Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Genes (Basel) ; 12(8)2021 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-34440304

RESUMO

GNAI2 (G protein subunit alpha i2) is a signaling modulator or transducer, involved in several transmembrane signaling systems, that plays a vital role in the melanogenesis signaling pathway. However, whether GNAI2 regulates cell proliferation and apoptosis in rabbit melanocytes is not known. We found that GNAI2 was differentially expressed in rabbits with different coat colors using qRT-PCR and Wes assays. Furthermore, it was observed that the rabbits with black skin had the highest GNAI2 levels, and those with white skin had the lowest expression. The coding sequence of GNAI2 was successfully cloned and inserted into pcDNA3.1 and pcDNA3.1-Myc vectors. It was observed that the GNAI2 protein was mainly localized in the cytoplasm using the indirect immunofluorescence staining assay. Overexpression of GNAI2 significantly increased melanin content, promoted melanocyte proliferation, and inhibited melanocyte apoptosis. On the contrary, the knockdown of GNAI2 using siRNA had the opposite effect. In addition, GNAI2 significantly increased the mRNA expression levels of the melanin-related genes TYR, GPNMB, PMEL, and DCT in rabbit melanocytes. The results suggested that GNAI2 regulated melanocyte development by promoting melanocyte proliferation and inhibiting apoptosis.


Assuntos
Apoptose/fisiologia , Proliferação de Células/fisiologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Melanócitos/citologia , Animais , Citoplasma/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Técnicas de Silenciamento de Genes , Cor de Cabelo , Melaninas/biossíntese , Melanócitos/metabolismo , Coelhos
2.
Mol Pharmacol ; 100(3): 217-223, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34135098

RESUMO

Regulators of G protein signaling (RGS) proteins modulate signaling by G protein-coupled receptors. Using a knock-in transgenic mouse model with a mutation in Gαo that does not bind RGS proteins (RGS-insensitive), we determined the effect of RGS proteins on presynaptic µ opioid receptor (MOR)-mediated inhibition of GABA release in the ventrolateral periaqueductal gray (vlPAG). The MOR agonists [d-Ala2, N-MePhe4, Gly-ol]-enkephalin (DAMGO) and met-enkephalin (ME) inhibited evoked inhibitory postsynaptic currents (eIPSCs) in the RGS-insensitive mice compared with wild-type (WT) littermates, respectively. Fentanyl inhibited eIPSCs similarly in both WT and RGS-insensitive mice. There were no differences in opioid agonist inhibition of spontaneous GABA release between the genotypes. To further probe the mechanism underlying these differences between opioid inhibition of evoked and spontaneous GABA release, specific myristoylated Gα peptide inhibitors for Gαo1 and Gαi1-3 that block receptor-G protein interactions were used to test the preference of agonists for MOR-Gα complexes. The Gαo1 inhibitor reduced DAMGO inhibition of eIPSCs, but Gαi1-3 inhibitors had no effect. Both Gαo1 and Gαi1-3 inhibitors separately reduced fentanyl inhibition of eIPSCs but had no effects on ME inhibition. Gαi1-3 inhibitors blocked the inhibitory effects of ME and fentanyl on miniature postsynaptic current (mIPSC) frequency, but both Gαo1 and Gαi1-3 inhibitors were needed to block the effects of DAMGO. Finally, baclofen-mediated inhibition of GABA release is unaffected in the RGS-insensitive mice and in the presence of Gαo1 and Gαi1-3 inhibitor peptides, suggesting that GABAB receptor coupling to G proteins in vlPAG presynaptic terminals is different than MOR coupling. SIGNIFICANCE STATEMENT: Presynaptic µ opioid receptors (MORs) in the ventrolateral periaqueductal gray are critical for opioid analgesia and are negatively regulated by RGS proteins. These data in RGS-insensitive mice provide evidence that MOR agonists differ in preference for Gαo versus Gαi and regulation by RGS proteins in presynaptic terminals, providing a mechanism for functional selectivity between agonists. The results further define important differences in MOR and GABAB receptor coupling to G proteins that could be exploited for new pain therapies.


Assuntos
Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Terminações Pré-Sinápticas/fisiologia , Receptores Opioides mu/fisiologia , Ácido gama-Aminobutírico/metabolismo , Analgésicos Opioides/farmacologia , Animais , Baclofeno/farmacologia , Feminino , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/antagonistas & inibidores , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/antagonistas & inibidores , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Modelos Animais , Proteínas RGS/metabolismo , Receptores de GABA-B/metabolismo , Receptores Opioides mu/agonistas
3.
Mol Metab ; 40: 101029, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32480042

RESUMO

OBJECTIVES: Typically, obesity results from an inappropriate balance between energy uptake from nutrient consumption and burning of calories, which leads to a pathological increase in fat mass. Obesity is a major cause of insulin resistance and diabetes. Inhibitory G proteins (Gαi) form a subfamily that is involved in the regulation of adipose tissue function. Among the three Gαi members, i.e. Gαi1, Gαi2, Gαi3, the Gαi2, protein is predominantly expressed in adipose tissue. However, the functions of the Gαi2 isoform in adipose tissue and its impact on the development of obesity are poorly understood. METHODS: By using AdipoqCreERT2 mice, we generated adipocyte-specific Gnai2-deficient mice to study Gαi2 function, specifically in white and brown adipocytes. These mice were fed either a control diet (CD) or a high fat diet (HFD). Mice were examined for obesity development, insulin resistance and glucose intolerance. We examined adipocyte morphology and the development of inflammation in the white adipose tissue. Finally, intracellular cAMP levels as an indicator of Gαi signaling and glycerol release as an indicator of lipolysis rates were measured to verify the impact of Gαi2 on the signaling pathway in brown and white adipocytes. RESULTS: An adipocyte-specific deficiency of Gαi2 significantly reduced diet-induced obesity, leading to decreased fat masses, smaller adipocytes and decreased inflammation in the white adipose tissue relative to littermate controls. Concurrently, oxygen consumption of brown adipocytes and in vivo measured energy expenditure were significantly enhanced. In addition, glucose tolerance and insulin sensitivity of HFD-fed adipocyte-specific Gnai2-deficient mice were improved compared to the respective controls. In the absence of Gαi2, adrenergic stimulation of intracellular adipocyte cAMP levels was increased, which correlated with increased lipolysis and energy expenditure. CONCLUSION: We conclude that adipocyte Gαi2 is a major regulator of adipocyte lipid content in diet-induced obesity by inhibiting adipocyte lipolysis in a cAMP-dependent manner resulting in increased energy expenditure.


Assuntos
Tecido Adiposo/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Obesidade/metabolismo , Adipócitos Marrons/metabolismo , Adipócitos Brancos/metabolismo , Tecido Adiposo/fisiologia , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Dieta Hiperlipídica , Metabolismo Energético , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Glucose/metabolismo , Intolerância à Glucose/metabolismo , Insulina/metabolismo , Resistência à Insulina/fisiologia , Lipólise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/genética , Consumo de Oxigênio
4.
J Neurochem ; 140(1): 82-95, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27787898

RESUMO

We analyzed the role of a heterotrimeric G-protein, Gi2, in the development of the cerebral cortex. Acute knockdown of the α-subunit (Gαi2) with in utero electroporation caused delayed radial migration of excitatory neurons during corticogenesis, perhaps because of impaired morphology. The migration phenotype was rescued by an RNAi-resistant version of Gαi2. On the other hand, silencing of Gαi2 did not affect axon elongation, dendritic arbor formation or neurogenesis at ventricular zone in vivo. When behavior analyses were conducted with acute Gαi2-knockdown mice, they showed defects in social interaction, novelty recognition and active avoidance learning as well as increased anxiety. Subsequently, using whole-exome sequencing analysis, we identified a de novo heterozygous missense mutation (c.680C>T; p.Ala227Val) in the GNAI2 gene encoding Gαi2 in an individual with periventricular nodular heterotopia and intellectual disability. Collectively, the phenotypes in the knockdown experiments suggest a role of Gαi2 in the brain development, and impairment of its function might cause defects in neuronal functions which lead to neurodevelopmental disorders.


Assuntos
Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Deficiência Intelectual/metabolismo , Heterotopia Nodular Periventricular/metabolismo , Animais , Aprendizagem da Esquiva/fisiologia , Células COS , Córtex Cerebral/diagnóstico por imagem , Chlorocebus aethiops , Feminino , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Humanos , Deficiência Intelectual/diagnóstico por imagem , Deficiência Intelectual/genética , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Heterotopia Nodular Periventricular/diagnóstico por imagem , Heterotopia Nodular Periventricular/genética , Gravidez
5.
J Am Heart Assoc ; 5(11)2016 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-27912212

RESUMO

BACKGROUND: We previously showed that the levels of both Giα-2 and Giα-3 proteins were augmented in spontaneously hypertensive rats (SHRs) before the onset of hypertension. In addition, intraperitoneal injection of pertussis toxin, which inactivates both Giα proteins, prevented the development of hypertension in SHRs. The aim of the present study was to determine the specific contributions of Giα-2 and Giα-3 proteins to the development of hypertension. METHODS AND RESULTS: Antisense oligodeoxynucleotide of Giα-2 and Giα-3 encapsulated in PEG/DOTAP/DOPE cationic liposomes were administrated intravenously into 3-week-old prehypertensive SHRs and Wistar Kyoto rats, whereas the control Wistar Kyoto rats and SHRs received PBS, empty liposomes, or sense. The knockdown of Giα-2 but not Giα-3 protein attenuated tachycardia and prevented the development of hypertension up to age 6 weeks; thereafter, blood pressure started increasing and reached the same level as that of untreated SHRs at 9 weeks. Furthermore, Giα-2 and Giα-3 antisense oligodeoxynucleotide treatments significantly decreased the enhanced levels of Giα-2 and Giα-3 proteins, respectively, and enhanced levels of superoxide anion and NADPH oxidase activity in heart, aorta, and kidney and hyperproliferation of vascular smooth muscle cells from SHRs aged 6 weeks. In addition, antisense oligodeoxynucleotide treatment with Giα-2 but not Giα-3 restored enhanced inhibition of adenylyl cyclase by oxotremorine to WKY levels. CONCLUSIONS: These results suggested that the enhanced expression of Giα-2 but not Giα-3 protein plays an important role in the pathogenesis of hypertension and tachycardia in SHRs.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Hipertensão/prevenção & controle , Oligodesoxirribonucleotídeos Antissenso/fisiologia , Taquicardia/prevenção & controle , Inibidores de Adenilil Ciclases/farmacologia , Animais , Aorta/metabolismo , Pressão Sanguínea/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/deficiência , Técnicas de Silenciamento de Genes , Frequência Cardíaca/fisiologia , Rim/metabolismo , Lipossomos/administração & dosagem , Masculino , Músculo Liso Vascular/metabolismo , Miocárdio/metabolismo , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Transdução de Sinais/fisiologia , Transfecção/métodos
6.
Nat Cell Biol ; 16(8): 758-69, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25016959

RESUMO

Asymmetric cell divisions allow stem cells to balance proliferation and differentiation. During embryogenesis, murine epidermis expands rapidly from a single layer of unspecified basal layer progenitors to a stratified, differentiated epithelium. Morphogenesis involves perpendicular (asymmetric) divisions and the spindle orientation protein LGN, but little is known about how the apical localization of LGN is regulated. Here, we combine conventional genetics and lentiviral-mediated in vivo RNAi to explore the functions of the LGN-interacting proteins Par3, mInsc and Gαi3. Whereas loss of each gene alone leads to randomized division angles, combined loss of Gnai3 and mInsc causes a phenotype of mostly planar divisions, akin to loss of LGN. These findings lend experimental support for the hitherto untested model that Par3-mInsc and Gαi3 act cooperatively to polarize LGN and promote perpendicular divisions. Finally, we uncover a developmental switch between delamination-driven early stratification and spindle-orientation-dependent differentiation that occurs around E15, revealing a two-step mechanism underlying epidermal maturation.


Assuntos
Proteínas de Transporte/fisiologia , Moléculas de Adesão Celular/fisiologia , Proteínas de Ciclo Celular/fisiologia , Células Epidérmicas , Epiderme/fisiologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Padronização Corporal , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Moléculas de Adesão Celular/antagonistas & inibidores , Moléculas de Adesão Celular/genética , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Divisão Celular/genética , Divisão Celular/fisiologia , Epiderme/embriologia , Feminino , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/antagonistas & inibidores , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/antagonistas & inibidores , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Gravidez , Interferência de RNA , Fuso Acromático/fisiologia
7.
J Hypertens ; 31(4): 747-57, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23391983

RESUMO

OBJECTIVE: We have previously demonstrated a role of GPCR-activated brain Gαi(2)-subunit protein-gated pathways in the natriuretic responses evoked by exogenous central α(2)-adrenoceptor activation and acute intravenous (i.v.) volume expansion in vivo. Our objective was to examine the role of brain Gαi(2) proteins in the integrated neural-humoral responses evoked by i.v. isovolumetric sodium loading, which does not alter mean arterial blood pressure or total blood volume, to maintain sodium homeostasis in conscious Sprague-Dawley rats. METHODS: Intact or chronic bilateral renal denervated (RDNX) rats were pretreated intracerebroventricularly (i.c.v.) with a scrambled or Gαi(2) oligodeoxynucleotide to selectively downregulate brain Gαi(2) proteins. On the day of study, an i.v. isovolumetric sodium load (1 mol/l NaCl) was administered. RESULTS: In naive and scrambled oligodeoxynucleotide groups, i.v. sodium loading evoked profound natriuresis, suppression of plasma renin activity (PRA) and global sympathoinhibition. Prior downregulation of brain Gαi(2) proteins significantly attenuated the natriuretic response [peak ΔUNaV (µeq/µl); scrambled 22 ± 2 vs. Gαi(2) 13 ± 2, P < 0.05] and abolished the sympathoinhibitory response [peak Δplasma norepinephrine (% control); SCR -72 ± 8 vs. Gαi(2) -7 ± 5, P < 0.05] without attenuating PRA suppression to sodium loading. In RDNX rats, Gαi(2) oligodeoxynucleotide pretreatment failed to attenuate the natriuretic response [peak ΔUNaV (µeq/µl); RDNX and scrambled 19 ± 3 vs. RDNX and Gαi(2) 20 ± 2] and only partially prevented the sympathoinhibitory response to i.v. sodium loading. CONCLUSION: These studies reveal a brain Gαi(2)-subunit protein-mediated (renin-angiotensin system-independent) sympathoinhibitory pathway that has a critical role in the central neural mechanisms activated to maintain fluid and electrolyte homeostasis.


Assuntos
Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Homeostase , Sódio/metabolismo , Animais , Sequência de Bases , Western Blotting , Cromatografia Líquida de Alta Pressão , Primers do DNA , Masculino , Norepinefrina/sangue , Ratos , Ratos Sprague-Dawley , Renina/sangue , Sódio/administração & dosagem , Sistema Nervoso Simpático/fisiologia
8.
J Neurochem ; 124(6): 782-94, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23373526

RESUMO

The Goα splice variants Go1α and Go2α are subunits of the most abundant G-proteins in brain, Go1 and Go2. Only a few interacting partners binding to Go1α have been described so far and splice variant-specific differences are not known. Using a yeast two-hybrid screen with constitutively active Go2α as bait, we identified Rap1GTPase activating protein (Rap1GAP) and Girdin as interacting partners of Go2α, which was confirmed by co-immunoprecipitation. Comparison of subcellular fractions from brains of wild type and Go2α-/- mice revealed no differences in the overall expression level of Girdin or Rap1GAP. However, we found higher amounts of active Rap1-GTP in brains of Go2α deficient mutants, indicating that Go2α may increase Rap1GAP activity, thereby effecting the Rap1 activation/deactivation cycle. Rap1 has been shown to be involved in neurite outgrowth and given a Rap1GAP-Go2α interaction, we found that the loss of Go2α affected axonal outgrowth. Axons of cultured cortical and hippocampal neurons prepared from embryonic Go2α-/- mice grew longer and developed more branches than those from wild-type mice. Taken together, we provide evidence that Go2α regulates axonal outgrowth and branching.


Assuntos
Axônios/fisiologia , GTP Fosfo-Hidrolases/fisiologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Animais , Células Cultivadas , Ativação Enzimática/fisiologia , GTP Fosfo-Hidrolases/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subunidades Proteicas/fisiologia
9.
J Immunol ; 190(1): 324-33, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23225882

RESUMO

Heterotrimeric G proteins of the Gα(i) family have been implicated in signaling pathways regulating cell migration in immune diseases. The Gα(i)-protein-coupled C5a receptor is a critical regulator of IgG FcR function in experimental models of immune complex (IC)-induced inflammation. By using mice deficient for Gα(i2) or Gα(i3), we show that Gα(i2) is necessary for neutrophil influx in skin and lung Arthus reactions and agonist-induced neutrophilia in the peritoneum, whereas Gα(i3) plays a less critical but variable role. Detailed analyses of the pulmonary IC-induced inflammatory response revealed several shared functions of Gα(i2) and Gα(i3), including mediating C5a anaphylatoxin receptor-induced activation of macrophages, involvement in alveolar production of chemokines, transition of neutrophils from bone marrow into blood, and modulation of CD11b and CD62L expression that account for neutrophil adhesion to endothelial cells. Interestingly, C5a-stimulated endothelial polymorphonuclear neutrophil transmigration, but not chemotaxis, is enhanced versus reduced in the absence of neutrophil Gα(i3) or Gα(i2), respectively, and knockdown of endothelial Gα(i2) caused decreased transmigration of wild-type neutrophils. These data demonstrate that Gα(i2) and Gα(i3) contribute to inflammation by redundant, overlapping, and Gα(i)-isoform-specific mechanisms, with Gα(i2) exhibiting unique functions in both neutrophils and endothelial cells that appear essential for polymorphonuclear neutrophil recruitment in IC disease.


Assuntos
Lesão Pulmonar Aguda/imunologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/patologia , Animais , Reação de Arthus/genética , Reação de Arthus/imunologia , Reação de Arthus/patologia , Adesão Celular/genética , Adesão Celular/imunologia , Quimiotaxia de Leucócito/genética , Quimiotaxia de Leucócito/imunologia , Modelos Animais de Doenças , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/citologia , Neutrófilos/imunologia , Neutrófilos/patologia , Regulação para Cima/genética , Regulação para Cima/imunologia
10.
Mol Cell ; 43(3): 418-31, 2011 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-21816348

RESUMO

Asymmetric cell division requires the establishment of cortical cell polarity and the orientation of the mitotic spindle along the axis of cell polarity. Evidence from invertebrates demonstrates that the Par3/Par6/aPKC and NuMA/LGN/Gαi complexes, which are thought to be physically linked by the adaptor protein mInscuteable (mInsc), play indispensable roles in this process. However, the molecular basis for the binding of LGN to NuMA and mInsc is poorly understood. The high-resolution structures of the LGN/NuMA and LGN/mInsc complexes presented here provide mechanistic insights into the distinct and highly specific interactions of the LGN TPRs with mInsc and NuMA. Structural comparisons, together with biochemical and cell biology studies, demonstrate that the interactions of NuMA and mInsc with LGN are mutually exclusive, with mInsc binding preferentially. Our results suggest that the Par3/mInsc/LGN and NuMA/LGN/Gαi complexes play sequential and partially overlapping roles in asymmetric cell division.


Assuntos
Antígenos Nucleares/química , Proteínas de Transporte/química , Proteínas de Ciclo Celular/química , Divisão Celular/fisiologia , Proteínas Associadas à Matriz Nuclear/química , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antígenos Nucleares/genética , Antígenos Nucleares/fisiologia , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/metabolismo , Moléculas de Adesão Celular/fisiologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiologia , Polaridade Celular , Cristalografia por Raios X , Escherichia coli/genética , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/química , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Humanos , Camundongos , Modelos Moleculares , Mutagênese , Proteínas Associadas à Matriz Nuclear/genética , Proteínas Associadas à Matriz Nuclear/fisiologia , Estrutura Terciária de Proteína , Transporte Proteico , Fuso Acromático/metabolismo
11.
J Neurochem ; 115(1): 234-46, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20649838

RESUMO

The α-subunit of Go2 is a regulator of dopamine (DA) homeostasis. Deletion of the protein results in an imbalance of the direct and indirect DA pathway by reducing D1 and increasing D2 receptors. As a result, cocaine-induced behavioral sensitization is abolished. Here we show that repeated amphetamine injections in Go2α-/- mice induced a similar D1/D2 receptor ratio shift as cocaine but surprisingly the knockouts developed normal behavioral sensitization. DA receptor signaling following either cocaine or amphetamine treatment was also similar in Go2α-/- mice suggesting another mechanism involved in the differential behavioral response. Evidence is increasing that DA-glutamate interactions in the striatum determine psychostimulant action. In this line, repeated amphetamine injections led to a twofold increase in the amount of the NMDA receptor subunit NR2B in Go2α-/- mice resulting in an enhanced inhibition of the indirect DA pathway. This effect is not seen after cocaine treatment. Furthermore, amphetamine but not cocaine treatment maintained the ratio between the glutamate receptor mGluR1/5 interacting proteins Homer and Homer1a in the knockouts thereby sustaining the direct pathway. We conclude that amphetamine provokes behavioral sensitization in Go2α-/- mice by an enhanced inhibition of the indirect pathway without disturbing the direct pathway thereby overcoming the imbalance in the DArgic system.


Assuntos
Anfetamina/farmacologia , Comportamento Animal/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Receptores de N-Metil-D-Aspartato/biossíntese , Animais , Western Blotting , Condicionamento Operante/efeitos dos fármacos , Dopamina/metabolismo , Antagonistas de Dopamina/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Deleção de Genes , Imunoprecipitação , Camundongos , Camundongos Knockout , Receptores de Dopamina D1/efeitos dos fármacos , Receptores de Dopamina D2/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Espiperona/metabolismo , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo
12.
Hepatology ; 51(3): 846-56, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20054866

RESUMO

UNLABELLED: The pathological relevance and significance of microRNAs (miRNAs) in hepatocarcinogenesis have attracted much attention in recent years; however, little is known about the underlying molecular mechanisms through which miRNAs are involved in the development and progression of hepatocellular carcinoma (HCC). In this study, we demonstrate that miR-30d is frequently up-regulated in HCC and that its expression is highly associated with the intrahepatic metastasis of HCC. Furthermore, the enhanced expression of miR-30d could promote HCC cell migration and invasion in vitro and intrahepatic and distal pulmonary metastasis in vivo, while silencing its expression resulted in a reduced migration and invasion. Galphai2 (GNAI2) was identified as the direct and functional target of miR-30d with integrated bioinformatics analysis and messenger RNA array assay. This regulation was further confirmed by luciferase reporter assays. In addition, our results, for the first time, showed that GNAI2 was frequently suppressed in HCC by way of quantitative reverse-transcription polymerase chain reaction and immunohistochemical staining assays. The increase of the GNAI2 expression significantly inhibits, whereas knockdown of the GNAI2 expression remarkably enhances HCC cell migration and invasion, indicating that GNAI2 functions as a metastasis suppressor in HCC. The restoration of GNAI2 can inhibit miR-30d-induced HCC cell invasion and metastasis. CONCLUSION: The newly identified miR-30d/GNAI2 axis elucidates the molecular mechanism of HCC cell invasion and metastasis and represents a new potential therapeutic target for HCC treatment.


Assuntos
Carcinoma Hepatocelular/genética , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Neoplasias Hepáticas/genética , MicroRNAs/fisiologia , Carcinoma Hepatocelular/secundário , Humanos , Neoplasias Hepáticas/patologia , Invasividade Neoplásica , Células Tumorais Cultivadas
14.
Circulation ; 117(11): 1378-87, 2008 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-18316484

RESUMO

BACKGROUND: A salient characteristic of dysfunctional myocardium progressing to heart failure is an upregulation of the adenylyl cyclase inhibitory guanine nucleotide (G) protein alpha subunit, G alpha(i2). It has not been determined conclusively whether increased Gi activity in the heart is beneficial or deleterious in vivo. Gi signaling has been implicated in the mechanism of cardioprotective agents; however, no in vivo evidence exists that any of the G alpha subunits are cardioprotective. We have created a novel molecular tool to specifically address the role of Gi proteins in normal and dysfunctional myocardium. METHODS AND RESULTS: We have developed a class-specific Gi inhibitor peptide, GiCT, composed of the region of G alpha(i2) that interacts specifically with G protein-coupled receptors. GiCT inhibits Gi signals specifically in vitro and in vivo, whereas Gs and Gq signals are not affected. In vivo expression of GiCT in transgenic mice effectively causes a "functional knockout" of cardiac G alpha(i2) signaling. Inducible, cardiac-specific GiCT transgenic mice display a baseline phenotype consistent with nontransgenic mice. However, when subjected to ischemia/reperfusion injury, GiCT transgenic mice demonstrate a significant increase in infarct size compared with nontransgenic mice (from 36.9+/-2.5% to 50.9+/-4.3%). Mechanistically, this post-ischemia/reperfusion phenotype includes increased myocardial apoptosis and resultant decreased contractile performance. CONCLUSIONS: Overall, our results demonstrate the in vivo utility of GiCT to dissect specific mechanisms attributed to Gi signaling in stressed myocardium. Our results with GiCT indicate that upregulation of G alpha(i2) is an adaptive protective response after ischemia to shield myocytes from apoptosis.


Assuntos
Apoptose/fisiologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Mitocôndrias Cardíacas/fisiologia , Isquemia Miocárdica/fisiopatologia , Miócitos Cardíacos/fisiologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Células Cultivadas/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/antagonistas & inibidores , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/química , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/prevenção & controle , Humanos , Isoproterenol/farmacologia , Camundongos , Camundongos Transgênicos , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Isquemia Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Estresse Oxidativo , Fragmentos de Peptídeos/genética , Ratos , Receptores Acoplados a Proteínas G/fisiologia , Proteínas Recombinantes de Fusão/fisiologia , Transdução de Sinais/fisiologia , Transdução Genética
17.
Cell Res ; 17(10): 858-68, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17893708

RESUMO

RIG-I (retinoid acid-inducible gene-I), a putative RNA helicase with a cytoplasmic caspase-recruitment domain (CARD), was identified as a pattern-recognition receptor (PRR) that mediates antiviral immunity by inducing type I interferon production. To further study the biological function of RIG-I, we generated Rig-I(-/-) mice through homologous recombination, taking a different strategy to the previously reported strategy. Our Rig-I(-/-) mice are viable and fertile. Histological analysis shows that Rig-I(-/-) mice develop a colitis-like phenotype and increased susceptibility to dextran sulfate sodium-induced colitis. Accordingly, the size and number of Peyer's patches dramatically decreased in mutant mice. The peripheral T-cell subsets in mutant mice are characterized by an increase in effector T cells and a decrease in naive T cells, indicating an important role for Rig-I in the regulation of T-cell activation. It was further found that Rig-I deficiency leads to the downregulation of G protein alpha i2 subunit (G alpha i2) in various tissues, including T and B lymphocytes. By contrast, upregulation of Rig-I in NB4 cells that are treated with ATRA is accompanied by elevated G alpha i2 expression. Moreover, G alpha i2 promoter activity is increased in co-transfected NIH3T3 cells in a Rig-I dose-dependent manner. All these findings suggest that Rig-I has crucial roles in the regulation of G alpha i2 expression and T-cell activation. The development of colitis may be, at least in part, associated with downregulation of G alpha i2 and disturbed T-cell homeostasis.


Assuntos
Colite/genética , RNA Helicases DEAD-box/genética , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Animais , Apoptose/genética , Apoptose/fisiologia , Northern Blotting , Western Blotting , Células Cultivadas , Colite/induzido quimicamente , Colite/patologia , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , RNA Helicases DEAD-box/fisiologia , Sulfato de Dextrana/toxicidade , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Camundongos , Camundongos Knockout , Células NIH 3T3 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/citologia , Linfócitos T/metabolismo
18.
Blood ; 110(10): 3773-9, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17699741

RESUMO

Chemokines, including CXCL1, participate in neutrophil recruitment by triggering the activation of integrins, which leads to arrest from rolling. The downstream signaling pathways which lead to integrin activation and neutophil arrest following G-protein-coupled receptor engagement are incompletely understood. To test whether Galpha(i2) is involved, mouse neutrophils in their native whole blood were investigated in mouse cremaster postcapillary venules and in flow chambers coated with P-selectin, ICAM-1, and CXCL1. Gnai2(-/-) neutrophils showed significantly reduced CXCL1-induced arrest in vitro and in vivo. Similar results were obtained with leukotriene B(4) (LTB(4)). Lethally irradiated mice reconstituted with Gnai2(-/-) bone marrow showed a similar defect in chemoattractant-induced arrest as that of Gnai2(-/-) mice. In thioglycollate-induced peritonitis and lipopolysaccaride (LPS)-induced lung inflammation, chimeric mice lacking Galpha(i2) in hematopoietic cells showed about 50% reduced neutrophil recruitment similar to that seen in Gnai2(-/-) mice. These data show that neutrophil Galpha(i2) is necessary for chemokine-induced arrest, which is relevant for neutrophil recruitment to sites of acute inflammation.


Assuntos
Ciclo Celular/efeitos dos fármacos , Quimiocinas/farmacologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Neutrófilos/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Lipopolissacarídeos , Camundongos , Camundongos Knockout , Neutrófilos/citologia , Peritonite/induzido quimicamente , Peritonite/patologia , Pneumonia/induzido quimicamente , Pneumonia/patologia , Tioglicolatos , Fosfolipases Tipo C/fisiologia , Vênulas/metabolismo , Vênulas/patologia
19.
Diabetes ; 56(9): 2319-27, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17575083

RESUMO

Ghrelin reportedly serves as a physiological regulator of insulin release. This study aimed to explore signaling mechanisms for insulinostatic ghrelin action in islet beta-cells, with special attention to heterotrimeric GTP-binding proteins and K(+) channels. Plasma insulin and growth hormone (GH) concentrations in rats were measured by enzyme-linked immunosorbent assay (ELISA). Islets were isolated from rats, ghrelin-knockout (Ghr-KO) mice, and wild-type mice by collagenase digestion, and insulin release was determined by ELISA. In rat single beta-cells, cytosolic Ca(2+) concentration ([Ca(2+)](i)) was measured by fura-2 microfluorometry, and membrane potentials and whole cell currents by patch-clamp technique. In rats, systemic ghrelin administration decreased plasma insulin concentrations, and this effect was blocked by treatment with pertussis toxin (PTX), whereas stimulation of GH release remained unaffected. In rat islets, ghrelin receptor antagonist increased and exogenous ghrelin suppressed glucose-induced insulin release in a PTX-sensitive manner. Glucose-induced insulin release from islets was greater in Ghr-KO than wild-type mice, and this enhanced secretion was blunted with PTX. Ghrelin PTX sensitively increased voltage-dependent K(+) (Kv) currents without affecting ATP-sensitive K(+) channels in rat beta-cells. In the presence of Kv channel blockers, ghrelin failed to suppress insulin release. Ghrelin attenuated glucose-induced action potentials and [Ca(2+)](i) increases in beta-cells. Suppressions of [Ca(2+)](i) increase and insulin release by ghrelin were blunted in beta-cells treated with PTX and with antisense oligonucleotide specific for G-protein Galpha(i2)-subunit. Ghrelin attenuates glucose-induced insulin release via PTX-sensitive Galpha(i2)-mediated activation of Kv channels and suppression of [Ca(2+)](i) in beta-cells, representing the unique signaling of ghrelin distinct from that for GH release.


Assuntos
Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Células Secretoras de Insulina/fisiologia , Insulina/metabolismo , Hormônios Peptídicos/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Transdução de Sinais/fisiologia , Animais , Cálcio/fisiologia , Grelina , Insulina/sangue , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Hormônios Peptídicos/deficiência , Hormônios Peptídicos/genética , Ratos , Ratos Wistar
20.
J Immunol ; 179(1): 439-48, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17579064

RESUMO

Signals generated by the engagement of chemoattractants with their cognate receptors orchestrate lymphocyte movements into and out of lymphoid organs and sites of inflammation. Yet, the role of chemokines in organizing lymphocyte movements in lymphoid organs is controversial. Recent evidence suggests that the extensive network of fibroblastic reticular cells within the T cell areas helps guide T cells. The expression of adhesion molecules and chemokines by fibroblastic reticular cells most likely facilitates their influence on T cell movements. Consistent with this hypothesis, CD4 T cells with defective chemokine receptor signaling move very differently within lymph nodes than do normal cells. For the imaging studies, we used CD4 T cells prepared from Gnai2(-/-) mice, which lack G(alphai2) expression. We first demonstrate that CD4 as well as CD8 T cells from these mice are markedly defective in chemokine receptor signaling. Gnai2(-/-) T cells have profound defects in chemokine-induced intracellular calcium mobilization, chemotaxis, and homing, whereas Gnai2(+/-) T cells exhibit modest defects. Intravital imaging revealed that within the inguinal lymph nodes Gnai2(-/-) CD4 T accumulate at the cortical ridge, poorly accessing the lymph node paracortex. They also lack the customary amoeboid-like cell movements and active membrane projections observed with normal CD4 T cells. These results demonstrate the importance of G(alphai2) for T lymphocyte chemokine receptor signaling and argue that local chemoattractants regulate the movement of CD4 T cells in lymph nodes.


Assuntos
Inibição de Migração Celular , Quimiotaxia de Leucócito/imunologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/biossíntese , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Linfonodos/imunologia , Linfonodos/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Sinalização do Cálcio/genética , Sinalização do Cálcio/imunologia , Quimiotaxia de Leucócito/genética , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Regulação da Expressão Gênica/imunologia , Linfonodos/patologia , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/deficiência , Isoformas de Proteínas/fisiologia , Receptores de Quimiocinas/antagonistas & inibidores , Receptores de Quimiocinas/biossíntese , Receptores de Quimiocinas/fisiologia , Subpopulações de Linfócitos T/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...