Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Clin. transl. oncol. (Print) ; 11(11): 715-720, nov. 2009. tab
Artigo em Inglês | IBECS | ID: ibc-123701

RESUMO

Transforming growth factor Beta (TGF-Beta) family members are polypeptidic cytokines with pleiotropic physiological properties. In relation to cancer, TGF-Beta exerts a dual tumour-suppressive and oncogenic effect, which is largely dependent on microenvironment stimuli. After activation of TGF-Beta signalling, two pathways can be activated: the canonical one through the mammalian Smad family or the non-canonical one activating, among others, the cellular mitogen-activated protein kinase (MAPK) signalling downstream, which interacts with Smad signalling. During tumorigenesis, cells of many cancer types often lose their response to the tumour-suppressive effects of TGF-Beta, which, in turn, has the opposite effect, acting as an autocrine tumour-promoting factor. In this review, we summarise the current knowledge about this intriguing cytokine, with special emphasis on its immunosuppressive actions (AU)


No disponible


Assuntos
Humanos , Animais , Masculino , Feminino , Imunossupressores/metabolismo , Neoplasias/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Linfócitos T Reguladores/metabolismo , Imunidade Inata , Modelos Biológicos , Transdução de Sinais , Proteínas Smad/metabolismo
2.
Br J Cancer ; 96(10): 1595-604, 2007 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-17486135

RESUMO

Prostate cancer represents a major concern in human oncology and the phytoalexin resveratrol (RES) inhibits growth and proliferation of prostate cancer cells through the induction of apoptosis. In addition, previous data indicate that in oestrogen-responsive human breast cancer cells, RES induces apoptosis by inhibition of the phosphoinositide-3-kinase (PI3K) pathway. Here, using androgen receptor (AR)-positive LNCaP and oestrogen receptor alpha (ERalpha)-expressing PC-3 prostate tumour cells, we have analysed whether the antiproliferative activity of RES takes place by inhibition of the AR- or ERalpha-dependent PI3K pathway. Although RES treatment (up to 150 microM) decreased AR and ERalpha protein levels, it did not affect AR and ERalpha interaction with p85-PI3K. Immunoprecipitation and kinase assays showed that RES inhibited AR- and ERalpha-dependent PI3K activities in LNCaP and PC-3, respectively. Consistently, lower PI3K activities correlated with decreased phosphorylation of downstream targets protein kinase B/AKT (PKB/AKT) and glycogen synthase kinase-3 (GSK-3). GSK-3 dephosphorylation could be responsible for the decreased cyclin D1 levels observed in both cell lines. Importantly, RES markedly decreased PKB/AKT phosphorylation in primary cultures from human prostate tumours, suggesting that the mechanism proposed here could take place in vivo. Thus, RES could have antitumoral activity in androgen-sensitive and androgen-non-sensitive human prostate tumours by inhibiting survival pathways such as that mediated by PI3K.


Assuntos
Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias da Próstata/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Receptores Androgênicos/metabolismo , Receptores de Estrogênio/metabolismo , Estilbenos/farmacologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Masculino , Modelos Biológicos , Fosforilação/efeitos dos fármacos , Neoplasias da Próstata/patologia , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Resveratrol , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas
3.
Carcinogenesis ; 27(5): 1099-104, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16410262

RESUMO

The transcription factor aryl hydrocarbon receptor (AhR) has relevant functions in cell proliferation. Interestingly, the AhR can either promote or inhibit proliferation depending on the cell phenotype. Although recent data reveal potential pathways for AhR signaling in cell proliferation, the mechanisms that regulate its activity in tumor cells remain unknown. Here, we have analyzed promoter hypermethylation as a potential mechanism controlling AhR expression in human tumor cells. AhR promoter CpG methylation was sporadic in a panel of 19 tumor cell lines except for the chronic myeloid leukemia (CML) K562 and the acute lymphoblastic leukemia (ALL) REH. When compared with normal lymphocytes, REH had very low constitutive AhR expression that could be attributed to promoter hypermethylation since treatment with the DNA demethylating agent 5-aza-2'-deoxycitidine (AZA) significantly increased AhR mRNA and protein. These results in leukemia-derived cell lines were further confirmed in primary ALL, where 33% of the patients (7/21) had AhR promoter hypermethylation. Chromatin immunoprecipitation (ChIP) showed that methylation impaired binding of the transcription factor Sp1 to the AhR promoter, thus providing a mechanism for AhR downregulation in REH cells. Therefore, promoter hypermethylation represents a novel epigenetic mechanism downregulating AhR activity in hematological malignancies such as ALL.


Assuntos
Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Regiões Promotoras Genéticas , Receptores de Hidrocarboneto Arílico/genética , Fator de Transcrição Sp1/genética , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Sequência de Bases , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Ilhas de CpG , Decitabina , Humanos , Dados de Sequência Molecular , Ligação Proteica
4.
Mol Cell Biol ; 21(5): 1700-9, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11238907

RESUMO

The aryl hydrocarbon receptor (AHR) is a transcription factor that is highly conserved during evolution and shares important structural features with the Drosophila developmental regulators Sim and Per. Although much is known about the mechanism of AHR activation by xenobiotics, little information is available regarding its activation by endogenous stimuli in the absence of exogenous ligand. In this study, using embryonic primary fibroblasts, we have analyzed the role of proteasome inhibition on AHR transcriptional activation in the absence of xenobiotics. Proteasome inhibition markedly reduced cytosolic AHR without affecting its total cellular content. Cytosolic AHR depletion was the result of receptor translocation into the nuclear compartment, as shown by transient transfection of a green fluorescent protein-tagged AHR and by immunoblot analysis of nuclear extracts. Gel retardation experiments showed that proteasome inhibition induced transcriptionally active AHR-ARNT heterodimers able to bind to a consensus xenobiotic-responsive element. Furthermore, nuclear AHR was transcriptionally active in vivo, as shown by the induction of the endogenous target gene CYP1A2. Synchronized to AHR activation, proteasome inhibition also induced a transient increase in AHR nuclear translocator (ARNT) at the protein and mRNA levels. Since nuclear levels of AHR and ARNT are relevant for AHR transcriptional activation, our data suggest that proteasome inhibition, through a transient increase in ARNT expression, could promote AHR stabilization and accumulation into the nuclear compartment. An elevated content of nuclear AHR could favor AHR-ARNT heterodimers able to bind to xenobiotic-responsive elements and to induce gene transcription in the absence of xenobiotics. Thus, depending on the cellular context, physiologically regulated proteasome activity could participate in the control of endogenous AHR functions.


Assuntos
Fibroblastos/metabolismo , Complexos Multienzimáticos/antagonistas & inibidores , Ativação Transcricional , Células 3T3 , Transporte Ativo do Núcleo Celular , Animais , Northern Blotting , Células Cultivadas , Cisteína Endopeptidases , Citocromo P-450 CYP1A2/metabolismo , Citosol/metabolismo , Fibroblastos/citologia , Proteínas de Fluorescência Verde , Immunoblotting , Ligantes , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Complexo de Endopeptidases do Proteassoma , RNA Mensageiro/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transcrição Gênica , Transfecção
5.
Nat Genet ; 23(2): 176-84, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10508513

RESUMO

Gadd45a-null mice generated by gene targeting exhibited several of the phenotypes characteristic of p53-deficient mice, including genomic instability, increased radiation carcinogenesis and a low frequency of exencephaly. Genomic instability was exemplified by aneuploidy, chromosome aberrations, gene amplification and centrosome amplification, and was accompanied by abnormalities in mitosis, cytokinesis and growth control. Unequal segregation of chromosomes due to multiple spindle poles during mitosis occurred in several Gadd45a -/- cell lineages and may contribute to the aneuploidy. Our results indicate that Gadd45a is one component of the p53 pathway that contributes to the maintenance of genomic stability.


Assuntos
Proteínas/genética , Animais , Apoptose/genética , Ciclo Celular/genética , Ciclo Celular/fisiologia , Divisão Celular/genética , Transformação Celular Neoplásica/genética , Senescência Celular , Centrossomo/metabolismo , Embrião de Mamíferos/citologia , Feminino , Fibroblastos/citologia , Fibroblastos/fisiologia , Fase G1 , Raios gama/efeitos adversos , Deleção de Genes , Genes ras/genética , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias/etiologia , Neoplasias/genética , Fenótipo , Proteínas/fisiologia , Hiperplasia do Timo/genética , Hiperplasia do Timo/patologia , Proteínas GADD45
6.
Toxicol Sci ; 47(1): 86-92, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10048156

RESUMO

The aryl hydrocarbon receptor (AhR) mediates many of the biological effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and transcriptional activation of genes encoding a number of xenobiotic metabolizing enzymes. Prenatal exposure of mice to TCDD causes severe alterations in embryo and fetal development, including hydronephrosis and cleft palate. However, the mechanisms underlying these effects are unclear. In this work, the teratogenicity of TCDD in AhR-null mice was evaluated to determine if this effect is mediated by the AhR. Homozygous wild-type (+/+) or AhR-null (-/-) female mice were mated with males of the same genotype overnight. On gestation day (GD)-10, mice were intubated orally with either corn oil (vehicle control) or 25 micrograms/kg TCDD. Fetuses were examined on GD18 for visceral and skeletal alterations. For non-TCDD-exposed litters, all developmental endpoints were comparable between genotypes, with the exception of a lower incidence of large interfrontal bones in (-/-) mice. For TCDD-exposed litters, (+/+) fetuses had a significantly greater incidence of cleft palate, hydronephrosis, small kidneys, tortuous ureters and greater dilation of the renal pelves and ureters compared to (-/-) fetuses. Interestingly, an increased resorption rate was observed in (-/-) fetuses exposed to TCDD. Results from this work demonstrate that fetal development per se is generally unaffected by the absence of the AhR or that other genes may have compensated for the loss of the AhR. More importantly, these data indicate that the AhR mediates TCDD-induced teratogenicity. Further, since a higher percentage of resorptions was observed in (-/-) litters from TCDD-treated dams, it is possible that AhR-independent mechanisms contribute to TCDD-induced developmental toxicity.


Assuntos
Anormalidades Múltiplas/induzido quimicamente , Dibenzodioxinas Policloradas/toxicidade , Receptores de Hidrocarboneto Arílico/fisiologia , Teratogênicos/toxicidade , Animais , Feminino , Homozigoto , Troca Materno-Fetal , Camundongos , Gravidez , Receptores de Hidrocarboneto Arílico/genética
7.
Br J Clin Pharmacol ; 46(2): 151-6, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9723824

RESUMO

AIMS: Dihydropyrimidine dehydrogenase (DPD) catalyses the reduction of pyrimidines, including the anticancer agent 5-fluorouracil (5FU). Impaired 5FU degradation, through low DPD activity, has led to severe, life-threatening or fatal toxicity after administration of 5FU. Complete DPD deficiency is associated with the inherited metabolic disease thymine uraciluria. Several mutations in the gene encoding DPD have recently been identified, but the phenotype-genotype concordance of these alterations in the general population has not been reported. METHODS: Mononuclear cells were isolated from whole blood and DPD activity was determined after ex vivo incubation with 14C-5FU followed by h.p.1.c. analysis of 5FU metabolites. Analysis of mutations in the DPD gene at an exon splice site, codons 534, 543, and 732, and a deletion at base 1897 (deltaC1897) were performed in 30 subjects with the lowest and 30 subjects with the highest enzyme activity using PCR-RFLP. RESULTS: DPD activity was measured in 226 Caucasian subjects and was highly variable (range 19.1-401.4 pmol min(-1)mg(-1) protein). Mutations were frequently observed at codons 543 (allele frequency 28%), 732 (allele frequency 5.8%), and 534 (allele frequency 0.8%), but were not associated with low DPD activity. There were no splice site or deltaC1897 mutations found in this population. CONCLUSIONS: The five mutations analysed in this study are insufficient for identification of patients at risk for 5FU toxicity or thymine uraciluria. Both the splice site mutation and deltaC1897 are relatively rare in the general Caucasian population. Therefore, identification of further molecular alterations is required to facilitate the use of DPD analysis in genetic diagnosis and cancer therapeutics.


Assuntos
Leucócitos Mononucleares/enzimologia , Oxirredutases/genética , População Branca/genética , Alelos , Di-Hidrouracila Desidrogenase (NADP) , Feminino , Humanos , Masculino , Oxirredutases/metabolismo , Mutação Puntual , Escócia , Fumar
8.
Mol Pharmacol ; 54(2): 313-21, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9687573

RESUMO

The aryl hydrocarbon receptor (AHR) is believed to mediate many of the toxic, carcinogenic, and teratogenic effects of environmental contaminants such as dioxins, polycyclic aromatic hydrocarbons, and polyhalogenated biphenyls. Ligands for the AHR have been shown to influence cell proliferation, differentiation, and apoptosis, but the mechanism by which the AHR affects the cell cycle is not known. Increased levels of mature transforming growth factor-beta (TGFbeta) has been correlated with reduced cell proliferation and increased rates of apoptosis and fibrosis. Based on the increase in portal fibrosis and small liver size observed in AHR-null (Ahr-/-) mice, the relationship between TGFbeta expression and apoptosis in this mouse line was analyzed. Livers from Ahr-/- mice had marked increase in active TGFbeta1 and TGFbeta3 proteins and elevated numbers of hepatocytes undergoing apoptosis compared with wild-type mice. Furthermore, increases in TGFbeta and apoptotic cells were found in the portal areas of the liver, where fibrosis is found in the Ahr-/- mice. In vitro, primary hepatocyte cultures from Ahr-/- mice exhibited a high number of cells in later stages of apoptosis and an elevated secretion of active TGFbeta into the media compared with cultures from wild-type mice, which have previously been shown to secrete only latent forms of the molecule. Conditioned media from Ahr-/- hepatocytes stimulated apoptosis in cultured hepatocytes from wild-type mice. Taken together, these findings suggest that the phenotypic abnormalities in Ahr-/- mice could be mediated in part by abnormal levels of active TGFbeta and altered cell cycle control.


Assuntos
Apoptose/fisiologia , Fígado/patologia , Receptores de Hidrocarboneto Arílico/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Meios de Cultivo Condicionados , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Vison , Receptores de Hidrocarboneto Arílico/genética
9.
Vet Pathol ; 34(6): 605-14, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9396142

RESUMO

We have analyzed the possible role of the aryl-hydrocarbon receptor (AHR) in the aging process of mice using a homozygous null mouse (Ahr-/-) line as a model. We studied 52 male and female Ahr-/- mice aged from 6-13 months. Forty-six percent died or were ill by 13 months of age. Ahr-/- mice developed age-related lesions in several organs, some of which were apparent after only 9 months of age. Cardiovascular alterations included cardiomyopathy (100%) with hypertrophy and focal fibrosis. Vascular hypertrophy and mild fibrosis were found in the portal areas of the liver (81%), and vascular hypertrophy and mineralization were common in the uterus (70%). Gastric hyperplasia that progressed with age into polyps was evident in the pylorus of 71% of the mice over 9 months of age. Ahr-/- mice had T-cell deficiency in their spleens but not in other lymphoid organs. The immune system deficiency described previously could be the origin for the rectal prolapse found in 48% of the null mice, associated with Helicobacter hepaticus infection. In the dorsal skin (53% incidence), severe, localized, interfollicular and follicular epidermal hyperplasia, with hyperkeratosis and acanthosis, and marked dermal fibrosis, associated with the presence of anagenic hair follicles, were also evident. None of these lesions were found in 42 control (Ahr +/+ or +/-) mice of similar ages. These observations suggest that the AHR protein, in the absence of an apparent exogenous (xenobiotic) ligand, plays an important role in physiology and homeostasis in major organs in mice, and further supports an evolutionary conserved role for this transcription factor.


Assuntos
Envelhecimento/patologia , Doenças Cardiovasculares/patologia , Hepatopatias/patologia , Receptores de Hidrocarboneto Arílico/deficiência , Dermatopatias/patologia , Gastropatias/patologia , Envelhecimento/fisiologia , Animais , Vasos Sanguíneos/patologia , Doenças Cardiovasculares/etiologia , Sistema Cardiovascular/patologia , Sistema Digestório/patologia , Modelos Animais de Doenças , Feminino , Folículo Piloso/ultraestrutura , Homeostase , Hipertrofia/etiologia , Hipertrofia/patologia , Hepatopatias/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Genéticos , Miocárdio/patologia , Fenótipo , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/fisiologia , Pele/patologia , Dermatopatias/etiologia , Baço/patologia , Gastropatias/etiologia , Linfócitos T/patologia , Útero/patologia
10.
Cancer Res ; 57(14): 2835-8, 1997 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-9230184

RESUMO

Livers from aryl hydrocarbon receptor-null mice showed a 3-fold increase in retinoids and a 65% decrease in retinoic acid metabolism. Levels of expression of the retinoic acid 4-hydroxylase, P450RAI, did not change, whereas cytochrome P4501A2 levels were lower in the null mouse, as shown earlier; however, this enzyme was found not to be active toward retinoic acid. These data suggest that aryl hydrocarbon receptor controls retinoic acid catabolism, through modulation of an unidentified target gene. Aldehyde dehydrogenases 1 and 2 were down-regulated markedly in the aryl hydrocarbon receptor-deficient mouse liver. 2,3,7,8-Tetrachlorodibenzo-p-dioxin induced cytochrome P4501A2 but not the aldehyde dehydrogenases in wild-type mice, suggesting that aryl hydrocarbon receptor is not involved directly in the down-regulation of this gene. Transglutaminase II, a retinoic acid-responsive gene product, was increased 2-fold, consistent with the liver fibrosis phenotype observed in the null mice. These findings suggest a molecular connection between xenobiotic-activated receptor signaling and retinoid homeostasis.


Assuntos
Fígado/metabolismo , Receptores de Hidrocarboneto Arílico/fisiologia , Retinoides/metabolismo , Tretinoína/metabolismo , Animais , Masculino , Camundongos , Camundongos Knockout , Dibenzodioxinas Policloradas/toxicidade , Receptores de Hidrocarboneto Arílico/genética
11.
Pharmacogenetics ; 7(2): 161-3, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9170156

RESUMO

Dihydropyrimidine dehydrogenase (DPD) is the initial and rate-limiting enzyme in pyrimidine catabolism. DPD deficiency is associated with an increased risk of toxicity in cancer patients receiving 5-fluorouracil (5-PU) treatment. DPD deficiency causes an inborn error of metabolism called thymine-uraciluria that is in some instances associated with convulsive disorders and developmental delay in children. We have studied the molecular mechanism accounting for DPD deficiency in a Pakistani pedigree having 2-year-old child with thymine-uraciluria and exhibiting some degree of motor impairment and developmental delay. A common splice mutation was found in the patient's dihydropyrimidine dehydrogenase (DPYD) gene that produces a mutant mRNA resulting in the complete lack of DPD protein and activity in lymphocytes and primary fibroblast. This trait segregated in the family following a typical Mendelian distribution. Surprisingly, the patient's brother also had thymine-uraciluria and was homozygous for the splicing mutation but was clinically asymptomatic. Sequence tagged sites (STS) linkage analyses within 5 megabases of telomeric and centromeric DNA surrounding the DPYD gene revealed no allelic polymorphism between the two brothers. These results suggest that DPD deficiency might not be the only cause of the more severe clinical phenotypes observed in certain thymine-uraciluria patients and that an incomplete correlation between phenotype and genotype is present in the population.


Assuntos
Oxirredutases/deficiência , Polimorfismo de Fragmento de Restrição , Erros Inatos do Metabolismo da Purina-Pirimidina/genética , Timina/urina , Uracila/urina , Pré-Escolar , Di-Hidrouracila Desidrogenase (NADP) , Genótipo , Humanos , Masculino , Dados de Sequência Molecular , Mutação , Paquistão/etnologia , Fenótipo , Splicing de RNA , Sitios de Sequências Rotuladas
12.
Toxicol Appl Pharmacol ; 140(1): 173-9, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8806883

RESUMO

Acute exposure of mammals to the environmental pollutant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) results in a diverse set of toxicologic and pathologic effects. The mechanism of some of these effects has been studied extensively in vitro and correlative studies have indicated the involvement of a transcription factor known as the aryl hydrocarbon receptor (AHR). However, a definitive association of the AHR with TCDD-mediated toxicity has been difficult to establish due to the diversity of effects and the ubiquitous expression of this receptor. In an effort to distinguish AHR-mediated TCDD toxicities from those resulting from alternative pathways, we have made use of the recently described AHR-deficient mouse that was generated by locus-specific homologous recombination in embryonic stem cells. Present studies demonstrate that AHR-deficient mice are relatively unaffected by doses of TCDD (2000 micrograms/kg) 10-fold higher than that found to induce severe toxic and pathologic effects in littermates expressing a functional AHR. Analyses of liver, thymus, heart, kidney, pancreas, spleen, lymph nodes, and uterus from AHR-deficient mice identified no significant TCDD-induced lesions. The resistance of AHR-deficient mice to TCDD-induced thymic atrophy appears restricted to processes involving AHR since the corticosteroid dexamethasone rapidly and efficiently induced cortical depletion in both AHR-deficient and normal littermate control mice. Taken together these results suggest that the pathological changes induced by TCDD in the liver and thymus are mediated entirely by the AHR. However, it is important to note that at high doses of TCDD, AHR-deficient mice displayed limited vasculitis and scattered single cell necrosis in their lungs and livers, respectively. The mechanism(s) responsible for these apparently receptor-independent processes remain unclear but may involve novel, alternative pathways for TCDD-induced toxicity.


Assuntos
Dibenzodioxinas Policloradas/toxicidade , Receptores de Hidrocarboneto Arílico/deficiência , Timo/efeitos dos fármacos , Animais , Dexametasona/uso terapêutico , Feminino , Injeções Intraperitoneais , Fígado/efeitos dos fármacos , Fígado/patologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Camundongos , Camundongos Knockout , Tamanho do Órgão/efeitos dos fármacos , Timo/patologia
14.
Mol Cell Biol ; 15(6): 3012-22, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7539101

RESUMO

To gain insight into the function of peroxisome proliferator-activated receptor (PPAR) isoforms in rodents, we disrupted the ligand-binding domain of the alpha isoform of mouse PPAR (mPPAR alpha) by homologous recombination. Mice homozygous for the mutation lack expression of mPPAR alpha protein and yet are viable and fertile and exhibit no detectable gross phenotypic defects. Remarkably, these animals do not display the peroxisome proliferator pleiotropic response when challenged with the classical peroxisome proliferators, clofibrate and Wy-14,643. Following exposure to these chemicals, hepatomegaly, peroxisome proliferation, and transcriptional-activation of target genes were not observed. These results clearly demonstrate that mPPAR alpha is the major isoform required for mediating the pleiotropic response resulting from the actions of peroxisome proliferators. mPPAR alpha-deficient animals should prove useful to further investigate the role of this receptor in hepatocarcinogenesis, fatty acid metabolism, and cell cycle regulation.


Assuntos
Microcorpos/fisiologia , Receptores Citoplasmáticos e Nucleares/deficiência , Fatores de Transcrição/deficiência , Animais , Sequência de Bases , Clofibrato/farmacologia , DNA Complementar , Fígado/fisiologia , Fígado/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microcorpos/efeitos dos fármacos , Microcorpos/ultraestrutura , Microscopia Eletrônica , Dados de Sequência Molecular , Pirimidinas/farmacologia , RNA/análise , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...