Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
Am J Physiol Lung Cell Mol Physiol ; 323(4): L450-L463, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-35972838

RESUMO

The enzyme, nitric oxide-sensitive guanylyl cyclase (NO-GC), is activated by binding NO to its prosthetic heme group and catalyzes the formation of cGMP. The NO-GC is primarily known to mediate vascular smooth muscle relaxation in the lung, and inhaled NO has been successfully used as a selective pulmonary vasodilator. In comparison, NO-GC's impact on the regulation of airway tone is less acknowledged and, most importantly, little is known about the issue that NO-GC signaling is accomplished by two isoforms: NO-GC1 and NO-GC2, implying the existence of distinct "cGMP pools." Herein, we investigated the functional role of the NO-GC isoforms in respiration by measuring lung function parameters of isoform-specific knockout (KO) mice using noninvasive and invasive techniques. Our data revealed the participation and ongoing influence of NO-GC1-derived cGMP in the regulation of airway tone by showing that respiratory resistance was enhanced in NO-GC1-KOs and increased more pronouncedly after the challenge with the bronchoconstrictor methacholine. The tissue resistance and stiffness of NO-GC1-KOs were also higher because of narrowed airways that cause tissue distortion. Contrariwise, NO-GC2-KOs displayed reduced tissue elasticity, elastic recoil, and airway reactivity to methacholine, which did not even increase in an ovalbumin model of asthma that induced hyperresponsiveness in NO-GC1-KOs. In addition, conscious NO-GC2-KOs showed a higher breathing rate with a shorter duration of inspiration and expiration time, which remained faster even in the presence of bronchoconstrictors that slow down breathing. Thus, we provide evidence of two distinct NO/cGMP pathways in airways, accomplished by either NO-GC1 or NO-GC2, adjusting differentially the airway reactivity.


Assuntos
Broncoconstritores , Guanilato Ciclase , Animais , GMP Cíclico/metabolismo , Guanilato Ciclase/metabolismo , Heme , Cloreto de Metacolina/farmacologia , Camundongos , Camundongos Knockout , Óxido Nítrico/metabolismo , Ovalbumina , Isoformas de Proteínas/metabolismo , Guanilil Ciclase Solúvel/metabolismo , Vasodilatadores
2.
Eur J Neurosci ; 55(1): 18-31, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34902209

RESUMO

In the central nervous system, the nitric oxide (NO)/cyclic guanosine monophosphate (cGMP) signalling cascade has an established role in fine-tuning of synaptic transmission. In the present study, we asked which isoform of NO-sensitive guanylyl cyclase, NO-GC1 or NO-GC2, is responsible for generation of N-methyl-d-aspartate (NMDA)- and AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid)-induced cGMP signals and which of the phosphodiesterases (PDEs) is responsible for degradation. To this end, we performed live cell fluorescence measurements of primary hippocampal neurons isolated from NO-GC isoform-deficient mice. Although both isoforms contributed to the NMDA- and AMPA-induced cGMP signals, NO-GC2 clearly played the predominant role. Whereas under PDE-inhibiting conditions the cGMP levels elicited by both glutamatergic ligands were comparable, NMDA-induced cGMP signals were clearly higher than the AMPA-induced ones in the absence of PDE inhibitors. Thus, AMPA-induced cGMP signals are more tightly controlled by PDE-mediated degradation than NMDA-induced signals. In addition, these findings are compatible with the existence of at least two different pools of cGMP in both of which PDE1 and PDE2-known to be highly expressed in the hippocampus-are mainly responsible for cGMP degradation. The finding that distinct pools of cGMP are equipped with different amounts of PDEs highlights the importance of PDEs for the shape of NO-induced cGMP signals in the central nervous system.


Assuntos
N-Metilaspartato , Óxido Nítrico , Animais , GMP Cíclico/metabolismo , Hipocampo/metabolismo , Camundongos , N-Metilaspartato/farmacologia , Óxido Nítrico/metabolismo , Diester Fosfórico Hidrolases/metabolismo , Isoformas de Proteínas/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/metabolismo
3.
Cereb Cortex ; 30(4): 2128-2143, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-31711126

RESUMO

The nitric oxide (NO)/cGMP signaling cascade has an established role in synaptic plasticity. However, with conventional methods, the underlying cGMP signals were barely detectable. Here, we set out to confirm the well-known NMDA-induced cGMP increases, to test the impact of AMPA on those signals, and to identify the relevant phosphodiesterases (PDEs) using a more sensitive fluorescence resonance energy transfer (FRET)-based method. Therefore, a "knock-in" mouse was generated that expresses a FRET-based cGMP indicator (cGi-500) allowing detection of cGMP concentrations between 100 nM and 3 µM. Measurements were performed in cultured hippocampal and cortical neurons as well as acute hippocampal slices. In hippocampal and cortical neurons, NMDA elicited cGMP signals half as high as the ones elicited by exogenous NO. Interestingly, AMPA increased cGMP independently of NMDA receptors and dependent on NO synthase (NOS) activation. NMDA- and AMPA-induced cGMP signals were not additive indicating that both pathways converge on the level of NOS. Accordingly, the same PDEs, PDE1 and PDE2, were responsible for degradation of NMDA- as well as AMPA-induced cGMP signals. Mechanistically, AMPAR induced calcium influx through L-type voltage-gated calcium channels leading to NOS and finally NO-sensitive guanylyl cyclase activation. Our results demonstrate that in addition to NMDA also AMPA triggers endogenous NO formation and hence cGMP production.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Córtex Cerebral/metabolismo , GMP Cíclico/metabolismo , Hipocampo/metabolismo , Óxido Nítrico/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia , Animais , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Técnicas de Cultura de Órgãos
4.
Nature ; 504(7480): 432-6, 2013 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-24213632

RESUMO

Myocardial infarction, a leading cause of death in the Western world, usually occurs when the fibrous cap overlying an atherosclerotic plaque in a coronary artery ruptures. The resulting exposure of blood to the atherosclerotic material then triggers thrombus formation, which occludes the artery. The importance of genetic predisposition to coronary artery disease and myocardial infarction is best documented by the predictive value of a positive family history. Next-generation sequencing in families with several affected individuals has revolutionized mutation identification. Here we report the segregation of two private, heterozygous mutations in two functionally related genes, GUCY1A3 (p.Leu163Phefs*24) and CCT7 (p.Ser525Leu), in an extended myocardial infarction family. GUCY1A3 encodes the α1 subunit of soluble guanylyl cyclase (α1-sGC), and CCT7 encodes CCTη, a member of the tailless complex polypeptide 1 ring complex, which, among other functions, stabilizes soluble guanylyl cyclase. After stimulation with nitric oxide, soluble guanylyl cyclase generates cGMP, which induces vasodilation and inhibits platelet activation. We demonstrate in vitro that mutations in both GUCY1A3 and CCT7 severely reduce α1-sGC as well as ß1-sGC protein content, and impair soluble guanylyl cyclase activity. Moreover, platelets from digenic mutation carriers contained less soluble guanylyl cyclase protein and consequently displayed reduced nitric-oxide-induced cGMP formation. Mice deficient in α1-sGC protein displayed accelerated thrombus formation in the microcirculation after local trauma. Starting with a severely affected family, we have identified a link between impaired soluble-guanylyl-cyclase-dependent nitric oxide signalling and myocardial infarction risk, possibly through accelerated thrombus formation. Reversing this defect may provide a new therapeutic target for reducing the risk of myocardial infarction.


Assuntos
Suscetibilidade a Doenças/metabolismo , Infarto do Miocárdio/metabolismo , Óxido Nítrico/metabolismo , Transdução de Sinais , Animais , Chaperonina com TCP-1/genética , Chaperonina com TCP-1/metabolismo , GMP Cíclico/metabolismo , Exoma/genética , Feminino , Predisposição Genética para Doença , Guanilato Ciclase/deficiência , Guanilato Ciclase/genética , Guanilato Ciclase/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos , Mutação/genética , Infarto do Miocárdio/genética , Infarto do Miocárdio/fisiopatologia , Linhagem , Ativação Plaquetária , Receptores Citoplasmáticos e Nucleares/deficiência , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Reprodutibilidade dos Testes , Solubilidade , Guanilil Ciclase Solúvel , Trombose/metabolismo , Vasodilatação
5.
Mol Pharmacol ; 93(2): 73-78, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29138269

RESUMO

Belonging to the class of so-called soluble guanylate cyclase (sGC) activators, cinaciguat and BAY 60-2770 are interesting therapeutic tools for the treatment of various cardiovascular pathologies. The drugs are supposed to preferentially stimulate oxidized or heme-depleted, but not native sGC. Since this concept has been challenged by studies demonstrating complete relaxation of nondiseased vessels, this study was designed to reinvestigate the mode of action in greater detail. To this purpose, the effect of cinaciguat was studied on vessel tone of porcine coronary arteries and rat thoracic aortas. Organ bath studies showed that the compound caused time- and concentration-dependent relaxation of precontracted vessels with a maximal effect observed at 90 minutes. The dilatory response was not affected by extensive washout of the drug. Cinaciguat-induced vasodilation was associated with a time- and concentration-dependent increase of cGMP levels. Experiments with purified sGC in the presence of Tween 20 showed that cinaciguat activates the heme-free enzyme in a concentration-dependent manner with an EC50 value of ∼0.2 µM and maximal cGMP formation at 10 µM. By contrast, the effect of cinaciguat on 1H-[1,2,4]oxadiazolo-[4,3-a]quinoxalin-1-one-oxidized (ferric) sGC was moderate, reaching ∼10%-15% of maximal activity. Dilution experiments of cinaciguat/Tween 20-preincubated sGC revealed the irreversible character of the drug. Assuming a sensitive balance between heme-free, ferric, and nitric oxide-sensitive ferrous sGC in cells and tissues, we propose that cinaciguat by virtue of its irreversible mode of action is capable of shifting this equilibrium toward the heme-free apo-sGC species.


Assuntos
Benzoatos/farmacologia , Inibidores Enzimáticos/farmacologia , Mimetismo Molecular , Protoporfirinas/metabolismo , Guanilil Ciclase Solúvel/antagonistas & inibidores , Vasodilatação/efeitos dos fármacos , Animais , Aorta Torácica/fisiologia , Bovinos , Vasos Coronários/metabolismo , GMP Cíclico/metabolismo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Ativação Enzimática , Estabilidade Enzimática , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Protoporfirinas/química , Ratos Sprague-Dawley , Guanilil Ciclase Solúvel/metabolismo , Suínos , Vasodilatadores/farmacologia
6.
Nitric Oxide ; 77: 44-52, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29684551

RESUMO

The intracellular messenger molecule cGMP has an established function in the regulation of numerous physiological events. Yet for the identification of further biological cGMP-mediated functions, precise information whether a cGMP response exists in a certain cell type or tissue is mandatory. In this review, the techniques to measure cGMP i.e. cGMP-formation, -degradation or levels are outlined and discussed. As a superior method to measure cGMP, the article focusses on FRET-based cGMP indicators, describes the different cGMP indicators and discusses their advantages and drawbacks. Finally, the successful applications of these cGMP indicators to measure cGMP responses in cells and tissues are outlined and summarized. Hopefully, with the availability of the FRET-based cGMP indicators, the knowledge about the cGMP responses in special cells or tissues is going to increase thereby allowing to assess further cGMP-mediated functional responses and possibly to address their pathophysiology with the available guanylyl cyclase activators, stimulators and PDE inhibitors.


Assuntos
GMP Cíclico/metabolismo , Transferência Ressonante de Energia de Fluorescência , Animais , GMP Cíclico/biossíntese , Guanilato Ciclase/metabolismo , Humanos , Óxido Nítrico/metabolismo , Transdução de Sinais
7.
Int J Mol Sci ; 19(8)2018 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-30087260

RESUMO

Impaired NO-cGMP signaling has been linked to several neurological disorders. NO-sensitive guanylyl cyclase (NO-GC), of which two isoforms-NO-GC1 and NO-GC2-are known, represents a promising drug target to increase cGMP in the brain. Drug-like small molecules have been discovered that work synergistically with NO to stimulate NO-GC activity. However, the effects of NO-GC stimulators in the brain are not well understood. In the present study, we used Förster/fluorescence resonance energy transfer (FRET)-based real-time imaging of cGMP in acute brain slices and primary neurons of cGMP sensor mice to comparatively assess the activity of two structurally different NO-GC stimulators, IWP-051 and BAY 41-2272, in the cerebellum, striatum and hippocampus. BAY 41-2272 potentiated an elevation of cGMP induced by the NO donor DEA/NO in all tested brain regions. Interestingly, IWP-051 potentiated DEA/NO-induced cGMP increases in the cerebellum and striatum, but not in the hippocampal CA1 area or primary hippocampal neurons. The brain-region-selective activity of IWP-051 suggested that it might act in a NO-GC isoform-selective manner. Results of mRNA in situ hybridization indicated that the cerebellum and striatum express NO-GC1 and NO-GC2, while the hippocampal CA1 area expresses mainly NO-GC2. IWP-051-potentiated DEA/NO-induced cGMP signals in the striatum of NO-GC2 knockout mice but was ineffective in the striatum of NO-GC1 knockout mice. These results indicate that IWP-051 preferentially stimulates NO-GC1 signaling in brain slices. Interestingly, no evidence for an isoform-specific effect of IWP-051 was observed when the cGMP-forming activity of whole brain homogenates was measured. This apparent discrepancy suggests that the method and conditions of cGMP measurement can influence results with NO-GC stimulators. Nevertheless, it is clear that NO-GC stimulators enhance cGMP signaling in the brain and should be further developed for the treatment of neurological diseases.


Assuntos
Encéfalo/metabolismo , GMP Cíclico/análise , Guanilato Ciclase/metabolismo , Óxido Nítrico/metabolismo , Animais , Células Cultivadas , GMP Cíclico/metabolismo , Transferência Ressonante de Energia de Fluorescência/métodos , Camundongos Knockout , Neuroimagem/métodos , Neurônios , Células de Purkinje
8.
Int J Mol Sci ; 19(4)2018 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-29570672

RESUMO

Nitric oxide (NO) modulates renal blood flow (RBF) and kidney function and is involved in blood pressure (BP) regulation predominantly via stimulation of the NO-sensitive guanylyl cyclase (NO-GC), existing in two isoforms, NO-GC1 and NO-GC2. Here, we used isoform-specific knockout (KO) mice and investigated their contribution to renal hemodynamics under normotensive and angiotensin II-induced hypertensive conditions. Stimulation of the NO-GCs by S-nitrosoglutathione (GSNO) reduced BP in normotensive and hypertensive wildtype (WT) and NO-GC2-KO mice more efficiently than in NO-GC1-KO. NO-induced increase of RBF in normotensive mice did not differ between the genotypes, but the respective increase under hypertensive conditions was impaired in NO-GC1-KO. Similarly, inhibition of endogenous NO increased BP and reduced RBF to a lesser extent in NO-GC1-KO than in NO-GC2-KO. These findings indicate NO-GC1 as a target of NO to normalize RBF in hypertension. As these effects were not completely abolished in NO-GC1-KO and renal cyclic guanosine monophosphate (cGMP) levels were decreased in both NO-GC1-KO and NO-GC2-KO, the results suggest an additional contribution of NO-GC2. Hence, NO-GC1 plays a predominant role in the regulation of BP and RBF, especially in hypertension. However, renal NO-GC2 appears to compensate the loss of NO-GC1, and is able to regulate renal hemodynamics under physiological conditions.


Assuntos
Guanilil Ciclase Solúvel/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , GMP Cíclico/metabolismo , Rim/efeitos dos fármacos , Rim/metabolismo , Camundongos , Camundongos Knockout , NG-Nitroarginina Metil Éster/metabolismo , Óxido Nítrico/metabolismo , Circulação Renal/efeitos dos fármacos , S-Nitrosoglutationa/farmacologia , Vasodilatação/efeitos dos fármacos
9.
Mol Pharmacol ; 92(4): 375-388, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28874607

RESUMO

Nitric oxide (NO) activates the NO-sensitive soluble guanylate cyclase (NO-GC, sGC) and triggers intracellular signaling pathways involving cGMP. For survival of cochlear hair cells and preservation of hearing, NO-mediated cascades have both protective and detrimental potential. Here we examine the cochlear function of mice lacking one of the two NO-sensitive guanylate cyclase isoforms [NO-GC1 knockout (KO) or NO-GC2 KO]. The deletion of NO-GC1 or NO-GC2 did not influence electromechanical outer hair cell (OHC) properties, as measured by distortion product otoacoustic emissions, neither before nor after noise exposure, nor were click- or noise-burst-evoked auditory brainstem response thresholds different from controls. Yet inner hair cell (IHC) ribbons and auditory nerve responses showed significantly less deterioration in NO-GC1 KO and NO-GC2 KO mice after noise exposure. Consistent with a selective role of NO-GC in IHCs, NO-GC ß1 mRNA was found in isolated IHCs but not in OHCs. Using transgenic mice expressing the fluorescence resonance energy transfer-based cGMP biosensor cGi500, NO-induced elevation of cGMP was detected in real-time in IHCs but not in OHCs. Pharmacologic long-term treatment with a NO-GC stimulator altered auditory nerve responses but did not affect OHC function and hearing thresholds. Interestingly, NO-GC stimulation exacerbated the loss of auditory nerve response in aged animals but attenuated the loss in younger animals. We propose NO-GC2 and, to some degree, NO-GC1 as targets for early pharmacologic prevention of auditory fiber loss (synaptopathy). Both isoforms provide selective benefits for hearing function by maintaining the functional integrity of auditory nerve fibers in early life rather than at old age.


Assuntos
Guanilato Ciclase/metabolismo , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Internas/patologia , Óxido Nítrico/metabolismo , Ruído/efeitos adversos , Receptores de Superfície Celular/metabolismo , Animais , Feminino , Células Ciliadas Auditivas Internas/efeitos dos fármacos , Isoenzimas/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Morfolinas/farmacologia , Pirimidinas/farmacologia , Ratos , Ratos Wistar , Receptores de Superfície Celular/agonistas , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Sinapses/patologia
10.
J Biol Chem ; 291(46): 24076-24084, 2016 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-27679490

RESUMO

Aldehyde dehydrogenase-2 (ALDH2) catalyzes vascular bioactivation of the antianginal drug nitroglycerin (GTN), resulting in activation of soluble guanylate cyclase (sGC) and cGMP-mediated vasodilation. We have previously shown that a minor reaction of ALDH2-catalyzed GTN bioconversion, accounting for about 5% of the main clearance-based turnover yielding inorganic nitrite, results in direct NO formation and concluded that this minor pathway could provide the link between vascular GTN metabolism and activation of sGC. However, lack of detectable NO at therapeutically relevant GTN concentrations (≤1 µm) in vascular tissue called into question the biological significance of NO formation by purified ALDH2. We addressed this issue and used a novel, highly sensitive genetically encoded fluorescent NO probe (geNOp) to visualize intracellular NO formation at low GTN concentrations (≤1 µm) in cultured vascular smooth muscle cells (VSMC) expressing an ALDH2 mutant that reduces GTN to NO but lacks clearance-based GTN denitration activity. NO formation was compared with GTN-induced activation of sGC. The addition of 1 µm GTN to VSMC expressing either wild-type or C301S/C303S ALDH2 resulted in pronounced intracellular NO elevation, with maximal concentrations of 7 and 17 nm, respectively. Formation of GTN-derived NO correlated well with activation of purified sGC in VSMC lysates and cGMP accumulation in intact porcine aortic endothelial cells infected with wild-type or mutant ALDH2. Formation of NO and cGMP accumulation were inhibited by ALDH inhibitors chloral hydrate and daidzin. The present study demonstrates that ALDH2-catalyzed NO formation is necessary and sufficient for GTN bioactivation in VSMC.


Assuntos
Aldeído-Desidrogenase Mitocondrial/metabolismo , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Óxido Nítrico/metabolismo , Nitroglicerina/farmacocinética , Aldeído-Desidrogenase Mitocondrial/antagonistas & inibidores , Aldeído-Desidrogenase Mitocondrial/genética , Substituição de Aminoácidos , Animais , Bovinos , Hidrato de Cloral/farmacologia , Humanos , Isoflavonas/farmacologia , Camundongos , Camundongos Knockout , Mutação de Sentido Incorreto , Nitroglicerina/farmacologia , Suínos
11.
Am J Pathol ; 186(8): 2220-2231, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27315776

RESUMO

Soluble guanylate cyclase (sGC), a key enzyme of the nitric oxide signaling pathway, is formed as a heterodimer by various isoforms of its α and ß subunit. GUCY1A3, encoding the α1 subunit, was identified as a risk gene for coronary artery disease and myocardial infarction, but its specific contribution to atherosclerosis remains unclear. This study sought to decipher the role of Gucy1a3 in atherosclerosis in mice. At age 32 weeks and after 20 weeks of standard or high-fat diet, Gucy1a3(-/-)/Ldlr(-/-) mice exhibited a significant reduction of the atherosclerotic plaque size at the aortic root and the aorta for high-fat diet animals as compared with Ldlr(-/-) control mice. Collagen content in plaques in the aortic root was reduced, suggesting an alteration of smooth muscle cell function. Proliferation and migration were reduced in Gucy1a3(-/-) primary aortic smooth muscle cells (AoSMCs), and proliferation was also reduced in human AoSMCs after inhibition of sGC by 1H-[1,2,4] oxadiazolo [4,3-a] quinoxalin-1-one. Gucy1a3 deficiency in AoSMCs prevents their phenotypic switching, as indicated by the differential expression of marker proteins. The inherited Gucy1a3(-/-) loss exerts an atheroprotective effect. We suggest that sGC activity promotes the phenotypic switching of smooth muscle cells from a contractile to a synthetic state, fostering the formation of atherosclerosis. Preventing this switch by sGC inhibition may provide a novel target in atherosclerotic disease.


Assuntos
Aterosclerose/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Guanilil Ciclase Solúvel/metabolismo , Animais , Aterosclerose/genética , Western Blotting , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Dieta Hiperlipídica , Modelos Animais de Doenças , Feminino , Genótipo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Reação em Cadeia da Polimerase em Tempo Real , Guanilil Ciclase Solúvel/genética
12.
J Biol Chem ; 290(19): 11936-47, 2015 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-25762721

RESUMO

Phosphodiesterase 10A (PDE10A) is a dual substrate PDE that can hydrolyze both cGMP and cAMP. In brain, PDE10A is almost exclusively expressed in the striatum. In several studies, PDE10A has been implicated in regulation of striatal output using either specific inhibitors or PDE10A knock-out mice and has been suggested as a promising target for novel antipsychotic drugs. In striatal medium spiny neurons, PDE10A is localized at the plasma membrane and in dendritic spines close to postsynaptic densities. In the present study, we identify PDE10A as the major cAMP PDE in mouse striatum and monitor PKA-dependent PDE10A phosphorylation. With recombinantly expressed PDE10A we demonstrate that phosphorylation does not alter PDE10A activity. In striatum, PDE10A was found to be associated with the A kinase anchoring protein AKAP150 suggesting the existence of a multiprotein signaling complex localizing PDE10A to a specific functional context at synaptic membranes. Furthermore, the cAMP effector PKA, the NMDA receptor subunits NR2A and -B, as well as PSD95, were tethered to the complex. In agreement, PDE10A was almost exclusively found in multiprotein complexes as indicated by migration in high molecular weight fractions in size exclusion chromatography. Finally, affinity of PDE10A to the signaling complexes formed around AKAP150 was reduced by PDE10A phosphorylation. The data indicate that phosphorylation of PDE10 has an impact on the interaction with other signaling proteins and adds an additional line of complexity to the role of PDE10 in regulation of synaptic transmission.


Assuntos
Corpo Estriado/metabolismo , AMP Cíclico/metabolismo , Regulação Enzimológica da Expressão Gênica , Guanilato Quinases/metabolismo , Proteínas de Membrana/metabolismo , Diester Fosfórico Hidrolases/metabolismo , Animais , Cálcio/química , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteína 4 Homóloga a Disks-Large , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Knockout , Fosforilação , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais
13.
J Pharmacol Exp Ther ; 356(1): 191-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26559126

RESUMO

In the regulation of vascular tone, the dilatory nitric oxide (NO)/cGMP pathway balances vasoconstriction induced by the renin-angiotensin and sympathetic nervous systems. NO-induced cGMP formation is catalyzed by two guanylyl cyclases (GC), NO-sensitive guanylyl cyclase 1 (NO-GC1) and NO-GC2, with indistinguishable enzymatic properties. In vascular smooth muscle cells, NO-GC1 is the major isoform and is responsible for more than 90% of cGMP formation. Despite reduced vasorelaxation, NO-GC1-deficient mice are not hypertensive. Here, the role of NO-GC1 in hypertension provoked by contractile agonists angiotensin II (Ang II) and norepinephrine (NE) was evaluated in NO-GC1-deficient mice. Hypertension induced by chronic Ang II treatment did not differ between wild-type (WT) and NO-GC1 knockout mice (KO). Also, attenuation of NO-dependent aortic relaxation induced by the Ang II treatment was similar in both genotypes and was most probably attributable to an increase of phosphodiesterase 1 expression. Analysis of plasma NE content-known to be influenced by Ang II-revealed lower NE in the NO-GC1 KO under Ang II-treated- and nontreated conditions. The finding indicates reduced sympathetic output and is underlined by the lower heart rate in the NO-GC1 KO. To find out whether the lack of higher blood pressure in the NO-GC1 KO is a result of reduced sympathetic activity counterbalancing the reduced vascular relaxation, mice were challenged with chronic NE application. As the resulting blood pressure was higher in the NO-GC1 KO than in WT, we conclude that the reduced sympathetic activity in the NO-GC1 KO prevents hypertension and postulate a possible sympatho-excitatory action of NO-GC1 counteracting NO-GC1's dilatory effect in the vasculature.


Assuntos
Angiotensina II , Guanilato Ciclase/fisiologia , Hipertensão/induzido quimicamente , Hipertensão/tratamento farmacológico , Receptores de Superfície Celular/fisiologia , Sistema Nervoso Simpático/fisiopatologia , Vasoconstritores , Animais , Pressão Sanguínea/genética , GMP Cíclico/metabolismo , Guanilato Ciclase/genética , Frequência Cardíaca/genética , Hipertensão/genética , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/efeitos dos fármacos , Norepinefrina/farmacologia , Diester Fosfórico Hidrolases/biossíntese , Receptores de Superfície Celular/genética , Vasodilatação/efeitos dos fármacos
14.
Basic Res Cardiol ; 111(4): 51, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27342234

RESUMO

Stimulators of the soluble guanylyl cyclase (sGC) are emerging therapeutic agents in cardiovascular diseases. Genetic alterations of the GUCY1A3 gene, which encodes the α1 subunit of the sGC, are associated with coronary artery disease. Studies investigating sGC stimulators in subjects with CAD and carrying risk-related variants in sGC are, however, lacking. Here, we functionally investigate the impact of coding GUCY1A3 variants on sGC activity and the therapeutic potential of sGC stimulators in vitro. In addition to a known loss-of-function variant, eight coding variants in GUCY1A3 were cloned and expressed in HEK 293 cells. Protein levels and dimerization capability with the ß1 subunit were analysed by immunoblotting and co-immunoprecipitation, respectively. All α1 variants found in MI patients dimerized with the ß1 subunit. Protein levels were reduced by 72 % in one variant (p < 0.01). Enzymatic activity was analysed using cGMP radioimmunoassay after stimulation with a nitric oxide (NO) donor. Five variants displayed decreased cGMP production upon NO stimulation (p < 0.001). The addition of the sGC stimulator BAY 41-2272 increased cGMP formation in all of these variants (p < 0.01). Except for the variant leading to decreased protein level, cGMP amounts reached the wildtype NO-induced level after addition of BAY 41-2272. In conclusion, rare coding variants in GUCY1A3 lead to reduced cGMP formation which can be rescued by a sGC stimulator in vitro. These results might therefore represent the starting point for discovery of novel treatment strategies for patients at risk with coding GUCY1A3 variants.


Assuntos
Doença da Artéria Coronariana/genética , GMP Cíclico/biossíntese , Guanilil Ciclase Solúvel/genética , Adulto , Animais , GMP Cíclico/genética , Predisposição Genética para Doença/genética , Células HEK293 , Humanos , Immunoblotting , Imunoprecipitação , Camundongos , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Reação em Cadeia da Polimerase , Polimorfismo de Nucleotídeo Único , Pirazóis/farmacologia , Piridinas/farmacologia , Radioimunoensaio , Guanilil Ciclase Solúvel/metabolismo , Adulto Jovem
15.
Nitric Oxide ; 54: 8-14, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26805578

RESUMO

Scavenging of nitric oxide (NO) often interferes with studies on NO signaling in cell-free preparations. We observed that formation of cGMP by NO-stimulated purified soluble guanylate cyclase (sGC) was virtually abolished in the presence of cytosolic preparations of porcine coronary arteries, with the scavenging activity localized in the tunica media (smooth muscle layer). Electrochemical measurement of NO release from a donor compound and light absorbance spectroscopy showed that cytosolic preparations contained a reduced heme protein that scavenged NO. This protein, which reacted with anti-human hemoglobin antibodies, was efficiently removed from the preparations by haptoglobin affinity chromatography. The cleared cytosols showed only minor scavenging of NO according to electrochemical measurements and did not decrease cGMP formation by NO-stimulated sGC. In contrast, the column flow-through caused a nearly 2-fold increase of maximal sGC activity (from 33.1 ± 1.6 to 54.9 ± 2.2 µmol × min(-1) × mg(-1)). The proteins retained on the affinity column were identified as hemoglobin α and ß subunits. The results indicate that hemoglobin, presumably derived from vasa vasorum erythrocytes, is present and scavenges NO in preparations of porcine coronary artery smooth muscle. Selective removal of hemoglobin-mediated scavenging unmasked stimulation of maximal NO-stimulated sGC activity by a soluble factor expressed in vascular tissue.


Assuntos
Vasos Coronários/metabolismo , Hemoglobinas/metabolismo , Óxido Nítrico/metabolismo , Túnica Média/metabolismo , Animais , Bovinos , GMP Cíclico/metabolismo , Citoglobina , Globinas/metabolismo , Haptoglobinas/metabolismo , Humanos , Técnicas In Vitro , Guanilil Ciclase Solúvel/metabolismo , Suínos
16.
Arterioscler Thromb Vasc Biol ; 35(9): 2011-9, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26205960

RESUMO

OBJECTIVE: In the vascular system, cyclic GMP (cGMP) in smooth muscle cells plays an important role for blood vessel relaxation. Intracellular concentrations of cGMP are thought to be determined by the action of cGMP-generating guanylyl cyclases (sensitive to nitric oxide or natriuretic peptides) and cGMP-degrading phosphodiesterases (PDE1, PDE3, and PDE5). Because functionally relevant cGMP elevations are not accessible to conventional methods, we applied real-time imaging with a fluorescence resonance energy transfer (FRET)-based cGMP indicator to follow nitric oxide- and natriuretic peptide-induced cGMP signals in living smooth muscle cells and analyzed the contribution of the miscellaneous cGMP-generating and cGMP-degrading enzymes. APPROACH AND RESULTS: By comparison of cGMP signals in living smooth muscle cells and vascular relaxation of aortic strips in organ bath experiments, we show for the first time that FRET-based cGMP indicators permit the measurement of functionally relevant cGMP signals. PDE5 was the major cGMP phosphodiesterase responsible for reducing nitric oxide- and natriuretic peptide-induced cGMP signals. In contrast, PDE3-involved in the degradation of lower cGMP concentrations-displayed a preference for natriuretic peptide-stimulated cGMP. Unexpectedly, we found that cGMP is transported out of the cells by the ABC transporter multidrug resistance-associated protein 4 and this export turned out to be of similar importance for intracellular cGMP signals as degradation by PDE5. Functionally, inhibition of cGMP export enhanced vascular relaxation as much as inhibition of PDE5. CONCLUSIONS: The findings indicate that cGMP export out of smooth muscle cells is a key player in the regulation of smooth muscle cGMP signals and blood vessel relaxation.


Assuntos
Aorta Torácica/fisiologia , GMP Cíclico/biossíntese , Músculo Liso Vascular/fisiologia , Vasodilatação/fisiologia , Animais , Aorta Torácica/citologia , Células Cultivadas , Líquido Intracelular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/citologia , Radioimunoensaio , Transdução de Sinais
17.
Cereb Cortex ; 24(7): 1923-36, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23448871

RESUMO

The nitric oxide (NO)/cyclic guanosine monophosphate (cGMP) signaling cascade participates in the modulation of synaptic transmission. The effects of NO are mediated by the NO-sensitive cGMP-forming guanylyl cyclases (NO-GCs), which exist in 2 isoforms with indistinguishable regulatory properties. The lack of long-term potentiation (LTP) in knock-out (KO) mice deficient in either one of the NO-GC isoforms indicates the contribution of both NO-GCs to LTP. Recently, we showed that the NO-GC1 isoform is located presynaptically in glutamatergic neurons and increases the glutamate release via hyperpolarization-activated cyclic nucleotide (HCN)-gated channels in the hippocampus. Electrophysiological analysis of hippocampal CA1 neurons in whole-cell recordings revealed a reduction of HCN currents and a hyperpolarizing shift of the activation curve in the NO-GC2 KOs associated with reduced resting membrane potentials. These features were mimicked in wild-type (WT) neurons with an NO-GC inhibitor. Analysis of glutamate receptors revealed a cGMP-dependent reduction of NMDA receptor currents in the NO-GC2 KO mice, which was mimicked in WT by HCN channel inhibition. Lowering extracellular Mg(2+) increased NMDA receptor currents in the NO-GC2 KO and allowed the induction of LTP that was absent at physiological Mg(2+). In sum, our data indicate that postsynaptic cGMP increases the N-methyl-D-aspartate (NMDA) receptor current by gating HCN channels and thereby is required for LTP.


Assuntos
Região CA1 Hipocampal/citologia , GMP Cíclico/metabolismo , Potenciação de Longa Duração/fisiologia , Neurônios/fisiologia , Óxido Nítrico/deficiência , Receptores de N-Metil-D-Aspartato/metabolismo , Anestésicos Locais/farmacologia , Animais , Animais Recém-Nascidos , GMP Cíclico/farmacologia , Estimulação Elétrica , Antagonistas de Aminoácidos Excitatórios/farmacologia , Guanilato Ciclase/genética , Guanilato Ciclase/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Técnicas In Vitro , Lidocaína/análogos & derivados , Lidocaína/farmacologia , Potenciação de Longa Duração/efeitos dos fármacos , Camundongos , Camundongos Knockout , Neurônios/efeitos dos fármacos , Óxido Nítrico/genética , Técnicas de Patch-Clamp , Pirimidinas/farmacologia , Tetraetilamônio/farmacologia
18.
Mol Pharmacol ; 84(3): 407-14, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23793290

RESUMO

Aldehyde dehydrogenase-2 (ALDH2) catalyzes vascular bioactivation of the antianginal drug nitroglycerin (GTN) to yield nitric oxide (NO) or a related species that activates soluble guanylate cyclase (sGC), resulting in cGMP-mediated vasodilation. Accordingly, established ALDH2 inhibitors attenuate GTN-induced vasorelaxation in vitro and in vivo. However, the ALDH2 hypothesis has not been reconciled with early studies demonstrating potent inhibition of the GTN response by diphenyleneiodonium (DPI), a widely used inhibitor of flavoproteins, in particular NADPH oxidases. We addressed this issue and investigated the effects of DPI on GTN-induced relaxation of rat aortic rings and the function of purified ALDH2. DPI (0.3 µM) inhibited the high affinity component of aortic relaxation to GTN without affecting the response to NO, indicating that the drug interfered with GTN bioactivation. Denitration and bioactivation of 1-2 µM GTN, assayed as 1,2-glycerol dinitrate formation and activation of purified sGC, respectively, were inhibited by DPI with a half-maximally active concentration of about 0.2 µM in a GTN-competitive manner. Molecular modeling indicated that DPI binds to the catalytic site of ALDH2, and this was confirmed by experiments showing substrate-competitive inhibition of the dehydrogenase and esterase activities of the enzyme. Our data identify ALDH2 as highly sensitive target of DPI and explain inhibition of GTN-induced relaxation by this drug observed previously. In addition, the data provide new evidence for the essential role of ALDH2 in GTN bioactivation and may have implications to other fields of ALDH2 research, such as hepatic ethanol metabolism and cardiac ischemia/reperfusion injury.


Assuntos
Aldeído Desidrogenase/antagonistas & inibidores , Proteínas Mitocondriais/antagonistas & inibidores , Nitroglicerina/metabolismo , Oniocompostos/farmacologia , Vasodilatadores/metabolismo , Aldeído Desidrogenase/química , Aldeído-Desidrogenase Mitocondrial , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/fisiologia , Domínio Catalítico , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Feminino , Humanos , Técnicas In Vitro , Masculino , Simulação de Acoplamento Molecular , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiologia , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Suínos , Vasodilatação/efeitos dos fármacos
19.
J Biol Chem ; 287(2): 1210-9, 2012 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-22105073

RESUMO

The most recently identified cyclic nucleotide phosphodiesterases, PDE10 and PDE11, contain a tandem of so-called GAF domains in their N-terminal regulatory regions. In PDE2 and PDE5, the GAF domains mediate cGMP stimulation; however, their function in PDE10 and PDE11 remains controversial. Although the GAF domains of PDE10 mediate cAMP-induced stimulation of chimeric adenylyl cyclases, cAMP binding did not stimulate the PDE10 holoenzyme. Comparable data about cGMP and the PDE11 GAF domains exist. Here, we identified synthetic ligands for the GAF domains of PDE10 and PDE11 to reduce interference of the GAF ligand with the catalytic reaction of PDE. With these ligands, GAF-mediated stimulation of the PDE10 and PDE11 holoenzymes is demonstrated for the first time. Furthermore, PDE10 is shown to be activated by cAMP, which paradoxically results in potent competitive inhibition of cGMP turnover by cAMP. PDE11, albeit susceptible to GAF-dependent stimulation, is not activated by the native cyclic nucleotides cAMP and cGMP. In summary, PDE11 can be stimulated by GAF domain ligands, but its native ligand remains to be identified, and PDE10 is the only PDE activated by cAMP.


Assuntos
AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Diester Fosfórico Hidrolases/metabolismo , AMP Cíclico/genética , GMP Cíclico/genética , Ativação Enzimática/fisiologia , Células HEK293 , Holoenzimas/genética , Holoenzimas/metabolismo , Humanos , Diester Fosfórico Hidrolases/genética , Estrutura Terciária de Proteína
20.
Sci Rep ; 13(1): 15833, 2023 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-37739972

RESUMO

Chronic smoking causes dysfunction of vascular endothelial cells, evident as a reduction of flow-mediated dilation in smokers, but the role of nicotine is still controversial. Given the increasing use of e-cigarettes and other nicotine products, it appears essential to clarify this issue. We studied extracts from cigarette smoke (CSE) and vapor from e-cigarettes (EVE) and heated tobacco (HTE) for their effects on vascular relaxation, endothelial nitric oxide signaling, and the activity of soluble guanylyl cyclase. The average nicotine concentrations of CSE, EVE, and HTE were 164, 800, and 85 µM, respectively. At a dilution of 1:3, CSE almost entirely inhibited the relaxation of rat aortas and porcine coronary arteries to acetylcholine and bradykinin, respectively, while undiluted EVE, with a 15-fold higher nicotine concentration, had no significant effect. With about 50% inhibition at 1:2 dilution, the effect of HTE was between CSE and EVE. Neither extract affected endothelium-independent relaxation to an NO donor. At the dilutions tested, CSE was not toxic to cultured endothelial cells but, in contrast to EVE, impaired NO signaling and inhibited NO stimulation of soluble guanylyl cyclase. Our results demonstrate that nicotine does not mediate the impaired endothelium-dependent vascular relaxation caused by smoking.


Assuntos
Vapor do Cigarro Eletrônico , Sistemas Eletrônicos de Liberação de Nicotina , Poluição por Fumaça de Tabaco , Ratos , Animais , Suínos , Nicotina/farmacologia , Células Endoteliais , Óxido Nítrico , Guanilil Ciclase Solúvel , Endotélio
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa