Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nitric Oxide ; 113-114: 23-30, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-33915274

RESUMO

PnPP-19 peptide has a primary sequence design based on molecular modeling studies of PnTx2-6 toxin. It comprises the amino acid residues that are potentially significant for the pharmacological action of PnTx2-6. Ex vivo and in vivo experiments in normotensive, hypertensive, or diabetic murine models have shown a significant improvement in penile erection after administration of PnPP-19. Given the potential use of PnPP-19 in pharmaceutical formulations to treat erectile dysfunction and the lack of information concerning its mode of action, the present work investigates its activities on the nitrergic system. PnPP-19 induced a significant increase in nitric oxide (NO) and cGMP levels in corpus cavernosum (cc). These effects were inhibited by l-NAME, a non-selective inhibitor of nitric oxide synthase (NOS); were partially inhibited by 7- Nitroindazole, a selective inhibitor of neuronal NOS (nNOS); and were abolished by L-NIL, a selective inhibitor of inducible NOS (iNOS). This potentiating effect was not affected by atropine. PnPP-19 also led to changes in mRNA levels, protein expression and phosphorylation at specific sites of NOS, in cc. Assays using cavernous tissue from knockout mice to endothelial NOS (eNOS), nNOS or iNOS showed that PnPP-19 potentiates relaxation only in eNOS-knockout mice, which suggests an essential role for nNOS. Surprisingly, iNOS enhanced the potentiation of erectile function evoked by PnPP-19. Our results demonstrate that this new synthetic peptide potentiates erectile function via nitric oxide activation and reinforce its role as a new pharmacological tool for the treatment of erectile dysfunction.


Assuntos
Disfunção Erétil/tratamento farmacológico , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Peptídeos/farmacologia , Animais , Biologia Computacional , Disfunção Erétil/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo I/deficiência , Óxido Nítrico Sintase Tipo I/genética , Óxido Nítrico Sintase Tipo II/deficiência , Óxido Nítrico Sintase Tipo II/genética , Peptídeos/síntese química , Peptídeos/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
2.
Am J Physiol Renal Physiol ; 317(3): F540-F546, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31215803

RESUMO

Voiding abnormalities are common among the sickle cell disease (SCD) population, among which overactive bladder (OAB) syndrome is observed at rates as high as 39%. Although detrusor overactivity is the most common cause of OAB, its molecular pathophysiology is not well elucidated. The nitric oxide (NO) signaling pathway has been implicated in the regulation of lower genitourinary tract function. In the present study, we evaluated the role of the NO signaling pathway in voiding function of transgenic SCD mice compared with combined endothelial and neuronal NO synthase gene-deficient mice, both serving as models of NO deficiency. Mice underwent void spot assay and cystometry, and bladder and urethral specimens were studied using in vitro tissue myography. Both mouse models exhibited increased void volumes; increased nonvoiding and voiding contraction frequencies; decreased bladder compliance; increased detrusor smooth muscle contraction responses to electrical field stimulation, KCl, and carbachol; and increased urethral smooth muscle relaxation responses to sodium nitroprusside compared with WT mice. In conclusion, our comprehensive behavioral and functional study of the SCD mouse lower genitourinary tract, in correlation with that of the NO-deficient mouse, reveals NO effector actions in voiding function and suggests that NO signaling derangements are associated with an OAB phenotype. These findings may allow further study of molecular targets for the characterization and evaluation of OAB.


Assuntos
Anemia Falciforme/complicações , Óxido Nítrico/metabolismo , Bexiga Urinária Hiperativa/etiologia , Bexiga Urinária/metabolismo , Urodinâmica , Anemia Falciforme/genética , Animais , Modelos Animais de Doenças , Hemoglobina A/genética , Hemoglobina A/metabolismo , Hemoglobinas/genética , Hemoglobinas/metabolismo , Humanos , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Muscular , Relaxamento Muscular , Óxido Nítrico Sintase Tipo I/deficiência , Óxido Nítrico Sintase Tipo I/genética , Óxido Nítrico Sintase Tipo III/deficiência , Óxido Nítrico Sintase Tipo III/genética , Transdução de Sinais , Bexiga Urinária/fisiopatologia , Bexiga Urinária Hiperativa/metabolismo , Bexiga Urinária Hiperativa/fisiopatologia
3.
Am J Physiol Renal Physiol ; 317(3): F547-F559, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31241990

RESUMO

The collecting duct (CD) concentrates the urine, thereby maintaining body water volume and plasma osmolality within a normal range. The endocrine hormone arginine vasopressin acts in the CD to increase water permeability via the vasopressin 2 receptor (V2R)-aquaporin (AQP) axis. Recent studies have suggested that autocrine factors may also contribute to the regulation of CD water permeability. Nitric oxide is produced predominantly by nitric oxide synthase 1 (NOS1) in the CD and acts as a diuretic during salt loading. The present study sought to determine whether CD NOS1 regulates diuresis during changes in hydration status. Male and female control and CD NOS1 knockout (CDNOS1KO) mice were hydrated (5% sucrose water), water deprived, or acutely challenged with the V2R agonist desmopressin. In male mice, water deprivation resulted in decreased urine flow and increased plasma osmolality, copeptin concentration, and kidney AQP2 abundance independent of CD NOS1. In female control mice, water deprivation reduced urine flow, increased plasma osmolality and copeptin, but did not significantly change total AQP2; however, there was increased basolateral AQP3 localization. Surprisingly, female CDNOS1KO mice while on the sucrose water presented with symptoms of dehydration. Fibroblast growth factor 21, an endocrine regulator of sweetness preference, was significantly higher in female CDNOS1KO mice, suggesting that this was reducing their drive to drink the sucrose water. With acute desmopressin challenge, female CDNOS1KO mice failed to appropriately concentrate their urine, resulting in higher plasma osmolality than controls. In conclusion, CD NOS1 plays only a minor role in urine-concentrating mechanisms.


Assuntos
Desidratação/enzimologia , Diurese , Capacidade de Concentração Renal , Túbulos Renais Coletores/enzimologia , Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico/metabolismo , Animais , Antidiuréticos/farmacologia , Aquaporina 2/genética , Aquaporina 2/metabolismo , Aquaporina 3/genética , Aquaporina 3/metabolismo , Desamino Arginina Vasopressina/farmacologia , Desidratação/fisiopatologia , Modelos Animais de Doenças , Diurese/efeitos dos fármacos , Feminino , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Capacidade de Concentração Renal/efeitos dos fármacos , Túbulos Renais Coletores/efeitos dos fármacos , Masculino , Camundongos Knockout , Óxido Nítrico Sintase Tipo I/deficiência , Óxido Nítrico Sintase Tipo I/genética , Estado de Hidratação do Organismo , Concentração Osmolar , Fatores Sexuais , Transdução de Sinais , Urodinâmica , Privação de Água
4.
J Proteome Res ; 17(3): 1031-1040, 2018 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-29394072

RESUMO

Priapism is a disorder in which prolonged penile erection persists uncontrollably, potentially leading to tissue damage. Priapism commonly afflicts patient populations with severely low nitric oxide (NO) bioavailability. Because NO is a primary mediator of erection, the molecular mechanisms involved in priapism pathophysiology associated with low NO bioavailability are not well-understood. The objective of this study was to identify dysregulated molecular targets and signaling pathways in penile tissue of a mouse model of low NO bioavailability that have potential relevance to priapism. Neuronal plus endothelial NO synthase double knockout mice (NOS1/3-/-) were used as a model of low NO bioavailability. Priapic-like activity was demonstrated in the NOS1/3-/- mice relative to wild-type (WT) mice by the measurement of prolonged erections following cessation of electrical stimulation of the cavernous nerve. Penile tissue was processed and analyzed by reverse-phase liquid chromatography tandem mass spectrometry. As a result, 1279 total proteins were identified and quantified by spectral counting, 46 of which were down-regulated and 110 of which were up-regulated in NOS1/3-/- versus WT (P < 0.05). Ingenuity Pathway Analysis of differentially expressed proteins revealed increased protein kinase A and G-protein coupled receptor signaling in NOS1/3-/- penises, which represent potential mechanisms contributing to priapism for secondary to low NO bioavailability.


Assuntos
Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo I/genética , Óxido Nítrico/metabolismo , Pênis/metabolismo , Priapismo/genética , Animais , Cromatografia de Fase Reversa , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Modelos Animais de Doenças , Estimulação Elétrica , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Regulação da Expressão Gênica , Ontologia Genética , Humanos , Masculino , Camundongos , Camundongos Knockout , Anotação de Sequência Molecular , Neurônios/metabolismo , Neurônios/patologia , Óxido Nítrico Sintase Tipo I/deficiência , Óxido Nítrico Sintase Tipo III/deficiência , Ereção Peniana/fisiologia , Pênis/irrigação sanguínea , Pênis/inervação , Priapismo/metabolismo , Priapismo/patologia , Priapismo/fisiopatologia , Proteoma/genética , Proteoma/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Nervos Esplâncnicos/metabolismo , Nervos Esplâncnicos/fisiopatologia , Espectrometria de Massas em Tandem
5.
Alcohol Clin Exp Res ; 42(9): 1627-1639, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29957842

RESUMO

BACKGROUND: Alcohol exposure during pregnancy can kill developing neurons and lead to fetal alcohol spectrum disorder (FASD). However, affected individuals differ in their regional patterns of alcohol-induced neuropathology. Because neuroprotective genes are expressed in spatially selective ways, their mutation could increase the vulnerability of some brain regions, but not others, to alcohol teratogenicity. The objective of this study was to determine whether a null mutation of neuronal nitric oxide synthase (nNOS) can increase the vulnerability of some brain regions, but not others, to alcohol-induced neuronal losses. METHODS: Immunohistochemistry identified brain regions in which nNOS is present or absent throughout postnatal development. Mice genetically deficient for nNOS (nNOS-/- ) and wild-type controls received alcohol (0.0, 2.2, or 4.4 mg/g/d) over postnatal days (PD) 4 to 9. Mice were sacrificed in adulthood (~PD 115), and surviving neurons in the olfactory bulb granular layer and brain stem facial nucleus were quantified stereologically. RESULTS: nNOS was expressed throughout postnatal development in olfactory bulb granule cells but was never expressed in the facial nucleus. In wild-type mice, alcohol reduced neuronal survival to similar degrees in both cell populations. However, null mutation of nNOS more than doubled alcohol-induced cell death in the olfactory bulb granule cells, while the mutation had no effect on the facial nucleus neurons. As a result, in nNOS-/- mice, alcohol caused substantially more cell loss in the olfactory bulb than in the facial nucleus. CONCLUSIONS: Mutation of the nNOS gene substantially increases vulnerability to alcohol-induced cell loss in a brain region where the gene is expressed (olfactory bulb), but not in a separate brain region, where the gene is not expressed (facial nucleus). Thus, differences in genotype may explain why some individuals are vulnerable to FASD, while others are not, and may determine the specific patterns of neuropathology in children with FASD.


Assuntos
Consumo de Bebidas Alcoólicas/genética , Etanol/toxicidade , Transtornos do Espectro Alcoólico Fetal/genética , Neurônios/efeitos dos fármacos , Óxido Nítrico Sintase Tipo I/genética , Bulbo Olfatório/efeitos dos fármacos , Consumo de Bebidas Alcoólicas/efeitos adversos , Consumo de Bebidas Alcoólicas/metabolismo , Consumo de Bebidas Alcoólicas/patologia , Animais , Animais Recém-Nascidos , Feminino , Transtornos do Espectro Alcoólico Fetal/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Neurônios/patologia , Óxido Nítrico Sintase Tipo I/deficiência , Bulbo Olfatório/metabolismo , Bulbo Olfatório/patologia , Gravidez , Distribuição Aleatória
6.
Cereb Cortex ; 27(8): 3918-3929, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27371763

RESUMO

Neuronal nitric oxide synthase is involved in diverse signaling cascades that regulate neuronal development and functions via S-Nitrosylation-mediated mechanism or the soluble guanylate cyclase (sGC)/cyclic guanosine monophosphate (cGMP) pathway activated by nitric oxide. Although it has been studied extensively in vitro and in invertebrate animals, effects on mammalian brain development and underlying mechanisms remain poorly understood. Here we report that genetic deletion of "Nos1" disrupts dendritic development, whereas pharmacological inhibition of the sGC/cGMP pathway does not alter dendritic growth during cerebral cortex development. Instead, nuclear distribution element-like (NDEL1), a protein that regulates dendritic development, is specifically S-nitrosylated at cysteine 203, thereby accelerating dendritic arborization. This post-translational modification is enhanced by N-methyl-D-aspartate receptor-mediated neuronal activity, the main regulator of dendritic formation. Notably, we found that disruption of S-Nitrosylation of NDEL1 mediates impaired dendritic maturation caused by developmental alcohol exposure, a model of developmental brain abnormalities resulting from maternal alcohol use. These results highlight S-Nitrosylation as a key activity-dependent mechanism underlying neonatal brain maturation and suggest that reduction of S-Nitrosylation of NDEL1 acts as a pathological factor mediating neurodevelopmental abnormalities caused by maternal alcohol exposure.


Assuntos
Proteínas de Transporte/metabolismo , Dendritos/metabolismo , Transtornos do Espectro Alcoólico Fetal/metabolismo , Córtex Pré-Frontal/metabolismo , Células Piramidais/metabolismo , Transmissão Sináptica/fisiologia , Animais , Proteínas de Transporte/genética , Dendritos/efeitos dos fármacos , Dendritos/patologia , Modelos Animais de Doenças , Transtornos do Espectro Alcoólico Fetal/patologia , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Óxido Nítrico Sintase Tipo I/deficiência , Óxido Nítrico Sintase Tipo I/genética , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/crescimento & desenvolvimento , Córtex Pré-Frontal/patologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/patologia
7.
Biochem Biophys Res Commun ; 493(4): 1560-1566, 2017 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-28974418

RESUMO

Neuronal nitric oxide synthase (nNOS) 1, mainly responsible for NO release in central nervous system (CNS) 2, plays a significant role in multiple physiological functions. However, the function of nNOS+ interneurons in fear learning has not been much explored. Here we focused on the medial ganglionic eminences (MGE) 3-derived nNOS+ interneurons in fear learning. To determine the origin of nNOS+ interneurons, we cultured neurons in vitro from MGE, cortex, lateral ganglionic eminence (LGE) 4, caudal ganglionic eminences (CGE) 5 and preoptic area (POA) 6. The results showed that MGE contained the most abundant precursors of nNOS+ interneurons. Moreover, donor cells from E12.5 embryos demonstrated the highest positive rate of nNOS+ interneurons compared with other embryonic periods (E11.5, E12, E13, E13.5 and E14). Additionally, these cells from E12.5 embryos showed long axonal and abundant dendritic arbors after 10 days culture, indicating the capability to disperse and integrate in host neural circuits after transplantation. To investigate the role of MGE-derived nNOS+ interneurons in fear learning, donor MGE cells were transplanted into dentate gyrus (DG) 7 of nNOS knock-out (nNOS-/-) or wild-type mice. Results showed that the transplantation of MGE cells promoted the acquisition of nNOS-/- but not the wild-type mice, suggesting the importance of nNOS+ neurons in fear acquisition. Moreover, we transplanted MGE cells from nNOS-/- mice or wild-type mice into DG of the nNOS-/- mice and found that only MGE cells from wild-type mice but not the nNOS-/- mice rescued the deficit in acquisition of the nNOS-/- mice, further confirming the positive role of nNOS+ neurons in fear learning.


Assuntos
Medo/fisiologia , Interneurônios/fisiologia , Eminência Mediana/fisiologia , Óxido Nítrico Sintase Tipo I/fisiologia , Animais , Comportamento Animal/fisiologia , Células Cultivadas , Giro Denteado/citologia , Giro Denteado/fisiologia , Giro Denteado/cirurgia , Interneurônios/citologia , Interneurônios/transplante , Aprendizagem/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Células-Tronco Neurais/transplante , Óxido Nítrico Sintase Tipo I/deficiência , Óxido Nítrico Sintase Tipo I/genética , Telencéfalo/citologia , Telencéfalo/embriologia
8.
Mol Psychiatry ; 21(3): 313-9, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26782056

RESUMO

As traditional antidepressants act only after weeks/months, the discovery that ketamine, an antagonist of glutamate/N-methyl-D-aspartate (NMDA) receptors, elicits antidepressant actions in hours has been transformative. Its mechanism of action has been elusive, though enhanced mammalian target of rapamycin (mTOR) signaling is a major feature. We report a novel signaling pathway wherein NMDA receptor activation stimulates generation of nitric oxide (NO), which S-nitrosylates glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Nitrosylated GAPDH complexes with the ubiquitin-E3-ligase Siah1 and Rheb, a small G protein that activates mTOR. Siah1 degrades Rheb leading to reduced mTOR signaling, while ketamine, conversely, stabilizes Rheb that enhances mTOR signaling. Drugs selectively targeting components of this pathway may offer novel approaches to the treatment of depression.


Assuntos
Antidepressivos/uso terapêutico , Depressão/tratamento farmacológico , Ketamina/uso terapêutico , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Neuropeptídeos/metabolismo , Proteólise/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Animais , Antidepressivos/farmacologia , Células Cultivadas , Córtex Cerebral/citologia , Cisteína/análogos & derivados , Cisteína/farmacologia , Modelos Animais de Doenças , Embrião de Mamíferos , Comportamento Exploratório/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Células HEK293 , Humanos , Ketamina/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Monoméricas de Ligação ao GTP/genética , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Neuropeptídeos/genética , Óxido Nítrico/genética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo I/deficiência , Óxido Nítrico Sintase Tipo I/genética , Óxido Nítrico Sintase Tipo II/deficiência , Óxido Nítrico Sintase Tipo II/genética , Gravidez , Proteína Enriquecida em Homólogo de Ras do Encéfalo , S-Nitrosotióis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Natação/psicologia , Serina-Treonina Quinases TOR/genética , Fatores de Tempo
9.
Circ Res ; 116(1): 46-55, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25326127

RESUMO

RATIONALE: Although nitric oxide (NO) signaling modulates cardiac function and excitation-contraction coupling, opposing results because of inconsistent experimental conditions, particularly with respect to temperature, confound the ability to elucidate NO signaling pathways. Here, we show that temperature significantly modulates NO effects. OBJECTIVE: To test the hypothesis that temperature profoundly affects nitroso-redox equilibrium, thereby affecting sarcoplasmic reticulum (SR) calcium (Ca(2+)) leak. METHODS AND RESULTS: We measured SR Ca(2+) leak in cardiomyocytes from wild-type (WT), NO/redox imbalance (neuronal nitric oxide synthase-deficient mice-1 [NOS1(-/-)]), and hyper S-nitrosoglutathione reductase-deficient (GSNOR(-/-)) mice. In WT cardiomyocytes, SR Ca(2+) leak increased because temperature decreased from 37°C to 23°C, whereas in NOS1(-/-) cells, the leak suddenly increased when the temperature surpassed 30°C. GSNOR(-/-) cardiomyocytes exhibited low leak throughout the temperature range. Exogenously added NO had a biphasic effect on NOS1(-/-) cardiomyocytes; reducing leak at 37°C but increasing it at subphysiological temperatures. Oxypurinol and Tempol diminished the leak in NOS1(-/-) cardiomyocytes. Cooling from 37°C to 23°C increased reactive oxygen species generation in WT but decreased it in NOS1(-/-) cardiomyocytes. Oxypurinol further reduced reactive oxygen species generation. At 23°C in WT cells, leak was decreased by tetrahydrobiopterin, an essential NOS cofactor. Cooling significantly increased SR Ca(2+) content in NOS1(-/-) cells but had no effect in WT or GSNOR(-/-). CONCLUSIONS: Ca(2+) leak and temperature are normally inversely proportional, whereas NOS1 deficiency reverses this effect, increasing leak and elevating reactive oxygen species production because temperature increases. Reduced denitrosylation (GSNOR deficiency) eliminates the temperature dependence of leak. Thus, temperature regulates the balance between NO and reactive oxygen species which in turn has a major effect on SR Ca(2+).


Assuntos
Cálcio/metabolismo , Óxido Nítrico Sintase Tipo I/deficiência , Retículo Sarcoplasmático/metabolismo , Transdução de Sinais/fisiologia , Temperatura , Animais , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução , Espécies Reativas de Oxigênio/metabolismo
10.
J Biol Chem ; 290(16): 10325-35, 2015 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-25716315

RESUMO

The failure to trigger mitophagy is implicated in the pathogenesis of familial Parkinson disease that is caused by PINK1 or Parkin mutations. According to the prevailing PINK1-Parkin signaling model, mitophagy is promoted by the mitochondrial translocation of Parkin, an essential PINK1-dependent step that occurs via a previously unknown mechanism. Here we determined that critical concentrations of NO was sufficient to induce the mitochondrial translocation of Parkin even in PINK1 deficiency, with apparent increased interaction of full-length PINK1 accumulated during mitophagy, with neuronal nitric oxide synthase (nNOS). Specifically, optimum levels of NO enabled PINK1-null dopaminergic neuronal cells to regain the mitochondrial translocation of Parkin, which appeared to be significantly suppressed by nNOS-null mutation. Moreover, nNOS-null mutation resulted in the same mitochondrial electron transport chain (ETC) enzyme deficits as PINK1-null mutation. The involvement of mitochondrial nNOS activation in mitophagy was further confirmed by the greatly increased interactions of full-length PINK1 with nNOS, accompanied by mitochondrial accumulation of phospho-nNOS (Ser(1412)) during mitophagy. Of great interest is that the L347P PINK1 mutant failed to bind to nNOS. The loss of nNOS phosphorylation and Parkin accumulation on PINK1-deficient mitochondria could be reversed in a PINK1-dependent manner. Finally, non-toxic levels of NO treatment aided in the recovery of PINK1-null dopaminergic neuronal cells from mitochondrial ETC enzyme deficits. In summary, we demonstrated the full-length PINK1-dependent recruitment of nNOS, its activation in the induction of Parkin translocation, and the feasibility of NO-based pharmacotherapy for defective mitophagy and ETC enzyme deficits in Parkinson disease.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Mitocôndrias/metabolismo , Mitofagia/genética , Óxido Nítrico Sintase Tipo I/genética , Proteínas Quinases/genética , Ubiquitina-Proteína Ligases/genética , Animais , Modelos Animais de Doenças , Neurônios Dopaminérgicos/patologia , Transporte de Elétrons , Fibroblastos/metabolismo , Fibroblastos/patologia , Regulação da Expressão Gênica , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo I/deficiência , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Cultura Primária de Células , Ligação Proteica , Proteínas Quinases/deficiência , Transporte Proteico , Transdução de Sinais , Ubiquitina-Proteína Ligases/metabolismo
11.
EMBO J ; 31(2): 417-28, 2012 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-22036948

RESUMO

Mobilization of intracellular Ca(2+) stores regulates a multitude of cellular functions, but the role of intracellular Ca(2+) release via the ryanodine receptor (RyR) in the brain remains incompletely understood. We found that nitric oxide (NO) directly activates RyRs, which induce Ca(2+) release from intracellular stores of central neurons, and thereby promote prolonged Ca(2+) signalling in the brain. Reversible S-nitrosylation of type 1 RyR (RyR1) triggers this Ca(2+) release. NO-induced Ca(2+) release (NICR) is evoked by type 1 NO synthase-dependent NO production during neural firing, and is essential for cerebellar synaptic plasticity. NO production has also been implicated in pathological conditions including ischaemic brain injury, and our results suggest that NICR is involved in NO-induced neuronal cell death. These findings suggest that NICR via RyR1 plays a regulatory role in the physiological and pathophysiological functions of the brain.


Assuntos
Sinalização do Cálcio/fisiologia , Cerebelo/fisiologia , Córtex Cerebral/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/fisiologia , Óxido Nítrico/fisiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia , Animais , Apoptose/efeitos dos fármacos , Cerebelo/citologia , Córtex Cerebral/citologia , Células HEK293 , Humanos , Técnicas In Vitro , Infarto da Artéria Cerebral Média/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Óxido Nítrico Sintase Tipo I/deficiência , Óxido Nítrico Sintase Tipo I/genética , Óxido Nítrico Sintase Tipo I/fisiologia , Técnicas de Patch-Clamp , Proteínas Recombinantes de Fusão/fisiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/biossíntese , Canal de Liberação de Cálcio do Receptor de Rianodina/deficiência , Canal de Liberação de Cálcio do Receptor de Rianodina/genética
12.
J Neurovirol ; 22(6): 747-762, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27178324

RESUMO

Memory deficits are characteristic of HIV-associated neurocognitive disorders (HAND) and co-occur with hippocampal pathology. The HIV-1 transactivator of transcription (Tat), a regulatory protein, plays a significant role in these events, but the cellular mechanisms involved are poorly understood. Within the hippocampus, diverse populations of interneurons form complex networks; even subtle disruptions can drastically alter synaptic output, resulting in behavioral dysfunction. We hypothesized that HIV-1 Tat would impair cognitive behavior and injure specific hippocampal interneuron subtypes. Male transgenic mice that inducibly expressed HIV-1 Tat (or non-expressing controls) were assessed for cognitive behavior or had hippocampal CA1 subregions evaluated via interneuron subpopulation markers. Tat exposure decreased spatial memory in a Barnes maze and mnemonic performance in a novel object recognition test. Tat reduced the percentage of neurons expressing neuronal nitric oxide synthase (nNOS) without neuropeptide Y immunoreactivity in the stratum pyramidale and the stratum radiatum, parvalbumin in the stratum pyramidale, and somatostatin in the stratum oriens, which are consistent with reductions in interneuron-specific interneuron type 3 (IS3), bistratified, and oriens-lacunosum-moleculare interneurons, respectively. The findings reveal that an interconnected ensemble of CA1 nNOS-expressing interneurons, the IS3 cells, as well as subpopulations of parvalbumin- and somatostatin-expressing interneurons are preferentially vulnerable to HIV-1 Tat. Importantly, the susceptible interneurons form a microcircuit thought to be involved in feedback inhibition of CA1 pyramidal cells and gating of CA1 pyramidal cell inputs. The identification of vulnerable CA1 hippocampal interneurons may provide novel insight into the basic mechanisms underlying key functional and neurobehavioral deficits associated with HAND.


Assuntos
Região CA1 Hipocampal/metabolismo , Disfunção Cognitiva/genética , Interneurônios/metabolismo , Óxido Nítrico Sintase Tipo I/genética , Parvalbuminas/genética , Somatostatina/genética , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética , Animais , Região CA1 Hipocampal/fisiopatologia , Cognição/fisiologia , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/fisiopatologia , Regulação da Expressão Gênica , Interneurônios/patologia , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Transgênicos , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Óxido Nítrico Sintase Tipo I/deficiência , Parvalbuminas/deficiência , Transdução de Sinais , Somatostatina/deficiência , Transgenes , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo
13.
J Mol Cell Cardiol ; 81: 54-61, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25595735

RESUMO

Excessive oxidative stress in the heart results in contractile dysfunction. While antioxidant therapies have been a disappointment clinically, exercise has shown beneficial results, in part by reducing oxidative stress. We have previously shown that neuronal nitric oxide synthase (nNOS) is essential for cardioprotective adaptations caused by exercise. We hypothesize that part of the cardioprotective role of nNOS is via the augmentation of the antioxidant defense with exercise by positively shifting the nitroso-redox balance. Our results show that nNOS is indispensable for the augmented anti-oxidant defense with exercise. Furthermore, exercise training of nNOS knockout mice resulted in a negative shift in the nitroso-redox balance resulting in contractile dysfunction. Remarkably, overexpressing nNOS (conditional cardiac-specific nNOS overexpression) was able to mimic exercise by increasing VO2max. This study demonstrates that exercise results in a positive shift in the nitroso-redox balance that is nNOS-dependent. Thus, targeting nNOS signaling may mimic the beneficial effects of exercise by combating oxidative stress and may be a viable treatment strategy for heart disease.


Assuntos
Contração Miocárdica/fisiologia , Miocárdio/metabolismo , Óxido Nítrico Sintase Tipo I/genética , Óxido Nítrico/biossíntese , Condicionamento Físico Animal , Adaptação Fisiológica , Animais , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Regulação da Expressão Gênica , Camundongos , Camundongos Knockout , Miocárdio/citologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Óxido Nítrico Sintase Tipo I/deficiência , Oxirredução , Estresse Oxidativo , Cultura Primária de Células , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
14.
J Immunol ; 190(4): 1767-77, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23335752

RESUMO

Adult skeletal muscle regeneration results from activation, proliferation, and fusion of muscle stem cells, such as myogenic precursor cells. Macrophages are consistently present in regenerating skeletal muscles and participate into the repair process. The signals involved in the cross-talk between various macrophage populations and myogenic precursor cells have been only partially identified. In this study, we show a key role of inducible NO synthase (iNOS), expressed by classically activated macrophages in the healing of skeletal muscle. We found that, after sterile injury, iNOS expression is required for effective regeneration of the tissue, as myogenic precursor cells in the muscle of injured iNOS(-/-) mice fail to proliferate and differentiate. We also found that iNOS modulates inflammatory cell recruitment: damaged muscles of iNOS(-/-) animals express significantly higher levels of chemokines such as MIP2, MCP1, MIP-1α, and MCP1, and display more infiltrating neutrophils after injury and a persistence of macrophages at later time points. Finally, we found that iNOS expression in the injured muscle is restricted to infiltrating macrophages. To our knowledge, these data thus provide the first evidence that iNOS expression by infiltrating macrophages contributes to muscle regeneration, revealing a novel mechanism of inflammation-dependent muscle healing.


Assuntos
Músculo Esquelético/enzimologia , Músculo Esquelético/lesões , Doenças Musculares/enzimologia , Doenças Musculares/imunologia , Óxido Nítrico Sintase Tipo II/fisiologia , Doença Aguda , Animais , Animais Recém-Nascidos , Movimento Celular/genética , Movimento Celular/imunologia , Modelos Animais de Doenças , Inflamação/enzimologia , Inflamação/imunologia , Inflamação/patologia , Macrófagos/enzimologia , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/imunologia , Doenças Musculares/patologia , Óxido Nítrico Sintase Tipo I/deficiência , Óxido Nítrico Sintase Tipo I/fisiologia , Óxido Nítrico Sintase Tipo II/deficiência , Óxido Nítrico Sintase Tipo II/genética , Regeneração/genética , Regeneração/imunologia , Cicatrização/genética , Cicatrização/imunologia
15.
J Physiol ; 592(21): 4627-38, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25194047

RESUMO

The secondary loss of neuronal nitric oxide synthase (nNOS) that occurs in dystrophic muscle is the basis of numerous, complex and interacting features of the dystrophic pathology that affect not only muscle itself, but also influence the interaction of muscle with other tissues. Many mechanisms through which nNOS deficiency contributes to misregulation of muscle development, blood flow, fatigue, inflammation and fibrosis in dystrophic muscle have been identified, suggesting that normalization in NO production could greatly attenuate diverse aspects of the pathology of muscular dystrophy through multiple regulatory pathways. However, the relative importance of the loss of nNOS from the sarcolemma versus the importance of loss of total nNOS from dystrophic muscle remains unknown. Although most current evidence indicates that nNOS localization at the sarcolemma is not required to achieve NO-mediated reductions of pathology in muscular dystrophy, the question remains open concerning whether membrane localization would provide a more efficient rescue from features of the dystrophic phenotype.


Assuntos
Distrofia Muscular de Duchenne/enzimologia , Distrofia Muscular de Duchenne/fisiopatologia , Óxido Nítrico Sintase Tipo I/deficiência , Animais , Distrofina/deficiência , Distrofina/genética , Distrofina/metabolismo , Humanos , Óxido Nítrico/metabolismo , Sarcolema/enzimologia
16.
J Gastroenterol Hepatol ; 29(10): 1800-7, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24720557

RESUMO

BACKGROUND AND AIM: Nitric oxide (NO) is an important inhibitory mediator of esophageal function, and its lack leads to typical features of achalasia. In contrast, the role of intramuscular interstitial cells of Cajal (ICC-IM) and vasoactive intestinal peptide (VIP) in lower esophageal sphincter (LES) function is still controversial. Therefore, we examined the function and morphology of the LES in vivo in NO-deficient (nNOS(-/-) ), ICC-IM-deficient (W/W(v) )-, and wild-type (WT) mice. METHODS: Esophageal manometry was performed with a micro-sized transducer catheter to quantify LES pressure, swallow evoked LES relaxation, and esophageal body motility. The LES morphology was examined by semiquantitative analysis of the immunoreactivity (reduction grade I-IV) of neuronal NOS (nNOS), ICC-IM, and VIP and their correlation with esophageal function. RESULTS: nNOS(-/-) in comparison to WT mice showed a significantly higher LES mean resting pressure with an impaired swallow induced relaxation, whereas W/W(v) mice had a hypotensive LES with decreased relaxation. W/W(v) and nNOS(-/-) mice demonstrated differing degrees of tubular esophageal dysfunction. The reduced immunoreactivity of nNOS correlated with an increased LES pressure and decreased LES relaxation, respectively. Cajal-cell reduction correlated with impaired LES relaxation, whereas VIP reduction revealed no correlation with esophageal function. CONCLUSIONS: The reduction of ICC-IM and nNOS can cause dysfunction of the LES and esophageal peristalsis, whereas VIP reduction seems to have no effect. ICC-IM and nNOS deficiency might be independent relevant causes of esophageal dysfunction similar to that seen in human achalasia.


Assuntos
Acalasia Esofágica/etiologia , Deleção de Genes , Células Intersticiais de Cajal/fisiologia , Óxido Nítrico Sintase Tipo I/deficiência , Óxido Nítrico Sintase Tipo I/genética , Animais , Acalasia Esofágica/fisiopatologia , Esfíncter Esofágico Inferior/fisiopatologia , Feminino , Humanos , Masculino , Manometria , Camundongos Endogâmicos , Óxido Nítrico/fisiologia , Peristaltismo , Peptídeo Intestinal Vasoativo/fisiologia
17.
Nature ; 456(7221): 511-5, 2008 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-18953332

RESUMO

Many neuromuscular conditions are characterized by an exaggerated exercise-induced fatigue response that is disproportionate to activity level. This fatigue is not necessarily correlated with greater central or peripheral fatigue in patients, and some patients experience severe fatigue without any demonstrable somatic disease. Except in myopathies that are due to specific metabolic defects, the mechanism underlying this type of fatigue remains unknown. With no treatment available, this form of inactivity is a major determinant of disability. Here we show, using mouse models, that this exaggerated fatigue response is distinct from a loss in specific force production by muscle, and that sarcolemma-localized signalling by neuronal nitric oxide synthase (nNOS) in skeletal muscle is required to maintain activity after mild exercise. We show that nNOS-null mice do not have muscle pathology and have no loss of muscle-specific force after exercise but do display this exaggerated fatigue response to mild exercise. In mouse models of nNOS mislocalization from the sarcolemma, prolonged inactivity was only relieved by pharmacologically enhancing the cGMP signal that results from muscle nNOS activation during the nitric oxide signalling response to mild exercise. Our findings suggest that the mechanism underlying the exaggerated fatigue response to mild exercise is a lack of contraction-induced signalling from sarcolemma-localized nNOS, which decreases cGMP-mediated vasomodulation in the vessels that supply active muscle after mild exercise. Sarcolemmal nNOS staining was decreased in patient biopsies from a large number of distinct myopathies, suggesting a common mechanism of fatigue. Our results suggest that patients with an exaggerated fatigue response to mild exercise would show clinical improvement in response to treatment strategies aimed at improving exercise-induced signalling.


Assuntos
Modelos Animais de Doenças , Exercício Físico/fisiologia , Fadiga/fisiopatologia , Óxido Nítrico Sintase Tipo I/metabolismo , Sarcolema/enzimologia , Animais , GMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5 , Edema/tratamento farmacológico , Edema/etiologia , Edema/prevenção & controle , Ativação Enzimática , Fadiga/patologia , Hemodinâmica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/citologia , Músculo Esquelético/enzimologia , Músculo Esquelético/fisiopatologia , Doenças Musculares/enzimologia , Doenças Musculares/patologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo I/deficiência , Óxido Nítrico Sintase Tipo I/genética , Inibidores da Fosfodiesterase 5 , Transporte Proteico , Transdução de Sinais
18.
Proc Natl Acad Sci U S A ; 108(42): 17544-9, 2011 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-21976486

RESUMO

Cells sense and respond to changes in oxygen concentration through gene regulatory processes that are fundamental to survival. Surprisingly, little is known about how anemia affects hypoxia signaling. Because nitric oxide synthases (NOSs) figure prominently in the cellular responses to acute hypoxia, we defined the effects of NOS deficiency in acute anemia. In contrast to endothelial NOS or inducible NOS deficiency, neuronal NOS (nNOS)(-/-) mice demonstrated increased mortality during anemia. Unlike wild-type (WT) animals, anemia did not increase cardiac output (CO) or reduce systemic vascular resistance (SVR) in nNOS(-/-) mice. At the cellular level, anemia increased expression of HIF-1α protein and HIF-responsive mRNA levels (EPO, VEGF, GLUT1, PDK1) in the brain of WT, but not nNOS(-/-) mice, despite comparable reductions in tissue PO(2). Paradoxically, nNOS(-/-) mice survived longer during hypoxia, retained the ability to regulate CO and SVR, and increased brain HIF-α protein levels and HIF-responsive mRNA transcripts. Real-time imaging of transgenic animals expressing a reporter HIF-α(ODD)-luciferase chimeric protein confirmed that nNOS was essential for anemia-mediated increases in HIF-α protein stability in vivo. S-nitrosylation effects the functional interaction between HIF and pVHL. We found that anemia led to nNOS-dependent S-nitrosylation of pVHL in vivo and, of interest, led to decreased expression of GSNO reductase. These findings identify nNOS effects on the HIF/pVHL signaling pathway as critically important in the physiological responses to anemia in vivo and provide essential mechanistic insight into the differences between anemia and hypoxia.


Assuntos
Anemia/fisiopatologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Adaptação Fisiológica , Anemia/genética , Animais , Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Débito Cardíaco , Células Endoteliais da Veia Umbilical Humana , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Óxido Nítrico Sintase Tipo I/deficiência , Óxido Nítrico Sintase Tipo I/genética , Oxigênio/sangue , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Resistência Vascular , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
19.
J Neurosci ; 32(43): 14994-9, 2012 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-23100421

RESUMO

Nitric oxide synthase-1 (NOS1) is involved in several forms of plasticity including hippocampal-dependent learning and memory, experience-dependent plasticity in the barrel cortex, and long-term potentiation (LTP) in the hippocampus and neocortex. NOS1 also contributes to ischemic damage during stroke and has a stronger deleterious effect in males than females. We therefore investigated whether the role of NOS1 in plasticity might also be sex specific. We tested LTP in the layer IV-II/III pathway between barrel columns and experience-dependent plasticity in the barrel cortex of αNOS1 knock-out mice and their wild-type littermates. We found that LTP was absent in male αNOS1 knock-out mice but not in females and that the residual LTP in females was not NO dependent. We also found that experience-dependent potentiation due to single whisker experience was significantly reduced in male αNOS1 knockouts but was unaffected in females. The αNOS1 knockout had a small effect on the development of the barrels, which were reduced in size by 20% compared with wild types, but this effect was not sex specific. We therefore conclude that neocortical plasticity mechanisms differ between males and females at the synaptic level, either in their basic plasticity induction pathways or in their ability to compensate for loss of αNOS1.


Assuntos
Córtex Cerebral/fisiologia , Potenciação de Longa Duração/fisiologia , Óxido Nítrico Sintase Tipo I/metabolismo , Caracteres Sexuais , Animais , Biofísica , Córtex Cerebral/citologia , Estimulação Elétrica , Feminino , Técnicas In Vitro , Potenciação de Longa Duração/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo I/deficiência , Privação Sensorial/fisiologia , Vibrissas/inervação
20.
J Neurosci ; 32(3): 932-45, 2012 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-22262891

RESUMO

Reproduction is controlled in the brain by a neural network that drives the secretion of gonadotropin-releasing hormone (GnRH). Various permissive homeostatic signals must be integrated to achieve ovulation in mammals. However, the neural events controlling the timely activation of GnRH neurons are not completely understood. Here we show that kisspeptin, a potent activator of GnRH neuronal activity, directly communicates with neurons that synthesize the gaseous transmitter nitric oxide (NO) in the preoptic region to coordinate the progression of the ovarian cycle. Using a transgenic Gpr54-null IRES-LacZ knock-in mouse model, we demonstrate that neurons containing neuronal NO synthase (nNOS), which are morphologically associated with kisspeptin fibers, express the kisspeptin receptor GPR54 in the preoptic region, but not in the tuberal region of the hypothalamus. The activation of kisspeptin signaling in preoptic neurons promotes the activation of nNOS through its phosphorylation on serine 1412 via the AKT pathway and mimics the positive feedback effects of estrogens. Finally, we show that while NO release restrains the reproductive axis at stages of the ovarian cycle during which estrogens exert their inhibitory feedback, it is required for the kisspeptin-dependent preovulatory activation of GnRH neurons. Thus, interactions between kisspeptin and nNOS neurons may play a central role in regulating the hypothalamic-pituitary-gonadal axis in vivo.


Assuntos
Hipotálamo/citologia , Kisspeptinas/metabolismo , Neurônios/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Ovulação/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Análise de Variância , Animais , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Ciclo Estral/efeitos dos fármacos , Ciclo Estral/genética , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Hipotálamo/efeitos dos fármacos , Kisspeptinas/deficiência , Kisspeptinas/farmacologia , Hormônio Luteinizante/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Óxido Nítrico Sintase Tipo I/deficiência , Ovulação/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Receptores Acoplados a Proteínas G/deficiência , Receptores de Kisspeptina-1 , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Esteroides/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA