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










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 11(1): 17189, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34433854

RESUMEN

Neuronal nitric oxide synthase (nNOS) neurons play a fundamental role in inhibitory neurotransmission, within the enteric nervous system (ENS), and in the establishment of gut motility patterns. Clinically, loss or disruption of nNOS neurons has been shown in a range of enteric neuropathies. However, the effects of nNOS loss on the composition and structure of the ENS remain poorly understood. The aim of this study was to assess the structural and transcriptional consequences of loss of nNOS neurons within the murine ENS. Expression analysis demonstrated compensatory transcriptional upregulation of pan neuronal and inhibitory neuronal subtype targets within the Nos1-/- colon, compared to control C57BL/6J mice. Conventional confocal imaging; combined with novel machine learning approaches, and automated computational analysis, revealed increased interconnectivity within the Nos1-/- ENS, compared to age-matched control mice, with increases in network density, neural projections and neuronal branching. These findings provide the first direct evidence of structural and molecular remodelling of the ENS, upon loss of nNOS signalling. Further, we demonstrate the utility of machine learning approaches, and automated computational image analysis, in revealing previously undetected; yet potentially clinically relevant, changes in ENS structure which could provide improved understanding of pathological mechanisms across a host of enteric neuropathies.


Asunto(s)
Sistema Nervioso Entérico/metabolismo , Óxido Nítrico Sintasa de Tipo I/genética , Animales , Sistema Nervioso Entérico/citología , Aprendizaje Automático , Ratones , Ratones Endogámicos C57BL , Red Nerviosa/citología , Red Nerviosa/metabolismo , Neuronas/citología , Neuronas/metabolismo , Óxido Nítrico Sintasa de Tipo I/deficiencia
2.
Nitric Oxide ; 113-114: 23-30, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-33915274

RESUMEN

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.


Asunto(s)
Disfunción Eréctil/tratamiento farmacológico , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Péptidos/farmacología , Animales , Biología Computacional , Disfunción Eréctil/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo I/deficiencia , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo II/deficiencia , Óxido Nítrico Sintasa de Tipo II/genética , Péptidos/síntesis química , Péptidos/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
3.
Neurotherapeutics ; 17(3): 1016-1030, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32632774

RESUMEN

Neuropathic pain is usually persistent due to maladaptive neuroplasticity-induced central sensitization and, therefore, necessitates long-term treatment. N-methyl-D-aspartate receptor (NMDAR)-mediated hypersensitivity in the spinal dorsal horn represents key mechanisms of central sensitization. Short-term use of NMDAR antagonists produces antinociceptive efficacy in animal pain models and in clinical practice by reducing central sensitization. However, how prolonged use of NMDAR antagonists affects central sensitization remains unknown. Surprisingly, we find that prolonged blockage of NMDARs does not prevent but aggravate nerve injury-induced central sensitization and produce analgesic tolerance, mainly due to reduced synaptic inhibition. The disinhibition that results from the continuous decrease in the production of nitric oxide from neuronal nitric oxide synthase, downstream signal of NMDARs, leads to the reduction of GABAergic inhibitory synaptic transmission by upregulating brain-derived neurotrophic factor expression and inhibiting the expression and function of potassium-chloride cotransporter. Together, our findings suggest that chronic blockage of NMDARs develops analgesic tolerance through the neuronal nitric oxide synthase-brain-derived neurotrophic factor-potassium-chloride cotransporter pathway. Thus, preventing the GABAergic disinhibition induced by nitric oxide reduction may be necessary for the long-term maintenance of the analgesic effect of NMDAR antagonists.


Asunto(s)
Analgésicos/administración & dosificación , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Neuronas GABAérgicas/metabolismo , Neuralgia/metabolismo , Óxido Nítrico Sintasa de Tipo I/deficiencia , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Analgésicos/efectos adversos , Animales , Maleato de Dizocilpina/administración & dosificación , Maleato de Dizocilpina/efectos adversos , Resistencia a Medicamentos/efectos de los fármacos , Resistencia a Medicamentos/fisiología , Antagonistas de Aminoácidos Excitadores/efectos adversos , Neuronas GABAérgicas/efectos de los fármacos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibición Neural/efectos de los fármacos , Inhibición Neural/fisiología , Neuralgia/tratamiento farmacológico , Neuralgia/genética , Óxido Nítrico/deficiencia , Óxido Nítrico/genética , Óxido Nítrico Sintasa de Tipo I/genética , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley
4.
Eur Neuropsychopharmacol ; 30: 30-43, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-28951000

RESUMEN

Nitric oxide (NO) is a gaseous neurotransmitter that has important behavioural functions in the vertebrate brain. In this study we compare the impact of decreased nitric NO signalling upon behaviour and neurobiology using both zebrafish and mouse. nitric oxide synthase mutant (nos1-/-) zebrafish show significantly reduced aggression and an increase in anxiety-like behaviour without altered production of the stress hormone cortisol. Nos1-/- mice also exhibit decreased aggression and are hyperactive in an open field test. Upon reduction of NO signalling, monoamine neurotransmitter metabolism is reduced as a consequence of decreased Monoamine oxidase activity. Treatment of nos1-/- zebrafish with the 5-HT receptor 1A agonist 8-OH-DPAT rescues aggression and some aspects of anxiety-like behaviour. Taken together, the interplay between NO and 5-HT appears to be critical to control behaviour. Our cross-species approach challenges the previous notion that reduced neuronal NOS leads to increased aggression. Rather, Nos1 knock-out can also lead to decreased aggression in some situations, a finding that may have implications for future translational research.


Asunto(s)
Agresión/fisiología , Ansiedad/metabolismo , Monoaminooxidasa/metabolismo , Óxido Nítrico Sintasa de Tipo I/deficiencia , Óxido Nítrico/metabolismo , Agresión/efectos de los fármacos , Agresión/psicología , Animales , Animales Modificados Genéticamente , Ansiedad/psicología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidores de la Monoaminooxidasa/farmacología , Serotonina/metabolismo , Agonistas del Receptor de Serotonina 5-HT1/farmacología , Pez Cebra
5.
Sleep ; 42(10)2019 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-31328777

RESUMEN

Slow-wave activity (SWA) is an oscillatory neocortical activity occurring in the electroencephalogram delta (δ) frequency range (~0.5-4 Hz) during nonrapid eye movement sleep. SWA is a reliable indicator of sleep homeostasis after acute sleep loss and is involved in memory processes. Evidence suggests that cortical neuronal nitric oxide synthase (nNOS) expressing neurons that coexpress somatostatin (SST) play a key role in regulating SWA. However, previous studies lacked selectivity in targeting specific types of neurons that coexpress nNOS-cells which are activated in the cortex after sleep loss. We produced a mouse model that knocks out nNOS expression in neurons that coexpress SST throughout the cortex. Mice lacking nNOS expression in SST positive neurons exhibited significant impairments in both homeostatic low-δ frequency range SWA production and a recognition memory task that relies on cortical input. These results highlight that SST+/nNOS+ neurons are involved in the SWA homeostatic response and cortex-dependent recognition memory.


Asunto(s)
Corteza Cerebral/metabolismo , Ritmo Delta/fisiología , Memoria/fisiología , Óxido Nítrico Sintasa de Tipo I/deficiencia , Reconocimiento en Psicología/fisiología , Somatostatina/deficiencia , Animales , Electroencefalografía/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neuronas/metabolismo , Óxido Nítrico Sintasa de Tipo I/genética , Sueño/fisiología , Somatostatina/genética
6.
Am J Physiol Renal Physiol ; 317(3): F547-F559, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31241990

RESUMEN

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.


Asunto(s)
Deshidratación/enzimología , Diuresis , Capacidad de Concentración Renal , Túbulos Renales Colectores/enzimología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico/metabolismo , Animales , Fármacos Antidiuréticos/farmacología , Acuaporina 2/genética , Acuaporina 2/metabolismo , Acuaporina 3/genética , Acuaporina 3/metabolismo , Desamino Arginina Vasopresina/farmacología , Deshidratación/fisiopatología , Modelos Animales de Enfermedad , Diuresis/efectos de los fármacos , Femenino , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Capacidad de Concentración Renal/efectos de los fármacos , Túbulos Renales Colectores/efectos de los fármacos , Masculino , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo I/deficiencia , Óxido Nítrico Sintasa de Tipo I/genética , Estado de Hidratación del Organismo , Concentración Osmolar , Factores Sexuales , Transducción de Señal , Urodinámica , Privación de Agua
7.
Am J Physiol Renal Physiol ; 317(3): F540-F546, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31215803

RESUMEN

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.


Asunto(s)
Anemia de Células Falciformes/complicaciones , Óxido Nítrico/metabolismo , Vejiga Urinaria Hiperactiva/etiología , Vejiga Urinaria/metabolismo , Urodinámica , Anemia de Células Falciformes/genética , Animales , Modelos Animales de Enfermedad , Hemoglobina A/genética , Hemoglobina A/metabolismo , Hemoglobinas/genética , Hemoglobinas/metabolismo , Humanos , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Muscular , Relajación Muscular , Óxido Nítrico Sintasa de Tipo I/deficiencia , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo III/deficiencia , Óxido Nítrico Sintasa de Tipo III/genética , Transducción de Señal , Vejiga Urinaria/fisiopatología , Vejiga Urinaria Hiperactiva/metabolismo , Vejiga Urinaria Hiperactiva/fisiopatología
8.
Brain Behav ; 8(6): e00985, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-30106246

RESUMEN

INTRODUCTION: Patients with dystrophinopathies show low levels of neuronal nitric oxide synthase (nNOS), due to reduced or absent dystrophin expression, as nNOS is attached to the dystrophin-associated protein complex. Deficient nNOS function leads to functional ischemia during muscle activity. Dystrophin-like proteins with nNOS attached have also been identified in the brain. This suggests that a mechanism of cerebral functional ischemia with attenuation of normal activation-related vascular response may cause changes in brain function. METHODS: The aim of this study was to investigate whether the brain response of patients with Becker muscular dystrophy (BMD) is dysfunctional compared to that of healthy controls. To investigate a potential change in brain activation response in patients with BMD, median nerve somatosensory evoked stimulation, with stimulation durations of 2, 4, and 10 s, was performed while recording electroencephalography and blood oxygen level-dependent (BOLD) functional magnetic resonance imaging. RESULTS: Results in 14 male patients with BMD (36.2 ± 9.9 years) were compared with those of 10 healthy controls (34.4 ± 10.9 years). Compared to controls, the patients with BMD showed sustained cortical electrical activity and a significant smaller BOLD activation in contralateral primary somatosensory cortex and bilaterally in secondary somatosensory cortex. In addition, significant activation differences were found after long duration (10 s) stimuli in thalamus. CONCLUSION: An altered neurovascular response in patients with BMD may increase our understanding of neurovascular coupling and the pathogenesis related to dystrophinopathy and nNOS.


Asunto(s)
Encéfalo/fisiología , Distrofia Muscular de Duchenne/fisiopatología , Óxido Nítrico Sintasa de Tipo I/deficiencia , Adulto , Encéfalo/irrigación sanguínea , Estudios de Casos y Controles , Estimulación Eléctrica/métodos , Electroencefalografía , Humanos , Imagen por Resonancia Magnética , Masculino , Nervio Mediano/fisiología , Oxígeno/sangre , Corteza Somatosensorial/fisiología
9.
Alcohol Clin Exp Res ; 42(9): 1627-1639, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29957842

RESUMEN

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.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Etanol/toxicidad , Trastornos del Espectro Alcohólico Fetal/genética , Neuronas/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo I/genética , Bulbo Olfatorio/efectos de los fármacos , Consumo de Bebidas Alcohólicas/efectos adversos , Consumo de Bebidas Alcohólicas/metabolismo , Consumo de Bebidas Alcohólicas/patología , Animales , Animales Recién Nacidos , Femenino , Trastornos del Espectro Alcohólico Fetal/patología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Óxido Nítrico Sintasa de Tipo I/deficiencia , Bulbo Olfatorio/metabolismo , Bulbo Olfatorio/patología , Embarazo , Distribución Aleatoria
10.
Sci Signal ; 11(522)2018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29559585

RESUMEN

Acetylation of the microtubule-associated protein tau promotes its polymerization into neurofibrillary tangles that are implicated in the pathology of Alzheimer's disease (AD). The gaseous neurotransmitter nitric oxide (NO) regulates cell signaling through the nitrosylation of proteins. We found that NO production and tau acetylation at Lys280 occurred in the brain tissue in mice and in cultured mouse cortical neurons in response to exposure to amyloid-ß1-42 (Aß1-42), a peptide that is also implicated in AD. An increased abundance of NO facilitated the S-nitrosylation (SNO) of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). S-nitrosylated GAPDH (GAPDH-SNO) promoted the acetylation and activation of the acetyltransferase p300 and facilitated the nitrosylation and inactivation of the deacetylase sirtuin 1 (SIRT1). The abundance of GAPDH-SNO was increased in postmortem brain samples from AD patients. Preventing the increase in GAPDH-SNO abundance in both cultured neurons and mice, either by overexpression of the nitrosylation mutant of GAPDH (GAPDH C150S) or by treatment with the GAPDH nitrosylation inhibitor CGP3466B (also known as omigapil), abrogated Aß1-42-induced tau acetylation, memory impairment, and locomotor dysfunction in mice, suggesting that this drug might be repurposed to treat patients with AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Óxido Nítrico Sintasa de Tipo I/deficiencia , Óxido Nítrico/metabolismo , Sirtuina 1/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Proteínas tau/metabolismo , Acetilación/efectos de los fármacos , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/toxicidad , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo I/genética , Oxepinas/farmacología , Fragmentos de Péptidos/toxicidad
11.
J Proteome Res ; 17(3): 1031-1040, 2018 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-29394072

RESUMEN

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.


Asunto(s)
Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico/metabolismo , Pene/metabolismo , Priapismo/genética , Animales , Cromatografía de Fase Inversa , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Estimulación Eléctrica , Células Endoteliales/metabolismo , Células Endoteliales/patología , Regulación de la Expresión Génica , Ontología de Genes , Humanos , Masculino , Ratones , Ratones Noqueados , Anotación de Secuencia Molecular , Neuronas/metabolismo , Neuronas/patología , Óxido Nítrico Sintasa de Tipo I/deficiencia , Óxido Nítrico Sintasa de Tipo III/deficiencia , Erección Peniana/fisiología , Pene/irrigación sanguínea , Pene/inervación , Priapismo/metabolismo , Priapismo/patología , Priapismo/fisiopatología , Proteoma/genética , Proteoma/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Nervios Esplácnicos/metabolismo , Nervios Esplácnicos/fisiopatología , Espectrometría de Masas en Tándem
12.
J Am Heart Assoc ; 6(10)2017 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-29066445

RESUMEN

BACKGROUND: During high sodium intake, the renin-angiotensin-aldosterone system is downregulated and nitric oxide signaling is upregulated in order to remain in sodium balance. Recently, we showed that collecting duct nitric oxide synthase 1ß is critical for fluid-electrolyte balance and subsequently blood pressure regulation during high sodium feeding. The current study tested the hypothesis that high sodium activation of the collecting duct nitric oxide synthase 1ß pathway is critical for maintaining sodium homeostasis and for the downregulation of the renin-angiotensin-aldosterone system-epithelial sodium channel axis. METHODS AND RESULTS: Male control and collecting duct nitric oxide synthase 1ß knockout (CDNOS1KO) mice were placed on low, normal, and high sodium diets for 1 week. In response to the high sodium diet, plasma sodium was significantly increased in control mice and to a significantly greater level in CDNOS1KO mice. CDNOS1KO mice did not suppress plasma aldosterone in response to the high sodium diet, which may be partially explained by increased adrenal AT1R expression. Plasma renin concentration was appropriately suppressed in both genotypes. Furthermore, CDNOS1KO mice had significantly higher intrarenal angiotensin II with high sodium diet, although intrarenal angiotensinogen levels and angiotensin-converting enzyme activity were similar between knockout mice and controls. In agreement with inappropriate renin-angiotensin-aldosterone system activation in the CDNOS1KO mice on a high sodium diet, epithelial sodium channel activity and sodium transporter abundance were significantly higher compared with controls. CONCLUSIONS: These data demonstrate that high sodium activation of collecting duct nitric oxide synthase 1ß signaling induces suppression of systemic and intrarenal renin-angiotensin-aldosterone system, thereby modulating epithelial sodium channel and other sodium transporter abundance and activity to maintain sodium homeostasis.


Asunto(s)
Aldosterona/sangre , Angiotensina II/sangre , Túbulos Renales Colectores/enzimología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Eliminación Renal , Sistema Renina-Angiotensina , Cloruro de Sodio Dietético/metabolismo , Animales , Activación Enzimática , Canales Epiteliales de Sodio/metabolismo , Genotipo , Homeostasis , Masculino , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo I/deficiencia , Óxido Nítrico Sintasa de Tipo I/genética , Fenotipo , Receptor de Angiotensina Tipo 1/metabolismo , Renina/sangre , Simportadores del Cloruro de Sodio/metabolismo
13.
Biochem Biophys Res Commun ; 493(4): 1560-1566, 2017 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-28974418

RESUMEN

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.


Asunto(s)
Miedo/fisiología , Interneuronas/fisiología , Eminencia Media/fisiología , Óxido Nítrico Sintasa de Tipo I/fisiología , Animales , Conducta Animal/fisiología , Células Cultivadas , Giro Dentado/citología , Giro Dentado/fisiología , Giro Dentado/cirugía , Interneuronas/citología , Interneuronas/trasplante , Aprendizaje/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Células-Madre Neurales/trasplante , Óxido Nítrico Sintasa de Tipo I/deficiencia , Óxido Nítrico Sintasa de Tipo I/genética , Telencéfalo/citología , Telencéfalo/embriología
14.
PLoS One ; 12(8): e0183090, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28800639

RESUMEN

MPTP-mouse model constitutes a well-known model of neuroinflammation and mitochondrial failure occurring in Parkinson's disease (PD). Although it has been extensively reported that nitric oxide (NO●) plays a key role in the pathogenesis of PD, the relative roles of nitric oxide synthase isoforms iNOS and nNOS in the nigrostriatal pathway remains, however, unclear. Here, the participation of iNOS/nNOS isoforms in the mitochondrial dysfunction was analyzed in iNOS and nNOS deficient mice. Our results showed that MPTP increased iNOS activity in substantia nigra and striatum, whereas it sharply reduced complex I activity and mitochondrial bioenergetics in all strains. In the presence of MPTP, mice lacking iNOS showed similar restricted mitochondrial function than wild type or mice lacking nNOS. These results suggest that iNOS-dependent elevated nitric oxide, a major pathological hallmark of neuroinflammation in PD, does not contribute to mitochondrial impairment. Therefore, neuroinflammation and mitochondrial dysregulation seem to act in parallel in the MPTP model of PD. Melatonin administration, with well-reported neuroprotective properties, counteracted these effects, preventing from the drastic changes in mitochondrial oxygen consumption, increased NOS activity and prevented reduced locomotor activity induced by MPTP. The protective effects of melatonin on mitochondria are also independent of its anti-inflammatory properties, but both effects are required for an effective anti-parkinsonian activity of the indoleamine as reported in this study.


Asunto(s)
Melatonina/farmacología , Mitocondrias/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo I/genética , Enfermedad de Parkinson Secundaria/tratamiento farmacológico , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Animales , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo I/deficiencia , Óxido Nítrico Sintasa de Tipo II/deficiencia , Consumo de Oxígeno/efectos de los fármacos , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/genética , Enfermedad de Parkinson Secundaria/patología , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Sustancia Negra/patología
16.
Orphanet J Rare Dis ; 12(1): 100, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28545481

RESUMEN

Duchenne Muscular Dystrophy is a rare and fatal neuromuscular disease in which the absence of dystrophin from the muscle membrane induces a secondary loss of neuronal nitric oxide synthase and the muscles capacity for endogenous nitric oxide synthesis. Since nitric oxide is a potent regulator of skeletal muscle metabolism, mass, function and regeneration, the loss of nitric oxide bioavailability is likely a key contributor to the chronic pathological wasting evident in Duchenne Muscular Dystrophy. As such, various therapeutic interventions to re-establish either the neuronal nitric oxide synthase protein deficit or the consequential loss of nitric oxide synthesis and bioavailability have been investigated in both animal models of Duchenne Muscular Dystrophy and in human clinical trials. Notably, the efficacy of these interventions are varied and not always translatable from animal model to human patients, highlighting a complex interplay of factors which determine the downstream modulatory effects of nitric oxide. We review these studies herein.


Asunto(s)
Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/enzimología , Donantes de Óxido Nítrico/administración & dosificación , Óxido Nítrico Sintasa de Tipo I/deficiencia , Animales , Disponibilidad Biológica , Ensayos Clínicos como Asunto/métodos , Quimioterapia Combinada , Humanos , Distrofia Muscular de Duchenne/genética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo I/genética , Pregnenodionas/administración & dosificación
17.
Cereb Cortex ; 27(8): 3918-3929, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27371763

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/metabolismo , Dendritas/metabolismo , Trastornos del Espectro Alcohólico Fetal/metabolismo , Corteza Prefrontal/metabolismo , Células Piramidales/metabolismo , Transmisión Sináptica/fisiología , Animales , Proteínas Portadoras/genética , Dendritas/efectos de los fármacos , Dendritas/patología , Modelos Animales de Enfermedad , Trastornos del Espectro Alcohólico Fetal/patología , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Óxido Nítrico Sintasa de Tipo I/deficiencia , Óxido Nítrico Sintasa de Tipo I/genética , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/crecimiento & desarrollo , Corteza Prefrontal/patología , Células Piramidales/efectos de los fármacos , Células Piramidales/patología
18.
Neurotherapeutics ; 14(2): 429-446, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27921261

RESUMEN

Duchenne muscular dystrophy arises from the loss of dystrophin and is characterized by calcium dysregulation, muscular atrophy, and metabolic dysfunction. The secondary reduction of neuronal nitric oxide synthase (nNOS) from the sarcolemma reduces NO production and bioavailability. As NO modulates glucose uptake, metabolism, and mitochondrial bioenergetics, we investigated whether an 8-week nitrate supplementation regimen could overcome metabolic dysfunction in the mdx mouse. Dystrophin-positive control (C57BL/10) and dystrophin-deficient mdx mice were supplemented with sodium nitrate (85 mg/l) in drinking water. Following the supplementation period, extensor digitorum longus and soleus were excised and radioactive glucose uptake was measured at rest (basal) and during contraction. Gastrocnemius was excised and mitochondrial respiration was measured using the Oroboros Oxygraph. Tibialis anterior was analyzed immunohistochemically for the presence of dystrophin, nNOS, nitrotyrosine, IgG and CD45+ cells, and histologically to assess areas of damage and regeneration. Glucose uptake in the basal and contracting states was normal in unsupplemented mdx muscles but was reduced following nitrate supplementation in mdx muscles only. The mitochondrial utilization of substrates was also impaired in mdx gastrocnemius during phosphorylating and maximal uncoupled respiration, and nitrate could not improve respiration in mdx muscle. Although nitrate supplementation reduced mitochondrial hydrogen peroxide emission, it induced mitochondrial uncoupling in red gastrocnemius, increased muscle fiber peroxynitrite (nitrotyrosine), and promoted skeletal muscle damage. Our novel data suggest that despite lower nNOS protein expression and likely lower NO production in mdx muscle, enhancing NO production with nitrate supplementation in these mice has detrimental effects on skeletal muscle. This may have important relevance for those with DMD.


Asunto(s)
Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Nitratos/administración & dosificación , Óxido Nítrico Sintasa de Tipo I/deficiencia , Animales , Distrofina/deficiencia , Transporte de Electrón , Glucosa/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Mitocondrias/fisiología , Especies Reactivas de Oxígeno
19.
Free Radic Biol Med ; 99: 472-484, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27609225

RESUMEN

Xanthine oxidoreductase (XOR) is generally known as the final enzyme in purine metabolism and as a source of reactive oxygen species (ROS). In addition, this enzyme has been suggested to mediate nitric oxide (NO) formation via reduction of inorganic nitrate and nitrite. This NO synthase (NOS)-independent pathway for NO generation is of particular importance during certain conditions when NO bioavailability is diminished due to reduced activity of endothelial NOS (eNOS) or increased oxidative stress, including aging and cardiovascular disease. The exact interplay between NOS- and XOR-derived NO generation is not fully elucidated yet. The aim of the present study was to investigate if eNOS deficiency is associated with changes in XOR expression and activity and the possible impact on nitrite, NO and ROS homeostasis. Plasma levels of nitrate and nitrite were similar between eNOS deficient (eNOS-/-) and wildtype (wt) mice. XOR activity was upregulated in eNOS-/- compared with wt, but not in nNOS-/-, iNOS-/- or wt mice treated with the non-selective NOS inhibitor L-NAME. Following an acute dose of nitrate, plasma nitrite increased more in eNOS-/- compared with wt, and this augmented response was abolished by the selective XOR inhibitor febuxostat. Livers from eNOS-/- displayed higher nitrite reducing capacity compared with wt, and this effect was attenuated by febuxostat. Dietary supplementation with nitrate increased XOR expression and activity, but concomitantly reduced superoxide generation. The latter effect was also seen in vitro after nitrite administration. Treatment with febuxostat elevated blood pressure in eNOS-/-, but not in wt mice. A high dose of dietary nitrate reduced blood pressure in naïve eNOS-/- mice, and again this effect was abolished by febuxostat. In conclusion, eNOS deficiency is associated with an upregulation of XOR facilitating the nitrate-nitrite-NO pathway and decreasing the generation of ROS. This interplay between XOR and eNOS is proposed to play a significant role in NO homeostasis and blood pressure regulation.


Asunto(s)
Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico/sangre , Xantina Deshidrogenasa/genética , Animales , Presión Sanguínea/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Febuxostat/farmacología , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Noqueados , NG-Nitroarginina Metil Éster/farmacología , Nitratos/sangre , Nitratos/farmacología , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo I/deficiencia , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/deficiencia , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/deficiencia , Nitritos/sangre , Nitritos/farmacología , Oxidación-Reducción , Transducción de Señal , Superóxidos/metabolismo , Xantina Deshidrogenasa/antagonistas & inhibidores , Xantina Deshidrogenasa/metabolismo
20.
J Neurovirol ; 22(6): 747-762, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27178324

RESUMEN

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.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Disfunción Cognitiva/genética , Interneuronas/metabolismo , Óxido Nítrico Sintasa de Tipo I/genética , Parvalbúminas/genética , Somatostatina/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Animales , Región CA1 Hipocampal/fisiopatología , Cognición/fisiología , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/fisiopatología , Regulación de la Expresión Génica , Interneuronas/patología , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Transgénicos , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Óxido Nítrico Sintasa de Tipo I/deficiencia , Parvalbúminas/deficiencia , Transducción de Señal , Somatostatina/deficiencia , Transgenes , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...