de GMP Cíclico/genética , Elementos Facilitadores Genéticos , Genes Reporter , Guanilato Ciclase/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutação , Fosforilação , Regiões Promotoras Genéticas , Ativação Transcricional , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno
14.
J Biol Chem ; 274(20): 14306-14, 1999 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-10318852

RESUMO

Phosphatidylinositol (PI) 3-kinase plays an important role in various insulin-stimulated biological responses including glucose transport, glycogen synthesis, and protein synthesis. However, the molecular link between PI 3-kinase and these biological responses is still unclear. We have investigated whether targeting of the catalytic p110 subunit of PI 3-kinase to cellular membranes is sufficient and necessary to induce PI 3-kinase dependent signaling responses, characteristic of insulin action. We overexpressed Myc-tagged, membrane-targeted p110 (p110(CAAX)), and wild-type p110 (p110(WT)) in 3T3-L1 adipocytes by adenovirus-mediated gene transfer. Overexpressed p110(CAAX) exhibited approximately 2-fold increase in basal kinase activity in p110 immunoprecipitates, that further increased to approximately 4-fold with insulin. Even at this submaximal PI 3-kinase activity, p110(CAAX) fully stimulated p70 S6 kinase, Akt, 2-deoxyglucose uptake, and Ras, whereas, p110(WT) had little or no effect on these downstream effects. Interestingly p110(CAAX) did not activate MAP kinase, despite its stimulation of p21(ras). Surprisingly, p110(CAAX) did not increase basal glycogen synthase activity, and inhibited insulin stimulated activity, indicative of cellular resistance to this action of insulin. p110(CAAX) also inhibited insulin stimulated, but not platelet-derived growth factor-stimulated mitogen-activated protein kinase phosphorylation, demonstrating that the p110(CAAX) induced inhibition of mitogen-activated protein kinase and insulin signaling is specific, and not due to some toxic or nonspecific effect on the cells. Moreover, p110(CAAX) stimulated IRS-1 Ser/Thr phosphorylation, and inhibited IRS-1 associated PI 3-kinase activity, without affecting insulin receptor tyrosine phosphorylation, suggesting that it may play an important role as a negative regulator for insulin signaling. In conclusion, our studies show that membrane-targeted PI 3-kinase can mimic a number of biologic effects normally induced by insulin. In addition, the persistent activation of PI 3-kinase induced by p110(CAAX) expression leads to desensitization of specific signaling pathways. Interestingly, the state of cellular insulin resistance is not global, in that some of insulin's actions are inhibited, whereas others are intact.


Assuntos
Resistência à Insulina , Insulina/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Células 3T3 , Animais , Transporte Biológico , Domínio Catalítico , Glucose/metabolismo , Glicogênio Sintase/metabolismo , Camundongos , Mimetismo Molecular , Proteínas de Transporte de Monossacarídeos , Fragmentos de Peptídeos/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais , Suínos
15.
Am J Physiol ; 276(4 Pt 1): G1005-15, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10198345

RESUMO

In the accompanying paper [Khare et al., Am. J. Physiol. 276 (Gastrointest. Liver Physiol. 39): G993-G1004, 1999], activation of protein kinase C-alpha (PKC-alpha) was shown to be involved in the stimulation of phospholipase D (PLD) by 1,25-dihydroxyvitamin D3 [1, 25(OH)2D3] and 12-O-tetradecanoylphorbol 13-acetate (TPA) in Caco-2 cells. Monomeric or heterotrimeric G proteins, as well as pp60(c-src) have been implicated in PLD activation. We therefore determined whether these signal transduction elements were involved in PLD stimulation by 1,25(OH)2D3 or TPA. Treatment with C3 transferase, which inhibits members of the Rho family of monomeric G proteins, markedly diminished the ability of 1,25(OH)2D3, but not TPA, to stimulate PLD. Brefeldin A, an inhibitor of ADP-ribosylation factor proteins, did not, however, significantly reduce the stimulation of PLD by either of these agents. Moreover, 1,25(OH)2D3, but not TPA, activated pp60(c-src) and treatment with PP1, a specific inhibitor of the pp60(c-src) family, blocked the ability of 1,25(OH)2D3 to activate PLD. Pretreatment of cells with pertussis toxin (PTx) markedly reduced the stimulation of PLD by either agonist. PTx, moreover, inhibited the stimulation of pp60(c-src) and PKC-alpha by 1,25(OH)2D3. PTx did not, however, block the membrane translocation of RhoA induced by 1,25(OH)2D3 or inhibit the stimulation of PKC-alpha by TPA. These findings, taken together with those of the accompanying paper, indicate that although 1,25(OH)2D3 and TPA each activate PLD in Caco-2 cells in part via PKC-alpha, their stimulation of PLD differs in a number of important aspects, including the requirement for pp60(c-src) and RhoA in the activation of PLD by 1,25(OH)2D3, but not TPA. Moreover, the requirement for different signal transduction elements by 1,25(OH)2D3 and TPA to induce the stimulation of PLD may potentially underlie differences in the physiological effects of these agents in Caco-2 cells.


Assuntos
Calcitriol/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Isoenzimas/metabolismo , Fosfolipase D/metabolismo , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Células CACO-2 , Ativação Enzimática , Proteínas de Ligação ao GTP/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Cinética , Modelos Biológicos , Proteína Quinase C-alfa , Transdução de Sinais , Proteína rhoA de Ligação ao GTP
16.
J Biol Chem ; 274(14): 9489-93, 1999 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-10092632

RESUMO

Nitric oxide (NO) regulates the expression of multiple genes but in most cases its precise mechanism of action is unclear. We used baby hamster kidney (BHK) cells, which have very low soluble guanylate cyclase and cGMP-dependent protein kinase (G-kinase) activity, and CS-54 arterial smooth muscle cells, which express these two enzymes, to study NO regulation of the human fos promoter. The NO-releasing agent Deta-NONOate (ethanamine-2,2'-(hydroxynitrosohydrazone)bis-) had no effect on a chloramphenicol acetyltransferase (CAT) reporter gene under control of the fos promoter in BHK cells transfected with an empty vector or in cells transfected with a G-kinase Ibeta expression vector. In BHK cells transfected with expression vectors for guanylate cyclase, Deta-NONOate markedly increased the intracellular cGMP concentration and caused a small (2-fold) increase in CAT activity; the increased CAT activity appeared to be from cGMP activation of cAMP-dependent protein kinase. In BHK cells co-transfected with guanylate cyclase and G-kinase expression vectors, CAT activity was increased 5-fold in the absence of Deta-NONOate and 7-fold in the presence of Deta-NONOate. Stimulation of CAT activity in the absence of Deta-NONOate appeared to be largely from endogenous NO since we found that: (i) BHK cells produced high amounts of NO; (ii) CAT activity was partially inhibited by a NO synthase inhibitor; and (iii) the inhibition by the NO synthase inhibitor was reversed by exogenous NO. In CS-54 cells, we found that NO increased fos promoter activity and that the increase was prevented by a guanylate cyclase inhibitor. In summary, we found that NO activates the fos promoter by a guanylate cyclase- and G-kinase-dependent mechanism.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Guanilato Ciclase/metabolismo , Óxido Nítrico/fisiologia , Transcrição Gênica , Animais , Células Cultivadas , Cricetinae , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteína Quinase Dependente de GMP Cíclico Tipo I , Ativação Enzimática , Humanos , Compostos Nitrosos/metabolismo , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-fos/genética , Solubilidade , Ativação Transcricional , Transfecção
17.
Mol Cell Biol ; 18(12): 6983-94, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9819386

RESUMO

Agents which increase the intracellular cyclic GMP (cGMP) concentration and cGMP analogs inhibit cell growth in several different cell types, but it is not known which of the intracellular target proteins of cGMP is (are) responsible for the growth-suppressive effects of cGMP. Using baby hamster kidney (BHK) cells, which are deficient in cGMP-dependent protein kinase (G-kinase), we show that 8-(4-chlorophenylthio)guanosine-3', 5'-cyclic monophosphate and 8-bromoguanosine-3',5'-cyclic monophosphate inhibit cell growth in cells stably transfected with a G-kinase Ibeta expression vector but not in untransfected cells or in cells transfected with a catalytically inactive G-kinase. We found that the cGMP analogs inhibited epidermal growth factor (EGF)-induced activation of mitogen-activated protein (MAP) kinase and nuclear translocation of MAP kinase in G-kinase-expressing cells but not in G-kinase-deficient cells. Ras activation by EGF was not impaired in G-kinase-expressing cells treated with cGMP analogs. We show that activation of G-kinase inhibited c-Raf kinase activation and that G-kinase phosphorylated c-Raf kinase on Ser43, both in vitro and in vivo; phosphorylation of c-Raf kinase on Ser43 uncouples the Ras-Raf kinase interaction. A mutant c-Raf kinase with an Ala substitution for Ser43 was insensitive to inhibition by cGMP and G-kinase, and expression of this mutant kinase protected cells from inhibition of EGF-induced MAP kinase activity by cGMP and G-kinase, suggesting that Ser43 in c-Raf is the major target for regulation by G-kinase. Similarly, B-Raf kinase was not inhibited by G-kinase; the Ser43 phosphorylation site of c-Raf is not conserved in B-Raf. Activation of G-kinase induced MAP kinase phosphatase 1 expression, but this occurred later than the inhibition of MAP kinase activation. Thus, in BHK cells, inhibition of cell growth by cGMP analogs is strictly dependent on G-kinase and G-kinase activation inhibits the Ras/MAP kinase pathway (i) by phosphorylating c-Raf kinase on Ser43 and thereby inhibiting its activation and (ii) by inducing MAP kinase phosphatase 1 expression.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteínas de Ciclo Celular , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Fosfoproteínas Fosfatases , Transdução de Sinais/fisiologia , Animais , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Cricetinae , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Fosfatase 1 de Especificidade Dupla , Ativação Enzimática/fisiologia , Fator de Crescimento Epidérmico/farmacologia , Genes ras/genética , Proteínas Imediatamente Precoces/metabolismo , Fosforilação , Proteína Fosfatase 1 , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Tionucleotídeos/farmacologia , Transfecção/genética
18.
Oncogene ; 17(17): 2211-23, 1998 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-9811452

RESUMO

Variant HL-60 cells resistant to differentiation induced by nitroprusside and cGMP analogs have normal guanylate cyclase and cGMP-dependent protein kinase (G-kinase) activity (J. Biol. Chem. 269, 32155-32161, 1994). We found decreased phosphorylation of a low molecular weight protein (pp23) in the variant cells and by co-migration on two-dimensional polyacrylamide gels, phosphopeptide mapping, immunoprecipitation and immunoblotting, we showed that pp23 was one of three post-translationally modified forms of Rap 1A expressed in HL-60 cells. Using an in vitro transcription/translation system, we studied each of the posttranslational processing steps of Rap 1A and we showed that pp23 represented fully processed Rap 1A. By immunoprecipitation, immunoblotting and 35S-methionine/cysteine incorporation, we showed that the variant cells were deficient in pp23, and thus in fully processed Rap 1A, but that these cells did express normal amounts of completely unprocessed Rap 1A and geranylgeranylated Rap 1A; the lack of Rap 1A processing beyond geranylgeranylation in the variant cells was not secondary to a change in Rap 1A's amino acid sequence. The variant cells had normal carboxyl methyltransferase activity suggesting they are deficient in proteolytic cleavage of Rap 1A. The deficient post-translational processing of Rap 1A had no effect on Rap 1A's subcellular distribution and we found no evidence for altered post-translational processing of H-Ras.


Assuntos
Células HL-60/química , Proteínas de Neoplasias/metabolismo , Fosfoproteínas/metabolismo , Processamento de Proteína Pós-Traducional , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Proteína Quinase Dependente de GMP Cíclico Tipo I , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Ativação Enzimática , Células HL-60/efeitos dos fármacos , Humanos , Indicadores e Reagentes/farmacologia , Peso Molecular , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Nitroprussiato/farmacologia , Mapeamento de Peptídeos , Fosfoproteínas/química , Fosfoproteínas/genética , Fosforilação , RNA Mensageiro/análise
19.
Oncogene ; 17(18): 2403-11, 1998 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-9811472

RESUMO

We have previously documented that transgenic mice expressing SV40 Tag regulated by the rat prostatic steroid-binding protein C3(1) 5'-flanking region display multistage mammary tumorigenesis. To delineate genetic changes associated with mammary tumor progression, comparative genomic hybridization (CGH) was performed. CGH revealed a consistent gain of the telomeric region of chromosome 6. This region contains the Ki-ras proto-oncogene. Analyses of genomic DNA by Southern blot demonstrated up to 40-fold amplification of the Ki-ras gene. Ki-ras amplification was detected in 12, 46 and 68% of tumors from 4, 5 and 6 month old mice, respectively, whereas no amplifications were found in any preneoplastic mammary tissues. Tumors bearing Ki-ras gene amplification exhibited high levels of Ki-ras RNA and protein. The over-expressed Ki-Ras protein in these tumors appeared functionally active as indicated by the elevated MAP kinase activity. These data demonstrate that while Ki-ras amplification might not be an early event, there is a strong association between Ki-ras amplification and over-expression and mammary tumor progression in this model. This study also shows that CGH is a powerful and useful technique for identifying chromosomal copy number changes during tumor progression, and that this model may provide a predictable in vivo system for studying gene amplification.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Amplificação de Genes , Genes ras/genética , Neoplasias Mamárias Animais/genética , Animais , Progressão da Doença , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Hibridização in Situ Fluorescente , Camundongos , Camundongos Transgênicos , Mutação , RNA Mensageiro/metabolismo , Receptor ErbB-2/metabolismo , Proteínas ras/metabolismo
20.
Mol Cell Biochem ; 185(1-2): 111-21, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9746217

RESUMO

We previously described the isolation of a variant subline of HL-60 cells that does not differentiate in response to nitric oxide (NO)-generating agents or to cGMP analogs. The variant cells have normal guanylate cyclase activity and normal NO-induced increases in the intracellular cGMP concentration. We now show that the variant cells have normal cGMP-dependent protein kinase (G-kinase) activity, both by an in vitro and in vivo assay, and using two-dimensional gel electrophoresis we have identified six G-kinase substrates in the parental cells. Of these six proteins, we found considerably less phosphorylation of one of the proteins in the variant cells than in parental cells, both in vitro and in intact cells, and by 35S-methionine/35S-cysteine incorporation we found much less of this protein in the variant cells than in parental cells. The protein is a shared substrate of cAMP-dependent protein kinase (A-kinase); since cAMP analogs still induce differentiation of the variant cells, it appears that the NO/cGMP/G-kinase and cAMP/A-kinase signal transduction pathways share some but not all of the same target proteins in inducing differentiation of HL-60 cells.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , GMP Cíclico/farmacologia , Células HL-60/metabolismo , Óxido Nítrico/metabolismo , Proteínas/metabolismo , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/efeitos dos fármacos , Cisteína/análise , Eletroforese em Gel Bidimensional/métodos , Células HL-60/efeitos dos fármacos , Células HL-60/enzimologia , Humanos , Metionina/análise , Fosforilação/efeitos dos fármacos , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/metabolismo , Proteínas/análise , Radioisótopos de Enxofre/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...