Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 205
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 117(35): 21723-21730, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32817560

RESUMO

G proteins are activated when they associate with G protein-coupled receptors (GPCRs), often in response to agonist-mediated receptor activation. It is generally thought that agonist-induced receptor-G protein association necessarily promotes G protein activation and, conversely, that activated GPCRs do not interact with G proteins that they do not activate. Here we show that GPCRs can form agonist-dependent complexes with G proteins that they do not activate. Using cell-based bioluminescence resonance energy transfer (BRET) and luminescence assays we find that vasopressin V2 receptors (V2R) associate with both Gs and G12 heterotrimers when stimulated with the agonist arginine vasopressin (AVP). However, unlike V2R-Gs complexes, V2R-G12 complexes are not destabilized by guanine nucleotides and do not promote G12 activation. Activating V2R does not lead to signaling responses downstream of G12 activation, but instead inhibits basal G12-mediated signaling, presumably by sequestering G12 heterotrimers. Overexpressing G12 inhibits G protein receptor kinase (GRK) and arrestin recruitment to V2R and receptor internalization. Formyl peptide (FPR1 and FPR2) and Smoothened (Smo) receptors also form complexes with G12 that are insensitive to nucleotides, suggesting that unproductive GPCR-G12 complexes are not unique to V2R. These results indicate that agonist-dependent receptor-G protein association does not always lead to G protein activation and may in fact inhibit G protein activation.


Assuntos
Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , AMP Cíclico/metabolismo , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Ligantes , Ligação Proteica/fisiologia , Receptores de Vasopressinas/metabolismo , Transdução de Sinais/fisiologia , Vasopressinas/metabolismo , beta-Arrestinas/metabolismo
2.
J Biol Chem ; 296: 100632, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33865855

RESUMO

Nonshivering thermogenesis is essential for mammals to maintain body temperature. According to the canonical view, temperature is sensed by cutaneous thermoreceptors and nerve impulses transmitted to the hypothalamus, which generates sympathetic signals to ß-adrenergic receptors in brown adipocytes. The energy for heat generation is primarily provided by the oxidation of fatty acids derived from triglyceride hydrolysis and cellular uptake. Fatty acids also activate the uncoupling protein, UCP1, which creates a proton leak that uncouples mitochondrial oxidative phosphorylation from ATP production, resulting in energy dissipation as heat. Recent evidence supports the idea that in response to mild cold, ß-adrenergic signals stimulate not only lipolysis and fatty acid oxidation, but also act through the mTORC2-Akt signaling module to stimulate de novo lipogenesis. This opposing anabolic effect is thought to maintain lipid fuel stores during increased catabolism. We show here, using brown fat-specific Gs-alpha knockout mice and cultured adipocytes that, unlike mild cold, severe cold directly cools brown fat and bypasses ß-adrenergic signaling to inhibit mTORC2. This cell-autonomous effect both inhibits lipogenesis and augments UCP1 expression to enhance thermogenesis. These findings suggest a novel mechanism for overriding ß-adrenergic-stimulated anabolic activities while augmenting catabolic activities to resolve the homeostatic crisis presented by severe cold.


Assuntos
Tecido Adiposo Marrom/metabolismo , Cromograninas/fisiologia , Temperatura Baixa , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Termogênese , Tecido Adiposo Marrom/citologia , Animais , Lipogênese , Masculino , Alvo Mecanístico do Complexo 2 de Rapamicina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Adrenérgicos beta/genética , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
3.
Mol Pharmacol ; 100(3): 203-216, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34158361

RESUMO

Norepinephrine (NE) controls many vital body functions by activating adrenergic receptors (ARs). Average core body temperature (CBT) in mice is 37°C. Of note, CBT fluctuates between 36 and 38°C within 24 hours, but little is known about the effects of CBT changes on the pharmacodynamics of NE. Here, we used Peltier element-controlled incubators and challenged murine hypothalamic mHypoA -2/10 cells with temperature changes of ±1°C. We observed enhanced NE-induced activation of a cAMP-dependent luciferase reporter at 36 compared with 38°C. mRNA analysis and subtype specific antagonists revealed that NE activates ß 2- and ß 3-AR in mHypoA-2/10 cells. Agonist binding to the ß 2-AR was temperature insensitive, but measurements of cytosolic cAMP accumulation revealed an increase in efficacy of 45% ± 27% for NE and of 62% ± 33% for the ß 2-AR-selective agonist salmeterol at 36°C. When monitoring NE-promoted cAMP efflux, we observed an increase in the absolute efflux at 36°C. However, the ratio of exported to cytosolic accumulated cAMP is higher at 38°C. We also stimulated cells with NE at 37°C and measured cAMP degradation at 36 and 38°C afterward. We observed increased cAMP degradation at 38°C, indicating enhanced phosphodiesterase activity at higher temperatures. In line with these data, NE-induced activation of the thyreoliberin promoter was found to be enhanced at 36°C. Overall, we show that physiologic temperature changes fine-tune NE-induced cAMP signaling in hypothalamic cells via ß 2-AR by modulating cAMP degradation and the ratio of intra- and extracellular cAMP. SIGNIFICANCE STATEMENT: Increasing cytosolic cAMP levels by activation of G protein-coupled receptors (GPCR) such as the ß 2-adrenergic receptor (AR) is essential for many body functions. Changes in core body temperature are fundamental and universal factors of mammalian life. This study provides the first data linking physiologically relevant temperature fluctuations to ß 2-AR-induced cAMP signaling, highlighting a so far unappreciated role of body temperature as a modulator of the prototypic class A GPCR.


Assuntos
AMP Cíclico/metabolismo , Citosol/metabolismo , Receptores Adrenérgicos beta 2/fisiologia , 1-Metil-3-Isobutilxantina/farmacologia , Fatores de Transcrição ARNTL/metabolismo , Aminopiridinas/farmacologia , Animais , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Hipotálamo/fisiologia , Camundongos , Neurônios/fisiologia , Norepinefrina/farmacologia , Receptores Adrenérgicos beta 2/biossíntese , Receptores Adrenérgicos beta 3/biossíntese , Receptores Adrenérgicos beta 3/fisiologia , Fatores de Transcrição STAT/metabolismo , Xinafoato de Salmeterol/farmacologia , Transdução de Sinais/fisiologia , Temperatura , Hormônio Liberador de Tireotropina/genética , Hormônio Liberador de Tireotropina/metabolismo
4.
J Mol Cell Cardiol ; 132: 49-59, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31071332

RESUMO

OBJECTIVE: Abdominal aortic aneurysm (AAA) is a life-threatening vascular disease without an effective pharmaceutical treatment. Genetic studies have proved the involvement of smooth muscle phenotype switch in the development of AAA. The alpha subunit of the heterotrimeric G stimulatory protein (Gsα) mediates receptor-stimulated production of cyclic adenosine monophosphate (cAMP). However, the role of smooth muscle Gsα in AAA formation remains unknown. APPROACH AND RESULTS: In this study, mice with knockout of smooth muscle-specific Gsα (GsαSMKO) were generated by cross-breeding Gsαflox/flox mice with SM22-CreERT2 transgenic mice, induced in adult mice by tamoxifen treatment. Gsα deficiency induced a smooth muscle phenotype switch from a contractile to a synthetic state. Mechanically, Gsα deletion reduced cAMP level and increased the level of human antigen R (HuR), which binds with the adenylate uridylate-rich elements of the 3' untranslated region of Krüppel-like factor 4 (KLF4) mRNA, thereby increasing the stability of KLF4. Moreover, genetic knockdown of HuR or KLF4 rescued the phenotype switch in Gsα-deficient smooth muscle cells. Furthermore, with acute infusion of angiotensin II, the incidence of AAA was markedly higher in ApoE-/-/GsαSMKO than ApoE-/-/Gsαflox/flox mice and induced increased elastic lamina degradation and aortic expansion. Finally, the levels of Gsα and SM α-actin were significantly lower while those of HuR and KLF4 were higher in human AAA samples than adjacent nonaneurysmal aortic sections. CONCLUSIONS: Gsα may play a protective role in AAA formation by regulating the smooth muscle phenotype switch and could be a potential therapeutic target for AAA disease.


Assuntos
Angiotensina II/toxicidade , Aneurisma da Aorta Abdominal/etiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Músculo Liso Vascular/patologia , Vasoconstritores/toxicidade , Animais , Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/patologia , Modelos Animais de Doenças , Humanos , Fator 4 Semelhante a Kruppel , Masculino , Camundongos , Camundongos Knockout para ApoE , Camundongos Transgênicos , Músculo Liso Vascular/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Fenótipo , Transdução de Sinais
5.
Gastroenterology ; 155(5): 1593-1607.e12, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30142336

RESUMO

BACKGROUND & AIMS: Mutations at hotspots in GNAS, which encodes stimulatory G-protein, α subunits, are detected in approximately 60% of intraductal papillary mucinous neoplasms (IPMNs) of the pancreas. We generated mice with KRAS-induced IPMNs that also express a constitutively active form of GNAS in pancreas and studied tumor development. METHODS: We generated p48-Cre; LSL-KrasG12D; Rosa26R-LSL-rtTA-TetO-GnasR201C mice (Kras;Gnas mice); pancreatic tissues of these mice express activated KRAS and also express a mutant form of GNAS (GNASR201C) upon doxycycline administration. Mice that were not given doxycycline were used as controls, and survival times were compared by Kaplan-Meier analysis. Pancreata were collected at different time points after doxycycline administration and analyzed by histology. Pancreatic ductal adenocarcinomas (PDACs) were isolated from mice and used to generate cell lines, which were analyzed by reverse transcription polymerase chain reaction, immunoblotting, immunohistochemistry, and colony formation and invasion assays. Full-length and mutant forms of yes-associated protein (YAP) were expressed in PDAC cells. IPMN specimens were obtained from 13 patients with IPMN undergoing surgery and analyzed by immunohistochemistry. RESULTS: All Kras;Gnas mice developed pancreatic cystic lesions that resemble human IPMNs; the grade of epithelial dysplasia increased with time. None of the control mice developed cystic lesions. Approximately one third of Kras;Gnas mice developed PDACs at a median of 30 weeks after doxycycline administration, whereas 33% of control mice developed PDACs. Expression of GNASR201C did not accelerate the development of PDACs compared with control mice. However, the neoplasms observed in Kras;Gnas mice were more differentiated, and expressed more genes associated with ductal phenotypes, than in control mice. PDACs isolated from Kras;Gnas mice had activation of the Hippo pathway; in cells from these tumors, phosphorylated YAP1 was sequestered in the cytoplasm, and this was also observed in human IPMNs with GNAS mutations. Sequestration of YAP1 was not observed in PDAC cells from control mice. CONCLUSIONS: In mice that express activated KRAS in the pancreas, we found expression of GNASR201C to cause development of more differentiated tumors, with gene expression pattern associated with the ductal phenotype. Expression of mutant GNAS caused phosphorylated YAP1 to be sequestered in the cytoplasm, altering tumor progression.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Carcinoma Ductal Pancreático/etiologia , Cromograninas/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Mutação , Neoplasias Císticas, Mucinosas e Serosas/etiologia , Neoplasias Pancreáticas/etiologia , Fosfoproteínas/fisiologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Transdução de Sinais/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/análise , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Animais , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Cromograninas/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Humanos , Camundongos , Fosfoproteínas/análise , Fosfoproteínas/antagonistas & inibidores , Proteínas Proto-Oncogênicas p21(ras)/fisiologia , Proteínas de Sinalização YAP
6.
Exp Cell Res ; 333(2): 289-302, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25704759

RESUMO

G protein-coupled receptor (GPCR) signaling in osteoblasts (OBs) is an important regulator of bone formation. We previously described a mouse model expressing Rs1, an engineered constitutively active Gs-coupled GPCR, under the control of the 2.3 kb Col I promoter. These mice showed a dramatic age-dependent increase in trabecular bone of femurs. Here, we further evaluated the effects of enhanced Gs signaling in OBs on intramembranous bone formation by examining calvariae of 1- and 9-week-old Col1(2.3)/Rs1 mice and characterized the in vivo gene expression specifically occurring in osteoblasts with activated Gs G protein-coupled receptor signaling, at the cellular level rather than in a whole bone. Rs1 calvariae displayed a dramatic increase in bone volume with partial loss of cortical structure. By immunohistochemistry, Osterix was detected in cells throughout the inter-trabecular space while Osteocalcin was expressed predominantly in cells along bone surfaces, suggesting the role of paracrine mediators secreted from OBs driven by 2.3 kb Col I promoter could influence early OB commitment, differentiation, and/or proliferation. Gene expression analysis of calvarial OBs revealed that genes affected by Rs1 signaling include those encoding proteins important for cell differentiation, cytokines and growth factors, angiogenesis, coagulation, and energy metabolism. The set of Gs-GPCRs and other GPCRs that may contribute to the observed skeletal phenotype and candidate paracrine mediators of the effect of Gs signaling in OBs were also determined. Our results identify novel detailed in vivo cellular changes of the anabolic response of the skeleton to Gs signaling in mature OBs.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Osteoblastos/metabolismo , Transcriptoma , Animais , Regeneração Óssea , Células Cultivadas , Fator 9 de Crescimento de Fibroblastos/genética , Fator 9 de Crescimento de Fibroblastos/metabolismo , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Osteogênese , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Crânio/patologia , Crânio/fisiopatologia , Fator de Transcrição Sp7 , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
7.
PLoS Genet ; 8(5): e1002706, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22589743

RESUMO

It has been suggested that imprinted genes are important in the regulation of sleep. However, the fundamental question of whether genomic imprinting has a role in sleep has remained elusive up to now. In this work we show that REM and NREM sleep states are differentially modulated by the maternally expressed imprinted gene Gnas. In particular, in mice with loss of imprinting of Gnas, NREM and complex cognitive processes are enhanced while REM and REM-linked behaviors are inhibited. This is the first demonstration that a specific overexpression of an imprinted gene affects sleep states and related complex behavioral traits. Furthermore, in parallel to the Gnas overexpression, we have observed an overexpression of Ucp1 in interscapular brown adipose tissue (BAT) and a significant increase in thermoregulation that may account for the REM/NREM sleep phenotypes. We conclude that there must be significant evolutionary advantages in the monoallelic expression of Gnas for REM sleep and for the consolidation of REM-dependent memories. Conversely, biallelic expression of Gnas reinforces slow wave activity in NREM sleep, and this results in a reduction of uncertainty in temporal decision-making processes.


Assuntos
Cognição/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Impressão Genômica , Sono REM/genética , Sono REM/fisiologia , Tecido Adiposo Marrom , Alelos , Animais , Temperatura Corporal , Regulação da Temperatura Corporal/genética , Regulação da Temperatura Corporal/fisiologia , Cromograninas , Metilação de DNA , Eletroencefalografia , Éxons , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Regulação da Expressão Gênica , Canais Iônicos , Camundongos , Proteínas Mitocondriais , Deleção de Sequência , Proteína Desacopladora 1 , Vigília
8.
J Neurosci ; 32(5): 1714-29, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22302812

RESUMO

Numerous physiological functions, including a role as a cell surface receptor, have been ascribed to Alzheimer's disease-associated amyloid precursor protein (APP). However, detailed analysis of intracellular signaling mediated by APP in neurons has been lacking. Here, we characterized intrinsic signaling associated with membrane-bound APP C-terminal fragments, which are generated following APP ectodomain release by α- or ß-secretase cleavage. We found that accumulation of APP C-terminal fragments or expression of membrane-tethered APP intracellular domain results in adenylate cyclase-dependent activation of PKA (protein kinase A) and inhibition of GSK3ß signaling cascades, and enhancement of axodendritic arborization in rat immortalized hippocampal neurons, mouse primary cortical neurons, and mouse neuroblastoma. We discovered an interaction between BBXXB motif of APP intracellular domain and the heterotrimeric G-protein subunit Gα(S), and demonstrate that Gα(S) coupling to adenylate cyclase mediates membrane-tethered APP intracellular domain-induced neurite outgrowth. Our study provides clear evidence that APP intracellular domain can have a nontranscriptional role in regulating neurite outgrowth through its membrane association. The novel functional coupling of membrane-bound APP C-terminal fragments with Gα(S) signaling identified in this study could impact several brain functions such as synaptic plasticity and memory formation.


Assuntos
Precursor de Proteína beta-Amiloide/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Membranas Intracelulares/fisiologia , Transdução de Sinais/fisiologia , Adenilil Ciclases/metabolismo , Adenilil Ciclases/fisiologia , Sequência de Aminoácidos , Precursor de Proteína beta-Amiloide/química , Animais , Células COS , Linhagem Celular Transformada , Linhagem Celular Tumoral , Membrana Celular/química , Membrana Celular/fisiologia , Proliferação de Células , Chlorocebus aethiops , Feminino , Subunidades alfa Gs de Proteínas de Ligação ao GTP/química , Membranas Intracelulares/química , Masculino , Camundongos , Dados de Sequência Molecular , Neuritos/fisiologia , Estrutura Terciária de Proteína , Ratos
9.
J Pharmacol Exp Ther ; 345(3): 404-18, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23516330

RESUMO

Parathyroid hormone (PTH) and parathyroid hormone-related peptide (PTHrP), acting through the osteoblast PTH1 receptor (PTH1R), play important roles in bone remodeling. Intermittent administration of PTH(1-34) (teriparatide) leads to bone formation, whereas continuous administration paradoxically leads to bone resorption. Activation of PTH1R promotes regulation of multiple signaling pathways, including G(s)/cAMP/protein kinase A, G(q)/calcium/protein kinase C, ß-arrestin recruitment, and extracellular signal-related kinase (ERK)1/2 phosphorylation, as well as receptor internalization, but their role in promoting anabolic and catabolic actions of PTH(1-34) are unclear. In the present investigation, a collection of PTH(1-34) and PTHrP(1-34) peptide analogs were evaluated in orthogonal human PTH1R (hPTH1R) functional assays capturing G(s)- and G(q)-signaling, ß-arrestin recruitment, ERK1/2 phosphorylation, and receptor internalization to further define the patterns of PTH1R signaling that they stimulate and further establish peptide domains contributing to agonist activity. Results indicate that both N- and C-terminal domains of PTH and PTHrP are critical for activation of signaling pathways. However, modifications of both regions lead to more substantial decreases in agonist potency and efficacy to stimulate G(q)-signaling, ß-arrestin recruitment, ERK1/2 phosphorylation, and receptor internalization than to stimulate G(s)-signaling. The substantial contribution of the peptide C-terminal domain in activation of hPTH1R signaling suggests a role in positioning of the peptide N-terminal region into the receptor J-domain. Several PTH and PTHrP peptides evaluated in this study promote different patterns of biased agonist signaling and may serve as useful tools to further elucidate therapeutically relevant PTH1R signaling in osteoblasts. With a better understanding of therapeutically relevant signaling, novel biased peptides with desired signaling could be designed for safer and more effective treatment of osteoporosis.


Assuntos
Hormônio Paratireóideo/farmacologia , Fragmentos de Peptídeos/farmacologia , Receptor Tipo 2 de Hormônio Paratireóideo/agonistas , Receptor Tipo 2 de Hormônio Paratireóideo/fisiologia , Transdução de Sinais/fisiologia , Algoritmos , Animais , Arrestina/fisiologia , Conservadores da Densidade Óssea/farmacologia , Células CHO , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Desenho de Fármacos , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Humanos , Fosfatos de Inositol/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Hormônio Paratireóideo/química , Proteína Relacionada ao Hormônio Paratireóideo/farmacologia , Fragmentos de Peptídeos/química , Fosforilação , Receptor Tipo 2 de Hormônio Paratireóideo/antagonistas & inibidores
10.
Exp Physiol ; 98(10): 1432-45, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23748904

RESUMO

Imbalances of energy homeostasis are often associated with cardiovascular complications. Previous work has shown that Gnasxl-deficient mice have a lean and hypermetabolic phenotype, with increased sympathetic stimulation of adipose tissue. The Gnasxl transcript from the imprinted Gnas locus encodes the trimeric G-protein subunit XLαs, which is expressed in brain regions that regulate energy homeostasis and sympathetic nervous system (SNS) activity. To determine whether Gnasxl knock-out (KO) mice display additional SNS-related phenotypes, we have now investigated the cardiovascular system. The Gnasxl KO mice were ∼20 mmHg hypertensive in comparison to wild-type (WT) littermates (P ≤ 0.05) and hypersensitive to the sympatholytic drug reserpine. Using telemetry, we detected an increased waking heart rate in conscious KOs (630 ± 10 versus 584 ± 12 beats min(-1), KO versus WT, P ≤ 0.05). Body temperature was also elevated (38.1 ± 0.3 versus 36.9 ± 0.4°C, KO versus WT, P ≤ 0.05). To investigate autonomic nervous system influences, we used heart rate variability analyses. We empirically defined frequency power bands using atropine and reserpine and verified high-frequency (HF) power and low-frequency (LF) LF/HF power ratio to be indicators of parasympathetic and sympathetic activity, respectively. The LF/HF power ratio was greater in KOs and more sensitive to reserpine than in WTs, consistent with elevated SNS activity. In contrast, atropine and exendin-4, a centrally acting agonist of the glucagon-like peptide-1 receptor, which influences cardiovascular physiology and metabolism, reduced HF power equally in both genotypes. This was associated with a greater increase in heart rate in KOs. Mild stress had a blunted effect on the LF/HF ratio in KOs consistent with elevated basal sympathetic activity. We conclude that XLαs is required for the inhibition of sympathetic outflow towards cardiovascular and metabolically relevant tissues.


Assuntos
Pressão Sanguínea/fisiologia , Temperatura Corporal , Subunidades alfa Gs de Proteínas de Ligação ao GTP/deficiência , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Frequência Cardíaca/fisiologia , Animais , Atropina/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Temperatura Corporal/efeitos dos fármacos , Cromograninas , Exenatida , Receptor do Peptídeo Semelhante ao Glucagon 1 , Frequência Cardíaca/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Peptídeos/farmacologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores de Glucagon/metabolismo , Reserpina/farmacologia , Estresse Psicológico , Peçonhas/farmacologia
11.
Dev Cell ; 13(5): 743-751, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17981141

RESUMO

An important problem in polarized morphogenesis is how polarized transport of membrane vesicles is spatiotemporally regulated. Here, we report that a local change in the transbilayer phospholipid distribution of the plasma membrane regulates the axis of polarized growth. Type 4 P-type ATPases Lem3p-Dnf1p and -Dnf2p are putative heteromeric phospholipid flippases in budding yeast that are localized to polarized sites on the plasma membrane. The lem3Delta mutant exhibits prolonged apical growth due to a defect in the switch to isotropic bud growth. In lem3Delta cells, the small GTPase Cdc42p remains polarized at the bud tip where phosphatidylethanolamine remains exposed on the outer leaflet. Intriguingly, phosphatidylethanolamine and phosphatidylserine stimulate GTPase-activating protein (GAP) activity of Rga1p and Rga2p toward Cdc42p, whereas PI(4,5)P(2) inhibits it. We propose that a redistribution of phospholipids to the inner leaflet of the plasma membrane triggers the dispersal of Cdc42p from the apical growth site, through activation of GAPs.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Proteínas Ativadoras de GTPase/fisiologia , Fosfolipídeos/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/fisiologia , Proteína cdc42 de Saccharomyces cerevisiae de Ligação ao GTP/fisiologia , Transportadores de Cassetes de Ligação de ATP , Adenosina Trifosfatases/metabolismo , Membrana Celular/metabolismo , Polaridade Celular , Proliferação de Células , Proteínas de Membrana Transportadoras/metabolismo , Mutação , Fosfatidiletanolaminas/metabolismo , Fosfatidilserinas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteína cdc42 de Saccharomyces cerevisiae de Ligação ao GTP/genética
12.
Minerva Endocrinol ; 37(2): 133-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22691887

RESUMO

Abnormalities in the cAMP/PKA signaling pathway have been linked to the formation of benign adrenal tumors, as well as a possible predisposition to adrenocortical cancer. Mutations in the G-protein coupled receptor are associated with McCune-Albright syndrome and ACTH-independent macronodular adrenal hyperplasia, while defects in cAMP-dependent protein kinase A can lead to the development of Carney's complex, as well as primary pigmented nodular adrenocortical disease (PPNAD), and micronodular adrenocortical hyperplasia (MAH). Defects in phosphodiesterases, which regulate cAMP levels, have also been demonstrated in PPNAD and MAH. The Wnt signaling pathway, which is involved in oncogenesis in a variety of tumors, has also been implicated in adrenocortical tumorigenesis. MicroRNA profiling has added to our understanding of the signaling pathways involved in tumor formation in the adrenal cortex. Will this all lead to the development of specific targets for pharmacologic therapies? In this article, we review the molecular genetics of adrenocortical tumors and refer to potential targets for pharmacologic therapy.


Assuntos
Neoplasias do Córtex Suprarrenal/genética , Carcinoma/genética , Transdução de Sinais/fisiologia , 3',5'-AMP Cíclico Fosfodiesterases/fisiologia , Neoplasias do Córtex Suprarrenal/tratamento farmacológico , Hormônio Adrenocorticotrópico/fisiologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Carcinoma/tratamento farmacológico , Cromograninas , AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/deficiência , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Humanos , MicroRNAs/fisiologia , Terapia de Alvo Molecular , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/fisiologia , Receptores da Corticotropina/deficiência , Receptores da Corticotropina/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteínas Wnt/fisiologia
13.
Clin Exp Nephrol ; 16(1): 17-24, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22124804

RESUMO

Control of the renin system by physiological mechanisms such as the baroreceptor or the macula densa (MD) is characterized by asymmetry in that the capacity for renin secretion and expression to increase is much larger than the magnitude of the inhibitory response. The large stimulatory reserve of the renin-angiotensin system may be one of the causes for the remarkable salt-conserving power of the mammalian kidney. Physiological stimulation of renin secretion and expression relies on the activation of regulatory pathways that converge on the cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) pathway. Mice with selective Gs-alpha (Gsα) deficiency in juxtaglomerular granular cells show a marked reduction of basal renin secretion, and an almost complete unresponsiveness of renin release to furosemide, hydralazine, or isoproterenol. Cyclooxygenase-2 generating prostaglandin E(2) (PGE(2)) and prostacyclin (PGI(2)) in MD and thick ascending limb cells is one of the main effector systems utilizing Gsα-coupled receptors to stimulate the renin-angiotensin system. In addition, ß-adrenergic receptors are critical for the expression of high basal levels of renin and for its release response to lowering blood pressure or MD sodium chloride concentration. Nitric oxide generated by nitric oxide synthases in the MD and in endothelial cells enhances cAMP-dependent signaling by stabilizing cAMP through cyclic guanosine monophosphate-dependent inhibition of phosphodiesterase 3. The stimulation of renin secretion by drugs that inhibit angiotensin II formation or action results from the convergent activation of cAMP probably through indirect augmentation of the activity of PGE(2) and PGI(2) receptors, ß-adrenergic receptors, and nitric oxide.


Assuntos
AMP Cíclico/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Renina/metabolismo , Transdução de Sinais/fisiologia , Animais , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Ciclo-Oxigenase 2/fisiologia , Diuréticos/farmacologia , Furosemida/farmacologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/deficiência , Sistema Justaglomerular/citologia , Sistema Justaglomerular/metabolismo , Túbulos Renais Distais/metabolismo , Camundongos , Óxido Nítrico Sintase/metabolismo , Prostaglandinas/fisiologia , Receptores Adrenérgicos beta/metabolismo , Sistema Renina-Angiotensina/fisiologia , Transdução de Sinais/efeitos dos fármacos
14.
Endocrinology ; 163(2)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34864945

RESUMO

Gonadotropin-releasing hormone (GnRH) regulates gonadal function via its stimulatory effects on gonadotropin production by pituitary gonadotrope cells. GnRH is released from the hypothalamus in pulses and GnRH pulse frequency differentially regulates follicle-stimulating hormone (FSH) and luteinizing hormone (LH) synthesis and secretion. The GnRH receptor (GnRHR) is a G protein-coupled receptor that canonically activates Gα q/11-dependent signaling on ligand binding. However, the receptor can also couple to Gα s and in vitro data suggest that toggling between different G proteins may contribute to GnRH pulse frequency decoding. For example, as we show here, knockdown of Gα s impairs GnRH-stimulated FSH synthesis at low- but not high-pulse frequency in a model gonadotrope-derived cell line. We next used a Cre-lox conditional knockout approach to interrogate the relative roles of Gα q/11 and Gα s proteins in gonadotrope function in mice. Gonadotrope-specific Gα q/11 knockouts exhibit hypogonadotropic hypogonadism and infertility, akin to the phenotypes seen in GnRH- or GnRHR-deficient mice. In contrast, under standard conditions, gonadotrope-specific Gα s knockouts produce gonadotropins at normal levels and are fertile. However, the LH surge amplitude is blunted in Gα s knockout females and postgonadectomy increases in FSH and LH are reduced both in males and females. These data suggest that GnRH may signal principally via Gα q/11 to stimulate gonadotropin production, but that Gα s plays important roles in gonadotrope function in vivo when GnRH secretion is enhanced.


Assuntos
Cromograninas/fisiologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Gonadotrofos/metabolismo , Gonadotropinas/metabolismo , Animais , Castração , Linhagem Celular , Cromograninas/genética , Feminino , Fertilidade/genética , Fertilidade/fisiologia , Subunidade beta do Hormônio Folículoestimulante/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Regulação da Expressão Gênica/fisiologia , Hormônio Liberador de Gonadotropina/fisiologia , Gonadotropinas/genética , Células HEK293 , Humanos , Hormônio Luteinizante/genética , Hormônio Luteinizante/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores LHRH/genética , Receptores LHRH/fisiologia , Maturidade Sexual , Transdução de Sinais/fisiologia
15.
Mol Metab ; 55: 101415, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34883278

RESUMO

OBJECTIVE: The goal of this study was to determine the glucometabolic effects of acute activation of Gs signaling in skeletal muscle (SKM) in vivo and its contribution to whole-body glucose homeostasis. METHODS: To address this question, we studied mice that express a Gs-coupled designer G protein-coupled receptor (Gs-DREADD or GsD) selectively in skeletal muscle. We also identified two Gs-coupled GPCRs that are endogenously expressed by SKM at relatively high levels (ß2-adrenergic receptor and CRF2 receptor) and studied the acute metabolic effects of activating these receptors in vivo by highly selective agonists (clenbuterol and urocortin 2 (UCN2), respectively). RESULTS: Acute stimulation of GsD signaling in SKM impaired glucose tolerance in lean and obese mice by decreasing glucose uptake selectively into SKM. The acute metabolic effects following agonist activation of ß2-adrenergic and, potentially, CRF2 receptors appear primarily mediated by altered insulin release. Clenbuterol injection improved glucose tolerance by increasing insulin secretion in lean mice. In SKM, clenbuterol stimulated glycogen breakdown. UCN2 injection resulted in decreased glucose tolerance associated with lower plasma insulin levels. The acute metabolic effects of UCN2 were not mediated by SKM Gs signaling. CONCLUSIONS: Selective activation of Gs signaling in SKM causes an acute increase in blood glucose levels. However, acute in vivo stimulation of endogenous Gs-coupled receptors enriched in SKM has only a limited impact on whole-body glucose homeostasis, most likely due to the fact that these receptors are also expressed by pancreatic islets where they modulate insulin release.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Músculo Esquelético/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Clembuterol/farmacologia , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Glucose/metabolismo , Intolerância à Glucose/metabolismo , Homeostase/efeitos dos fármacos , Insulina/metabolismo , Resistência à Insulina/fisiologia , Secreção de Insulina/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/fisiologia , Obesidade/metabolismo , Receptores Adrenérgicos beta 2/metabolismo
16.
J Neurosci ; 30(13): 4562-72, 2010 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-20357107

RESUMO

A central question in insect chemoreception is whether signaling occurs via G-proteins. Two families of seven-transmembrane-domain chemoreceptors, the odor (Or) and gustatory receptor (Gr) families, have been identified in Drosophila (Clyne et al., 1999, 2000; Vosshall et al., 1999). Ors mediate odor responses, whereas two Grs, Gr21a and Gr63a, mediate CO2 response (Hallem et al., 2004; Jones et al., 2007; Kwon et al., 2007). Using single-sensillum recordings, we systematically investigate the role of Galpha proteins in vivo, initially with RNA interference constructs, competitive peptides, and constitutively active Galpha proteins. The results do not support a role for Galpha proteins in odor sensitivity. In parallel experiments, manipulations of Galpha(q), but not other Galpha proteins, affected CO2 response. Transient, conditional, and ectopic expression analyses consistently supported a role for Galpha(q) in the response of CO2-sensing neurons, but not odor-sensing neurons. Genetic mosaic analysis confirmed that odor responses are normal in the absence of Galpha(q). Ggamma30A is also required for normal CO2 response. The simplest interpretation of these results is that Galpha(q) and Ggamma30A play a role in the response of CO2-sensing neurons, but are not required for Or-mediated odor signaling.


Assuntos
Células Quimiorreceptoras/fisiologia , Drosophila/fisiologia , Subunidades alfa de Proteínas de Ligação ao GTP/fisiologia , Subunidades gama da Proteína de Ligação ao GTP/fisiologia , Animais , Dióxido de Carbono/fisiologia , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Subunidades gama da Proteína de Ligação ao GTP/genética , Mutação , Odorantes , Condutos Olfatórios , Olfato
17.
J Neurosci ; 30(13): 4660-6, 2010 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-20357116

RESUMO

Clinical pain conditions may remain responsive to opiate analgesics for extended periods, but such persistent acute pain can undergo a transition to an opiate-resistant chronic pain state that becomes a much more serious clinical problem. To test the hypothesis that cellular mechanisms of chronic pain in the primary afferent also contribute to the development of opiate resistance, we used a recently developed model of the transition of from acute to chronic pain, hyperalgesic priming. Repeated intradermal administration of the potent and highly selective mu-opioid agonist, [d-Ala(2),N-MePhe(4),gly-ol]-enkephalin (DAMGO), to produce tolerance for its inhibition of prostaglandin E(2) hyperalgesia, simultaneously produced hyperalgesic priming. Conversely, injection of an inflammogen, carrageenan, used to produce priming produced DAMGO tolerance. Both effects were prevented by inhibition of protein kinase Cepsilon (PKCepsilon). Carrageenan also induced opioid dependence, manifest as mu-opioid receptor antagonist (d-Phe-Cys-Tyr-d-Trp-Orn-Thr-Pen-Thr-NH(2))-induced hyperalgesia that, like priming, was PKCepsilon and G(i) dependent. These findings suggest that the transition from acute to chronic pain, and development of mu-opioid receptor tolerance and dependence may be linked by common cellular mechanisms in the primary afferent.


Assuntos
Tolerância a Medicamentos , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Dor/fisiopatologia , Doença Aguda , Analgésicos Opioides/farmacologia , Animais , Carragenina/farmacologia , Doença Crônica , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Hiperalgesia/etiologia , Hiperalgesia/imunologia , Hiperalgesia/fisiopatologia , Inflamação/complicações , Masculino , Nociceptores/metabolismo , Transtornos Relacionados ao Uso de Opioides/imunologia , Dor/etiologia , Dor/imunologia , Proteína Quinase C-épsilon/antagonistas & inibidores , Proteína Quinase C-épsilon/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Transdução de Sinais , Síndrome de Abstinência a Substâncias/fisiopatologia
18.
J Biol Chem ; 285(15): 11114-20, 2010 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-20133939

RESUMO

The alpha subunit of stimulatory G protein (G alpha(s)) activates adenylyl cyclase, which catalyzes cAMP production, and regulates many physiological aspects, such as cardiac regulation and endocrine systems. Ric-8B (resistance to inhibitors of cholinesterase 8B) has been identified as the G alpha(s)-binding protein; however, its role in G(s) signaling remains obscure. In this study, we present evidence that Ric-8B specifically and positively regulates G(s) signaling by stabilizing the G alpha(s) protein. An in vitro biochemical study suggested that Ric-8B does not possess guanine nucleotide exchange factor activity. However, knockdown of Ric-8B attenuated beta-adrenergic agonist-induced cAMP accumulation, indicating that Ric-8B positively regulates G(s) signaling. Interestingly, overexpression and knockdown of Ric-8B resulted in an increase and a decrease in the G alpha(s) protein, respectively, without affecting the G alpha(s) mRNA level. We found that the G alpha(s) protein is ubiquitinated and that this ubiquitination is inhibited by Ric-8B. This Ric-8B-mediated inhibition of G alpha(s) ubiquitination requires interaction between Ric-8B and G alpha(s) because Ric-8B splicing variants, which are defective for G alpha(s) binding, failed to inhibit the ubiquitination. Taken together, these results suggest that Ric-8B plays a critical and specific role in the control of G alpha(s) protein levels by modulating G alpha(s) ubiquitination and positively regulates G(s) signaling.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Proteínas de Ligação ao GTP/química , Proteínas Nucleares/fisiologia , Ubiquitinação , Animais , Linhagem Celular , AMP Cíclico/metabolismo , Cicloeximida/farmacologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/química , Fatores de Troca do Nucleotídeo Guanina , Humanos , Camundongos , Células NIH 3T3 , Proteínas Nucleares/química , Ligação Proteica , Estrutura Terciária de Proteína , Transdução de Sinais , Ubiquitina/química , Ubiquitina-Proteína Ligases/química
19.
J Immunol ; 182(5): 3243-51, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19234222

RESUMO

Extracellular acidification inhibited LPS-induced TNF-alpha protein production, which was associated with an inhibition of TNF-alpha mRNA expression, in mouse peritoneal macrophages. The LPS-induced cytokine production was also inhibited by G(s) protein-coupled receptor agonists prostaglandin E(1) and isoproterenol. Among OGR1 family proton-sensing GTP-binding regulatory protein-coupled receptors, TDAG8, OGR1, and G2A are expressed in the cells. The inhibitory action by acidic pH on TNF-alpha production was significantly attenuated in macrophages from TDAG8(Tp/Tp) mice but not in those from OGR1(geo/geo) mice. Moreover, small interfering RNA specific to TDAG8, but not to G2A, clearly attenuated the acidification-induced inhibition of TNF-alpha production. On the other hand, the down-regulation or deficiency of TDAG8 hardly affected prostaglandin E(1)- or isoproterenol-induced actions. LPS-induced IL-6 production was also inhibited by extracellular acidification in a manner that was sensitive to TDAG8 expression. The acidic pH-induced inhibitory action on the cytokine production was significantly reversed either by a small interfering RNA specific to G(s) proteins or by a protein kinase A (PKA)-specific inhibitor H89. Indeed, a PKA-specific cAMP derivative inhibited LPS-induced cytokine production. Moreover, acidification induced cAMP accumulation in a TDAG8-specific way. We conclude that TDAG8, at least partly, mediates the extracellular acidification-induced inhibition of proinflammatory cytokine production through the G(s) protein/cAMP/PKA signaling pathway in mouse macrophages.


Assuntos
Citocinas/antagonistas & inibidores , Citocinas/biossíntese , Espaço Extracelular/metabolismo , Mediadores da Inflamação/antagonistas & inibidores , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/patologia , Prótons , Receptores Acoplados a Proteínas G/fisiologia , Animais , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Espaço Extracelular/imunologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Concentração de Íons de Hidrogênio , Mediadores da Inflamação/metabolismo , Macrófagos Peritoneais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/imunologia
20.
Proc Natl Acad Sci U S A ; 105(4): 1209-14, 2008 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-18212126

RESUMO

Osteoblasts are essential for maintaining bone mass, avoiding osteoporosis, and repairing injured bone. Activation of osteoblast G protein-coupled receptors (GPCRs), such as the parathyroid hormone receptor, can increase bone mass; however, the anabolic mechanisms are poorly understood. Here we use "Rs1," an engineered GPCR with constitutive G(s) signaling, to evaluate the temporal and skeletal effects of G(s) signaling in murine osteoblasts. In vivo, Rs1 expression induces a dramatic anabolic skeletal response, with midfemur girth increasing 1,200% and femur mass increasing 380% in 9-week-old mice. Bone volume, cellularity, areal bone mineral density, osteoblast gene markers, and serum bone turnover markers were also elevated. No such phenotype developed when Rs1 was expressed after the first 4 weeks of postnatal life, indicating an exquisite temporal sensitivity of osteoblasts to Rs1 expression. This pathway may represent an important determinant of bone mass and may open future avenues for enhancing bone repair and treating metabolic bone diseases.


Assuntos
Densidade Óssea/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/biossíntese , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Osteoblastos/química , Osteoblastos/metabolismo , Engenharia de Proteínas , Receptores 5-HT4 de Serotonina/biossíntese , Receptores 5-HT4 de Serotonina/genética , Sequência de Aminoácidos , Animais , Linhagem Celular , Feminino , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Humanos , Ligantes , Masculino , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Engenharia de Proteínas/métodos , Agonistas do Receptor 5-HT4 de Serotonina , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA