Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Opin Pharmacol ; 38: 59-64, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29525720

RESUMO

In spite of the recent advancement in the molecular characterization of malignant gliomas and medulloblastomas, the treatment of primary brain tumors remains suboptimal. The use of small molecule inhibitors of intracellular signaling pathways, inhibitors of angiogenesis, and immunotherapic agents is limited by systemic adverse effects, limited brain penetration, and, in some cases, lack of efficacy. Thus, adjuvant chemo-therapy and radiotherapy still remain the gold standard in the treatment of grade-IV astrocytoma (glioblastoma multiforme) and medulloblastoma. We review evidence that supports the development of mGlu3 receptor antagonists as add-on drugs in the treatment of malignant gliomas. These drugs appear to display pleiotropic effect on tumor cells, affecting proliferation, differentiation, and response to chemotherapy. mGlu1 and mGlu4 receptors could also be targeted by potential anticancer agents in the treatment of malignant gliomas and medulloblastoma, but extensive research is required for target validation.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Meduloblastoma/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Humanos , Meduloblastoma/tratamento farmacológico
2.
PLoS One ; 4(8): e6591, 2009 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-19672295

RESUMO

Metabotropic glutamate (mGlu) receptors have been considered potential targets for the therapy of experimental parkinsonism. One hypothetical advantage associated with the use of mGlu receptor ligands is the lack of the adverse effects typically induced by ionotropic glutamate receptor antagonists, such as sedation, ataxia, and severe learning impairment. Low doses of the mGlu2/3 metabotropic glutamate receptor agonist, LY379268 (0.25-3 mg/kg, i.p.) increased glial cell line-derived neurotrophic factor (GDNF) mRNA and protein levels in the mouse brain, as assessed by in situ hybridization, real-time PCR, immunoblotting, and immunohistochemistry. This increase was prominent in the striatum, but was also observed in the cerebral cortex. GDNF mRNA levels peaked at 3 h and declined afterwards, whereas GDNF protein levels progressively increased from 24 to 72 h following LY379268 injection. The action of LY379268 was abrogated by the mGlu2/3 receptor antagonist, LY341495 (1 mg/kg, i.p.), and was lost in mGlu3 receptor knockout mice, but not in mGlu2 receptor knockout mice. In pure cultures of striatal neurons, the increase in GDNF induced by LY379268 required the activation of the mitogen-activated protein kinase and phosphatidylinositol-3-kinase pathways, as shown by the use of specific inhibitors of the two pathways. Both in vivo and in vitro studies led to the conclusion that neurons were the only source of GDNF in response to mGlu3 receptor activation. Remarkably, acute or repeated injections of LY379268 at doses that enhanced striatal GDNF levels (0.25 or 3 mg/kg, i.p.) were highly protective against nigro-striatal damage induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine in mice, as assessed by stereological counting of tyrosine hydroxylase-positive neurons in the pars compacta of the substantia nigra. We speculate that selective mGlu3 receptor agonists or enhancers are potential candidates as neuroprotective agents in Parkinson's disease, and their use might circumvent the limitations associated with the administration of exogenous GDNF.


Assuntos
Corpo Estriado/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/biossíntese , Neurônios/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Aminoácidos/farmacologia , Animais , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Corpo Estriado/citologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Hibridização In Situ , Camundongos , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , Receptores de Glutamato Metabotrópico/agonistas
3.
J Neurochem ; 106(2): 551-9, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18410509

RESUMO

Trophic deprivation contributes to astrocyte damage that occurs in acute and chronic neurodegenerative disorders. Unraveling the underlying mechanisms may pave way to novel cytoprotective strategies. Cultured mouse astrocytes responded to trophic deprivation with a large and transient increase in the expression of p21(ras), which was secondary to an enhanced formation of reactive oxygen species (ROS) detected by cytofluorimetric analysis after preloading with 2',7'-dichlorofluorescein diacetate. The increase in p21(ras) levels was largely attenuated by the reducing agent, N-acetylcysteine, which was proven to reduce ROS formation in astrocytes subjected to serum deprivation. We extended the analysis to the Ha-Ras isoform, which has been implicated in mechanisms of cytotoxicity. We found that serum deprivation enhanced the expression and activity of Ha-Ras without changing Ha-Ras mRNA levels. The increase in Ha-Ras levels was sensitive to the protein synthesis inhibitor, cycloheximide, suggesting that serum deprivation increases translation of preformed Ha-Ras mRNA. The late decline in Ha-Ras levels observed after 60 min was prevented by the proteasome inhibitor, MG132, as well as by the selective mitogen-activated protein kinase (MAPK) inhibitor, PD98059. Serum deprivation led to the activation of the MAPK pathway in cultured astrocytes, as shown by an increase in phosphorylated extracellular signal-regulated kinase 1/2 levels after 5 and 30 min. Finally, using the siRNA technology, we found that an acute knock-down of Ha-Ras was protective against astrocyte damage induced by serum deprivation. We conclude that cultured astrocytes respond to trophic deprivation with an increased expression in Ha-Ras, which is limited by the concomitant activation of the MAPK pathway, but is nevertheless involved in the pathophysiology of cell damage.


Assuntos
Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Meios de Cultura Livres de Soro/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Oncogênica p21(ras)/metabolismo , Análise de Variância , Animais , Células Cultivadas , Córtex Cerebral/citologia , Cicloeximida/farmacologia , Inibidores Enzimáticos/farmacologia , Fluoresceínas/metabolismo , Camundongos , Proteína Oncogênica p21(ras)/genética , Inibidores da Síntese de Proteínas/farmacologia , RNA Interferente Pequeno/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sais de Tetrazólio , Tiazóis , Fatores de Tempo
4.
J Neurochem ; 104(6): 1588-98, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17988238

RESUMO

We examined the effect of Wnt1 and Wnt7a on cell proliferation using undifferentiated PC12 cells, which originate from the neural crest and are widely employed as a neuronal cell model. Heterologous expression of Wnt1 enhanced [3H]thymidine incorporation and expression of cyclin D1 and cylin E in PC12 cells. Opposite effects were observed in PC12 cells expressing Wnt7a. Searching for the mechanisms underlying the opposite effects of Wnt1 and Wnt7a on PC12 cell proliferation, we examined the activation of the canonical beta-catenin/T-cell-lymphoid enhancer-binding protein transcription factor pathway and the 'calcium pathway' by co-transfecting the cells with a reporter gene controlled by either T-cell-lymphoid enhancer-binding protein transcription factor or the calcium-activated transcription factor, NFAT. Wnt1 and Wnt7a activated both pathways, but to a different extent. While Wnt1 preferentially activated the calcium pathway, Wnt7a mainly activated the canonical pathway. Pharmacological inhibition of protein kinase C, which is a component of the calcium pathway, abrogated the increase in cell proliferation induced by Wnt1 without affecting the antiproliferative action of Wnt7a. The action of Wnt7a was instead occluded by lithium ions, which mimic the activation of the canonical pathway, and was largely reduced by Dickkopf-1, which acts as an inhibitor of the canonical pathway. In addition, expression of a constitutively active mutant of beta-catenin potently activated the canonical Wnt pathway and reduced [3H]thymidine incorporation. These data challenge the view that the canonical Wnt pathway invariably supports cell growth and suggest that, at least in PC12 cells, cell proliferation is regulated by the balance between the calcium/protein kinase C pathway and the canonical pathway.


Assuntos
Neurônios/metabolismo , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/fisiologia , Proteínas Wnt/metabolismo , Proteína Wnt1/metabolismo , beta Catenina/metabolismo , Animais , Cálcio/metabolismo , Divisão Celular/fisiologia , Elementos Facilitadores Genéticos/genética , Inibidores Enzimáticos/farmacologia , Luciferases/genética , Neurônios/citologia , Células PC12 , Proteína Quinase C/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Ratos , Timidina/farmacocinética , Transfecção , Trítio , Proteínas Wnt/genética , Proteína Wnt1/genética
5.
Neuropharmacology ; 53(4): 473-80, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17675103

RESUMO

Functional mGlu receptor subtypes are found in stem/progenitor cells, and regulate proliferation, differentiation, and survival of these cells. Activation of mGlu5 receptors supports self-renewal of embryonic stem cells, which are pluripotent cells isolated from the blastocyst capable of generating all the body's cell lineages, including germ cells. Differentiation of embryonic stem cells into embryoid bodies is associated with the induction of mGlu4 receptors, the activation of which drives cell differentiation towards the mesoderm and endoderm lineages. Different mGlu receptor subtypes, mGlu3 and mGlu5 receptors in particular, are found in neural stem cells (stem cells resident in the CNS that give rise to neurons, astrocytes or oligodendrocytes) isolated from the developing brain or from regions of persistent neurogenesis of the adult brain (e.g. the subventricular zone lining the wall of the lateral ventricles). The evidence that activation of mGlu3 and mGlu5 receptors stimulates proliferation of these cells is particularly interesting because of the similarities between neural stem cells and putative cancer stem cells that support the growth of malignant gliomas. A link among mGlu receptors, stem cells and cancer is supported by the finding that mGlu4 receptors are expressed by cerebellar granule cell neuroprogenitors, which are the putative cells of origin of medulloblastomas. The study of mGlu receptors in stem/progenitor cells has potential applications in the optimisation of protocols of cell expansion and differentiation aimed at cell replacement strategies, and may gain new insights into the pathophysiology of neurodevelopmental disorders and brain tumours.


Assuntos
Células-Tronco Embrionárias/fisiologia , Receptores de Glutamato Metabotrópico/fisiologia , Células-Tronco/fisiologia , Adulto , Animais , Encéfalo/embriologia , Humanos , Receptor de Glutamato Metabotrópico 5
6.
Am J Nephrol ; 27(4): 360-5, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17556836

RESUMO

BACKGROUND: Anorexia is frequently found in end-stage renal disease and is a reliable predictor of morbidity and mortality in hemodialysis (HD) patients. The pathogenesis of anorexia is complex and the appetite-modulating hormone ghrelin could be involved. Two forms of circulating ghrelin have been described: acylated ghrelin (<10% of circulating ghrelin) which promotes food intake, and des-acyl ghrelin which induces a negative energy balance. The aim of this cross-sectional study is to clarify whether anorexia and body weight change in HD patients relate to plasma des-acyl ghrelin levels. METHODS: 34 HD patients and 15 healthy controls were studied. The presence of anorexia was assessed by a questionnaire. Serum des-acyl ghrelin was measured in HD patients and in 15 body mass index-, sex- and age-matched controls by ELISA. Energy intake was assessed by a 3-day dietary diary, and fat-free mass (FFM) was evaluated by body impedance analysis. Data have been statistically analyzed and are presented as mean +/- SD. RESULTS: 14 patients (41%) were found to be anorexic, and 20 patients (59%) non-anorexic. Energy intake (kcal/day) was significantly lower in anorexic than in non-anorexic patients (1,682 +/- 241 vs. 1,972.50 +/- 490; p < 0.05). FFM (%) was lower in anorexic than in non-anorexic patients (65.8 +/- 4.4 vs. 70.9 +/- 8.7; p = 0.05). Plasma des-acyl ghrelin levels (fmol/ml) were significantly higher in HD patients than in controls (214.88 +/- 154.24 vs. 128.93 +/- 51.07; p < 0.05), and in anorexic HD patients than in non-anorexic (301.7 +/- 162.4 vs. 159.1 +/- 115.5; p < 0.01). CONCLUSION: Anorexia is highly prevalent among HD patients and des-acyl ghrelin could be involved in its pathogenesis.


Assuntos
Anorexia/sangue , Falência Renal Crônica/sangue , Hormônios Peptídicos/sangue , Diálise Renal/efeitos adversos , Idoso , Anorexia/etiologia , Estudos de Casos e Controles , Estudos Transversais , Feminino , Grelina , Humanos , Falência Renal Crônica/complicações , Falência Renal Crônica/terapia , Masculino , Pessoa de Meia-Idade
7.
J Neurochem ; 99(1): 299-307, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16987252

RESUMO

We have shown that endogenous activation of type 5 metabotropic glutamate (mGlu5) receptors supports the maintenance of a pluripotent, undifferentiated state in D3 mouse embryonic stem cells cultured in the presence of leukaemia inhibitory factor (LIF). Here, we examined the interaction between LIF and mGlu5 receptors using as a read-out the immediate early gene, c-Myc. The selective mGlu5 receptor antagonist, 2-methyl-6-(phenylenthynyl)pyridine (MPEP; 1 mum), reduced the increase in c-Myc protein levels induced by LIF by enhancing c-Myc ubiquitination. A reduction in c-Myc levels was also observed following small interfering RNA-mediated mGlu5 receptor gene silencing. MPEP reduced glycogen synthase kinase-3beta phosphorylation on Ser9, but increased phosphorylation of the phosphatidylinositol-3-kinase (PI-3-K) substrate, AKT. In our hands, activated PI-3-K reduced the stability of c-Myc, because (i) the PI-3-K inhibitor, LY294002, prevented the reduction in c-Myc levels induced by MPEP; and (ii) over-expression of AKT promoted c-Myc ubiquitination. All effects of MPEP were mimicked by protein kinase C (PKC) inhibitors and reversed by the PKC activator, tetradecanoylphorbol-13-acetate. We conclude that endogenous activation of mGlu5 receptors sustains the increase in c-Myc induced by LIF in embryonic stem cells by inhibiting both glycogen synthase kinase-3beta and PI-3-K, both effects resulting from the activation of PKC.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Genes myc , Interleucina-6/fisiologia , Receptores de Glutamato Metabotrópico/fisiologia , Células-Tronco/fisiologia , Animais , Células Cultivadas , Embrião de Mamíferos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Fator Inibidor de Leucemia , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Piridinas/farmacologia , Receptor de Glutamato Metabotrópico 5 , Células-Tronco/citologia
8.
J Neurosci ; 26(32): 8388-97, 2006 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-16899734

RESUMO

Moving from the evidence that activation of type 4 metabotropic glutamate (mGlu4) receptors inhibits proliferation and promotes differentiation of cerebellar granule cell neuroprogenitors, we examined the expression and function of mGlu4 receptors in medulloblastoma cells. mGlu4 receptors were expressed in 46 of 60 human medulloblastoma samples. Expression varied in relation to the histotype (nodular desmoplastic>classic>>large-cell anaplastic) and was inversely related to tumor severity, spreading, and recurrence. mGlu4 receptors were also found in D283med, D341med, and DAOY medulloblastoma cell lines, where receptor activation with the selective enhancer PHCCC inhibited adenylyl cyclase and the phosphatidylinositol-3-kinase pathway without affecting the mitogen-activated protein kinase, Sonic Hedgehog, and Wnt pathways. Interestingly, mGlu4 receptor activation reduced DNA synthesis and cell proliferation in all three cell lines. This effect was abrogated by the phosphatidylinositol-3-kinase inhibitor LY294002 [2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one]. In in vivo experiments, repeated subcutaneous injections of N-phenyl-7-(hydroxyimino)cyclopropa[b]chromen-1a-carboxamide (PHCCC) reduced the growth of D283med and DAOY cell xenografts in nude mice. More remarkably, subcutaneous or intracranial injections of PHCCC during the first week of life prevented the development of medulloblastomas in mice lacking one Patched-1 allele and x-irradiated 1 d after birth. These data suggest that mGlu4 receptor enhancers are promising drugs for the treatment of medulloblastomas.


Assuntos
Benzopiranos/administração & dosagem , Meduloblastoma/metabolismo , Meduloblastoma/prevenção & controle , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Meduloblastoma/patologia , Camundongos , Transdução de Sinais/efeitos dos fármacos , Resultado do Tratamento
9.
Neuropharmacology ; 51(3): 606-11, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16806298

RESUMO

The mGlu5 receptor is the only metabotropic glutamate receptor subtype expressed by mouse embryonic stem (ES) cells grown under non-differentiating conditions [Cappuccio, I., Spinanti, P. Porcellini, A., Desiderati, F., De Vita, T., Storto, M., Capobianco, L., Battaglia, G., Nicoletti, F., Melchiorri, D., 2005. Endogenous activation of mGlu5 metabotropic glutamate receptors supports self-renewal of cultured mouse embryonic stem cells. Neuropharmacology 1, 196-205]. We now report that ES cells differentiating into embryoid bodies (EBs) progressively lose mGlu5 receptors and begin to express mGlu4 receptors at both mRNA and proteinc level. A 4-day treatment of EBs with the mGlu4 receptor agonist, L-2-amino-4-phosphonobutanoate (L-AP4), increased mRNA levels of the mesoderm marker, brachyury and the endoderm marker, H19, and decreased the expression of the transcript for the primitive ectoderm marker, fibroblast-growth factor-5 (FGF-5). These effects were prevented by the mGlu4 receptor antagonists, alpha-methylserine-O-phosphate (MSOP). Plating of EBs for 4 days in vitro in ITSFn medium induced cell differentiation towards a neural lineage, as reflected by the expression of the intermediate filament protein, nestin, and the homeobox protein, Dlx-2. Pharmacological activation of mGlu4 receptors during cell incubation in ITSFn medium increased the expression of both neural markers. Similar results were obtained when neural differentiation was induced by exposure of EBs to retinoic acid. These data suggest that differentiation of cultured ES cells is associated with changes in the expression pattern of mGlu receptors and that activation of mGlu4 receptors affects cell differentiation in a context-dependent manner.


Assuntos
Diferenciação Celular/fisiologia , Embrião de Mamíferos/citologia , Receptores de Glutamato Metabotrópico/fisiologia , Células-Tronco/fisiologia , Proteínas Adaptadoras de Transporte Vesicular/farmacologia , Aminobutiratos/farmacologia , Análise de Variância , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Interações Medicamentosas/fisiologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Proteínas Fetais/genética , Proteínas Fetais/metabolismo , Fator 5 de Crescimento de Fibroblastos/genética , Fator 5 de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Glicina/análogos & derivados , Glicina/farmacologia , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica/métodos , Camundongos , Fosfosserina/farmacologia , RNA Longo não Codificante , RNA Mensageiro/biossíntese , RNA não Traduzido/genética , RNA não Traduzido/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Células-Tronco/efeitos dos fármacos , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/metabolismo , Tretinoína/farmacologia
10.
Neuropharmacology ; 49 Suppl 1: 196-205, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16023153

RESUMO

Cultured mouse embryonic stem (ES) cells maintained under undifferentiated conditions (i.e. grown in medium containing 15% FCS and leukemia inhibitory factor--LIF) expressed mGlu5 metabotropic glutamate receptors. Activation of these receptors with quisqualate increased [Ca2+]i but only when cultures were deprived of extracellular glutamate, indicating that the receptor was saturated by the endogenous glutamate. Pharmacological blockade of mGlu5 receptors with 2-methyl-6-(phenylethynyl)pyridine (MPEP) or antisense-induced knock-down of mGlu5 receptors decreased the expression of the two main transcription factors that sustain ES cell self-renewal, i.e. Oct-4 and Nanog, as assessed by real-time PCR and immunoblotting. Exposure of ES cell cultures to MPEP also reduced alkaline phosphatase activity, a marker of undifferentiated ES cells. These data support a critical role for mGlu receptors in early development showing that mGlu5 receptors are expressed by ES cells and their activation sustains ES cell self-renewal in culture.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Receptores de Glutamato Metabotrópico/metabolismo , Células-Tronco/fisiologia , Alanina Transaminase/farmacologia , Animais , Northern Blotting/métodos , Western Blotting/métodos , Encéfalo/citologia , Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Células Cultivadas , Cromatografia Líquida de Alta Pressão/métodos , Interações Medicamentosas , Embrião de Mamíferos , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios , Citometria de Fluxo/métodos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Ácido Glutâmico/análise , Interleucina-6/farmacologia , Fator Inibidor de Leucemia , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Piridinas/farmacologia , Ácido Quisquálico/farmacologia , RNA Mensageiro/biossíntese , Receptor de Glutamato Metabotrópico 5 , Receptores de Glutamato Metabotrópico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Células-Tronco/efeitos dos fármacos , Timidina/metabolismo , Fatores de Tempo , Trítio/metabolismo
11.
Peptides ; 23(3): 555-60, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11836006

RESUMO

To investigate the functional relevance of a regulatory mutation affecting the enhancer element PEA2 of the rat ANP gene we transfected rat cardiomyocytes and aortic endothelial cells with either the mutant or the wild-type ANP promoter construct (-683 +54) and performed CAT assays both at baseline and in response to Phenylephrine and Angiotensin II. In the myocardial cells we also determined the DNA/nuclear protein interaction through electrophoretic mobility shift assay. These studies showed a significantly lower degree of ANP transcription in the presence of the mutant PEA2 site, thus demonstrating its functional significance and the biological relevance of ANP gene structural alterations.


Assuntos
Fator Natriurético Atrial/genética , Regulação da Expressão Gênica , Regiões Promotoras Genéticas/genética , Transcrição Gênica , Animais , Proteínas de Ligação a DNA/metabolismo , Mutação , Ratos , Ratos Wistar , Fator de Transcrição AP-2 , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...