Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Biomed Sci ; 24(1): 68, 2017 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-28870220

RESUMO

BACKGROUND: The type VI adenylyl cyclase (AC6) is a main contributor of cAMP production in the heart. The amino acid (aa) sequence of AC6 is highly homologous to that of another major cardiac adenylyl cyclase, AC5, except for its N-terminus (AC6-N, aa 1-86). Activation of AC6, rather than AC5, produces cardioprotective effects against heart failure, while the underlying mechanism remains to be unveiled. Using an AC6-null (AC6-/-) mouse and a knockin mouse with AC6-N deletion (AC6 ΔN/ΔN), we aimed to investigate the cardioprotective mechanism of AC6 in the heart. METHODS: Western blot analysis and immunofluorescence staining were performed to determine the intracellular distribution of AC6, AC6-ΔN (a truncated AC6 lacking the first 86 amino acids), and STAT3 activation. Activities of AC6 and AC6-ΔN in the heart were assessed by cAMP assay. Apoptosis of cardiomyocytes were evaluated by the TUNEL assay and a propidium iodine-based survival assay. Fibrosis was examined by collagen staining. RESULTS: Immunofluorescence staining revealed that cardiac AC6 was mainly anchored on the sarcolemmal membranes, while AC6-ΔN was redistributed to the sarcoplasmic reticulum. AC6ΔN/ΔN and AC6-/- mice had more apoptotic myocytes and cardiac remodeling than WT mice in experimental models of isoproterenol (ISO)-induced myocardial injury. Adult cardiomyocytes isolated from AC6ΔN/ΔN or AC6-/- mice survived poorly after exposure to ISO, which produced no effect on WT cardiomyocytes under the condition tested. Importantly, ISO treatment induced cardiac STAT3 phosphorylation/activation in WT mice, but not in AC6ΔN/ΔN and AC6-/- mice. Pharmacological blockage of PKA-, Src-, or STAT3- pathway markedly reduced the survival of WT myocytes in the presence of ISO, but did not affect those of AC6ΔN/ΔN and AC6-/- myocytes, suggesting an important role of AC6 in mediating cardioprotective action through the activation of PKA-Src-STAT3-signaling. CONCLUSIONS: Collectively, AC6-N controls the anchorage of cardiac AC6 on the sarcolemmal membrane, which enables the coupling of AC6 with the pro-survival PKA-STAT3 pathway. Our findings may facilitate the development of novel therapies for heart failure.


Assuntos
Adenilil Ciclases/genética , Miócitos Cardíacos/metabolismo , Transdução de Sinais , Adenilil Ciclases/metabolismo , Animais , Apoptose/efeitos dos fármacos , Insuficiência Cardíaca/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Substâncias Protetoras/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo
2.
J Biol Chem ; 289(3): 1257-70, 2014 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-24293369

RESUMO

The A2A adenosine receptor (A2AR) is a G protein-coupled receptor and a major target of caffeine. The A2AR gene encodes alternative transcripts that are initiated from at least two independent promoters. The different transcripts of the A2AR gene contain the same coding region and 3'-untranslated region and different 5'-untranslated regions that are highly conserved among species. We report here that in addition to the production of the A2AR protein, translation from an upstream, out-of-frame AUG of the rat A2AR gene produces a 134-amino acid protein (designated uORF5). An anti-uORF5 antibody recognized a protein of the predicted size of uORF5 in PC12 cells and rat brains. Up-regulation of A2AR transcripts by hypoxia led to increased levels of both the A2AR and uORF5 proteins. Moreover, stimulation of A2AR increased the level of the uORF5 protein via post-transcriptional regulation. Expression of the uORF5 protein suppressed the AP1-mediated transcription promoted by nerve growth factor and modulated the expression of several proteins that were implicated in the MAPK pathway. Taken together, our results show that the rat A2AR gene encodes two distinct proteins (A2AR and uORF5) in an A2AR-dependent manner. Our study reveals a new example of the complexity of the mammalian genome and provides novel insights into the function of A2AR.


Assuntos
Regiões 5' não Traduzidas/fisiologia , Regulação da Expressão Gênica/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Fases de Leitura Aberta/fisiologia , Receptor A2A de Adenosina/biossíntese , Transdução de Sinais/fisiologia , Regiões 3' não Traduzidas/fisiologia , Animais , Hipóxia Celular/fisiologia , Células HEK293 , Humanos , Células PC12 , Ratos , Ratos Sprague-Dawley , Receptor A2A de Adenosina/genética
3.
Carcinogenesis ; 35(10): 2164-74, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24832085

RESUMO

Embryonal tumors of the central nervous system represent a highly malignant tumor group of medulloblastoma (MB), atypical teratoid/rhabdoid tumor (AT/RT) and primitive neuroectodermal tumor that frequently afflict children. AT/RT is often misdiagnosed as MB/primitive neuroectodermal tumor but with higher recurrence and lower survival rates. Pathogenesis of AT/RT is largely unknown. In this study, we report both the miRNome and transcriptome traits in AT/RT and MB by using small RNA sequencing and gene expression microarray analyses. Our findings demonstrate that the miR-221/222-encoded micro RNAs are abundantly expressed in AT/RT but not in MB, which contribute substantially to the malignancy of embryonal tumors. miR-221/222 targeted SUN2, a newly discovered tumor suppressor, directly to increase cell proliferation and tumor malignancy in vitro and in vivo. Immunohistochemistry against SUN2 in a tissue microarray of 33 AT/RT and 154 MB tumor specimens also detected less SUN2 protein in AT/RT. Collectively, this study uncovers a novel tumor suppressor, SUN2, plays a critical role in miR-221/222-mediated AT/RT malignancy as well as supports miR-221/222 and SUN2 represent new promising targets for more active therapies in AT/RT. In addition, our miRNome and transcriptome data also provide a roadmap for further embryonal tumor research.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , MicroRNAs/genética , Neoplasias Embrionárias de Células Germinativas/genética , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidade , Neoplasias Cerebelares/genética , Neoplasias Cerebelares/mortalidade , Criança , Pré-Escolar , Regulação para Baixo , Feminino , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Meduloblastoma/genética , Meduloblastoma/mortalidade , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos NOD , Neoplasias Embrionárias de Células Germinativas/mortalidade , Tumores Neuroectodérmicos Primitivos/genética , Tumores Neuroectodérmicos Primitivos/mortalidade , Tumor Rabdoide/genética , Tumor Rabdoide/mortalidade , Ensaios Antitumorais Modelo de Xenoenxerto
4.
PLoS One ; 12(6): e0179758, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28640846

RESUMO

Tissue angiogenesis is intimately regulated during embryogenesis and postnatal development. Defected angiogenesis contributes to aberrant development and is the main complication associated with ischemia-related diseases. We previously identified the increased expression of RUNX1T1 in umbilical cord blood-derived endothelial colony-forming cells (ECFCs) by gene expression microarray. However, the biological relevance of RUNX1T1 in endothelial lineage is not defined clearly. Here, we demonstrate RUNX1T1 regulates the survival, motility and tube forming capability of ECFCs and EA.hy926 endothelial cells by loss-and gain-of function assays, respectively. Second, embryonic vasculatures and quantity of bone marrow-derived angiogenic progenitors are found to be reduced in the established Runx1t1 heterozygous knockout mice. Finally, a central RUNX1T1-regulated signature is uncovered and VEGFA, BMP4 as well as TGF-ß2 are demonstrated to mediate RUNX1T1-orchested angiogenic activities. Taken together, our results reveal that RUNX1T1 serves as a common angiogenic driver for vaculogenesis and functionality of endothelial lineage cells. Therefore, the discovery and application of pharmaceutical activators for RUNX1T1 will improve therapeutic efficacy toward ischemia by promoting neovascularization.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/metabolismo , Neovascularização Fisiológica , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta2/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Vasos Sanguíneos/fisiologia , Sangue Fetal/citologia , Técnicas de Inativação de Genes , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Camundongos , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Proteína 1 Parceira de Translocação de RUNX1 , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
5.
Sci Rep ; 6: 22529, 2016 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-26932446

RESUMO

The calcium-sensitive type VI adenylyl cyclase (AC6) is a membrane-bound adenylyl cyclase (AC) that converts ATP to cAMP under stimulation. It is a calcium-inhibited AC and integrates negative inputs from Ca(2+) and multiple other signals to regulate the intracellular cAMP level. In the present study, we demonstrate that AC6 functions upstream of CREB and negatively controls neuronal plasticity in the hippocampus. Genetic removal of AC6 leads to cyclase-independent and N-terminus of AC6 (AC6N)-dependent elevation of CREB expression, and enhances the expression of GluN2B-containing NMDA receptors in hippocampal neurons. Consequently, GluN2B-dependent calcium signaling and excitatory postsynaptic current, long-term depression, and spatial reversal learning are enhanced in the hippocampus of AC6(-/-) mice without altering the gross anatomy of the brain. Together, our results suggest that AC6 negatively regulates neuronal plasticity by modulating the levels of CREB and GluN2B in the hippocampus.


Assuntos
Adenilil Ciclases/metabolismo , Aprendizagem , Depressão Sináptica de Longo Prazo/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Adenilil Ciclases/genética , Animais , Hipocampo/enzimologia , Hipocampo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
6.
Brain Res Mol Brain Res ; 111(1-2): 61-73, 2003 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-12654506

RESUMO

By a combination of PCR and DNA walking technique, we isolated a 4.8-kb DNA fragment containing a 4.3 kb 5'-flanking region and a 0.5-kb 5'-untranslated region of the rat A(2A) adenosine receptor (A(2A)-R) gene. Various lengths of the 5'-flanking region of the A(2A)-R gene were inserted into an expression vector and transfected into several different cell lines for promoter analysis. Our results reveal that a consensus NF1 element (designated as A(2A)-R/NF1), located between bases -2846 and -2827 of the A(2A)-R gene, functions as a repressor for A(2A)-R promoters in the rat brain-derived type-2 astrocyte cell line (RBA2), which expresses no A(2A)-R. Electrophoretic gel mobility shift assay (EMSA) revealed that two A(2A)-R/NF1-protein complexes of RBA2 nuclear extract were formed. Supershift experiments using an anti-NF1 antibody suggest that NF1 proteins exist in both A(2A)-R/NF1-protein complexes. Furthermore, mutations in the conserved NF1 binding site of this A(2A)-R/NF1 element disturbed DNA-protein formation. Thus, NF1 proteins appear to mediate this cell line-specific suppression of A(2A)-R promoters in RBA2 cells. The importance of NF1 proteins in regulating A(2A)-R promoters was further confirmed in another cell line (Siha) which expresses no endogenous A(2A)-R. Moreover, addition of the A(2A)-R/NF1element upstream of an irrelevant thymidine kinase (TK) promoter suppressed its promoter activity in Siha cells, but not in RBA2 cells. Thus, the NF1-mediated inhibition of the A(2A)-R promoter was promoter- and cell line-specific. In summary, we have defined a distal negative element (A(2A)-R/NF1) that plays a functional role in modulating the expression of A(2A)-R.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/genética , Sistema Nervoso Central/metabolismo , Proteínas de Ligação a DNA , Regulação da Expressão Gênica/genética , Genes Reguladores/genética , Regiões Promotoras Genéticas/genética , Receptores Purinérgicos P1/genética , Proteínas Repressoras/genética , Fatores de Transcrição , Região 5'-Flanqueadora/genética , Animais , Anticorpos , Astrócitos/metabolismo , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Humanos , Imuno-Histoquímica , Substâncias Macromoleculares , Fatores de Transcrição NFI , Neurônios/metabolismo , Proteínas Nucleares , Células PC12 , Ratos , Receptor A2A de Adenosina , Proteína 1 de Ligação a Y-Box
7.
Exp Neurol ; 248: 10-5, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23726959

RESUMO

Visceral functions are regulated by a basal sympathetic nerve discharge (SND), also known as 'sympathetic tone'. We demonstrate herein that AC6 existed in tyrosine hydroxylase-positive rostral ventrolateral medulla neurons in the brainstem. Adenylyl cyclase (AC) assays showed lower basal and pituitary adenylate cyclase-activating peptide-evoked AC activities in the brainstem of AC6-null mice, indicating that AC6 is a prominent AC isozyme in the brainstem. Furthermore, two independent lines of AC6-null mice exhibited a much higher SND, recorded from splanchnic sympathetic nerves of neonatal brainstem-spinal cord preparations, than wildtype mice. An assay of urine noradrenaline confirmed this observation. Collectively, AC6 plays a critical role in the regulation of sympathetic tone.


Assuntos
Adenilil Ciclases/metabolismo , Tronco Encefálico/metabolismo , Medula Espinal/metabolismo , Nervos Esplâncnicos/metabolismo , Sistema Nervoso Simpático/metabolismo , Adenilil Ciclases/genética , Animais , Animais Recém-Nascidos , Camundongos , Camundongos Knockout , Norepinefrina/urina
8.
Mol Cell Biol ; 31(24): 4874-86, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21986494

RESUMO

3'-5'-Cyclic AMP (cAMP) is an important second messenger which regulates neurite outgrowth. We demonstrate here that type VI adenylyl cyclase (AC6), an enzyme which catalyzes cAMP synthesis, regulates neurite outgrowth by direct interaction with a binding protein (Snapin) of Snap25 at the N terminus of AC6 (AC6-N). We first showed that AC6 expression increased during postnatal brain development. In primary hippocampal neurons and Neuro2A cells, elevated AC6 expression suppressed neurite outgrowth, whereas the downregulation or genetic removal of AC6 promoted neurite extension. An AC6 variant (AC6-N5) that contains the N terminus of AC5 had no effect, indicating the importance of AC6-N. The downregulation of endogenous Snapin or the overexpression of a Snapin mutant (Snap(Δ33-51)) that does not bind to AC6, or another Snapin mutant (Snapin(S50A)) that does not interact with Snap25, reversed the inhibitory effect of AC6. Pulldown assays and immunoprecipitation-AC assays revealed that the complex formation of AC6, Snapin, and Snap25 is dependent on AC6-N and the phosphorylation of Snapin. The overexpression of Snap25 completely reversed the action of AC6. Collectively, in addition to cAMP production, AC6 plays a complex role in modulating neurite outgrowth by redistributing localization of the SNARE apparatus via its interaction with Snapin.


Assuntos
Adenilil Ciclases/metabolismo , Neuritos/metabolismo , Proteína 25 Associada a Sinaptossoma/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Adenilil Ciclases/genética , Animais , Western Blotting , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Células Cultivadas , Regulação para Baixo , Eletroforese em Gel de Poliacrilamida , Regulação da Expressão Gênica no Desenvolvimento , Hipocampo/citologia , Hipocampo/metabolismo , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Camundongos Knockout , Mutação , Fatores de Crescimento Neural/análise , Fosforilação , Plasmídeos , Ratos , Ratos Sprague-Dawley , Proteínas SNARE/genética , Proteínas SNARE/metabolismo , Proteína 25 Associada a Sinaptossoma/genética , Proteínas de Transporte Vesicular/genética
9.
FEBS Lett ; 584(13): 2883-90, 2010 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-20466003

RESUMO

Adenylyl cyclase (AC) type VI (AC6) is a calcium-inhibitable enzyme which produces cAMP upon stimulation. Herein, we characterized the specific role of AC6 in the kidneys using two AC6-knockout mouse lines. Immunohistochemical staining revealed that AC6 exists in the tubular parts of the nephron and collecting duct. Activities of AC evoked by forskolin or a selective agonist of the V2 vasopressin receptor were lower in the kidneys of AC6-null mice compared to those of wildtype mice. Results of a metabolic cage assay and dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) showed for the first time that AC6 plays a critical role in regulating water homeostasis.


Assuntos
Adenilil Ciclases/metabolismo , Água/metabolismo , Adenilil Ciclases/genética , Animais , AMP Cíclico/metabolismo , Imuno-Histoquímica , Túbulos Renais Coletores/metabolismo , Túbulos Renais Coletores/fisiologia , Imageamento por Ressonância Magnética , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
J Neurochem ; 93(2): 310-20, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15816854

RESUMO

Huntington's disease (HD) is an autosomal dominant neurodegenerative disease caused by a CAG trinucleotide expansion in exon 1 of the Huntingtin (Htt) gene. We show herein that in an HD transgenic mouse model (R6/2), daily administration of CGS21680 (CGS), an A(2A) adenosine receptor (A(2A)-R)-selective agonist, delayed the progressive deterioration of motor performance and prevented a reduction in brain weight. 3D-microMRI analysis revealed that CGS reversed the enlarged ventricle-to-brain ratio of R6/2 mice, with particular improvements in the left and right ventricles. (1)H-MRS showed that CGS significantly reduced the increased choline levels in the striatum. Immunohistochemical analyses further demonstrated that CGS reduced the size of ubiquitin-positive neuronal intranuclear inclusions (NIIs) in the striatum of R6/2 mice and ameliorated mutant Htt aggregation in a striatal progenitor cell line overexpressing mutant Htt with expanded polyQ. Moreover, chronic CGS treatment normalized the elevated blood glucose levels and reduced the overactivation of a major metabolic sensor [5'AMP-activated protein kinase (AMPK)] in the striatum of R6/2 mice. Since AMPK is a master switch for energy metabolism, modulation of energy dysfunction caused by the mutant Htt might contribute to the beneficial effects of CGS. Collectively, CGS is a potential drug candidate for the treatment of HD.


Assuntos
Adenosina/análogos & derivados , Adenosina/uso terapêutico , Modelos Animais de Doenças , Doença de Huntington/genética , Doença de Huntington/prevenção & controle , Fenetilaminas/uso terapêutico , Agonistas do Receptor A2 de Adenosina , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Doença de Huntington/patologia , Masculino , Camundongos , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos
11.
J Biol Chem ; 279(44): 46271-9, 2004 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-15319443

RESUMO

In the present study, we used the N terminus (amino acids 1 approximately 160) of type VI adenylyl cyclase (ACVI) as bait to screen a mouse brain cDNA library and identified Snapin as a novel ACVI-interacting molecule. Snapin is a binding protein of SNAP25, a component of the SNARE complex. Co-immunoprecipitation analyses confirmed the interaction between Snapin and full-length ACVI. Mutational analysis revealed that the interaction domains of ACVI and Snapin were located within amino acids 1 approximately 86 of ACVI and 33-51 of Snapin, respectively. Co-localization of ACVI and Snapin was observed in primary hippocampal neurons. Moreover, expression of Snapin specifically eliminated protein kinase C (PKC)-mediated suppression of ACVI, but not that of cAMP-dependent protein kinase (PKA) or calcium. Mutation of the potential PKC and PKA phosphorylation sites of Snapin did not affect the ability of Snapin to reverse the PKC inhibitory effect on ACVI. Phosphorylation of Snapin by PKC or PKA therefore might not be crucial for Snapin action on ACVI. In contrast, Snapin(Delta33-51), which harbors an internal deletion of amino acids 33-51 did not affect PKC-mediated inhibition of ACVI, supporting that amino acids 33-51 of Snapin comprises the ACVI-interacting region. Consistently, Snapin exerted no effect on PKC-mediated inhibition of an ACVI mutant (ACVI-DeltaA87), which lacked the Snapin-interacting region (amino acids 1-86). Snapin thus reverses its action via direct interaction with the N terminus of ACVI. Collectively, we demonstrate herein that in addition to its association with the SNARE complex, Snapin also functions as a regulator of an important cAMP synthesis enzyme in the brain.


Assuntos
Adenilil Ciclases/metabolismo , Proteínas de Transporte Vesicular/fisiologia , Adenilil Ciclases/química , Sequência de Aminoácidos , Animais , Cálcio/fisiologia , Células Cultivadas , AMP Cíclico/biossíntese , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Humanos , Isoenzimas/metabolismo , Camundongos , Dados de Sequência Molecular , Proteína Quinase C/fisiologia , Ratos , Ratos Sprague-Dawley
12.
Eur J Neurosci ; 18(7): 1786-96, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14622213

RESUMO

We isolated and characterized a 4.8-kb 5' flanking region of the rat A2A adenosine receptor (A2A-R) gene in the present study. Promoter activity was observed with this DNA fragment in PC12 cells and C6 cells which contain endogenous A2A-Rs. A fusion fragment consisting of the 4.8-kb promoter-proximal DNA fragment of the A2A-R gene, and the coding region of lacZ was utilized to produce mice harbouring the fusion gene. In three independent founder lines, proteins and transcripts of the transgene were found in many areas of the central nervous system (CNS), but not in three peripheral tissues examined. Double immunohistochemical analyses revealed that the transgene was coexpressed with endogenous A2A-R and proper neuronal markers in the brain. Specifically, the transgene in the striatum was found in the enkephalin-containing GABAergic neurons and in the cholinergic neurons as was found for the endogenous A2A-R. However, a selectively enriched striatal expression of the transgene was not found as was observed for the endogenous A2A-R. Collectively, the 4.8-kb promoter-proximal DNA fragment of the rat A2A-R gene contains important element(s) to direct its expression in the CNS where functional A2A-R are found, but were not sufficient to confer the highly concentrated expression of the striatal A2A-R. Furthermore, expressions of A2A-R and the transgene were found in both neurons and astrocytes, suggesting that adenosine might mediate its function through A2A-R in both cell types.


Assuntos
Região 5'-Flanqueadora , Sistema Nervoso Central/metabolismo , Regiões Promotoras Genéticas , Receptor A2A de Adenosina/genética , Adenilil Ciclases/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Southern Blotting , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Sistema Nervoso Central/anatomia & histologia , Sistema Nervoso Central/efeitos dos fármacos , Fragmentação do DNA , Expressão Gênica/efeitos dos fármacos , Genes Reporter/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Células PC12 , Reação em Cadeia da Polimerase , RNA/metabolismo , Ratos , Receptor A2A de Adenosina/química , Receptor A2A de Adenosina/metabolismo , Transfecção , Tubulina (Proteína)/metabolismo , beta-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA