Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dev Biol ; 445(1): 54-67, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30385274

RESUMO

The role of agrin, Lrp4 and MuSK, key organizers of neuromuscular synaptogenesis, in the developing CNS is only poorly understood. We investigated the role of these proteins in cultured mouse embryonic cortical neurons from wildtype and from Lrp4- and MuSK-deficient mice. Neurons from Lrp4-deficient mice had fewer but longer primary dendrites and a decreased density of puncta containing excitatory and inhibitory synapse-associated proteins. Neurons from MuSK-deficient mice had an altered dendritic branching pattern but no change in the density of puncta stained by antibodies against synapse-associated proteins. Transfection of TM-agrin compensated the dendritic branching deficits in Lrp4-deficient but not in MuSK-deficient neurons. TM-agrin transfection increased the density of excitatory synaptic puncta in MuSK-deficient but not in Lrp4-deficient mice and reduced the number of inhibitory synaptic puncta irrespective of MuSK and Lrp4 expression. Addition of purified soluble agrin to microisland cultures of cortical neurons revealed an Lrp4-dependent increase in the size and density of glutamatergic synaptic puncta and in mEPSC but not in mIPSC frequency and amplitude. Thus, agrin induced an Lrp4-independent increase in dendritic branch complexity, an Lrp4-dependent increase of excitatory synaptic puncta and an Lrp4- and MuSK-independent decrease in the density of puncta containing inhibitory synapse-associated proteins. These results establish selective roles for agrin, Lrp4 and MuSK during dendritogenesis and synaptogenesis in cultured CNS neurons.


Assuntos
Agrina/metabolismo , Junção Neuromuscular/metabolismo , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de LDL/metabolismo , Sinapses/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Sistema Nervoso Central/patologia , Dendritos/metabolismo , Feminino , Proteínas Relacionadas a Receptor de LDL , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese
2.
Syst Biol (Stevenage) ; 152(4): 185-92, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16986259

RESUMO

A four-state cell-cycle model with explicit G1-phase representation, termed the quiescent-cell model (QCM), has been proposed to represent biologically the G1-phase specific effect of the chemotherapeutic tamoxifen. The QCM was used to model untreated and tamoxifen-treated tumour xenograft data from the literature with equivalent accuracy to previously developed tumour growth models. Open-loop analysis demonstrated that perturbations to the two newly introduced parameters, kG01 and kG10, significantly altered untreated tumour growth predictions. However, the sensitivity did not carry over to closed-loop simulations, where alterations to kD and kGS proved most significant in determining overall controller performance. Additional mismatch studies comparing controllers designed using the QCM to controllers designed with the Gompertz model and saturating-rate, cell-cycle model returned similar performance for a step-wise tumour reduction case study, but the quiescent-cell controller delivered a more aggressive treatment regimen. More importantly, the Gompertz and saturating-rate, cell-cycle controllers were unable to follow a reference trajectory when measurement updates were made biweekly, with both controllers returning tamoxifen dose schedules alternating between the maximum and minimum allowable dose.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/fisiopatologia , Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/efeitos dos fármacos , Quimioterapia Assistida por Computador/métodos , Modelos Biológicos , Proteínas de Neoplasias/metabolismo , Animais , Antineoplásicos Hormonais/administração & dosagem , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Camundongos , Transdução de Sinais/efeitos dos fármacos , Tamoxifeno/administração & dosagem
3.
Hypertension ; 38(6): 1337-41, 2001 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11751714

RESUMO

Epidermal growth factor (EGF) causes contraction in arteries from deoxycorticosterone acetate (DOCA)-salt hypertensive rats but not in normotensive sham rats. We hypothesized that an increase in the number of EGF receptors (EGFRs) in arteries from DOCA-salt rats enables the observed contraction to EGF to occur. DOCA-salt rats had a systolic blood pressure >170 mm Hg, whereas all sham rats had a systolic blood pressure <125 mm Hg. Thoracic aorta were removed for measurement of isometric force, EGFR mRNA levels, and EGFR protein levels. EGF caused a significant contraction in endothelium-denuded aorta from DOCA-salt rats (38+/-7% of maximal phenylephrine-induced [10 micromol/L] contraction) compared with aorta from sham rats (4+/-2%). The EGFR tyrosine kinase-specific inhibitors 4,5-dianilinophthalimide (10 micromol/L) and AG1478 (250 nmol/L) reduced contraction in aorta from DOCA-salt by 85+/-14% and 65+/-10%, respectively. EGFR mRNA in DOCA-salt aorta was increased 4.2-fold compared with that in sham aorta. However, Western analyses of membrane-enriched and whole-tissue lysate of aorta from sham and DOCA-salt revealed no statistical difference in the density of EGFR protein between sham and DOCA-salt aorta. These data refute our hypothesis and suggest that a change downstream of EGFR is responsible for enabling EGF-induced contraction in hypertension.


Assuntos
Artérias/fisiologia , Receptores ErbB/fisiologia , Hipertensão/fisiopatologia , Sódio na Dieta/administração & dosagem , Sistema Vasomotor/fisiologia , Animais , Desoxicorticosterona , Relação Dose-Resposta a Droga , Receptores ErbB/administração & dosagem , Hipertensão/induzido quimicamente , Técnicas In Vitro , Masculino , Ratos , Ratos Sprague-Dawley , Sistema Vasomotor/efeitos dos fármacos
4.
Am J Physiol Regul Integr Comp Physiol ; 281(3): R878-86, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11507004

RESUMO

The present studies test the hypothesis that contraction to EGF is dependent on mineralocorticoids and/or an elevation in systolic blood pressure (SBP). Endothelium-denuded thoracic aortas from sham normotensive, N(omega)-nitro-L-arginine (L-NNA) hypertensive, Wistar-Kyoto (WKY), and spontaneously hypertensive rats (SHR) were used in isolated tissue-bath experiments. Maximal contraction to epidermal growth factor [EGF; percentage of phenylephrine (PE; 10 umol/l)-induced contraction] was greater in strips from L-NNA (32 +/- 5%) and SHR (53 +/- 8%) rats compared with sham and WKY rats (17 +/- 1 and 12 +/- 4%, respectively). Wistar-Furth rats became only mildly hypertensive when given DOCA salt (134 +/- 6 mmHg) compared with Wistar rats (176 +/- 9 mmHg), but aortas from both strains had a similarly enhanced contraction to EGF (approximately 9 times the maximal contraction of sham aorta). Furthermore, in vitro incubation of aortas from Wistar and Wistar-Furth rats with aldosterone (10 nmol/l) increased EGF-receptor mRNA expression by >50%. These data indicate that arterial contraction to EGF may occur independent of hypertension and be stimulated by mineralocorticoids.


Assuntos
Fator de Crescimento Epidérmico/farmacologia , Hipertensão/fisiopatologia , Mineralocorticoides/farmacologia , Regulação para Cima , Vasoconstrição/efeitos dos fármacos , Aldosterona/farmacologia , Animais , Aorta Torácica , Pressão Sanguínea/efeitos dos fármacos , Desoxicorticosterona , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Hipertensão/induzido quimicamente , Técnicas In Vitro , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WF , Ratos Sprague-Dawley , Ratos Wistar , Cloreto de Sódio , Especificidade da Espécie , Regulação para Cima/efeitos dos fármacos
5.
J Biol Chem ; 276(19): 15794-800, 2001 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-11279064

RESUMO

Na(+)/H(+) exchanger isoform-1 (NHE1), the ubiquitous form of the Na(+)/H(+) exchanger, has increased activity in hypertensive patients and in animal models of hypertension. Furthermore, NHE1 is activated in cells stimulated with growth factors. We showed previously that activation of the exchanger is dependent on phosphorylation of serine 703 (Ser(P)(703)) by p90 ribosomal S6 kinase (RSK). Because the NHE1 sequence at Ser(P)(703) (RIGSDP) is similar to a consensus sequence (RSXSXP) specific for 14-3-3 ligands, we evaluated whether serum stimulated 14-3-3 binding to NHE1. Five different GST-NHE1 fusion proteins spanning amino acids 515-815 were phosphorylated by RSK and used as ligands in a far Western analysis; only those containing Ser(P)(703) exhibited high affinity 14-3-3 binding. In PS127A cells (NHE1-overexpressing Chinese hamster fibroblasts) stimulated with 20% serum, NHE1 co-precipitation with GST-14-3-3 fusion protein increased at 5 min (5.2 +/- 0.4-fold versus control; p < 0.01) and persisted at 40 min (3.9 +/- 0.3-fold; p < 0.01). We confirmed that binding occurs at the RIGSDP motif using PS120 (NHE1 null) cells transfected with S703A-NHE1 or P705A-NHE1 (based on data indicating that 14-3-3 binding requires phosphoserine and +2 proline). Serum failed to stimulate association of 14-3-3 with these mutants. A GST-NHE1 fusion protein was phosphorylated by RSK and used as a ligand to assess the effect of 14-3-3 on protein phosphatase 1-mediated dephosphorylation of Ser(P)(703). GST-14-3-3 limited dephosphorylation (66% of initial state at 60 min) compared with GST alone (27% of initial state; p < 0.01). The protective effect of GST-14-3-3 was lost in the GST-NHE1 P705A mutant. Finally, the base-line rate of pH recovery in acid-loaded cells was equal in unstimulated cells expressing wild-type or P705A-NHE1. However, activation of NHE1 by serum was dramatically inhibited in cells expressing P705A-NHE1 compared with wild-type (0.13 +/- 0.02 versus 0.48 +/- 0.06 mmol of H(+)/min/liter, p < 0.01). These data suggest that 14-3-3 binding to NHE1 participates in serum-stimulated exchanger activation, a new function for 14-3-3.


Assuntos
Trocadores de Sódio-Hidrogênio/química , Trocadores de Sódio-Hidrogênio/metabolismo , Sódio/metabolismo , Tirosina 3-Mono-Oxigenase/química , Tirosina 3-Mono-Oxigenase/metabolismo , Proteínas 14-3-3 , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sangue , Linhagem Celular , Sequência Consenso , Cricetinae , Cricetulus , Meios de Cultura , Primers do DNA , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Estrutura Secundária de Proteína , Proteínas Recombinantes/metabolismo , Trocadores de Sódio-Hidrogênio/genética , Transfecção
6.
J Pharmacol Exp Ther ; 291(3): 1179-87, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10565840

RESUMO

5-Hydroxytryptamine (5-HT) activates the extracellular signal-regulated kinase (Erk) mitogen-activated protein kinases (MAPKs) in the vasculature, resulting in contraction. The mechanisms by which this occurs are unclear. G protein-coupled receptors can activate Erk MAPK pathways through a variety of mechanisms, including stimulation of Src, phosphoinositide-3 kinase (PI-3-K), protein kinase C (PKC), or the epidermal growth factor (EGF) receptor tyrosine kinase. We hypothesize that 5-HT uses one or more of these pathways. In isolated strips of rat aorta, the MAPK/Erk kinase inhibitor U0126 (50 microM), Src inhibitor PP1 (0.5 microM), PKC inhibitors calphostin C (1 microM) and chelerythrine (10 microM), and the PI-3-K inhibitor LY294002 (1-20 microM) reduced 5-HT-induced contraction. The EGF receptor tyrosine kinase inhibitor AG1478 (0.25-1 microM) was without effect. Thus, 5-HT activates PKC, Src, and possibly PI-3-K to result in contraction. In rat aortic myocytes, 5-HT (1 microM) activated Erk MAPK proteins 2- to 3-fold over basal values; activation was reduced by U0126, PP1, and LY294002 and unaffected by calphostin C or chelerythrine, wortmannin, or AG1478. The lack of effect of EGF receptor tyrosine kinase and PI-3-K inhibitors was confirmed in that the EGF receptor immunoprecipitated from 5-HT-exposed cells did not display an increase in autophosphorylation, nor did 5-HT significantly increase activation of Akt/protein kinase B, a downstream substrate for PI-3-K. These data suggest that the rat aortic 5-HT(2A) receptor uses Src but not PKC, PI-3-K, or the EGF receptor tyrosine kinase in stimulating Erk MAPK activation.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Liso Vascular/enzimologia , Receptores de Serotonina/efeitos dos fármacos , Agonistas do Receptor de Serotonina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/enzimologia , Western Blotting , Inibidores Enzimáticos/farmacologia , Técnicas In Vitro , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Masculino , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Músculo Liso Vascular/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Testes de Precipitina , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/metabolismo , Receptor 5-HT2A de Serotonina
7.
Am J Physiol ; 276(3): H976-83, 1999 03.
Artigo em Inglês | MEDLINE | ID: mdl-10070082

RESUMO

We have tested the hypothesis that growth factor signaling pathways are augmented in hypertension, a disease associated with vascular smooth muscle cell growth. Thoracic aorta was dissected from deoxycorticosterone acetate-salt (DOCA-salt) and one kidney, one clip (1K, 1C) hypertensive rats and from sham normotensive rats for use in isolated tissue bath experiments. Systolic blood pressure was significantly higher in DOCA-salt and 1K, 1C than in normotensive sham rats: 192 +/- 7, 185 +/- 10, and 117 +/- 4 mmHg, respectively. Although virtually no contraction to epidermal growth factor (EGF) was observed in endothelium-denuded sham rat aorta [1 +/- 1% phenylephrine (PE) (10 micromol/l)-induced contraction], the maximal EGF-induced contraction was 45 +/- 7% in endothelium-denuded aorta from DOCA-salt hypertensive rats and 39 +/- 7% in aorta from 1K, 1C rats. Although slightly attenuated, a contraction to EGF was still observed in endothelium-intact aortic strips from 28-day DOCA-salt hypertensive rats. We also conducted concentration-response curves to EGF on days 1, 3, 5, 7, 14, and 21 of DOCA-salt therapy. A significant contraction to EGF in aorta from DOCA-salt rats was observed on day 14, when DOCA-salt rats had significantly higher blood pressure than sham rats: 188 +/- 6 and 122 +/- 3 mmHg, respectively. Transforming growth factor-alpha, an agonist of the EGF receptor, contracted DOCA-salt rat aorta (30 +/- 7% PE-induced contraction) but not sham aorta (3 +/- 3%). The EGF receptor tyrosine kinase inhibitor 4,5-dianilinophthalimide (10 micromol/l), the mitogen-activated protein kinase kinase inhibitor PD-098059 (10 micromol/l), and the L-type voltage-gated calcium channel inhibitor diltiazem (1 mol/l), but not the cyclooxygenase inhibitor indomethacin (10 micromol/l), virtually abolished EGF-induced contraction (85, 98, and 99% reduction, respectively). These data support a striking difference in EGF signaling between normotensive and hypertensive animals. Furthermore, they provide evidence that growth factors should be considered vasoconstrictors as well as growth modulators in hypertension.


Assuntos
Fator de Crescimento Epidérmico/farmacologia , Hipertensão/fisiopatologia , Vasoconstritores/farmacologia , Animais , Aorta/efeitos dos fármacos , Desoxicorticosterona , Hipertensão/induzido quimicamente , Hipertensão Renovascular/fisiopatologia , Masculino , Ratos , Ratos Sprague-Dawley , Valores de Referência , Cloreto de Sódio , Vasoconstrição/fisiologia
8.
J Pharmacol Exp Ther ; 286(3): 1431-8, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9732408

RESUMO

Angiotensin II (Ang II) is a potent pressor hormone, a stimulus for vascular smooth muscle hypertrophy and an activator of multiple tyrosine kinases. The physiological effects of Ang II are mediated through activation of AT1 and AT2 receptors, receptors that have been coupled to tyrosine kinase(s) and tyrosine phosphatases, respectively. Agonists of G protein-coupled receptors, of which Ang II is one, have recently been shown to stimulate smooth muscle contraction in part via activation tyrosine kinases. We tested the hypothesis that Ang II-induced contraction in the rat aorta was dependent on activation of tyrosine kinase(s) and specifically investigated the role of the tyrosine kinase mitogen-activated protein kinase kinase (MEK), a kinase important to the mitogen activated protein kinase (MAPK) pathway. Rat thoracic aortic strips denuded of endothelium and cultured aortic smooth muscle cells were used in isolated tissue baths for measurement of isometric contractile force and Western analyses of protein tyrosyl-phosphorylation. Ang II (0.1-100 nM)-induced contraction in the aorta was completely blocked by the AT1 receptor antagonist losartan (1 microM) but unaffected by the AT2 receptor antagonist PD123319 (100 nM) or tyrosine phosphatase inhibitor sodium orthovanadate (1 microM), indicating an AT1 receptor mediates aortic contraction to Ang II. Neither the tyrosine kinase inhibitor genistein (5 microM), inactive tyrosine kinase inhibitor daidzein (5 microM) nor MEK inhibitor PD098059 (10 microM) reduced Ang II-induced contraction; the concentrations of inhibitors used maximally reduced contraction stimulated by other agonists of G protein-coupled receptors such as serotonin. Moreover, Ang II-induced contraction was not altered by the combination of PD098059 and PD123319, indicating that it is unlikely AT2 receptor stimulation masks activation of the MAPK pathway through AT1 receptor activation. The nonflavone tyrosine kinase inhibitor tyrphostin B42 (30 microM) reduced Ang II-induced maximal contraction (to 11.2% control) but, unlike the other tyrosine kinase inhibitors, also reduced KCl-induced contraction (to 55.2% control), indicating a probable nonselectivity of tyrphostin B42. Ang IIinduced maximal contraction was reduced by the L-type voltage gated calcium channel antagonist nifedipine (50 nM), consistent with the activation of calcium channels by Ang II. In cultured rat aortic smooth muscle cells, Ang II (0.1-1000 nM) stimulated concentration-dependent tyrosyl-phosphorylation of the extracellular signal regulated kinase (Erk) mitogen activated protein kinases (maximal stimulation, fold basal: Erk-1 = 17-fold, Erk-2 = 3-fold), indicating that Ang II can activate MEK. Losartan (1 microM) abolished Ang II (10 nM)-induced Erk tyrosyl-phosphorylation and PD098059 (10 microM), which did not diminish Ang II-induced aortic contraction, reduced Ang II (10 nM)-stimulated phosphorylation of Erk-2 by 72%. Finally, Ang II (1 microM) increased tyrosyl-phosphorylation of the Erk proteins in isolated aorta exposed to Ang II for 5 min. Thus, while Ang II can stimulate both MEK activation and vascular contraction via interaction with AT1 receptors, stimulation of MEK does not appear to be important for Ang II-induced contraction. These findings dissociate the process of Ang II-stimulated Erk protein tyrosyl-phosphorylation from Ang II-induced contraction in the rat aorta.


Assuntos
Angiotensina II/farmacologia , Proteínas Quinases/metabolismo , Tirfostinas , Vasoconstrição/efeitos dos fármacos , Animais , Aorta/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Flavonoides/farmacologia , Genisteína/farmacologia , Técnicas In Vitro , MAP Quinase Quinase 1 , Masculino , Quinases de Proteína Quinase Ativadas por Mitógeno , Músculo Liso Vascular/enzimologia , Nitrilas/farmacologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Tirosina Quinases/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de Angiotensina/fisiologia
9.
J Pharmacol Exp Ther ; 284(1): 346-55, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9435197

RESUMO

Vascular 5-Hydroxytryptamine2A (5-HT2A) receptor signaling and contraction has been associated with the activation of L-type calcium channels, phospholipase C (PLC) and, as we previously demonstrated, tyrosine kinase activation. We hypothesize the 5-HT2A receptor activates all three pathways independently to elicit contraction and that one of the tyrosine kinases activated by 5-HT is mitogen-activated protein kinase kinase (MEK). Endothelium-denuded rat thoracic aorta was mounted into isolated tissue baths for measurement of isometric contractile force. 5-HT, alpha-methyl-5-HT and 2,5-dimethoxy-4-iodoamphetamine all contracted the rat aorta, whereas the 5-HT2A receptor antagonist ketanserin (30 nM) blocked contraction to 5-HT. The tyrosine kinase inhibitor genistein (5 microM) shifted contraction to 5-HT, alpha-methyl-5-HT and DOI approximately 10-fold to the right, whereas daidzein (5 microM), the inactive isomer of genistein, was unable to shift 5-HT-induced contraction. PD098059 (10 microM), an inhibitor of MEK, shifted contraction to 5-HT approximately 7-fold to the right. We next examined the integration of tyrosine kinase activation in 5-HT2A receptor signaling. 5-HT-induced contraction was reduced individually by the PLC inhibitor 2-nitro-4-carboxyphenyl-N,N-diphenylcarbamate (NCDC; 100 microM) or the Ca++ channel inhibitor nifedipine (50 nM); the remaining response to 5-HT was reduced by further addition of either genistein or PD098059. When nifedipine and NCDC were used in combination, a part of the contraction to 5-HT remained: this contraction was further reduced by genistein or PD098059. In cultured aortic smooth muscle cells, 5-HT (0.01-100 microM) stimulated tyrosyl-phosphorylation of 42- and 44-kDa proteins identified as Erk MAPKs; this phosphorylation was reduced by PD098059 (10 microM). Neither nifedipine nor NCDC reduced 5-HT (1 microM)-stimulated Erk MAPK tyrosyl-phosphorylation, but the combination of nifedipine, NCDC and PD098059 abolished 5-HT (1 microM)-stimulated Erk MAPK tyrosyl-phosphorylation. Taken together, these studies indicate that stimulation of a vascular 5-HT2A receptor activates Ca++ channels and PLC as well as MEK to cause rat aortic contraction and that MEK activation is at least partially independent of the two pathways classically associated with 5-HT2A receptors.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Músculo Liso Vascular/efeitos dos fármacos , Receptores de Serotonina/fisiologia , Transdução de Sinais , Animais , Canais de Cálcio/fisiologia , Canais de Cálcio Tipo L , Ativação Enzimática , Genisteína/farmacologia , Masculino , Músculo Liso Vascular/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley , Receptor 5-HT2A de Serotonina , Serotonina/farmacologia , Fosfolipases Tipo C/fisiologia , Vasoconstrição/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...