Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 522
Filtrar
1.
Reproduction ; 150(4): 331-41, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26159830

RESUMO

Estradiol (E2) accelerates egg transport by a nongenomic action, requiring activation of estrogen receptor (ER) and successive cAMP and IP3 production in the rat oviduct. Furthermore, E2 increases IP3 production in primary cultures of oviductal smooth muscle cells. As smooth muscle cells are the mechanical effectors for the accelerated oocyte transport induced by E2 in the oviduct, herein we determined the mechanism by which E2 increases IP3 in these cells. Inhibition of protein synthesis by Actinomycin D did not affect the E2-induced IP3 increase, although this was blocked by the ER antagonist ICI182780 and the inhibitor of phospholipase C (PLC) ET-18-OCH3. Immunoelectron microscopy for ESR1 or ESR2 showed that these receptors were associated with the plasma membrane, indicating compatible localization with E2 nongenomic actions in the smooth muscle cells. Furthermore, ESR1 but not ESR2 agonist mimicked the effect of E2 on the IP3 level. Finally, E2 stimulated the activity of a protein associated with the contractile tone, calcium/calmodulin-dependent protein kinase II (CaMKII), in the smooth muscle cells. We conclude that E2 increases IP3 by a nongenomic action operated by ESR1 and that involves the activation of PLC in the smooth muscle cells of the rat oviduct. This E2 effect is associated with CaMKII activation in the smooth muscle cells, suggesting that IP3 and CaMKII are involved in the contractile activity necessary to accelerate oviductal egg transport.


Assuntos
Estradiol/farmacologia , Inositol 1,4,5-Trifosfato/biossíntese , Miócitos de Músculo Liso/metabolismo , Oviductos/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/antagonistas & inibidores , Feminino , Técnicas In Vitro , Camundongos , Contração Muscular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Oviductos/efeitos dos fármacos , Ratos Sprague-Dawley , Fosfolipases Tipo C/antagonistas & inibidores
2.
J Mol Biol ; 427(17): 2734-47, 2015 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-25916191

RESUMO

Activation of the phospholipase, PLCγ1, is critical for proper T cell signaling following antigen receptor engagement. In T cells, the Tec family kinase, interleukin-2-induced tyrosine kinase (ITK), phosphorylates PLCγ1 at tyrosine 783 (Y783) leading to activation of phospholipase function and subsequent production of the second messengers inositol 1,4,5-trisphosphate and diacylglycerol. In this work, we demonstrate that PLCγ1 can be primed for ITK-mediated phosphorylation on Y783 by a specific region of the adaptor protein, SLP-76. The SLP-76 phosphotyrosine-containing sequence, pY(173)IDR, does not conform to the canonical recognition motif for an SH2 domain yet binds with significant affinity to the C-terminal SH2 domain of PLCγ1 (SH2C). The SLP-76 pY(173) motif competes with the autoinhibited conformation surrounding the SH2C domain of PLCγ1 leading to exposure of the ITK recognition element on the PLCγ1 SH2 domain and release of the target tyrosine, Y783. These data contribute to the evolving model for the molecular events occurring early in the T cell activation process.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fosfolipase C gama/metabolismo , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinases/metabolismo , Linfócitos T/imunologia , Domínios de Homologia de src/genética , Animais , Sítios de Ligação , Ativação Enzimática , Inositol 1,4,5-Trifosfato/biossíntese , Ativação Linfocitária/imunologia , Camundongos , Fosforilação , Ligação Proteica , Ratos , Transdução de Sinais/imunologia , Domínios de Homologia de src/imunologia
3.
Elife ; 4: e06195, 2015 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-25781633

RESUMO

Bradykinin (Bk) is a potent inflammatory mediator that causes hyperalgesia. The action of Bk on the sensory system is well documented but its effects on motoneurons, the final pathway of the motor system, are unknown. By a combination of patch-clamp recordings and two-photon calcium imaging, we found that Bk strongly sensitizes spinal motoneurons. Sensitization was characterized by an increased ability to generate self-sustained spiking in response to excitatory inputs. Our pharmacological study described a dual ionic mechanism to sensitize motoneurons, including inhibition of a barium-sensitive resting K(+) conductance and activation of a nonselective cationic conductance primarily mediated by Na(+). Examination of the upstream signaling pathways provided evidence for postsynaptic activation of B2 receptors, G protein activation of phospholipase C, InsP3 synthesis, and calmodulin activation. This study questions the influence of motoneurons in the assessment of hyperalgesia since the withdrawal motor reflex is commonly used as a surrogate pain model.


Assuntos
Bradicinina/farmacologia , Gânglios Espinais/efeitos dos fármacos , Hiperalgesia/metabolismo , Neurônios Motores/efeitos dos fármacos , Dor/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Cálcio/metabolismo , Calmodulina/genética , Calmodulina/metabolismo , Feminino , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Expressão Gênica , Hiperalgesia/induzido quimicamente , Hiperalgesia/genética , Hiperalgesia/patologia , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Inositol 1,4,5-Trifosfato/biossíntese , Masculino , Imagem Molecular , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Dor/induzido quimicamente , Dor/genética , Dor/patologia , Técnicas de Patch-Clamp , Canais de Potássio/genética , Canais de Potássio/metabolismo , Ratos , Ratos Wistar , Receptor B2 da Bradicinina/genética , Receptor B2 da Bradicinina/metabolismo , Transdução de Sinais , Canais de Sódio/genética , Canais de Sódio/metabolismo , Medula Espinal/citologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Fosfolipases Tipo C/genética , Fosfolipases Tipo C/metabolismo
4.
Dev Biol ; 401(2): 188-205, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25748412

RESUMO

This review emphasizes how lipids regulate membrane fusion and the proteins involved in three developmental stages: oocyte maturation to the fertilizable egg, fertilization and during first cleavage. Decades of work show that phosphatidic acid (PA) releases intracellular calcium, and recent work shows that the lipid can activate Src tyrosine kinase or phospholipase C during Xenopus fertilization. Numerous reports are summarized to show three levels of increase in lipid second messengers inositol 1,4,5-trisphosphate and sn 1,2-diacylglycerol (DAG) during the three different developmental stages. In addition, possible roles for PA, ceramide, lysophosphatidylcholine, plasmalogens, phosphatidylinositol 4-phosphate, phosphatidylinositol 5-phosphate, phosphatidylinositol 4,5-bisphosphate, membrane microdomains (rafts) and phosphatidylinositol 3,4,5-trisphosphate in regulation of membrane fusion (acrosome reaction, sperm-egg fusion, cortical granule exocytosis), inositol 1,4,5-trisphosphate receptors, and calcium release are discussed. The role of six lipases involved in generating putative lipid second messengers during fertilization is also discussed: phospholipase D, autotaxin, lipin1, sphingomyelinase, phospholipase C, and phospholipase A2. More specifically, proteins involved in developmental events and their regulation through lipid binding to SH3, SH4, PH, PX, or C2 protein domains is emphasized. New models are presented for PA activation of Src (through SH3, SH4 and a unique domain), that this may be why the SH2 domain of PLCγ is not required for Xenopus fertilization, PA activation of phospholipase C, a role for PA during the calcium wave after fertilization, and that calcium/calmodulin may be responsible for the loss of Src from rafts after fertilization. Also discussed is that the large DAG increase during fertilization derives from phospholipase D production of PA and lipin dephosphorylation to DAG.


Assuntos
Cálcio/metabolismo , Fusão de Membrana/fisiologia , Fosfolipase D/metabolismo , Fosfolipases Tipo C/metabolismo , Xenopus laevis/embriologia , Animais , Diglicerídeos/biossíntese , Fertilização/fisiologia , Inositol 1,4,5-Trifosfato/biossíntese , Metabolismo dos Lipídeos/fisiologia , Ácidos Fosfatídicos/metabolismo , Quinases da Família src/metabolismo
5.
Mol Hum Reprod ; 20(10): 938-47, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25057041

RESUMO

Mature mammalian oocytes undergo a prolonged series of cytoplasmic calcium (Ca(2+)) oscillations at fertilization that are the cause of oocyte activation. The Ca(2+) oscillations in mammalian oocytes are driven via inositol 1,4,5-trisphosphate (IP3) generation. Microinjection of the sperm-derived phospholipase C-zeta (PLCζ), which generates IP3, causes the same pattern of Ca(2+) oscillations as observed at mammalian fertilization and it is thought to be the physiological agent that triggers oocyte activation. However, another sperm-specific protein, 'post-acrosomal WW-domain binding protein' (PAWP), has also been reported to elicit activation when injected into mammalian oocytes, and to produce a Ca(2+) increase in frog oocytes. Here we have investigated whether PAWP can induce fertilization-like Ca(2+) oscillations in mouse oocytes. Recombinant mouse PAWP protein was found to be unable to hydrolyse phosphatidylinositol 4,5-bisphosphate in vitro and did not cause any detectable Ca(2+) release when microinjected into mouse oocytes. Microinjection with cRNA encoding either the untagged PAWP, or yellow fluorescent protein (YFP)-PAWP, or luciferase-PAWP fusion proteins all failed to trigger Ca(2+) increases in mouse oocytes. The lack of response in mouse oocytes was despite PAWP being robustly expressed at similar or higher concentrations than PLCζ, which successfully initiated Ca(2+) oscillations in every parallel control experiment. These data suggest that sperm-derived PAWP is not involved in triggering Ca(2+) oscillations at fertilization in mammalian oocytes.


Assuntos
Cálcio/metabolismo , Proteínas de Transporte/metabolismo , Oócitos/metabolismo , Fosfoinositídeo Fosfolipase C/metabolismo , Proteínas de Plasma Seminal/metabolismo , Espermatozoides/metabolismo , Animais , Proteínas de Bactérias , Sinalização do Cálcio , Proteínas de Transporte/administração & dosagem , Feminino , Inositol 1,4,5-Trifosfato/biossíntese , Proteínas Luminescentes , Masculino , Camundongos , Microinjeções , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfoinositídeo Fosfolipase C/administração & dosagem , RNA Complementar/administração & dosagem , RNA Complementar/genética , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/metabolismo , Proteínas de Plasma Seminal/administração & dosagem , Interações Espermatozoide-Óvulo
6.
Biomed Res Int ; 2013: 560141, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24073410

RESUMO

Secretory leukoprotease inhibitor (SLPI) is an anti-inflammatory protein present in respiratory secretions. Whilst epithelial cell SLPI is extensively studied, neutrophil associated SLPI is poorly characterised. Neutrophil function including chemotaxis and degranulation of proteolytic enzymes involves changes in cytosolic calcium (Ca(2+)) levels which is mediated by production of inositol 1,4,5-triphosphate (IP3) in response to G-protein-coupled receptor (GPCR) stimuli. The aim of this study was to investigate the intracellular function of SLPI and the mechanism-based modulation of neutrophil function by this antiprotease. Neutrophils were isolated from healthy controls (n = 10), individuals with cystic fibrosis (CF) (n = 5) or chronic obstructive pulmonary disease (COPD) (n = 5). Recombinant human SLPI significantly inhibited fMet-Leu-Phe (fMLP) and interleukin(IL)-8 induced neutrophil chemotaxis (P < 0.05) and decreased degranulation of matrix metalloprotease-9 (MMP-9), hCAP-18, and myeloperoxidase (MPO) (P < 0.05). The mechanism of inhibition involved modulation of cytosolic IP3 production and downstream Ca(2+) flux. The described attenuation of Ca(2+) flux was overcome by inclusion of exogenous IP3 in electropermeabilized cells. Inhibition of IP3 generation and Ca(2+) flux by SLPI may represent a novel anti-inflammatory mechanism, thus strengthening the attractiveness of SLPI as a potential therapeutic molecule in inflammatory airway disease associated with excessive neutrophil influx including CF, non-CF bronchiectasis, and COPD.


Assuntos
Anti-Inflamatórios/metabolismo , Fibrose Cística/patologia , Inositol 1,4,5-Trifosfato/biossíntese , Espaço Intracelular/metabolismo , Neutrófilos/metabolismo , Doença Pulmonar Obstrutiva Crônica/patologia , Inibidor Secretado de Peptidases Leucocitárias/metabolismo , Adulto , Anti-Inflamatórios/farmacologia , Cálcio/metabolismo , Degranulação Celular/efeitos dos fármacos , Quimiotaxia/efeitos dos fármacos , Fibrose Cística/metabolismo , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Citosol/efeitos dos fármacos , Citosol/metabolismo , Feminino , Humanos , Fatores Imunológicos/metabolismo , Fatores Imunológicos/farmacologia , Espaço Intracelular/efeitos dos fármacos , Masculino , Modelos Biológicos , Ativação de Neutrófilo/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Neutrófilos/fisiologia , Oxirredução/efeitos dos fármacos , Doença Pulmonar Obstrutiva Crônica/metabolismo , Proteínas Recombinantes/farmacologia
7.
Neuro Endocrinol Lett ; 34(4): 302-8, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23803874

RESUMO

OBJECTIVE: Mechanism(s) responsible for VPA-induced effects on reproductive axis activity are not fully recognized. Previously we reported that VPA suppressed only GnRH-stimulated but not the basal LH release from rat anterior pituitary (AP) cells in vitro. Since the inhibitory effect of VPA was exerted only in GnRH-activated cells, potential VPA impact on GnRH-R-coupled IP3/PKC signaling could not be excluded. In this study the effect of VPA on IPs synthesis in non-stimulated and GnRH-treated rat AP cells was examined. MATERIAL AND METHODS: In the first experiment 5 × 105 cells/ml were incubated for 3h with VPA (10 nM-10 µM), PMA (100 nM), GnRH (100 nM), PMA (100 nM) + VPA (10 nM-10 µM), GnRH (100 nM) + VPA (10 nM-10 µM). In the second experiment cells were preincubated for 24h with 1µCi myo-[23 H]-inositol, then for 30 min with 10 mM LiCl and finally for 3hr with GnRH (100 nM) VPA (1 µM, 10 µM), GnRH (100 nM) + VPA (1 µM, 10 µM). LH concentration was measured by RIA and intracellular IPs accumulation by ion-exchange chromatography analysis. RESULTS: VPA diminished GnRH-stimulated LH release without affecting PMA-induced LH release at any dose tested. Moreover, VPA-induced increase of IPs accumulation occurred in both non-stimulated and GnRH-treated cells and intensity of cellular response was similar in both groups. CONCLUSION: VPA affects IP3/PKC pathway activity through its up-regulatory effect on IPs synthesis in AP cells. VPA-induced inhibition of GnRH-stimulated LH release from gonadotrope cells appears to be the result of still unrecognized cellular mechanism.


Assuntos
GABAérgicos/farmacologia , Hormônio Liberador de Gonadotropina/farmacologia , Inositol 1,4,5-Trifosfato/biossíntese , Adeno-Hipófise/citologia , Ácido Valproico/farmacologia , Animais , Células Cultivadas , Cromatografia por Troca Iônica , Feminino , Gonadotrofos/efeitos dos fármacos , Gonadotrofos/metabolismo , Hormônio Luteinizante/efeitos dos fármacos , Hormônio Luteinizante/metabolismo , Adeno-Hipófise/efeitos dos fármacos , Adeno-Hipófise/metabolismo , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima
8.
Gastroenterology ; 145(1): 232-241, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23542070

RESUMO

BACKGROUND & AIMS: The cyclic adenosine monophosphate (cAMP) and Ca(2+) signaling pathways synergize to regulate many physiological functions. However, little is known about the mechanisms by which these pathways interact. We investigated the synergy between these signaling pathways in mouse pancreatic and salivary gland ducts. METHODS: We created mice with disruptions in genes encoding the solute carrier family 26, member 6 (Slc26a6(-/-) mice) and inositol 1,4,5-triphosphate (InsP3) receptor-binding protein released with InsP3 (Irbit(-/-)) mice. We investigated fluid secretion by sealed pancreatic ducts and the function of Slc26a6 and the cystic fibrosis transmembrane conductance regulator (CFTR) in HeLa cells and in ducts isolated from mouse pancreatic and salivary glands. Slc26a6 activity was assayed by measuring intracellular pH, and CFTR activity was assayed by measuring Cl(-) current. Protein interactions were determined by immunoprecipitation analyses. RESULTS: Irbit mediated the synergistic activation of CFTR and Slc26a6 by Ca(2+) and cAMP. In resting cells, Irbit was sequestered by InsP3 receptors (IP3Rs) in the endoplasmic reticulum. Stimulation of Gs-coupled receptors led to phosphorylation of IP3Rs, which increased their affinity for InsP3 and reduced their affinity for Irbit. Subsequent weak stimulation of Gq-coupled receptors, which led to production of low levels of IP3, caused dissociation of Irbit from IP3Rs and allowed translocation of Irbit to CFTR and Slc26a6 in the plasma membrane. These processes stimulated epithelial secretion of electrolytes and fluid. These pathways were not observed in pancreatic and salivary glands from Irbit(-/-) or Slc26a6(-/-) mice, or in salivary gland ducts expressing mutant forms of IP3Rs that could not undergo protein kinase A-mediated phosphorylation. CONCLUSIONS: Irbit promotes synergy between the Ca(2+) and cAMP signaling pathways in cultured cells and in pancreatic and salivary ducts from mice. Defects in this pathway could be involved in cystic fibrosis, pancreatitis, or Sjögren syndrome.


Assuntos
Adenosil-Homocisteinase/fisiologia , Cálcio/metabolismo , AMP Cíclico/fisiologia , Transdução de Sinais/fisiologia , Animais , Antiporters/metabolismo , Transporte Biológico , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Epitélio/metabolismo , Inositol 1,4,5-Trifosfato/biossíntese , Receptores de Inositol 1,4,5-Trifosfato/fisiologia , Camundongos , Ductos Pancreáticos/metabolismo , Fosforilação , Ductos Salivares/metabolismo , Transportadores de Sulfato
9.
Am J Physiol Lung Cell Mol Physiol ; 304(3): L143-51, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23204067

RESUMO

An increase in intracellular calcium concentration ([Ca(2+)](i)) in pulmonary arterial smooth muscle cells (PASMCs) induces hypoxic cellular responses in the lungs; however, the underlying molecular mechanisms remain incompletely understood. We report, for the first time, that acute hypoxia significantly enhances phospholipase C (PLC) activity in mouse resistance pulmonary arteries (PAs), but not in mesenteric arteries. Western blot analysis and immunofluorescence staining reveal the expression of PLC-γ1 protein in PAs and PASMCs, respectively. The activity of PLC-γ1 is also augmented in PASMCs following hypoxia. Lentiviral shRNA-mediated gene knockdown of mitochondrial complex III Rieske iron-sulfur protein (RISP) to inhibit reactive oxygen species (ROS) production prevents hypoxia from increasing PLC-γ1 activity in PASMCs. Myxothiazol, a mitochondrial complex III inhibitor, reduces the hypoxic response as well. The PLC inhibitor U73122, but not its inactive analog U73433, attenuates the hypoxic vasoconstriction in PAs and hypoxic increase in [Ca(2+)](i) in PASMCs. PLC-γ1 knockdown suppresses its protein expression and the hypoxic increase in [Ca(2+)](i). Hypoxia remarkably increases inositol 1,4,5-trisphosphate (IP(3)) production, which is blocked by U73122. The IP(3) receptor (IP(3)R) antagonist 2-aminoethoxydiphenyl borate (2-APB) or xestospongin-C inhibits the hypoxic increase in [Ca(2+)](i). PLC-γ1 knockdown or U73122 reduces H(2)O(2)-induced increase in [Ca(2+)](i) in PASMCs and contraction in PAs. 2-APB and xestospongin-C produce similar inhibitory effects. In conclusion, our findings provide novel evidence that hypoxia activates PLC-γ1 by increasing RISP-dependent mitochondrial ROS production in the complex III, which causes IP(3) production, IP(3)R opening, and Ca(2+) release, playing an important role in hypoxic Ca(2+) and contractile responses in PASMCs.


Assuntos
Cálcio/metabolismo , Hipóxia/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fosfolipase C gama/metabolismo , Artéria Pulmonar/metabolismo , Animais , Compostos de Boro/farmacologia , Complexo III da Cadeia de Transporte de Elétrons/antagonistas & inibidores , Complexo III da Cadeia de Transporte de Elétrons/genética , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Estrenos/farmacologia , Expressão Gênica , Hipóxia/genética , Hipóxia/patologia , Inositol 1,4,5-Trifosfato/antagonistas & inibidores , Inositol 1,4,5-Trifosfato/biossíntese , Receptores de Inositol 1,4,5-Trifosfato/antagonistas & inibidores , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Lentivirus/genética , Compostos Macrocíclicos/farmacologia , Metacrilatos/farmacologia , Camundongos , Contração Muscular/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/patologia , Oxazóis/farmacologia , Fosfolipase C gama/antagonistas & inibidores , Fosfolipase C gama/genética , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/patologia , Pirrolidinonas/farmacologia , RNA Interferente Pequeno/genética , Espécies Reativas de Oxigênio/agonistas , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Tiazóis/farmacologia , Vasoconstrição/efeitos dos fármacos
10.
Am J Physiol Endocrinol Metab ; 303(8): E1006-14, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22912364

RESUMO

Sunitinib is an oral, small molecule multitargeted receptor tyrosine kinase inhibitor with antiangiogenic and antitumor activity that primarily targets vascular endothelial growth factor receptors (VEGFRs). Although sunitinib is an active agent for the treatment of malignant pheochromocytomas, it is unclear whether sunitinib acts through only antiangiogenic mechanisms or also directly targets tumor cells. We previously showed that sunitinib directly induced apoptosis of PC-12 cells. To further confirm these direct effects, we examined the effects of sunitinib on tyrosine hydroxylase (TH) (the rate-limiting enzyme in catecholamine biosynthesis) activity and catecholamine secretion in PC-12 cells and the underlying mechanisms. Sunitinib inhibited TH activity in a dose-dependent manner, and decreased TH protein levels. Consistent with this finding, sunitinib decreased TH phosphorylation at Ser(31) and Ser(40) and significantly decreased catecholamine secretion. VEGFR-2 knockdown attenuated these effects, including inhibition of TH activity and catecholamine secretion, suggesting that they were mediated by VEGFR-2. Sunitinib significantly decreased phospholipase C (PLC)-γ phosphorylation and subsequent protein kinase C (PKC) activity. Because Ser(40) phosphorylation significantly affects TH activity and is known to be regulated by PKC, sunitinib may inhibit Ser(40) phosphorylation via the VEGFR-2/PLC-γ/PKC pathway. Additionally, sunitinib markedly decreased the activity of extracellular signal-regulated kinase (ERK), but not c-Jun NH(2)-terminal kinase or p38 mitogen-activated protein kinase. Therefore, sunitinib may reduce TH Ser(31) phosphorylation through inhibition of the VEGFR-2/PLC-γ/PKC/Raf/mitogen-activated protein kinase/extracellular signal-regulated kinase kinase/ERK pathway. Sunitinib also significantly reduced inositol 1,4,5-trisphosphate production. However, because PC-12 cells do not precisely reflect the pathogenesis of malignant cells, we confirmed the key findings in a human neuroblastoma cell line, SK-N-SH. In conclusion, sunitinib directly inhibits catecholamine synthesis and secretion in pheochromocytoma PC-12 cells.


Assuntos
Inibidores da Angiogênese/farmacologia , Catecolaminas/biossíntese , Catecolaminas/metabolismo , Indóis/farmacologia , Fosfolipase C gama/metabolismo , Pirróis/farmacologia , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Western Blotting , Linhagem Celular Tumoral , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Inositol 1,4,5-Trifosfato/biossíntese , Células PC12 , Feocromocitoma/metabolismo , Proteína Quinase C/metabolismo , RNA Interferente Pequeno/genética , Ratos , Transdução de Sinais/efeitos dos fármacos , Sunitinibe , Tirosina 3-Mono-Oxigenase/metabolismo
11.
J Pharmacol Exp Ther ; 342(2): 486-96, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22588257

RESUMO

The involvement of inositol 1,4,5-trisphosphate (IP(3)) formation in the voltage-dependent regulation of intracellular Ca(2+) concentration ([Ca(2+)](i)) was examined in smooth muscle cells of the porcine coronary artery. Slow ramp depolarization from -90 to 0 mV induced progressive [Ca(2+)](i) increase. The slope was reduced or increased in the presence of Cd(2+) or (±)-1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-[trifluoromethyl]-phenyl)pyridine-3-carboxlic acid methyl ester (Bay K 8644), respectively. The decrease in [Ca(2+)](i) via the membrane hyperpolarization induced by K(+) channel openers (levcromakalim and Evans blue) under current clamp was identical to that under voltage clamp. The step hyperpolarization from -40 to -80 mV reduced [Ca(2+)](i) uniformly over the whole-cell area with a time constant of ∼10 s. The [Ca(2+)](i) at either potential was unaffected by heparin, an inhibitor of IP(3) receptors. Alternatively, [Ca(2+)](i) rapidly increased in the peripheral regions by depolarization from -80 to 0 mV and stayed at that level (∼400 nM) during a 60-s pulse. When the pipette solution contained IP(3) pathway blockers [heparin, 2-aminoethoxydiphenylborate, xestospongin C, or 1-[6-[((17ß)-3-methoxyestra-1,3,5[10]-trien-17-yl)amino]hexyl]-1H-pyrrole-2,5-dione (U73122)], the peak [Ca(2+)](i) was unchanged, but the sustained [Ca(2+)](i) was gradually reduced by ∼250 nM within 30 s. In the presence of Cd(2+), a long depolarization period slightly increased the [Ca(2+)](i), which was lower than that in the presence of heparin alone. In coronary arterial myocytes, the sustained increase in the [Ca(2+)](i) during depolarization was partly caused by the Ca(2+) release mediated by the enhanced formation of IP(3). The initial [Ca(2+)](i) elevation triggered by the Ca(2+) influx though voltage-dependent Ca(2+) channels may be predominantly responsible for the activation of phospholipase C for IP(3) formation.


Assuntos
Cálcio/metabolismo , Vasos Coronários/metabolismo , Inositol 1,4,5-Trifosfato/biossíntese , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Animais , Canais de Cálcio/metabolismo , Vasos Coronários/efeitos dos fármacos , Heparina/farmacologia , Inositol 1,4,5-Trifosfato/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/antagonistas & inibidores , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Canais de Potássio/metabolismo , Suínos
12.
Int J Biochem Cell Biol ; 44(5): 722-7, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22326998

RESUMO

The current paradigm of G protein coupled receptor signaling involves a classical pathway being the activation of phospholipase C and the generation of 1,4,5-inositol trisphosphate, signaling through ß-arrestin scaffold molecules and the transactivation of tyrosine kinase growth factor receptors. Transactivation greatly expands the range of signaling pathways and responses attributable to the receptor. Recently it has been revealed that G protein coupled receptor agonists can also transactivate the serine/threonine kinase cell surface receptor for transforming growth factor-ß (Alk5). This leads to the generation of carboxyl terminal phosphorylated Smad2 which is the immediate downstream product of the activated Alk5. Thus, the current paradigm of G protein coupled signaling can be expanded to include the transactivation of the serine kinase receptor Alk5. These insights expand the possibilities for outcomes of therapeutically targeting GPCRs where more substantive and prolonged actions such as the synthesis of extracellular matrix may be affected.


Assuntos
Proteínas Serina-Treonina Quinases/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Transdução de Sinais/genética , Ativação Transcricional/genética , Arrestinas/genética , Arrestinas/metabolismo , Humanos , Inositol 1,4,5-Trifosfato/biossíntese , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteína Smad2/genética , Proteína Smad2/metabolismo , Fosfolipases Tipo C/genética , Fosfolipases Tipo C/metabolismo , beta-Arrestinas
13.
Biophys J ; 101(3): 554-64, 2011 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-21806923

RESUMO

The quantification of spontaneous calcium (Ca(2+)) oscillations (SCOs) in astrocytes presents a challenge because of the large irregularities in the amplitudes, durations, and initiation times of the underlying events. In this article, we use a stochastic context to account for such SCO variability, which is based on previous models for cellular Ca(2+) signaling. First, we found that passive Ca(2+) influx from the extracellular space determine the basal concentration of this ion in the cytosol. Second, we demonstrated the feasibility of estimating both the inositol 1,4,5-trisphosphate (IP(3)) production levels and the average number of IP(3) receptor channels in the somatic clusters from epifluorescent Ca(2+) imaging through the combination of a filtering strategy and a maximum-likelihood criterion. We estimated these two biophysical parameters using data from wild-type adult mice and age-matched transgenic mice overexpressing the 695-amino-acid isoform of human Alzheimer ß-amyloid precursor protein. We found that, together with an increase in the passive Ca(2+) influx, a significant reduction in the sensitivity of G protein-coupled receptors might lie beneath the abnormalities in the astrocytic Ca(2+) signaling, as was observed in rodent models of Alzheimer's disease. This study provides new, to our knowledge, indices for a quantitative analysis of SCOs in normal and pathological astrocytes.


Assuntos
Doença de Alzheimer/patologia , Astrócitos/patologia , Sinalização do Cálcio , Modelos Biológicos , Incerteza , Doença de Alzheimer/metabolismo , Animais , Astrócitos/metabolismo , Cálcio/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Citosol/metabolismo , Retículo Endoplasmático/metabolismo , Espaço Extracelular/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Inositol 1,4,5-Trifosfato/biossíntese , Inositol 1,4,5-Trifosfato/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Camundongos , Imagem Molecular , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo
14.
Jpn J Vet Res ; 59(1): 5-14, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21476485

RESUMO

In an attempt to explore the functioning of nitric oxide (NO) in pancreatic exocrine cells, we have recently obtained several lines of circumstantial evidence indicating that one of molecular targets of NO is phospholipase C (PLC), the activation of which leads to an increase in the cytosolic Ca2+ concentration ([Ca2+]i) via inositol 1, 4, 5-trisphosphate, IP3. However, whether IP3 is actually produced by NO has not yet been substantiated. The present study was therefore designed to directly measure the intracellular IP3, concentration ([IP3]i) for better understanding of the underlying mechanisms with the help of pharmacological tools. [IP3]i was measured using a fluorescence polarization technique (HitHunter). We obtained the following results: 1) varying concentrations of an NO donor, sodium nitroprusside (SNP), elevated [IP3]i, 2) this elevation was completely inhibited in the presence of the soluble guanylyl cyclase (sGC) inhibitor, 1H-[1, 2, 4] oxadiazolo [4, 3-a] quinoxalin-1-one (ODQ), 3) varying concentrations of the cGMP analogue, 8-Br-cGMP, also increased [IP3]i, 4) the cGMP analogue-induced IP3 production was abolished by pretreatment with either a PLC inhibitor, U73122, or a G-protein inhibitor, GP2A, and 5) KT5823, a potent and highly selective inhibitor of cGMP-dependent protein kinase G (PKG), also abolished the IP3 production induced by 8-Br-cGMP. These results suggest that the NO-induced [Ca2+]i increase is triggered by an increase in [IP3]i located downstream from intracellular cGMP elevation. In this intracellular pathway, each sGC, cGMP-dependent PKG, G-protein and PLC were suggested to be involved. The present work provides new insights into the intracellular signaling accelerated by NO. NO triggers a [Ca2+]I increase via cGMP and IP3 in pancreatic acinar cells.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Inositol 1,4,5-Trifosfato/biossíntese , Óxido Nítrico/metabolismo , Pâncreas Exócrino/metabolismo , Animais , Cálcio/metabolismo , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , GMP Cíclico/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Doadores de Óxido Nítrico/farmacologia , Nitroprussiato/farmacologia , Pâncreas Exócrino/efeitos dos fármacos , Pâncreas Exócrino/enzimologia , Ratos , Ratos Wistar , Fosfolipases Tipo C/metabolismo
15.
Endocrinology ; 152(4): 1691-701, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21285318

RESUMO

FSH binds to its receptor (FSHR) on target cells in the ovary and testis, to regulate oogenesis and spermatogenesis, respectively. The signaling cascades activated after ligand binding are extremely complex and have been shown to include protein kinase A, mitogen-activated protein kinase, phosphatidylinositol 3-kinase/protein kinase B, and inositol 1,4,5-trisphosphate-mediated calcium signaling pathways. The adapter protein APPL1 (Adapter protein containing Pleckstrin homology domain, Phosphotyrosine binding domain and Leucine zipper motif), which has been linked to an assortment of other signaling proteins, was previously identified as an interacting protein with FSHR. Thus, alanine substitution mutations in the first intracellular loop of FSHR were generated to determine which residues are essential for FSHR-APPL1 interaction. Three amino acids were essential; when any one of them was altered, APPL1 association with FSHR mutants was abrogated. Two of the mutants (L377A and F382A) that displayed poor cell-surface expression were not studied further. Substitution of FSHR-K376A did not affect FSH binding or agonist-stimulated cAMP production in either transiently transfected human embryonic kidney cells or virally transduced human granulosa cells (KGN). In the KGN line, as well as primary cultures of rat granulosa cells transduced with wild type or mutant receptor, FSH-mediated progesterone or estradiol production was not affected by the mutation. However, in human embryonic kidney cells inositol 1,4,5-trisphosphate production was curtailed and KGN cells transduced with FSHR-K376A evidenced reduced Ca(2+) mobilization from intracellular stores after FSH treatment.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cálcio/metabolismo , Inositol 1,4,5-Trifosfato/biossíntese , Receptores do FSH/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Western Blotting , Linhagem Celular , AMP Cíclico/metabolismo , Estradiol/metabolismo , Feminino , Hormônio Foliculoestimulante/metabolismo , Humanos , Imunoprecipitação , Mutação , Progesterona/metabolismo , Ligação Proteica , Ratos , Receptores do FSH/genética
16.
ACS Chem Biol ; 6(3): 223-8, 2011 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-21158426

RESUMO

Phospholipase C isozymes (PLCs) catalyze the conversion of the membrane lipid phosphatidylinositol 4,5-bisphosphate (PIP(2)) into two second messengers, inositol 1,4,5-trisphosphate and diacylglycerol. This family of enzymes are key signaling proteins that regulate the physiological responses of many extracellular stimuli such as hormones, neurotransmitters, and growth factors. Aberrant regulation of PLCs has been implicated in various diseases including cancer and Alzheimer's disease. How, when, and where PLCs are activated under different cellular contexts are still largely unknown. We have developed a fluorogenic PLC reporter, WH-15, that can be cleaved in a cascade reaction to generate fluorescent 6-aminoquinoline. When applied in enzymatic assays with either pure PLCs or cell lysates, this reporter displays more than a 20-fold fluorescence enhancement in response to PLC activity. Under assay conditions, WH-15 has comparable K(m) and V(max) with the endogenous PIP(2). This novel reporter will likely find broad applications that vary from imaging PLC activity in live cells to high-throughput screening of PLC inhibitors.


Assuntos
Aminoquinolinas/química , Ensaios Enzimáticos/métodos , Corantes Fluorescentes/química , Inositol 1,4,5-Trifosfato/análogos & derivados , Sondas Moleculares/química , Fosfolipases Tipo C/análise , Fosfolipases Tipo C/metabolismo , Aminoquinolinas/metabolismo , Biocatálise , Linhagem Celular , Membrana Celular/química , Membrana Celular/metabolismo , Diglicerídeos/biossíntese , Diglicerídeos/química , Corantes Fluorescentes/metabolismo , Humanos , Inositol 1,4,5-Trifosfato/biossíntese , Inositol 1,4,5-Trifosfato/química , Inositol 1,4,5-Trifosfato/metabolismo , Isoenzimas/análise , Isoenzimas/antagonistas & inibidores , Isoenzimas/química , Isoenzimas/metabolismo , Cinética , Sondas Moleculares/metabolismo , Estrutura Molecular , Peso Molecular , Fosfatidilinositol 4,5-Difosfato/química , Fosfatidilinositol 4,5-Difosfato/metabolismo , Estereoisomerismo , Fosfolipases Tipo C/antagonistas & inibidores , Fosfolipases Tipo C/química
17.
Int J Biochem Cell Biol ; 43(3): 393-400, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21095240

RESUMO

The ability of magnetic fields (MFs) to promote/increase Ca(2+) influx into cells is widely recognized, but the underlying mechanisms remain obscure. Here we analyze how static MFs of 6 mT modulates thapsigargin-induced Ca(2+) movements in non-excitable U937 monocytes, and how this relates to the anti-apoptotic effect of MFs. Magnetic fields do not affect thapsigargin-induced Ca(2+) mobilization from endoplasmic reticulum, but significantly increase the resulting Ca(2+) influx; this increase requires intracellular signal transduction actors including G protein, phospholipase C, diacylglycerol lipase and nitric oxide synthase, and behaves as a non-capacitative Ca(2+) entry (NCCE), a type of influx with an inherent signaling function, rather than a capacitative Ca(2+) entry (CCE). All treatments abrogating the extra Ca(2+) influx also abrogate the anti-apoptotic effect of MFs, demonstrating that MF-induced NCCE elicits an anti-apoptotic survival pathway.


Assuntos
Cálcio/metabolismo , Magnetismo , Transdução de Sinais , Fosfolipases Tipo C/metabolismo , Apoptose/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Proteínas de Ligação ao GTP/metabolismo , Humanos , Inositol 1,4,5-Trifosfato/biossíntese , Células Jurkat , Lipase Lipoproteica/metabolismo , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tapsigargina/farmacologia , Células U937
18.
Biochem Biophys Res Commun ; 394(3): 464-9, 2010 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-20144588

RESUMO

The beta subunits of voltage-dependent calcium channels are known to modify calcium channel currents through pore-forming alpha1 subunits. The beta3 subunit is expressed in the adrenal gland and participates in forming various calcium channel types. We performed a series of experiments in beta3-null mice to determine the role of the beta3 subunit in catecholamine release from the adrenal chromaffin system. Protein levels of N-type channel forming CaV2.2 and L-type forming CaV1.2 decreased. The beta3-null mice showed a decreased baroreflex, suggesting decreased sympathetic tonus, whereas plasma catecholamine levels did not change. Pulse-voltage stimulation revealed significantly increased amperometrical currents in beta3-null mice, while patch-clamp recordings showed a significant reduction in Ca(2+)-currents due to reduced L- and N-type currents, indicating facilitated exocytosis. A biochemical analysis revealed increased InsP3 production. In conclusion, our results indicate the importance of the beta3 subunit in determining calcium channel characteristics and catecholamine release in adrenal chromaffin cells.


Assuntos
Glândulas Suprarrenais/metabolismo , Canais de Cálcio/fisiologia , Células Cromafins/metabolismo , Epinefrina/metabolismo , Sistema Nervoso Simpático/fisiologia , Animais , Barorreflexo , Pressão Sanguínea/fisiologia , Canais de Cálcio/genética , Catecolaminas/sangue , Inositol 1,4,5-Trifosfato/biossíntese , Camundongos , Camundongos Knockout , Sistema Nervoso Simpático/metabolismo
19.
Acta Physiol (Oxf) ; 198(3): 387-92, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19583704

RESUMO

AIM: Fluorescent protein-based indicators have enabled measurement of intracellular signals previously nearly inaccessible for studies. However, indicators showing intracellular translocation upon response suffer from serious limitations, especially the very time-consuming data collection. We therefore set out in this study to evaluate whether fixing and counting cells showing translocation could mend this issue. METHODS: Altogether three different genetically encoded indicators for diacylglycerol and inositol-1,4,5-trisphosphate were transiently expressed in Chinese hamster ovary cells stably expressing human OX(1) orexin receptors. Upon stimulation with orexin-A, the cells were fixed with six different protocols. RESULTS: Different protocols showed clear differences in their ability to preserve the indicator's localization (i.e. translocation after stimulus) and its fluorescence, and the best results for each indicator were obtained with a different protocol. The concentration-response data obtained with cell counting are mostly comparable to the real-time translocation and biochemical data. CONCLUSION: The counting method, as used here, works at single time point and looses the single-cell-quantitative aspect. However, it also has some useful properties. First, it easily allows processing of a 100- to 1000-fold higher cell numbers than real-time imaging producing statistically consistent population-quantitative data much faster. Secondly, it does not require expensive real-time imaging equipment. Fluorescence in fixed cells can also be quantitated, though this analysis would be more time-consuming than cell counting. Thirdly, in addition to the quantitative data collection, the method could be applied for identifying responsive cells. This might be very useful in identification of e.g. orexin-responding neurones in a large population of non-responsive cells in primary cultures.


Assuntos
Diglicerídeos/biossíntese , Inositol 1,4,5-Trifosfato/biossíntese , Métodos , Ovário/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropeptídeos/metabolismo , Transdução de Sinais/fisiologia , Animais , Transporte Biológico , Contagem de Células , Linhagem Celular , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Citosol/metabolismo , Diagnóstico por Imagem , Estudos de Viabilidade , Feminino , Fixadores , Fluorescência , Humanos , Indicadores e Reagentes/farmacocinética , Receptores de Orexina , Concentração Osmolar , Ovário/citologia , Fatores de Tempo , Distribuição Tecidual
20.
J Pharmacol Exp Ther ; 331(3): 917-24, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19741149

RESUMO

We have recently reported that left atrial injections of the thromboxane A(2) (TXA(2)) mimetic, (5Z)-7-[(1R,4S,5S,6R)-6-[(1E,3S)-3-hydroxy-1-octenyl]-2 -oxabicyclo[2.2.1]hept-5-yl]-5-heptenoic acid (U46619), induced ventricular arrhythmias in the anesthetized rabbit. Data from this study led us to hypothesize that TXA(2) may be inducing direct actions on the myocardium to induce these arrhythmias. The aim of this study was to further elucidate the mechanism responsible for these arrhythmias. We report that TXA(2)R is expressed at both the gene and protein levels in atrial and ventricular samples of adult rabbits. In addition, TXA(2)R mRNA was identified in single, isolated ventricular cardiac myocytes. Furthermore, treatment of isolated cardiac myocytes with U46619 increased intracellular calcium in a dose-dependent manner and these increases were blocked by the specific TXA(2)R antagonist, 7-(3-((2-((phenylamino)carbonyl)hydrazino)methyl)-7-oxabicyclo(2.2.1)hept-2-yl)-5-heptenoic acid (SQ29548). Pretreatment of myocytes with an inhibitor of inositol trisphosphate (IP(3)) formation, gentamicin, or with an inhibitor of IP(3) receptors, 2-aminoethoxydiphenylborate (2-APB), blocked the increase in intracellular calcium. In vivo pretreatment of anesthetized rabbits with either gentamicin or 2-APB subsequently inhibited the formation of ventricular arrhythmias elicited by U46619. These data support the hypothesis that TXA(2) can induce arrhythmias via a direct action on cardiac myocytes. Furthermore, these arrhythmogenic actions were blocked by inhibitors of the IP(3) pathway. In summary, this study provides novel evidence for direct TXA(2)-induced cardiac arrhythmias and provides a rationale for IP(3) as a potential target for the treatment of TXA(2)-mediated arrhythmias.


Assuntos
Arritmias Cardíacas/metabolismo , Cálcio/metabolismo , Inositol 1,4,5-Trifosfato/antagonistas & inibidores , Miócitos Cardíacos/metabolismo , Tromboxano A2/fisiologia , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Animais , Arritmias Cardíacas/prevenção & controle , Western Blotting , Compostos Bicíclicos Heterocíclicos com Pontes , Células Cultivadas , Ácidos Graxos Insaturados , Átrios do Coração/metabolismo , Ventrículos do Coração/metabolismo , Hidrazinas/farmacologia , Inositol 1,4,5-Trifosfato/biossíntese , Receptores de Inositol 1,4,5-Trifosfato/antagonistas & inibidores , Receptores de Inositol 1,4,5-Trifosfato/biossíntese , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Coelhos , Receptores de Tromboxano A2 e Prostaglandina H2/antagonistas & inibidores , Receptores de Tromboxano A2 e Prostaglandina H2/biossíntese , Receptores de Tromboxano A2 e Prostaglandina H2/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Tromboxano A2/agonistas , Tromboxano A2/antagonistas & inibidores , Vasoconstritores/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...