Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Platelets ; 35(1): 2313359, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38353233

RESUMO

Cyclic guanosine monophosphate (cGMP) is a second messenger produced by the NO-sensitive guanylyl cyclase (NO-GC). The NO-GC/cGMP pathway in platelets has been extensively studied. However, its role in regulating the biomechanical properties of platelets has not yet been addressed and remains unknown. We therefore investigated the stiffness of living platelets after treatment with the NO-GC stimulator riociguat or the NO-GC activator cinaciguat using scanning ion conductance microscopy (SICM). Stimulation of human and murine platelets with cGMP-modulating drugs decreased cellular stiffness and downregulated P-selectin, a marker for platelet activation. We also quantified changes in platelet shape using deep learning-based platelet morphometry, finding that platelets become more circular upon treatment with cGMP-modulating drugs. To test for clinical applicability of NO-GC stimulators in the context of increased thrombogenicity risk, we investigated the effect of riociguat on platelets from human immunodeficiency virus (HIV)-positive patients taking abacavir sulfate (ABC)-containing regimens. Our results corroborate a functional role of the NO-GC/cGMP pathway in platelet biomechanics, indicating that biomechanical properties such as stiffness or shape could be used as novel biomarkers in clinical research.


Increased platelet activation and development of thrombosis has been linked to a dysfunctional NO-GC/cGMP signaling pathway. How this pathway affects platelet stiffness, however, has not been studied yet. For the first time, we used novel microscopy techniques to investigate stiffness and shape of platelets in human and murine blood samples treated with cGMP modifying drugs. Stiffness contains information about biomechanical properties of the cytoskeleton, and shape quantifies the spreading behavior of platelets. We showed that the NO-GC/cGMP signaling pathway affects platelet stiffness, shape, and activation in human and murine blood. HIV-positive patients are often treated with medication that may disrupt the NO-GC/cGMP signaling pathway, leading to increased cardiovascular risk. We showed that treatment with cGMP-modifying drugs altered platelet shape and aggregation in blood from HIV-negative volunteers but not from HIV-positive patients treated with medication. Our study suggests that platelet stiffness and shape can be biomarkers for estimating cardiovascular risk.


Assuntos
Plaquetas , Transdução de Sinais , Humanos , Camundongos , Animais , Fenômenos Biomecânicos , Plaquetas/metabolismo , Guanilato Ciclase/metabolismo , Guanilato Ciclase/farmacologia , Ativação Plaquetária , GMP Cíclico/metabolismo , GMP Cíclico/farmacologia , Óxido Nítrico/metabolismo , Agregação Plaquetária
2.
Kidney Int ; 104(3): 508-525, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37356621

RESUMO

Natriuretic peptides exert not only blood-lowering but also kidney-protective effects through guanylyl cyclase-A (GC-A), a natriuretic peptide receptor. Signaling through GC-A has been shown to protect podocytes from aldosterone-induced glomerular injury, and a p38 mitogen-activated protein kinase (MAPK) inhibitor reduced glomerular injury in aldosterone-infused podocyte-specific GC-A knockout mice. To explore the role of p38 MAPK in podocytes, we constructed podocyte-specific p38 MAPK and GC-A double knockout mice (pod-double knockout mice). Unexpectedly, aldosterone-infused and high salt-fed (B-ALDO)-treated pod-double knockout mice resulted in elevated serum creatinine, massive albuminuria, macrophage infiltration, foot process effacement, nephrin and podocin reduction, and additionally, intra-capillary fibrin thrombi, indicating endothelial injury. Microarray analysis showed increased plasminogen activator inhibitor-1 (PAI-1) in glomeruli of B-ALDO-treated pod-double knockout mice. In B-ALDO-treated pod-double knockout mice, PAI-1 increased in podocytes, and treatment with PAI-1 neutralizing antibody ameliorated intra-capillary thrombus formation. In vitro, deletion of p38 MAPK by the CRISPR/Cas9 system and knockdown of GC-A in human cultured podocytes upregulated PAI-1 and transforming growth factor- ß1 (TGF-ß1). When p38 MAPK knockout podocytes, transfected with a small interfering RNA to suppress GC-A, were co-cultured with glomerular endothelial cells in a transwell system, the expression of TGF-ß1 was increased in glomerular endothelial cells. PAI-1 inhibition ameliorated both podocyte and endothelial injury in the transwell system signifying elevated PAI-1 in podocytes is a factor disrupting normal podocyte-endothelial crosstalk. Thus, our results indicate that genetic dual deletion of p38 MAPK and GC-A in podocytes accelerates both podocyte and endothelial injuries, suggesting these two molecules play indispensable roles in podocyte function.


Assuntos
Podócitos , Trombose , Animais , Humanos , Camundongos , Aldosterona/farmacologia , Aldosterona/metabolismo , Células Endoteliais/metabolismo , Guanilato Ciclase/metabolismo , Guanilato Ciclase/farmacologia , Camundongos Knockout , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Inibidor 1 de Ativador de Plasminogênio/farmacologia , Podócitos/metabolismo , Trombose/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Proteína Quinase 14 Ativada por Mitógeno
3.
J Urol ; 190(2): 746-56, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23454157

RESUMO

PURPOSE: Because neuronal released endogenous H2S has a key role in relaxation of the bladder outflow region, we investigated the mechanisms involved in H2S dependent inhibitory neurotransmission to the pig bladder neck. MATERIALS AND METHODS: Bladder neck strips were mounted in myographs for isometric force recording and simultaneous measurement of intracellular Ca(2+) and tension. RESULTS: On phenylephrine contracted preparations electrical field stimulation and the H2S donor GYY4137 evoked frequency and concentration dependent relaxation, which was reduced by desensitizing capsaicin sensitive primary afferents with capsaicin, and the blockade of adenosine 5'-triphosphate dependent K(+) channels, cyclooxygenase and cyclooxygenase-1 with glibenclamide, indomethacin and SC560, respectively. Inhibition of vanilloid, transient receptor potential A1, transient receptor potential vanilloid 1, vasoactive intestinal peptide/pituitary adenylyl cyclase-activating polypeptide and calcitonin gene-related peptide receptors with capsazepine, HC030031, AMG9810, PACAP6-38 and CGRP8-37, respectively, also decreased electrical field stimulation and GYY4137 responses. H2S relaxation was not changed by guanylyl cyclase, protein kinase A, or Ca(2+) activated or voltage gated K(+) channel inhibitors. GYY4137 inhibited the contractions induced by phenylephrine and by K(+) enriched (80 mM) physiological saline solution. To a lesser extent it decreased the phenylephrine and K(+) induced increases in intracellular Ca(2+). CONCLUSIONS: H2S produces pig bladder neck relaxation via activation of adenosine 5'-triphosphate dependent K(+) channel and by smooth muscle intracellular Ca(2+) desensitization dependent mechanisms. H2S also promotes the release of sensory neuropeptides and cyclooxygenase-1 pathway derived prostanoids from capsaicin sensitive primary afferents via transient receptor potential A1, transient receptor potential vanilloid 1 and/or related ion channel activation.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Sulfeto de Hidrogênio/farmacologia , Canais KATP/metabolismo , Músculo Liso/efeitos dos fármacos , Células Receptoras Sensoriais/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Bexiga Urinária/inervação , Bexiga Urinária/metabolismo , Acetanilidas/farmacologia , Acrilamidas/farmacologia , Animais , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Capsaicina/análogos & derivados , Capsaicina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/farmacologia , Estimulação Elétrica , Glibureto/farmacologia , Guanilato Ciclase/farmacologia , Indometacina/farmacologia , Morfolinas/farmacologia , Compostos Organotiofosforados/farmacologia , Fenilefrina/farmacologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Purinas/farmacologia , Pirazóis/farmacologia , Suínos
4.
Digestion ; 87(2): 102-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23327898

RESUMO

BACKGROUND: Portal pressure (PP) results from the interplay of vasoconstrictors and vasodilators. Recently, we have shown that Kupffer cell (KC) activation increases PP. AIMS: The role of the vasodilating compounds nitric oxide (NO) and carbon monoxide (CO) was studied. The hypothesis of the present study was that these vasodilators counteract the PP increase following KC activation. METHODS: Livers of rats weighing 180-200 g were isolated and perfused. KCs were activated by zymosan A (cell wall particles from yeast; 150 µg/ml). The effects of NO and guanylate cyclase (GC) were evaluated by the NO synthase inhibitor N(G)-nitro-L-arginine methylester (L-NAME; 0.3 mM, and the GC inhibitor 4H-8-bromo-1,2,4-oxadiazolo(3,4-d)benz(b)(1,4)oxazin-1-one (NS-2028, 1.0 µM); the effects of the heme oxygenase (HO) derived compound CO were evaluated by direct administration of CO or inhibition of HO by zinc protoporphyrin IX (ZnPP IX, 1.0 µM). RESULTS: In isolated perfused rat livers, administration of L-NAME or NS-2028 further raised PP increase following KC activation. This effect could be reduced by the cGMP analogue 8-Br-cGMP. Inhibition of HO caused marked amplification of PP increase in zymosan-treated organs. CO prevented this PP increase cGMP independently. Interestingly, KC activation and simultaneous inhibition of HO augmented the production of prostaglandins D2 and F2α and of thromboxane A2. Accordingly, indomethacin blunted the increase of PP in zymosan/ZnPP-treated livers. CONCLUSIONS: NO restricts the initial PP increase after KC activation by GC-mediated cGMP. CO from heme degradation limits the increase of PP after KC activation eicosanoid dependently, but cGMP independently.


Assuntos
Monóxido de Carbono/farmacologia , Heme Oxigenase (Desciclizante)/antagonistas & inibidores , Células de Kupffer/metabolismo , Óxido Nítrico/farmacologia , Pressão na Veia Porta/efeitos dos fármacos , Prostaglandinas/metabolismo , Animais , Inibidores Enzimáticos/farmacologia , Guanilato Ciclase/metabolismo , Guanilato Ciclase/farmacologia , Heme Oxigenase (Desciclizante)/metabolismo , Fígado/metabolismo , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Oxidiazóis/farmacologia , Oxazinas/farmacologia , Protoporfirinas/farmacologia , Ratos , Ratos Sprague-Dawley
5.
Cell Rep Med ; 4(4): 100992, 2023 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-37023747

RESUMO

Diabetic kidney disease (DKD) is the most common cause of renal failure. Therapeutics development is hampered by our incomplete understanding of animal models on a cellular level. We show that ZSF1 rats recapitulate human DKD on a phenotypic and transcriptomic level. Tensor decomposition prioritizes proximal tubule (PT) and stroma as phenotype-relevant cell types exhibiting a continuous lineage relationship. As DKD features endothelial dysfunction, oxidative stress, and nitric oxide depletion, soluble guanylate cyclase (sGC) is a promising DKD drug target. sGC expression is specifically enriched in PT and stroma. In ZSF1 rats, pharmacological sGC activation confers considerable benefits over stimulation and is mechanistically related to improved oxidative stress regulation, resulting in enhanced downstream cGMP effects. Finally, we define sGC gene co-expression modules, which allow stratification of human kidney samples by DKD prevalence and disease-relevant measures such as kidney function, proteinuria, and fibrosis, underscoring the relevance of the sGC pathway to patients.


Assuntos
Diabetes Mellitus , Nefropatias Diabéticas , Humanos , Ratos , Animais , Guanilil Ciclase Solúvel/metabolismo , Guanilil Ciclase Solúvel/farmacologia , Guanilil Ciclase Solúvel/uso terapêutico , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/patologia , Guanilato Ciclase/genética , Guanilato Ciclase/metabolismo , Guanilato Ciclase/farmacologia , Rim/metabolismo , Fibrose
6.
Psychopharmacology (Berl) ; 239(10): 3297-3311, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35978221

RESUMO

RATIONALE: Re-exposing an animal to an environment previously paired with an aversive stimulus evokes large alterations in behavioral and cardiovascular parameters. Dorsal hippocampus (dHC) receives important cholinergic inputs from the basal forebrain, and respective acetylcholine (ACh) levels are described to influence defensive behavior. Activation of muscarinic M1 and M3 receptors facilitates autonomic and behavioral responses along threats. Evidence show activation of cholinergic receptors promoting formation of nitric oxide (NO) and cyclic guanosine monophosphate (cGMP) in dHC. Altogether, the action of ACh and NO on conditioned responses appears to converge within dHC. OBJECTIVES: As answer about how ACh and NO interact to modulate defensive responses has so far been barely addressed, we aimed to shed additional light on this topic. METHODS: Male Wistar rats had guide cannula implanted into the dHC before being submitted to the contextual fear conditioning (3footshocks/085 mA/2 s). A catheter was implanted in the femoral artery the next day for cardiovascular recordings. Drugs were delivered into dHC 10 min before contextual re-exposure, which occurred 48 h after the conditioning procedure. RESULTS: Neostigmine (Neo) amplified the retrieval of conditioned responses. Neo effects (1 nmol) were prevented by the prior infusion of a M1-M3 antagonist (fumarate), a neuronal nitric oxide synthase inhibitor (NPLA), a NO scavenger (cPTIO), a guanylyl cyclase inhibitor (ODQ), and a NMDA antagonist (AP-7). Pretreatment with a selective M1 antagonist (pirenzepine) only prevented the increase in autonomic responses induced by Neo. CONCLUSION: The results show that modulation in the retrieval of contextual fear responses involves coordination of the dHC M1-M3/NO/cGMP/NMDA pathway.


Assuntos
N-Metilaspartato , Óxido Nítrico , Acetilcolina , Animais , Colinérgicos/farmacologia , Medo/fisiologia , Fumaratos/farmacologia , Guanosina Monofosfato/farmacologia , Guanilato Ciclase/metabolismo , Guanilato Ciclase/farmacologia , Hipocampo , Masculino , N-Metilaspartato/farmacologia , Neostigmina/farmacologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Pirenzepina/farmacologia , Ratos , Ratos Wistar , Receptores Colinérgicos/metabolismo , Receptores de N-Metil-D-Aspartato , Transmissão Sináptica
7.
Vascul Pharmacol ; 146: 107092, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35907614

RESUMO

Hinokitiol is a natural bioactive compound with numerous pharmacological properties. Here, we aimed to examine hinokitiol's effects on vascular relaxation. Cumulative relaxation responses to hinokitiol were assessed in isolated aortae from normotensive and angiotensin II-induced hypertensive rats in the presence and absence of selective inhibitors. Hinokitiol produced vasodilation of phenylephrine preconstricted aortae using both normotensive and hypertensive rats. In normotensive rats, hinokitiol's vasodilation was reduced by endothelial denudation and nitric oxide synthase (NOS), guanylate cyclase, and cyclooxygenase inhibition. Also, hinokitiol vasodilation was attenuated by ß-receptors, adenylate cyclase, Ca2+-activated K+ channels and hyperpolarization inhibition. Moreover, hinokitiol exhibited a blocking activity on Ca2+ mobilization through voltage dependent Ca2+ channels (VDCC). However, its effect was not changed by muscarinic receptor and Sarc-K+ ATP channels blocking but was enhanced by blocking voltage-dependent K+ channels. However, in angiotensin II-induced hypertension, hinokitiol vasodilating activity was attenuated by NOS inhibition and it blocked Ca2+ mobilization through VDCC, while its vasodilation was partially attenuated by Sarc-K+ ATP channels blocking. However, the vasodilating effect of hinokitiol was not attenuated by either cyclooxygenase, ß-receptor, Ca2+-activated K+ channels, or voltage-dependent potassium channels inhibition, but was enhanced by blocking hyperpolarization. Hinokitiol's vasodilating effect in normotensive and hypertensive vessels is mediated through both endothelium-dependent and endothelium-independent mechanisms.


Assuntos
Hipertensão , Vasodilatação , Trifosfato de Adenosina/metabolismo , Adenilil Ciclases/metabolismo , Adenilil Ciclases/farmacologia , Angiotensina II/metabolismo , Angiotensina II/farmacologia , Animais , Canais de Cálcio Tipo L/metabolismo , Endotélio Vascular/metabolismo , Guanilato Ciclase/metabolismo , Guanilato Ciclase/farmacologia , Óxido Nítrico Sintase , Fenilefrina/farmacologia , Canais de Potássio/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Prostaglandina-Endoperóxido Sintases/farmacologia , Ratos
8.
Cardiovasc Res ; 118(12): 2703-2717, 2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-34550322

RESUMO

AIMS: Intimal hyperplasia is a common feature of vascular remodelling disorders. Accumulation of synthetic smooth muscle cell (SMC)-like cells is the main underlying cause. Current therapeutic approaches including drug-eluting stents are not perfect due to the toxicity on endothelial cells and novel therapeutic strategies are needed. Our preliminary screening for dysregulated cyclic nucleotide phosphodiesterases (PDEs) in growing SMCs revealed the alteration of PDE10A expression. Herein, we investigated the function of PDE10A in SMC proliferation and intimal hyperplasia both in vitro and in vivo. METHODS AND RESULTS: RT-qPCR, immunoblot, and in situ proximity ligation assay were performed to determine PDE10A expression in synthetic SMCs and injured vessels. We found that PDE10A mRNA and/or protein levels are up-regulated in cultured SMCs upon growth stimulation, as well as in intimal cells in injured mouse femoral arteries. To determine the cellular functions of PDE10A, we focused on its role in SMC proliferation. The anti-mitogenic effects of PDE10A on SMCs were evaluated via cell counting, BrdU incorporation, and flow cytometry. We found that PDE10A deficiency or inhibition arrested the SMC cell cycle at G1-phase with a reduction of cyclin D1. The anti-mitotic effect of PDE10A inhibition was dependent on cGMP-dependent protein kinase Iα (PKGIα), involving C-natriuretic peptide (CNP) and particulate guanylate cyclase natriuretic peptide receptor 2 (NPR2). In addition, the effects of genetic depletion and pharmacological inhibition of PDE10A on neointimal formation were examined in a mouse model of femoral artery wire injury. Both PDE10A knockout and inhibition decreased injury-induced intimal thickening in femoral arteries by at least 50%. Moreover, PDE10A inhibition decreased ex vivo remodelling of cultured human saphenous vein segments. CONCLUSIONS: Our findings indicate that PDE10A contributes to SMC proliferation and intimal hyperplasia at least partially via antagonizing CNP/NPR2/cGMP/PKG1α signalling and suggest that PDE10A may be a novel drug target for treating vascular occlusive disease.


Assuntos
Músculo Liso Vascular , Lesões do Sistema Vascular , Animais , Bromodesoxiuridina/metabolismo , Bromodesoxiuridina/farmacologia , Proliferação de Células , Células Cultivadas , GMP Cíclico/metabolismo , Proteína Quinase Dependente de GMP Cíclico Tipo I/metabolismo , Ciclina D1/metabolismo , Células Endoteliais/metabolismo , Guanilato Ciclase/metabolismo , Guanilato Ciclase/farmacologia , Humanos , Hiperplasia/metabolismo , Hiperplasia/patologia , Camundongos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Diester Fosfórico Hidrolases/metabolismo , RNA Mensageiro/metabolismo , Remodelação Vascular , Lesões do Sistema Vascular/tratamento farmacológico , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/metabolismo
9.
J Thromb Haemost ; 20(11): 2465-2474, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35950928

RESUMO

Platelets are the "guardians" of the blood circulatory system. At sites of vessel injury, they ensure hemostasis and promote immunity and vessel repair. However, their uncontrolled activation is one of the main drivers of thrombosis. To keep circulating platelets in a quiescent state, the endothelium releases platelet antagonists including nitric oxide (NO) that acts by stimulating the intracellular receptor guanylyl cyclase (GC). The latter produces the second messenger cyclic guanosine-3',5'-monophosphate (cGMP) that inhibits platelet activation by stimulating protein kinase G, which phosphorylates hundreds of intracellular targets. Intracellular cGMP pools are tightly regulated by a fine balance between GC and phosphodiesterases (PDEs) that are responsible for the hydrolysis of cyclic nucleotides. Phosphodiesterase type 5 (PDE5) is a cGMP-specific PDE, broadly expressed in most tissues in humans and rodents. In clinical practice, PDE5 inhibitors (PDE5i) are used as first-line therapy for erectile dysfunction, pulmonary artery hypertension, and lower urinary tract symptoms. However, several studies have shown that PDE5i may ameliorate the outcome of various other conditions, like heart failure and stroke. Interestingly, NO donors and cGMP analogs increase the capacity of anti-platelet drugs targeting the purinergic receptor type Y, subtype 12 (P2Y12) receptor to block platelet aggregation, and preclinical studies have shown that PDE5i inhibits platelet functions. This review summarizes the molecular mechanisms underlying the effect of PDE5i on platelet activation and aggregation focusing on the therapeutic potential of PDE5i in platelet disorders, and the outcomes of a combined therapy with PDE5i and NO donors to inhibit platelet activation.


Assuntos
Óxido Nítrico , Inibidores da Fosfodiesterase 5 , Humanos , Masculino , Plaquetas/metabolismo , GMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/farmacologia , Guanosina/metabolismo , Guanosina/farmacologia , Guanilato Ciclase/metabolismo , Guanilato Ciclase/farmacologia , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/metabolismo , Nucleotídeos Cíclicos/metabolismo , Nucleotídeos Cíclicos/farmacologia , Inibidores da Fosfodiesterase 5/farmacologia , Inibidores da Fosfodiesterase 5/uso terapêutico , Inibidores da Fosfodiesterase 5/metabolismo , Inibidores da Agregação Plaquetária/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Proteínas Quinases/metabolismo
10.
J Exp Med ; 199(3): 347-56, 2004 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-14744991

RESUMO

Insulin-induced vasodilatation is sensitive to nitric oxide (NO) synthase (NOS) inhibitors. However, insulin is unable to relax isolated arteries or to activate endothelial NOS in endothelial cells. Since insulin can enhance platelet endothelial NOS activity, we determined whether insulin-induced vasodilatation can be attributed to a NO-dependent, platelet-mediated process. Insulin failed to relax endothelium-intact rings of porcine coronary artery. The supernatant from insulin-stimulated human platelets induced complete relaxation, which was prevented by preincubation of platelets with a NOS inhibitor, the soluble guanylyl cyclase inhibitor, NS 2028, or the G kinase inhibitor, KT 5823, and was abolished by an adenosine A2A receptor antagonist. Insulin induced the release of adenosine trisphosphate (ATP), adenosine, and serotonin from platelet-dense granules in a NO-dependent manner. This response was not detected using insulin-stimulated platelets from endothelial NOS-/- mice, although a NO donor elicited ATP release. Insulin-induced ATP release from human platelets correlated with the association of syntaxin 2 with the vesicle-associated membrane protein 3 but was not associated with the activation of alphaIIbbeta3 integrin. Thus, insulin elicits the release of vasoactive concentrations of ATP and adenosine from human platelets via a NO-G kinase-dependent signaling cascade. The mechanism of dense granule secretion involves the G kinase-dependent association of syntaxin 2 with vesicle-associated membrane protein 3.


Assuntos
Plaquetas/fisiologia , Endotélio Vascular/fisiologia , Insulina/farmacologia , Óxido Nítrico/sangue , Vasodilatação/fisiologia , Adenosina/sangue , Trifosfato de Adenosina/sangue , Plaquetas/efeitos dos fármacos , Carbazóis/farmacologia , Endotélio Vascular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Guanilato Ciclase/farmacologia , Humanos , Técnicas In Vitro , Indóis/farmacologia , Oxidiazóis/farmacologia , Oxazinas/farmacologia , Inibidores de Proteínas Quinases , Serotonina/sangue , Vasodilatação/efeitos dos fármacos
11.
J Pharmacol Exp Ther ; 333(1): 210-7, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20040579

RESUMO

We tested the hypothesis that changes in arterial blood flow modify the function of endothelial Ca2+-activated K+ channels [calcium-activated K+ channel (K(Ca)), small-conductance calcium-activated K+ channel (SK3), and intermediate calcium-activated K+ channel (IK1)] before arterial structural remodeling. In rats, mesenteric arteries were exposed to increased [+90%, high flow (HF)] or reduced blood flow [-90%, low flow (LF)] and analyzed 24 h later. There were no detectable changes in arterial structure or in expression level of endothelial nitric-oxide synthase, SK3, or IK1. Arterial relaxing responses to acetylcholine and 3-oxime-6,7-dichlore-1H-indole-2,3-dione (NS309; activator of SK3 and IK1) were measured in the absence and presence of endothelium, NO, and prostanoid blockers, and 6,12,19,20,25,26-hexahydro-5,27:13,18:21,24-trietheno-11,7-metheno-7H-dibenzo [b,n] [1,5,12,16]tetraazacyclotricosine-5,13-diium dibromide (UCL 1684; inhibitor of SK3) or 1-[(2-chlorophenyl)diphenylmethyl]-1H-pyrazole (TRAM-34; inhibitor of IK1). In LF arteries, endothelium-dependent relaxation was markedly reduced, due to a reduction in the endothelium-derived hyperpolarizing factor (EDHF) response. In HF arteries, the balance between the NO/prostanoid versus EDHF response was unaltered. However, the contribution of IK1 to the EDHF response was enhanced, as indicated by a larger effect of TRAM-34 and a larger residual NS309-induced relaxation in the presence of UCL 1684. Reduction of blood flow selectively blunts EDHF relaxation in resistance arteries through inhibition of the function of K(Ca) channels. An increase in blood flow leads to a more prominent role of IK1 channels in this relaxation.


Assuntos
Endotélio Vascular/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/biossíntese , Artérias Mesentéricas/metabolismo , Canais de Potássio Cálcio-Ativados/biossíntese , Acetilcolina/farmacologia , Animais , Fatores Biológicos/fisiologia , Inibidores de Ciclo-Oxigenase/farmacologia , Endotélio Vascular/efeitos dos fármacos , Guanilato Ciclase/farmacologia , Indóis/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/agonistas , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/antagonistas & inibidores , Masculino , Artérias Mesentéricas/efeitos dos fármacos , Contração Muscular , Relaxamento Muscular , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiologia , Óxido Nítrico Sintase/antagonistas & inibidores , Oximas/farmacologia , Canais de Potássio Cálcio-Ativados/agonistas , Canais de Potássio Cálcio-Ativados/antagonistas & inibidores , Ratos , Ratos Endogâmicos WKY , Receptores Citoplasmáticos e Nucleares/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa , Guanilil Ciclase Solúvel , Circulação Esplâncnica , Estresse Mecânico
12.
Scand J Gastroenterol ; 43(4): 447-55, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18365910

RESUMO

OBJECTIVE: The peptide hormones guanylin and uroguanylin and their receptor, guanylate cyclase C (GC-C), are expressed in pancreatic duct cells. In colon cancer, guanylin peptides are shown to exert strong anti-tumor activity through the GC-C pathway. The objective of this study was to analyze the role of guanylin and uroguanylin in human pancreatic cancer. MATERIAL AND METHODS: Quantitative real-time polymerase chain reaction (QRT-PCR) was used to show the expression of guanylin, uroguanylin and GC-C in specimens of human pancreatic cancer, chronic pancreatitis donor and in pancreatic tumor cell lines. The presence of guanylins and GC-C in tumor cell lines and in pancreatic cancer tissues was shown by immunofluorescence and immunohistochemistry. The effect of guanylin and uroguanylin on cell cycle and cell death of pancreatic cancer cells was investigated by fluorescence activated cell sorter (FACS) analysis using annexin and propidium iodide. In addition, the growth inhibitory effect of guanylins on pancreatic cancer cells was assessed using the MTT assay. RESULTS: Guanylin, uroguanylin and GC-C were expressed at mRNA and protein levels in pancreatic cancer and cancer cell lines. As shown by QRT-PCR, GC-C expression was significantly up-regulated in pancreatic cancer compared with that in healthy pancreatic tissues (p<0.00001) and chronic pancreatitis (p<0.05). Guanylin and uroguanylin were not up-regulated in pancreatic cancer. The MTT assay revealed significant inhibition of pancreatic cancer cell proliferation by uroguanylin in a dose-dependent fashion, whereas Panc1 and Capan1 cell lines were significantly inhibited already at the lowest uroguanylin concentration (2 nM, p<0.05). CONCLUSIONS: Our data suggest therapeutic properties of uroguanylin in pancreatic cancer via GC-C-dependent mechanisms. In addition, determination of GC-C expression might be a useful marker for differentiation between pancreatic cancer and chronic pancreatitis.


Assuntos
Carcinoma Ductal Pancreático/patologia , Divisão Celular/efeitos dos fármacos , Peptídeos Natriuréticos/farmacologia , Neoplasias Pancreáticas/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/uso terapêutico , Apoptose , Biomarcadores Tumorais/análise , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral/metabolismo , Linhagem Celular Tumoral/patologia , Doença Crônica , Feminino , Hormônios Gastrointestinais/metabolismo , Hormônios Gastrointestinais/farmacologia , Guanilato Ciclase/metabolismo , Guanilato Ciclase/farmacologia , Humanos , Masculino , Pessoa de Meia-Idade , Peptídeos Natriuréticos/metabolismo , Peptídeos Natriuréticos/uso terapêutico , Pâncreas/metabolismo , Neoplasias Pancreáticas/metabolismo , Pancreatite/patologia , Reação em Cadeia da Polimerase
13.
Mol Cancer Ther ; 5(5): 1190-6, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16731751

RESUMO

The nonsteroidal anti-inflammatory drug sulindac is metabolized to sulindac sulfone (exisulind), an antineoplastic agent that inhibits growth and induces apoptosis in solid tumors. In colon cancer cells, the antineoplastic effects of exisulind have been attributed, in part, to induction of cyclic guanosine 3',5'-monophosphate (cGMP) signaling through inhibition of cGMP-specific phosphodiesterases, which elevates intracellular cGMP, and novel expression of cGMP-dependent protein kinase (PKG) Ibeta, the presumed downstream effector mediating apoptosis. Here, inhibition of proliferation and induction of cell death by exisulind was dissociated from cGMP signaling in human colon cancer cells. Accumulation of intracellular cGMP produced by an exogenous cell-permeant analogue of cGMP or a potent agonist of guanylyl cyclase C yielded cytostasis without cell death. Surprisingly, the antiproliferative effects of induced cGMP accumulation were paradoxically less than additive, rather than synergistic, when combined with exisulind. Further, although exisulind induced expression of PKG Ibeta, it did not elevate intracellular cGMP and its efficacy was not altered by inhibition or activation of PKG I. Rather, PKG I induced by exisulind may mediate desensitization of cytostasis induced by cGMP. Thus, cytotoxic effects of exisulind are independent of cGMP signaling in human colon cancer cells. Moreover, combination therapies, including exisulind and agents that induce cGMP signaling, may require careful evaluation in patients with colon cancer.


Assuntos
Antineoplásicos/farmacologia , Neoplasias do Colo/metabolismo , Guanilato Ciclase/farmacologia , Transdução de Sinais , Sulindaco/análogos & derivados , Células CACO-2 , Morte Celular , Proliferação de Células/efeitos dos fármacos , GMP Cíclico/metabolismo , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Ligantes , Modelos Biológicos , Receptores de Enterotoxina , Receptores Acoplados a Guanilato Ciclase , Receptores de Peptídeos , Transdução de Sinais/efeitos dos fármacos , Sulindaco/farmacologia , Sulindaco/toxicidade , Células Tumorais Cultivadas
14.
J Appl Physiol (1985) ; 100(5): 1666-73, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16424074

RESUMO

ACh regulates arousal, and the present study was designed to provide insight into the neurochemical mechanisms modulating ACh release in the pontine reticular formation. Nitric oxide (NO)-releasing beads microinjected into the pontine reticular formation of C57BL/6J (B6) mice significantly (P < 0.0001) increased ACh release. Microdialysis delivery of the NO donor N-ethyl-2-(1-ethyl-2-hydroxy-2-nitrosohydrazino)-ethanamine (NOC-12) to the mouse pontine reticular formation also caused a concentration-dependent increase in ACh release (P < 0.001). These are the first neurochemical data showing that ACh release in the pontine reticular formation of the B6 mouse is modulated by NO. The signal transduction cascade through which NO modulates ACh release in the pontine reticular formation has not previously been characterized. Therefore, an additional series of studies quantified the effects of a soluble guanylate cyclase (sGC) inhibitor, 1H-[1,2,4]oxadiazolo-[4,3-a]quinoxalin-1-one (ODQ), on ACh release in the cat medial pontine reticular formation. During naturally occurring states of sleep and wakefulness, but not anesthesia, ODQ caused a significant (P < 0.001) decrease in ACh release. These results show for the first time that NO modulates ACh in the medial pontine reticular formation of the cat via an NO-sensitive sGC signal transduction cascade. Isoflurane and halothane anesthesia have been shown to decrease ACh release in the medial pontine reticular formation. The finding that ODQ did not alter ACh release during isoflurane or halothane anesthesia demonstrates that these anesthetics disrupt the NO-sensitive sGC-cGMP pathway. Considered together, results from the mouse and cat indicate that NO modulates ACh release in arousal-promoting regions of the pontine reticular formation via an NO-sensitive sGC-cGMP pathway.


Assuntos
Acetilcolina/metabolismo , Nível de Alerta/fisiologia , Guanilato Ciclase/fisiologia , Óxido Nítrico/fisiologia , Ponte/metabolismo , Animais , Gatos , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Relação Dose-Resposta a Droga , Guanilato Ciclase/antagonistas & inibidores , Guanilato Ciclase/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos , Óxido Nítrico/farmacologia , Compostos Nitrosos/farmacologia , Oxidiazóis/farmacologia , Quinoxalinas/farmacologia , Transdução de Sinais/fisiologia , Sono/fisiologia , Fatores de Tempo
15.
Auton Neurosci ; 126-127: 193-201, 2006 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-16580888

RESUMO

We previously identified an action of nitric oxide (NO) within the nucleus tractus solitarii (NTS) that attenuates the cardiac component of the baroreceptor reflex. In the present study we have tested the hypothesis that angiotensin II (AngII), acting on angiotensin type 1 receptors (AT1R), can release NO within the NTS and that its actions are mediated by soluble guanylate cyclase (sGC). Utilising cryogenic electron paramagnetic resonance (EPR), we have detected NO release in brainstem samples following AngII, but not saline, microinjections into the NTS. In these experiments, we confirmed that both AngII and a NO donor (diethylamine NONOate) in the NTS both depressed the baroreflex bradycardia. In additional studies, we showed that the latter effects were both sensitive to blockade of sGC using 1H-[1,2,4]oxadiazolo-[4,3-a]quinoxalin-1-one (ODQ). To initiate studies to resolve the cellular source of NO released by angiotensin II in the NTS, we performed immunohistochemical/electron microscopy studies on the distribution of AT1R. We found AT1R located on NTS neurones and blood vessels. Since a rise in intracellular calcium [Ca]i levels is prerequisite for nNOS activation, we imaged responses in [Ca]i in NTS neurones during exposure to AngII in vitro using confocal microscopy. Our data indicate a paucity of neurones showing changes in [Ca]i when exposed to AngII (200 nM). We suggest that AngII-induced release of NO is from non-neuronal sites. With the presence of AT1R on blood vessel endothelial cells we propose that AngII released NO in the NTS is due to activation of endothelial nitric oxide synthase located within the endothelium. The present study supports the novel concept that AngII can trigger NO release in the NTS by a mechanism of vascular-neuronal signalling that affects central neuronal networks regulating cardiovascular function.


Assuntos
Angiotensina II/farmacologia , Óxido Nítrico/metabolismo , Núcleo Solitário/efeitos dos fármacos , Núcleo Solitário/enzimologia , Animais , Barorreflexo/efeitos dos fármacos , Interações Medicamentosas , Espectroscopia de Ressonância de Spin Eletrônica/métodos , Guanilato Ciclase/antagonistas & inibidores , Guanilato Ciclase/farmacologia , Hidrazinas/farmacologia , Imuno-Histoquímica/métodos , Técnicas In Vitro , Masculino , Microscopia Imunoeletrônica/métodos , Doadores de Óxido Nítrico/farmacologia , Óxidos de Nitrogênio/farmacologia , Oxidiazóis/farmacologia , Ratos , Ratos Wistar , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 1 de Angiotensina/ultraestrutura , Núcleo Solitário/metabolismo , Núcleo Solitário/ultraestrutura
16.
Curr Vasc Pharmacol ; 3(1): 41-53, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15638781

RESUMO

In the vasculature it is well established that cGMP is involved in the relaxant response to nitric oxide (NO) and NO donors. However, there is an increasing evidence that alternative/additional pathways that are cGMP-independent may also exist. A key criterion for a response to NO or a NO donor drug to be classified as cGMP-independent is lack of (or incomplete) inhibition by the selective inhibitor of soluble guanylate cyclase, ODQ (1H-[1,2,4]oxadiazole[4,3-a]quinoxalin-1-one). In many blood vessels cGMP-independent mechanisms contribute to the vasorelaxation, and in certain vascular beds cGMP-independent relaxation may be the predominant mechanism of action of NO and NO donors. NO donor drugs that generate NO "spontaneously", like authentic NO (i.e. solutions of NO gas), appear to exhibit a larger component of cGMP-independent vasorelaxation than do those drugs that require bioactivation in the tissue. The long lasting inhibition of responses to vasoconstrictors by S-nitrosothiols, persisting after removal of these NO donors, may be a cGMP-independent process, at least in some vessels. The mechanisms involved in the inhibition of vascular growth by NO and NO donors are predominantly cGMP-independent, as are the mechanisms responsible for the effects of NO donors on apoptosis in vascular smooth muscle and endothelial cells. The ability of NO and NO donors to inhibit platelet aggregation has a significant cGMP-independent component. cGMP-independent pathways are most often, though not exclusively, seen at high concentrations (microM - mM) of NO and NO donors. Hence, in relation to the actions of endogenous NO, these pathways may be particularly important in settings when the inducible isoform of NO-synthase is expressed. Furthermore, cGMP-independent pathways are enhanced in animal models of atherosclerosis and ischaemia. This suggests that it may be possible to target cGMP-independent mechanisms with selected NO donors in disease states.


Assuntos
Plaquetas/efeitos dos fármacos , Vasos Sanguíneos/efeitos dos fármacos , GMP Cíclico/antagonistas & inibidores , GMP Cíclico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico/metabolismo , Animais , Austrália , Plaquetas/metabolismo , Vasos Sanguíneos/anatomia & histologia , Vasos Sanguíneos/patologia , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Guanilato Ciclase/antagonistas & inibidores , Guanilato Ciclase/metabolismo , Guanilato Ciclase/farmacologia , Humanos , Doadores de Óxido Nítrico/classificação , Doadores de Óxido Nítrico/metabolismo
17.
FASEB J ; 18(10): 1108-10, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15132985

RESUMO

Nitric oxide (NO) is thought to play an important role as a signaling molecule in embryonic and adult cardiomyocytes; however, its involvement in muscarinic signaling is still unclear. The aim of the present work was to analyze the muscarinic modulation of the L-type Ca2+ current (ICa) in early- and late-stage embryonic ventricular cardiomyocytes. Muscarinic stimulation depressed basal ICa by 30.1 +/- 3.2% (n=27) in early-stage cardiomyocytes. Pharmacological evidence suggested that the muscarinic modulation was mediated through generation of NO, activation of cGMP-dependent phosphodiesterase (PDE) 2, and ensuing lowering of cyclic AMP/protein kinase A (cAMP/PKA) levels. Conversely, in late-stage cardiomyocytes, muscarinic regulation of ICa occurred in a NO-independent manner via inhibition of prestimulated adenylyl cyclase (AC). To unequivocally prove the involvement of NO and to identify the nitric oxide synthase (NOS) isoform(s), we analyzed muscarinic signaling in embryonic ventricular cardiomyocytes of NOS2 (-/-) and NOS3 (-/-) mice. The early-stage NOS3 (-/-) cardiomyocytes lacked muscarinic modulation, whereas it was preserved in NOS2 (-/-) cells. Moreover, at the late embryonic stage, muscarinic modulation of ICa was intact in both strains. Thus, NO is the key regulator of muscarinic signaling in the early embryonic ventricle, whereas at later stages, signaling occurs through a NO-independent pathway.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Cálcio/metabolismo , Coração Fetal/fisiologia , Óxido Nítrico/fisiologia , Transdução de Sinais/fisiologia , Animais , Animais não Endogâmicos , Carbacol/farmacologia , Colinérgicos/farmacologia , Proteína Quinase Tipo II Dependente de AMP Cíclico , Proteínas Quinases Dependentes de AMP Cíclico/farmacologia , Feminino , Guanilato Ciclase/antagonistas & inibidores , Guanilato Ciclase/farmacologia , Ventrículos do Coração/citologia , Ventrículos do Coração/embriologia , Transporte de Íons/efeitos dos fármacos , Camundongos , Camundongos Knockout , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/fisiologia , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Toxina Pertussis/farmacologia , Inibidores de Fosfodiesterase/farmacologia , Receptores Muscarínicos/efeitos dos fármacos , Receptores Muscarínicos/fisiologia , Transdução de Sinais/efeitos dos fármacos , ômega-N-Metilarginina/farmacologia
18.
Clin Cancer Res ; 9(13): 4891-6, 2003 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-14581363

RESUMO

PURPOSE: To determine the role of gelatinase in the hepatic metastatic process of colorectal cancer, we correlated gelatinolytic activity in colorectal tumor tissue both to the presence of intravasated colorectal epithelial cells and to the formation of liver metastasis. EXPERIMENTAL DESIGN: The gelatinolytic activity was analyzed in tumor tissue samples from 68 colorectal cancer patients by using gelatin substrate zymography. The presence of intravasated colorectal epithelial cells was defined as detection of guanylyl cyclase C mRNA in blood sampled from drainage vein of a tumor-bearing colorectal segment. RESULTS: Forty of 68 patients were noted to have guanylyl cyclase C mRNA expression in their drainage venous blood. Fifteen patients were noted to have liver metastasis at the time of surgery, and another 15 patients developed liver metastasis during median follow-up period of 53 months. Either individual or total gelatinolytic activity in colorectal tumor tissue failed to predict the presence of intravasated colorectal epithelial cells in the drainage venous blood or formation of liver metastasis. Presence of both intravasated colorectal epithelial cells and high total gelatinolytic activity in colorectal tumor tissue, however, is a strong predictor of liver metastasis (P = 0.004). CONCLUSION: Our data suggested that gelatinolytic activity in colorectal tumor tissues may facilitate the hepatic metastatic process in the steps after intravasation but not during or before intravasation.


Assuntos
Neoplasias Colorretais/patologia , Gelatinases/fisiologia , Guanilato Ciclase/farmacologia , Neoplasias Hepáticas/secundário , Metástase Neoplásica , Adulto , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular Tumoral , Neoplasias Colorretais/enzimologia , Feminino , Gelatinases/metabolismo , Guanilato Ciclase/biossíntese , Humanos , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/patologia , Masculino , Pessoa de Meia-Idade , RNA/metabolismo , RNA Mensageiro/metabolismo , Receptores de Enterotoxina , Receptores Acoplados a Guanilato Ciclase , Receptores de Peptídeos/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sensibilidade e Especificidade , Fatores de Tempo , Resultado do Tratamento
19.
Cardiovasc Res ; 51(1): 51-8, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11399247

RESUMO

OBJECTIVE: : In sinoatrial (SA) node cells, nitric oxide (NO) exerts a dual effect on the hyperpolarization-activated current, I(f), i.e. in basal conditions NO enhances I(f) whereas in the presence of beta-adrenergic stimulation it decreases it. Recent studies have shown that I(f) is present in ventricular myocytes from hypertrophied or failing hearts where it may promote abnormal automaticity. Since these pathological conditions are associated with increased sympathetic tone and upregulation of myocardial NO production, we set out to investigate whether I(f) is similarly modulated by NO in hypertrophied ventricular myocytes. METHODS: Left ventricular myocytes were isolated from 18-20-month-old spontaneously hypertensive rats (SHRs). Membrane current was measured under whole-cell or amphotericin-perforated patch-clamp conditions, at 35 degrees C. RESULTS: Application of diethylamine-NO (DEA-NO, 1-100 microM) did not alter the amplitude or voltage dependence of activation of I(f) under basal conditions (half-activation voltage, V(h): control -82.9+/-2.6, DEA-NO -84.0+/-2.6 mV). Similarly, I(f) was not affected by the inhibition of endogenous NO production (L-NMMA, 500 microM) or guanylate cyclase (ODQ, 10 microM). Forskolin (10 microM) or isoprenaline (100 nM) elicited a positive shift in V(h) but subsequent application of DEA-NO did not further affect the properties of I(f). CONCLUSIONS: Our results show that, unlike in SA node cells, in SHR ventricular myocytes basal and adrenergically stimulated I(f) is not modulated by exogenous NO or by constitutive NO or cGMP production.


Assuntos
Arritmias Cardíacas/fisiopatologia , Cardiomegalia/fisiopatologia , Hidrazinas/farmacologia , Doadores de Óxido Nítrico/farmacologia , Nó Sinoatrial/efeitos dos fármacos , Adenilil Ciclases/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Anfotericina B/farmacologia , Análise de Variância , Animais , Colforsina/farmacologia , Inibidores Enzimáticos/farmacologia , Guanilato Ciclase/antagonistas & inibidores , Guanilato Ciclase/farmacologia , Isoproterenol/farmacologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Óxido Nítrico Sintase/antagonistas & inibidores , Óxidos de Nitrogênio , Oxidiazóis/farmacologia , Técnicas de Patch-Clamp , Penicilamina/análogos & derivados , Penicilamina/farmacologia , Piridinas/farmacologia , Ratos , Ratos Endogâmicos SHR , ômega-N-Metilarginina/farmacologia
20.
Cell Calcium ; 23(6): 405-12, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9924632

RESUMO

We investigated the mechanism of guanosine 3',5'-monophosphate (cGMP) production in rabbit parotid acinar cells. Methacholine, a muscarinic cholinergic agonist, stimulated cGMP production in a dose-dependent manner but not isoproterenol, a beta-adrenergic receptor stimulant. Methacholine-stimulated cGMP production has been suggested to be coupled to Ca2+ mobilization, because intracellular Ca2+ elevating reagents, such as thapsigargin and the Ca2+ ionophore A23187, mimicked the effect of methacholine. The cGMP production induced by Ca2+ mobilization has also been suggested to be coupled to nitric oxide (NO) generation because the effects of methacholine, thapsigargin and A23187 on cGMP production were blocked by NG-nitro-L-arginine methyl ester (L-NAME), a specific inhibitor of nitric oxide synthase (NOS), and hemoglobin, a scavenger of nitric oxide (NO). Sodium nitroprusside (SNP), a NO donor, stimulated cGMP production. Furthermore, methacholine stimulated NO generation, and NOS activity in the cytosolic fraction in rabbit parotid acinar cells was exclusively dependent on Ca2+. These findings suggest that cGMP production induced by the activation of muscarinic cholinergic receptors is coupled to NO generation via Ca2+ mobilization.


Assuntos
Cálcio/metabolismo , GMP Cíclico/biossíntese , Óxido Nítrico/biossíntese , Glândula Parótida/metabolismo , Aminoquinolinas/farmacologia , Animais , Arginina/metabolismo , Calcimicina/farmacologia , Cálcio/farmacologia , Citrulina/metabolismo , Inibidores Enzimáticos/metabolismo , Guanilato Ciclase/farmacologia , Cloreto de Metacolina/farmacologia , Modelos Biológicos , NG-Nitroarginina Metil Éster/farmacologia , Coelhos , Receptores Muscarínicos/metabolismo , Tapsigargina/farmacologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA