Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros











Intervalo de año de publicación
1.
Neuropsychobiology ; 83(2): 101-113, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38744261

RESUMEN

INTRODUCTION: The brain-derived neurotrophic factor (BDNF) and transcription nuclear factor erythroid 2-related factor-2 (NRF-2) play an important role in Alzheimer's disease (AD). However, the interactive involvement of BDNF and NRF-2 in respect to antioxidant mechanisms in different parts of the AD brain is still unclear. Considering the above condition, used S-nitrosoglutathione (GSNO) to examine whether it modulates the BDNF and NRF-2 levels to activate signaling pathway to promote antioxidant levels in AD brains. METHOD: AD was induced by intracerebroventricular infusion of streptozotocin (ICV-STZ, 3 mg/kg) in Wistar rats. The effect of GSNO was analyzed by evaluating the retention of memory in months 1, 2, and 3. After the behavior study, rats were sacrificed and accessed the amyloid beta (Aß)-40, Aß42, glutathione (GSH), BDNF, and NRF-2 levels in the hippocampus, cortex, and amygdala tissue. RESULTS: Pretreatment with GSNO (50 µg/kg/intraperitoneal/day) restored the BDNF, and NRF-2 levels toward normalcy as compared with ICV-STZ + saline-treated animals. Also, GSNO treatment reversed the oxidative stress and increased the GSH levels toward normal levels. Further, reduced Aß levels and neuronal loss in different brain regions. As a result, GSNO treatment improved the cognitive deficits in ICV-STZ-treated rats. CONCLUSION: The results showed that endogenous nitric oxide donor GSNO improved the cognitive deficits and ICV-STZ-induced AD pathological conditions, possibly via attenuating the oxidative stress. Hence, the above finding supported that GSNO treatment may activate BDNF and NRF-2 antioxidant signaling pathways in the AD brain to normalize oxidative stress, which is the main causative factor for ICV-STZ-induced AD pathogenesis.


Asunto(s)
Enfermedad de Alzheimer , Factor Neurotrófico Derivado del Encéfalo , Modelos Animales de Enfermedad , Trastornos de la Memoria , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Ratas Wistar , S-Nitrosoglutatión , Transducción de Señal , Estreptozocina , Animales , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Estrés Oxidativo/efectos de los fármacos , S-Nitrosoglutatión/farmacología , S-Nitrosoglutatión/administración & dosificación , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/efectos de los fármacos , Ratas , Transducción de Señal/efectos de los fármacos , Masculino , Estreptozocina/farmacología , Estreptozocina/administración & dosificación , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/tratamiento farmacológico , Antioxidantes/farmacología , Antioxidantes/administración & dosificación
2.
Biomed Pharmacother ; 174: 116540, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38579400

RESUMEN

Rheumatoid arthritis (RA) is characterized by high level of reactive oxygen species (ROS) and proinflammatory cytokines, which facilitate the activation of the inflammatory signaling such as NF-κB pathway and exacerbate the development of inflammation. Herein, we designed a nanodrug by encapsulating the NO donor S-nitrosoglutathione (GSNO) into an emulsion and coating the surface with a polydopamine (PDA) layer to yield GSNO@PDA, which simultaneously scavenged the extra ROS and suppressed NF-κB signaling for potent RA treatment. To enhance the cellular uptake and NO generation efficiency, dextran sulfate (DS) and Cu2+ were anchored on the surface of GSNO@PDA to obtain the final formulation GSNO@PDA@DS. Our results demonstrated that GSNO@PDA@DS were successfully prepared and the modification of DS effectively boosted the cellular uptake of GSNO@PDA@DS. Moreover, GSNO@PDA@DS lowered cellular ROS and elevated intracellular NO, resulting in a decrease of M1 phenotype, inhibition of NF-κB pathway and down-regulation of proinflammatory cytokine tumor necrosis factor-α (TNF-α). Further in vivo studies confirmed that GSNO@PDA@DS significantly relieved symptoms and bone erosion by regulating the microenvironment of RA, highlighting the potential of GSNO@PDA@DS for RA therapy through ROS scavenging and NO-mediated suppression of inflammatory signaling.


Asunto(s)
Artritis Reumatoide , FN-kappa B , Donantes de Óxido Nítrico , Polímeros , Especies Reactivas de Oxígeno , S-Nitrosoglutatión , Especies Reactivas de Oxígeno/metabolismo , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/metabolismo , Animales , Donantes de Óxido Nítrico/farmacología , Donantes de Óxido Nítrico/administración & dosificación , Ratones , FN-kappa B/metabolismo , S-Nitrosoglutatión/farmacología , S-Nitrosoglutatión/administración & dosificación , Células RAW 264.7 , Polímeros/química , Indoles/farmacología , Indoles/administración & dosificación , Depuradores de Radicales Libres/farmacología , Depuradores de Radicales Libres/administración & dosificación , Sinergismo Farmacológico , Masculino , Transducción de Señal/efectos de los fármacos , Sulfato de Dextran , Factor de Necrosis Tumoral alfa/metabolismo , Óxido Nítrico/metabolismo , Sistemas de Liberación de Medicamentos/métodos
3.
Biomolecules ; 11(11)2021 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-34827584

RESUMEN

The COVID-19 pandemic has escalated the occurrence of hypoxia including thrombotic stroke worldwide, for which nitric oxide (NO) therapy seems very promising and translatable. Therefore, various modes/routes of NO-delivery are now being tested in different clinical trials for safer, faster, and more effective interventions against ischemic insults. Intravenous (IV) infusion of S-Nitrosoglutathione (GSNO), the major endogenous molecular pool of NO, has been reported to protect against mechanical cerebral ischemia-reperfusion (IR); however, it has been never tested in any kind of "clinically" relevant thromboembolic stroke models with or without comorbidities and in combination with the thrombolytic reperfusion therapy. Moreover, "IV-effects" of higher dose of GSNO following IR-injury have been contradicted to augment stroke injury. Herein, we tested the hypothesis that nebulization of low-dose GSNO will not alter blood pressure (BP) and will mitigate stroke injury in diabetic mice via enhanced cerebral blood flow (CBF) and brain tissue oxygenation (PbtO2). GSNO-nebulization (200 µg/kgbwt) did not alter BP, but augmented the restoration of CBF, improved behavioral outcomes and reduced stroke injury. Moreover, GSNO-nebulization increased early reoxygenation of brain tissue/PbtO2 as measured at 6.5 h post-stroke following thrombolytic reperfusion, and enervated unwanted effects of late thrombolysis in diabetic stroke. We conclude that the GSNO-nebulization is safe and effective for enhancing collateral microvascular perfusion in the early hours following stroke. Hence, nebulized-GSNO therapy has the potential to be developed and translated into an affordable field therapy against ischemic events including strokes, particularly in developing countries with limited healthcare infrastructure.


Asunto(s)
Complicaciones de la Diabetes/tratamiento farmacológico , Diabetes Mellitus/tratamiento farmacológico , Hemorragia/prevención & control , S-Nitrosoglutatión/administración & dosificación , Accidente Cerebrovascular/complicaciones , Terapia Trombolítica/efectos adversos , Animales , Conducta Animal , Presión Sanguínea , Barrera Hematoencefálica , COVID-19/epidemiología , Hemorragia/complicaciones , Hipoxia , Infusiones Intravenosas , Flujometría por Láser-Doppler , Masculino , Ratones , Ratones Endogámicos C57BL , Microcirculación , Nebulizadores y Vaporizadores , Fármacos Neuroprotectores/farmacología , Perfusión , Daño por Reperfusión/tratamiento farmacológico , Riesgo , Estrés Mecánico
4.
Biochem Pharmacol ; 168: 352-365, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31374192

RESUMEN

The epigenetic signatures associated with cognitive decline driven by lack of estrogen in post-menopausal state, is not well-understood. The present study is an attempt to unravel the epigenetic mechanisms involved in cognitive impairment preceding ovariectomy in mice and evaluate the protective effects of S-nitrosoglutathione (GSNO). A significant decline in cognitive functions was observed in mice following ovariectomy as assessed by Morris water maze and Novel object recognition test. Administration of GSNO (100 µg/kg body weight, orally) daily for 4 weeks was found to ameliorate cognitive deficits observed in ovariectomised (OVX) mice. The activity of histone acetyl-transferase (HAT) was significantly disrupted in cortex and hippocampus of OVX mice. This was accompanied by increased activity of histone deacetylase (HDAC) and increased levels of HDAC-2, HDAC-3 causing lowered acetylated histone (H)3 levels. Reduced H3 acetylation triggers epigenetic repression of brain derived neurotrophic factor (BDNF) in cortex and hippocampus of OVX mice that may be responsible for neuronal damage and cognitive impairment. GSNO supplementation to OVX mice was able to reinstate HAT(CBP/p300) and HDAC balance through S-nitrosylation. GSNO restored histone acetylation at BDNF promoters (pII, pIV) thereby ameliorating BDNF levels and improving brain morphology and cognition. The study suggests that GSNO improves cognitive function in OVX mice by modulating epigenetic programming.


Asunto(s)
Disfunción Cognitiva/prevención & control , Epigénesis Genética/efectos de los fármacos , S-Nitrosoglutatión/farmacología , Animales , Conducta Animal/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/sangre , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Inmunoprecipitación de Cromatina , Estradiol/sangre , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ovariectomía , S-Nitrosoglutatión/administración & dosificación
5.
Br J Pharmacol ; 176(12): 2049-2062, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30822355

RESUMEN

BACKGROUND AND PURPOSE: Angiotensin II (AngII) and NO regulate the cerebral circulation. AngII AT1 receptors exert ligand-dependent and ligand-independent (myogenic tone [MT]) vasoconstriction of cerebral vessels. NO induces post-translational modifications of proteins such as S-nitrosation (redox modification of cysteine residues). In cultured cells, S-nitrosation decreases AngII's affinity for the AT1 receptor. The present work evaluated the functional consequences of S-nitrosation on both AngII-dependent and AngII-independent cerebrovascular responses. EXPERIMENTAL APPROACH: S-Nitrosation was induced in rat isolated middle cerebral arteries by pretreatment with the NO donors, S-nitrosoglutathione (GSNO) or sodium nitroprusside (SNP). Agonist-dependent activation of AT1 receptors was evaluated by obtaining concentration-response curves to AngII. Ligand-independent activation of AT1 receptors was evaluated by calculating MT (active vs. passive diameter) at pressures ranging from 20 to 200 mmHg in the presence or not of a selective AT1 receptor inverse agonist. KEY RESULTS: GSNO or SNP completely abolished the AngII-dependent AT1 receptor-mediated vasoconstriction of cerebral arteries. GSNO had no impact on responses to other vasoconstrictors sharing (phenylephrine, U46619) or not (5-HT) the same signalling pathway. MT was reduced by GSNO, and the addition of losartan did not further decrease MT, suggesting that GSNO blocks AT1 receptor-dependent MT. Ascorbate (which reduces S-nitrosated compounds) restored the response to AngII but not the soluble GC inhibitor ODQ, suggesting that these effects are mediated by S-nitrosation rather than by S-nitrosylation. CONCLUSIONS AND IMPLICATIONS: In rat middle cerebral arteries, GSNO pretreatment specifically affects the AT1 receptor and reduces both AngII-dependent and AngII-independent activation, most likely through AT1 receptor S-nitrosation.


Asunto(s)
Arterias Cerebrales/efectos de los fármacos , Receptor de Angiotensina Tipo 1/metabolismo , S-Nitrosoglutatión/farmacología , Angiotensina II/farmacología , Animales , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Ligandos , Masculino , Óxido Nítrico/metabolismo , Nitrosación/efectos de los fármacos , Ratas , Ratas Wistar , S-Nitrosoglutatión/administración & dosificación , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
6.
Eur J Pharm Biopharm ; 132: 94-102, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30223029

RESUMEN

Methicillin-resistant Staphylococcus aureus (MRSA)-infected wounds have become a significant clinical issue worldwide. Recently, nitric oxide (NO) has emerged as a potent antibacterial agent against MRSA infections and a wound-healing enhancer. Nevertheless, clinical applications of NO have been largely restricted by its gaseous state and short half-life. In this study, our aim was to develop S-nitrosoglutathione (GSNO, an endogenous NO donor)-loaded poly(lactic-co-glycolic acid) [PLGA] microparticles (GSNO-MPs) that release NO over a prolonged period, to accelerate the healing of MRSA-infected wounds with less frequent dosing. GSNO was successfully encapsulated into PLGA microparticles by a solid-in-oil-in-water emulsion solvent evaporation method. Scanning electron microscopy and X-ray diffraction analyses confirmed the successful fabrication of GSNO-MPs. The latter released NO in a prolonged manner over 7 days and exerted a remarkable antibacterial activity against MRSA in a concentration- and time-dependent manner. Moreover, GSNO-MPs had good antibacterial efficacy and were found to accelerate wound healing in a mouse model of MRSA-infected wounds. Therefore, NO-releasing MPs devised in this study may be a promising option for the treatment of cutaneous wounds infected by drug-resistant bacteria such as MRSA.


Asunto(s)
S-Nitrosoglutatión/administración & dosificación , Infecciones Estafilocócicas/tratamiento farmacológico , Cicatrización de Heridas/efectos de los fármacos , Animales , Antibacterianos/administración & dosificación , Antibacterianos/farmacología , Preparaciones de Acción Retardada , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Portadores de Fármacos/química , Masculino , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Microscopía Electrónica de Rastreo , Microesferas , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/administración & dosificación , Donantes de Óxido Nítrico/farmacología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , S-Nitrosoglutatión/farmacología , Infecciones Estafilocócicas/microbiología , Factores de Tiempo , Difracción de Rayos X
7.
Neurochem Int ; 118: 205-216, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29792953

RESUMEN

Diabetes is associated with increased blood brain barrier (BBB) permeability resulting in neurological deficits. The present study investigated the role of S-nitrosoglutathione (GSNO) on tight junction proteins and cell adhesion molecules in streptozotocin-induced diabetic mice. Diabetes was induced by intraperitoneal injection of streptozotocin (40 mg/kg body weight) for 5 days in mice. GSNO was administered daily (100 µg/kg body weight, orally) for 8 weeks after the induction of diabetes. A significant decline was observed in the cognitive ability of diabetic animals assessed using radial arm maze test. A significant increase was observed in nitrotyrosine levels in cortex and hippocampus of diabetic mice. Relative mRNA and protein expression of tight junction proteins viz; zona occludens-1 (ZO-1) and occludin were significantly lower in the microvessels isolated from cortex and hippocampus of diabetic animals, whereas expression of claudin-5 was unaltered. Immunofluorescence of tight junction proteins confirmed loss of ZO-1 and occludin in the diabetic brain. Furthermore, significant increase in interstitial cell adhesion molecule (ICAM)-1 and vascular cell adhesion molecule (VCAM)-1 mRNA and protein levels was observed in diabetic animals. Ultrastructure of microvessels from diabetic brain was also altered thereby confirming BBB disruption. GSNO administration to diabetic animals, on the other hand, was able to ameliorate loss of ZO-1 and occludin as well as normalize ICAM-1 and VCAM-1 expression, restore BBB integrity, and improve cognitive deficits. The findings clearly suggest that GSNO is a therapeutic molecule with potential to protect BBB and prevent diabetes induced neurological deficits.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Moléculas de Adhesión Celular/biosíntesis , Hiperglucemia/metabolismo , Fármacos Neuroprotectores/administración & dosificación , S-Nitrosoglutatión/administración & dosificación , Proteínas de Uniones Estrechas/biosíntesis , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/prevención & control , Hiperglucemia/patología , Hiperglucemia/prevención & control , Masculino , Ratones
8.
J Neurophysiol ; 120(2): 720-728, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29742029

RESUMEN

The nitric oxide (NO)/cyclic GMP signaling pathway has been suggested to be important in the formation of olfactory memory in insects. However, the molecular targets of the NO signaling cascade in the central neurons associated with olfactory learning and memory have not been fully analyzed. In this study, we investigated the effects of NO donors on single voltage-dependent Na+ channels in intrinsic neurons, called Kenyon cells, in the mushroom bodies in the brain of the cricket. Step depolarization on cell-attached patch membranes induces single-channel currents with fast-activating and -inactivating brief openings at the beginning of the voltage steps followed by more persistently recurring brief openings all along the 150-ms pulses. Application of the NO donor S-nitrosoglutathione (GSNO) increased the number of channel openings of both types of single Na+ channels. This excitatory effect of GSNO on the activity of these Na+ channels was diminished by KT5823, an inhibitor of protein kinase G (PKG), indicating an involvement of PKG in the downstream pathway of NO. Application of KT5823 alone decreased the activity of the persistent Na+ channels without significant effects on the fast-inactivating Na+ channels. The membrane-permeable cGMP analog 8Br-cGMP increased the number of channel openings of both types of single Na+ channels, similar to the action of NO. Taken together, these results indicate that NO acts as a critical modulator of both fast-inactivating and persistent Na+ channels and that persistent Na+ channels are constantly upregulated by the endogenous cGMP/PKG signaling cascade. NEW & NOTEWORTHY This study clarified that nitric oxide (NO) increases the activity of both fast-inactivating and persistent Na+ channels via the cGMP/PKG signaling cascade in cricket Kenyon cells. The persistent Na+ channels are also found to be upregulated constantly by endogenous cGMP/PKG signaling. On the basis of the present results and the results of previous studies, we propose a hypothetical model explaining NO production and NO-dependent memory formation in cricket large Kenyon cells.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/fisiología , GMP Cíclico/fisiología , Gryllidae/fisiología , Cuerpos Pedunculados/fisiología , Neuronas/fisiología , Óxido Nítrico/fisiología , Canales de Sodio Activados por Voltaje/fisiología , Animales , Masculino , Potenciales de la Membrana , Modelos Neurológicos , Donantes de Óxido Nítrico/administración & dosificación , S-Nitrosoglutatión/administración & dosificación , Transducción de Señal
9.
Neurotoxicology ; 66: 58-65, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29522778

RESUMEN

The present study evaluated the effects of s-nitrosoglutathione (GSNO), a nitrosothiol and sustained NO releaser, on experimental model of sporadic Alzheimer`s disease (sAD) in rats. Levels of Aß40, Aß42 and BDNF were assessed in brain hippocampal homogenates for correlative purposes. Intracerebroventricular-Streptozotocin (icv-STZ) induced increased escape latencies (acquisition) and reduced time in target quadrant (probe trial) in Morris Water Maze (MWM) test at 3 months post icv-STZ administration. These behavioural changes were associated with increased Aß depositions and lowered BDNF levels in brain hippocampal homogenates. Pre-treatment with GSNO (50 µg/kg/day), reduced the icv-STZ induced cognitive deficits in acquisition and probe trials in the MWM. The icv-STZ induced elevations in Aß40 and Aß42 and reduced levels of BDNF in hippocampal homogenates were also attenuated after GSNO treatment in these rats. The NO-precursor, l-arginine (100 mg/kg) induced similar effects on behavioural and biochemical parameters tested but was marginally less consistent as compared to those seen with GSNO. The results suggest that GSNO ameliorates the cognitive deficits and associated brain biochemical changes in this experimental model of sporadic AD, and NO-BDNF interactions could play crucial role in these effects.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/psicología , Fármacos Neuroprotectores/administración & dosificación , Óxido Nítrico/metabolismo , S-Nitrosoglutatión/administración & dosificación , Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Donantes de Óxido Nítrico/metabolismo , Fragmentos de Péptidos/metabolismo , Ratas Wistar , S-Nitrosoglutatión/metabolismo , Estreptozocina/administración & dosificación
10.
Antioxid Redox Signal ; 27(7): 433-451, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28006950

RESUMEN

AIMS: Cigarette smoke (CS)-mediated acquired cystic fibrosis transmembrane conductance regulator (CFTR)-dysfunction, autophagy-impairment, and resulting inflammatory-oxidative/nitrosative stress leads to chronic obstructive pulmonary disease (COPD)-emphysema pathogenesis. Moreover, nitric oxide (NO) signaling regulates lung function decline, and low serum NO levels that correlates with COPD severity. Hence, we aim to evaluate here the effects and mechanism(s) of S-nitrosoglutathione (GSNO) augmentation in regulating inflammatory-oxidative stress and COPD-emphysema pathogenesis. RESULTS: Our data shows that cystic fibrosis transmembrane conductance regulator (CFTR) colocalizes with aggresome bodies in the lungs of COPD subjects with increasing emphysema severity (Global Initiative for Chronic Obstructive Lung Disease [GOLD] I - IV) compared to nonemphysema controls (GOLD 0). We further demonstrate that treatment with GSNO or S-nitrosoglutathione reductase (GSNOR)-inhibitor (N6022) significantly inhibits cigarette smoke extract (CSE; 5%)-induced decrease in membrane CFTR expression by rescuing it from ubiquitin (Ub)-positive aggresome bodies (p < 0.05). Moreover, GSNO restoration significantly (p < 0.05) decreases CSE-induced reactive oxygen species (ROS) activation and autophagy impairment (decreased accumulation of ubiquitinated proteins in the insoluble protein fractions and restoration of autophagy flux). In addition, GSNO augmentation inhibits protein misfolding as CSE-induced colocalization of ubiquitinated proteins and LC3B (in autophagy bodies) is significantly reduced by GSNO/N6022 treatment. We verified using the preclinical COPD-emphysema murine model that chronic CS (Ch-CS)-induced inflammation (interleukin [IL]-6/IL-1ß levels), aggresome formation (perinuclear coexpression/colocalization of ubiquitinated proteins [Ub] and p62 [impaired autophagy marker], and CFTR), oxidative/nitrosative stress (p-Nrf2, inducible nitric oxide synthase [iNOS], and 3-nitrotyrosine expression), apoptosis (caspase-3/7 activity), and alveolar airspace enlargement (Lm) are significantly (p < 0.05) alleviated by augmenting airway GSNO levels. As a proof of concept, we demonstrate that GSNO augmentation suppresses Ch-CS-induced perinuclear CFTR protein accumulation (p < 0.05), which restores both acquired CFTR dysfunction and autophagy impairment, seen in COPD-emphysema subjects. INNOVATION: GSNO augmentation alleviates CS-induced acquired CFTR dysfunction and resulting autophagy impairment. CONCLUSION: Overall, we found that augmenting GSNO levels controls COPD-emphysema pathogenesis by reducing CS-induced acquired CFTR dysfunction and resulting autophagy impairment and chronic inflammatory-oxidative stress. Antioxid. Redox Signal. 27, 433-451.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Enfisema Pulmonar/tratamiento farmacológico , S-Nitrosoglutatión/administración & dosificación , Humo/efectos adversos , Animales , Autofagia/efectos de los fármacos , Benzamidas/farmacología , Línea Celular , Humanos , Ratones , Estrés Oxidativo/efectos de los fármacos , Agregado de Proteínas/efectos de los fármacos , Enfisema Pulmonar/inducido químicamente , Enfisema Pulmonar/inmunología , Pirroles/farmacología , S-Nitrosoglutatión/farmacología , Productos de Tabaco
11.
PLoS One ; 11(4): e0153716, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27116554

RESUMEN

S-nitrosoglutathione (GSNO) is a nitric oxide (NO) donor, which exerts antioxidant, anti-inflammatory, and microbicidal actions. Intragingival application of GSNO was already shown to decrease alveolar bone loss, inflammation and oxidative stress in an experimental periodontal disease (EPD) model. In the present study, we evaluated the potential therapeutic effect of topical applications of hydroxypropylmethylcellulose (HPMC)/GSNO solutions on EPD in Wistar rats. EPD was induced by placing a sterilized nylon (3.0) thread ligature around the cervix of the second left upper molar of the animals, which received topical applications of a HPMC solutions containing GSNO 2 or 10 mM or vehicle (HPMC solution), 1 h prior to the placement of the ligature and then twice daily until sacrifice on day 11. Treatment with HPMC/GSNO 10 mM solution significantly reduced alveolar bone loss, oxidative stress and TNF-α e IL-1ß levels in the surrounding gingival tissue, and led to a decreased transcription of RANK and TNF-α genes and elevated bone alkaline phosphatase, compared to the HPMC group. In conclusion, topical application of HPMC/GSNO solution is a potential treatment to reduce inflammation and bone loss in periodontal disease.


Asunto(s)
Derivados de la Hipromelosa/administración & dosificación , Enfermedades Periodontales/tratamiento farmacológico , S-Nitrosoglutatión/administración & dosificación , Administración Tópica , Pérdida de Hueso Alveolar/tratamiento farmacológico , Pérdida de Hueso Alveolar/metabolismo , Pérdida de Hueso Alveolar/patología , Animales , Modelos Animales de Enfermedad , Encía/efectos de los fármacos , Encía/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Interleucina-1beta/metabolismo , Masculino , Donantes de Óxido Nítrico/administración & dosificación , Óxido Nítrico Sintasa de Tipo II/metabolismo , Enfermedades Periodontales/metabolismo , Enfermedades Periodontales/patología , Ratas , Ratas Wistar , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Soluciones , Fosfatasa Ácida Tartratorresistente/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
12.
PLoS One ; 10(12): e0144659, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26646285

RESUMEN

Treatment of stroke, especially during the first hours or days, is still lacking. S-nitrosoglutathione (GSNO), a cerebroprotective agent with short life time, may help if administered early with a sustain delivery while avoiding intensive reduction in blood pressure. We developed in situ forming implants (biocompatible biodegradable copolymer) and microparticles (same polymer and solvent emulsified with an external oily phase) of GSNO to lengthen its effects and allow cerebroprotection after a single subcutaneous administration to Wistar rats. Arterial pressure was recorded for 3 days (telemetry, n = 14), whole-blood platelet aggregation up to 13 days (aggregometry, n = 58), and neurological score, cerebral infarct size and edema volume for 2 days after obstruction of the middle cerebral artery by autologous blood clots (n = 30). GSNO-loaded formulations (30 mg/kg) induced a slighter and longer hypotension (-10 vs. -56 ± 6 mmHg mean arterial pressure, 18 h vs. 40 min) than free GSNO at the same dose. The change in pulse pressure (-50%) lasted even up to 42 h for microparticles. GSNO-loaded formulations (30 mg/kg) prevented the transient 24 h hyper-aggregability observed with free GSNO and 7.5 mg/kg-loaded formulations. When injected 2 h after stroke, GSNO-loaded microparticles (30 mg/kg) reduced neurological score at 24 (-62%) and 48 h (-75%) vs. empty microparticles and free GSNO 7.5 mg/kg and, compared to free GSNO, divided infarct size by 10 and edema volume by 8 at 48 h. Corresponding implants reduced infarct size and edema volume by 2.5 to 3 times. The longer (at least 2 days) but slight effects on arterial pressures show sustained delivery of GSNO-loaded formulations (30 mg/kg), which prevent transient platelet hyper-responsiveness and afford cerebroprotection against the consequences of stroke. In conclusion, in situ GSNO-loaded formulations are promising candidates for the treatment of stroke.


Asunto(s)
Fármacos Neuroprotectores/uso terapéutico , S-Nitrosoglutatión/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Presión Sanguínea/efectos de los fármacos , Modelos Animales de Enfermedad , Inyecciones Subcutáneas , Masculino , Microesferas , Fármacos Neuroprotectores/administración & dosificación , Agregación Plaquetaria/efectos de los fármacos , Ratas , Ratas Wistar , S-Nitrosoglutatión/administración & dosificación , Telemetría
13.
Int J Pharm ; 495(1): 354-361, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26319632

RESUMEN

S-nitrosoglutathione (GSNO) is a nitric oxide (NO) donor with therapeutic potential for cardiovascular disease treatment. Chronic oral treatment with GSNO is limited by high drug sensitivity to the environment and limited oral bioavailability, requiring the development of delivery systems able to sustain NO release. The present work describes new platforms based on polymer nanocomposite particles for the delivery of GSNO. Five types of optimized nanocomposite particles have been developed (three based on chitosan, two based on alginate sodium). Those nanocomposite particles encapsulate GSNO with high efficiency from 64% to 70% and an average size of 13 to 61 µm compatible with oral delivery. Sustained release of GSNO in vitro was achieved. Indeed, chitosan nanocomposites discharged their payload within 24h; whereas alginate nanocomposites released GSNO more slowly (10% of GSNO was still remaining in the dosage form after 24h). Their cytocompatibility toward intestinal Caco-2 cells (MTT assay) was acceptable (IC50: 6.07 ± 0.07-9.46 ± 0.08 mg/mL), demonstrating their suitability as oral delivery systems for GSNO. These delivery systems presented efficient GSNO loading and sustained release as well as cytocompatibility, showing their promise as a means of improving the oral bioavailability of GSNO and as a potential new treatment.


Asunto(s)
Alginatos/química , Quitosano/química , Nanocompuestos/química , Donantes de Óxido Nítrico/administración & dosificación , S-Nitrosoglutatión/administración & dosificación , Administración Oral , Células CACO-2 , Química Farmacéutica/métodos , Preparaciones de Acción Retardada , Portadores de Fármacos/química , Liberación de Fármacos , Estabilidad de Medicamentos , Ácido Glucurónico/química , Ácidos Hexurónicos/química , Humanos , Tamaño de la Partícula
14.
Clin Hemorheol Microcirc ; 60(4): 397-404, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-24958331

RESUMEN

Glutathione is an abundant molecule inside erythrocyte, originating S-nitrosoglutathione (GSNO) by reacting with nitric oxide (NO). GSNO has been regarded as a store and transporter of NO, with significant interest as a potential therapeutic agent, acting as an NO donor.NO metabolism inside the erythrocyte generates several derivatives, which can be altered by external and internal stimuli such as acetylcholine (ACh), a natural substrate of acetylcholinesterase (AChE). In spite of the knowledge gained in the last decades concerning NO efflux in erythrocytes little is known regarding erythrocyte GSNO efflux, which has also a significant role in microcirculation. Hence, the objective of this research was to evaluate the efflux of GSNO, concomitant with the efflux of NO, after stimulation with AChE effectors. To achieve these goals, the in vitro effect of AChE modulators - ACh and timolol - in erythrocyte NO and GSNO were studied. Timolol is an erythrocyte AChE inhibitor. Venous blood samples were collected from 18 healthy Caucasian men. For each blood sample, erythrocyte suspensions were obtained and incubated in the absence (controls) and presence of ACh and timolol maleate (10 µM final concentration of each modulator). Both timolol and ACh induced significant GSNO efflux in the erythrocyte when compared to the control; however the efflux was lower in the presence of timolol compared to ACh. Although erythrocyte NO efflux in presence of timolol is similar to the control, the efflux decreased when compared to the ACh treatment. The presence of timolol induces significant decrease of intra-erythrocyte GSNO levels, relative to control and ACh treatment. In conclusion, when erythrocytes were stimulated with ACh or timolol, GSNO efflux occurred associated with NO efflux. These new results bring new insight into the metabolism of erythrocyte NO and new possible therapeutic applications for GSNO.


Asunto(s)
Acetilcolina/farmacología , Eritrocitos/efectos de los fármacos , Óxido Nítrico/sangre , S-Nitrosoglutatión/uso terapéutico , Humanos , Masculino , S-Nitrosoglutatión/administración & dosificación
15.
Biofizika ; 60(6): 1157-65, 2015.
Artículo en Ruso | MEDLINE | ID: mdl-26841511

RESUMEN

The anti-tumor activity of the binuclear form of dinitrosyl iron complexes with glutathione against Lewis lung carcinoma, found earlier upon intraperitoneal administration of the complexes, was also observed when this preparation was injected subcutaneously. A 100 µM/kg subcutaneous dose of the complex being used daily (as calculated per one iron atom in binuclear dinitrosyl iron complexes) for 10 or 15 days, inhibited the tumor growth by 43%. The effect was observed during the first two weeks after tumor transplantation. After that, the tumors began to grow at the rate equal to or even higher than that one for control animals. The mean survival time for treated mice exceeded the control values by 30%. Binuclear dinitrosyl iron complexes administered intraperitoneally was also effective against Ca-755 adenocarcinoma. However, in this case the mean survival time for treated animals increased only by 7%. The anti-tumor activity of S-nitrosoglutathione against Lewis lung carcinoma growth inhibition by 70% and Ca-755 adenocarcinoma growth inhibition by 90% was also shown. However, unlike binuclear dinitrosyl iron complexes the anti-tumor effect of S-nitrosoglutathione decreased when a daily dose of the compound increased (from 200 to 400 µM/kg) The initial anti-tumor effect of binuclear dinitrosyl iron complexes and S-nitrosoglutathione is suggested to be due to NO released from both compounds. A subsequent suppression of the effect is determined by the development of anti-nitrosative and anti-oxidant defense systems in tumors.


Asunto(s)
Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Glutatión/administración & dosificación , Hierro/administración & dosificación , Óxidos de Nitrógeno/administración & dosificación , S-Nitrosoglutatión/administración & dosificación , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Animales , Carcinoma Pulmonar de Lewis/patología , Cisteína/química , Cisteína/metabolismo , Glutatión/química , Humanos , Hierro/química , Ratones , Óxido Nítrico/química , Óxido Nítrico/metabolismo , Óxidos de Nitrógeno/química , S-Nitrosoglutatión/química
16.
PLoS One ; 9(12): e113378, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25478918

RESUMEN

INTRODUCTION: Mucositis induced by anti-neoplastic drugs is an important, dose-limiting and costly side-effect of cancer therapy. AIM: To evaluate the effect of the topical application of S-nitrosoglutathione (GSNO), a nitric oxide donor, on 5-fluorouracil (5-FU)-induced oral mucositis in hamsters. MATERIALS AND METHODS: Oral mucositis was induced in male hamsters by two intraperitoneal administrations of 5-FU on the first and second days of the experiment (60 and 40 mg/kg, respectively) followed by mechanical trauma on the fourth day. Animals received saline, HPMC or HPMC/GSNO (0.1, 0.5 or 2.0 mM) 1 h prior to the 5-FU injection and twice a day for 10 or 14 days. Samples of cheek pouches were harvested for: histopathological analysis, TNF-α and IL-1ß levels, immunohistochemical staining for iNOS, TNF-α, IL-1ß, Ki67 and TGF-ß RII and a TUNEL assay. The presence and levels of 39 bacterial taxa were analyzed using the Checkerboard DNA-DNA hybridization method. The profiles of NO released from the HPMC/GSNO formulations were characterized using chemiluminescence. RESULTS: The HPMC/GSNO formulations were found to provide sustained release of NO for more than 4 h at concentration-dependent rates of 14 to 80 nmol/mL/h. Treatment with HPMC/GSNO (0.5 mM) significantly reduced mucosal damage, inflammatory alterations and cell death associated with 5-FU-induced oral mucositis on day 14 but not on day 10. HPMC/GSNO administration also reversed the inhibitory effect of 5-FU on cell proliferation on day 14. In addition, we observed that the chemotherapy significantly increased the levels and/or prevalence of several bacterial species. CONCLUSION: Topical HPMC/GSNO accelerates mucosal recovery, reduces inflammatory parameters, speeds up re-epithelization and decreases levels of periodontopathic species in mucosal ulcers.


Asunto(s)
Inflamación/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , S-Nitrosoglutatión/administración & dosificación , Estomatitis/tratamiento farmacológico , Administración Tópica , Animales , Cricetinae , Modelos Animales de Enfermedad , Fluorouracilo/efectos adversos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/genética , Inflamación/patología , Interleucina-1beta/biosíntesis , Masculino , Neoplasias/patología , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Estomatitis/inducido químicamente , Estomatitis/genética , Estomatitis/patología , Factor de Crecimiento Transformador beta/biosíntesis , Factor de Necrosis Tumoral alfa/biosíntesis
17.
Eur J Pharmacol ; 741: 37-44, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25058904

RESUMEN

It has been established that intraperitoneal bolus administration of S-nitrosoglutathione (GS-NO) (12.5µmoles/kg; 10 injections in 10 days), beginning with day 4 after transplantation of two 2-mm autologous fragments of endometrial tissue onto the inner surface of the abdominal wall of rats with surgically induced (experimenta) endometriosis failed to prevent further growth of endometrioid (EMT) and additive tumors, while treatment of animals with dinitrosyl iron complexes (DNIC) with glutathione (12.5µmoles/kg, 10 injections in 10 days) suppressed tumor growth virtually completely. The histological analysis of EMT samples of GS-NO-treated rats revealed pathological changes characteristic of control (non-treated with GS-NO or DNIC) rats with experimental endometriosis. EPR studies established the presence of the active form of ribonucleotide reductase, a specific marker for rapidly proliferating tumors, in EMT samples of both control and GS-NO-treated animals. Noteworthy, in small-size EMT and adjacent tissues of DNIC-treated rats the active form of ribonucleotide reductase and pathological changes were not found.


Asunto(s)
Endometriosis/patología , Endometriosis/prevención & control , Glutatión/administración & dosificación , Hierro/administración & dosificación , Óxidos de Nitrógeno/administración & dosificación , S-Nitrosoglutatión/administración & dosificación , Animales , Combinación de Medicamentos , Femenino , Ratas , Ratas Wistar , Resultado del Tratamiento
19.
PLoS One ; 9(6): e97897, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24887420

RESUMEN

This study examines the role of s-nitrosylation in the growth of ovarian cancer using cell culture based and in vivo approaches. Using the nitrosylating agent, S-nitrosoglutathione (GSNO), a physiological nitric oxide molecule, we show that GSNO treatment inhibited proliferation of chemoresponsive and chemoresistant ovarian cancer cell lines (A2780, C200, SKVO3, ID8, OVCAR3, OVCAR4, OVCAR5, OVCAR7, OVCAR8, OVCAR10, PE01 and PE04) in a dose dependent manner. GSNO treatment abrogated growth factor (HB-EGF) induced signal transduction including phosphorylation of Akt, p42/44 and STAT3, which are known to play critical roles in ovarian cancer growth and progression. To examine the therapeutic potential of GSNO in vivo, nude mice bearing intra-peritoneal xenografts of human A2780 ovarian carcinoma cell line (2 × 10(6)) were orally administered GSNO at the dose of 1 mg/kg body weight. Daily oral administration of GSNO significantly attenuated tumor mass (p<0.001) in the peritoneal cavity compared to vehicle (phosphate buffered saline) treated group at 4 weeks. GSNO also potentiated cisplatin mediated tumor toxicity in an A2780 ovarian carcinoma nude mouse model. GSNO's nitrosylating ability was reflected in the induced nitrosylation of various known proteins including NFκB p65, Akt and EGFR. As a novel finding, we observed that GSNO also induced nitrosylation with inverse relationship at tyrosine 705 phosphorylation of STAT3, an established player in chemoresistance and cell proliferation in ovarian cancer and in cancer in general. Overall, our study underlines the significance of S-nitrosylation of key cancer promoting proteins in modulating ovarian cancer and proposes the therapeutic potential of nitrosylating agents (like GSNO) for the treatment of ovarian cancer alone or in combination with chemotherapeutic drugs.


Asunto(s)
Neoplasias Ováricas/tratamiento farmacológico , S-Nitrosoglutatión/uso terapéutico , Administración Oral , Animales , Biotina/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , ADN/metabolismo , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones Desnudos , Invasividad Neoplásica , Nitrosación/efectos de los fármacos , Neoplasias Ováricas/patología , Unión Proteica/efectos de los fármacos , S-Nitrosoglutatión/administración & dosificación , S-Nitrosoglutatión/farmacología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Ensayo de Tumor de Célula Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA