Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 174(4): 831-842.e12, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-30057115

RESUMEN

Overnutrition disrupts circadian metabolic rhythms by mechanisms that are not well understood. Here, we show that diet-induced obesity (DIO) causes massive remodeling of circadian enhancer activity in mouse liver, triggering synchronous high-amplitude circadian rhythms of both fatty acid (FA) synthesis and oxidation. SREBP expression was rhythmically induced by DIO, leading to circadian FA synthesis and, surprisingly, FA oxidation (FAO). DIO similarly caused a high-amplitude circadian rhythm of PPARα, which was also required for FAO. Provision of a pharmacological activator of PPARα abrogated the requirement of SREBP for FAO (but not FA synthesis), suggesting that SREBP indirectly controls FAO via production of endogenous PPARα ligands. The high-amplitude rhythm of PPARα imparted time-of-day-dependent responsiveness to lipid-lowering drugs. Thus, acquisition of rhythmicity for non-core clock components PPARα and SREBP1 remodels metabolic gene transcription in response to overnutrition and enables a chronopharmacological approach to metabolic disorders.


Asunto(s)
Ritmo Circadiano , Dieta/efectos adversos , Hígado/metabolismo , Obesidad/metabolismo , PPAR alfa/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Animales , Regulación de la Expresión Génica , Metabolismo de los Lípidos , Lipogénesis , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/patología , PPAR alfa/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética
2.
J Neurovirol ; 27(3): 367-378, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33876414

RESUMEN

In the brain, both HIV-1 and methamphetamine (meth) use result in increases in oxidative and nitrosative stress. This redox stress is thought to contribute to the pathogenesis of HIV-associated neurocognitive disorder (HAND) and further worsening cognitive activity in the setting of drug abuse. One consequence of such redox stress is aberrant protein S-nitrosylation, derived from nitric oxide, which may disrupt normal protein activity. Here, we report an improved, mass spectrometry-based technique to assess S-nitrosylated protein in human postmortem brains using selective enrichment of S-nitrosocysteine residues with an organomercury resin. The data show increasing S-nitrosylation of tricarboxylic acid (TCA) enzymes in the setting of HAND and HAND/meth use compared with HIV+ control brains without CNS pathology. The consequence is systematic inhibition of multiple TCA cycle enzymes, resulting in energy collapse that can contribute to the neuronal and synaptic damage observed in HAND and meth use.


Asunto(s)
Ciclo del Ácido Cítrico/efectos de los fármacos , Disfunción Cognitiva/metabolismo , Infecciones por VIH/metabolismo , Metanfetamina/efectos adversos , Procesamiento Proteico-Postraduccional , Trastornos Relacionados con Sustancias/metabolismo , Autopsia , Bancos de Muestras Biológicas , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/patología , Ciclo del Ácido Cítrico/genética , Disfunción Cognitiva/complicaciones , Disfunción Cognitiva/patología , Disfunción Cognitiva/virología , Cisteína/análogos & derivados , Cisteína/metabolismo , Infecciones por VIH/complicaciones , Infecciones por VIH/patología , Infecciones por VIH/virología , VIH-1/crecimiento & desarrollo , VIH-1/patogenicidad , Humanos , Masculino , Persona de Mediana Edad , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Mitocondrias/patología , Neuronas/efectos de los fármacos , Neuronas/enzimología , Neuronas/patología , Óxido Nítrico/metabolismo , S-Nitrosotioles/metabolismo , Trastornos Relacionados con Sustancias/complicaciones , Trastornos Relacionados con Sustancias/patología , Trastornos Relacionados con Sustancias/virología , Sinapsis/efectos de los fármacos , Sinapsis/patología
3.
Nitric Oxide ; 117: 1-6, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34536587

RESUMEN

Cysteine S-nitrosation mediates NO signaling and protein function under pathophysiological conditions. Herein, we provide a detailed protocol regarding the organic mercury chemoselective enrichment of S-nitrosated proteins and peptides. We discuss key aspects of the enrichment strategy and provide technical tips for the best performance of the experimental protocol.


Asunto(s)
Mercurio/química , Nitratos , Proteínas , Proteómica/métodos , Cromatografía , Cisteína/análisis , Cisteína/aislamiento & purificación , Cisteína/metabolismo , Nitratos/análisis , Nitratos/aislamiento & purificación , Nitratos/metabolismo , Óxido Nítrico/metabolismo , Nitrosación , Péptidos/análisis , Péptidos/aislamiento & purificación , Péptidos/metabolismo , Proteínas/análisis , Proteínas/aislamiento & purificación , Proteínas/metabolismo
4.
Nitric Oxide ; 106: 17-23, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33080411

RESUMEN

BACKGROUND: Chronic Kidney Disease (CKD) patients exhibit a reduced exercise capacity that impacts quality of life. Dietary nitrate supplementation has been shown to have favorable effects on exercise capacity in disease populations by reducing the oxygen cost of exercise. This study investigated whether dietary nitrates would acutely improve exercise capacity in CKD patients. METHODS AND RESULTS: In this randomized, double-blinded crossover study, 12 Stage 3-4 CKD patients (Mean ± SEM: Age, 60 ± 5yrs; eGFR, 50.3 ± 4.6 ml/min/1.73 m2) received an acute dose of 12.6 mmol of dietary nitrate in the form of concentrated beetroot juice (BRJ) and a nitrate depleted placebo (PLA). Skeletal muscle mitochondrial oxidative function was assessed using near-infrared spectroscopy. Cardiopulmonary exercise testing was performed on a cycle ergometer, with intensity increased by 25 W every 3 min until volitional fatigue. Plasma nitric oxide (NO) metabolites (NOm; nitrate, nitrite, low molecular weight S-nitrosothiols, and metal bound NO) were determined by gas-phase chemiluminescence. Plasma NOm values were significantly increased following BRJ (BRJ vs. PLA: 1074.4 ± 120.4 µM vs. 28.4 ± 6.6 µM, p < 0.001). Total work performed (44.4 ± 10.6 vs 39.6 ± 9.9 kJ, p = 0.03) and total exercise time (674 ± 85 vs 627 ± 86s, p = 0.04) were significantly greater following BRJ. Oxygen consumption at the ventilatory threshold was also improved by BRJ (0.90 ± 0.08 vs. 0.74 ± 0.06 L/min, p = 0.04). These changes occurred in the absence of improved skeletal muscle mitochondrial oxidative capacity (p = 0.52) and VO2peak (p = 0.35). CONCLUSIONS: Our findings demonstrate that inorganic nitrate can acutely improve exercise capacity in CKD patients. The effects of chronic nitrate supplementation on CKD related exercise intolerance should be investigated in future studies.


Asunto(s)
Tolerancia al Ejercicio/efectos de los fármacos , Nitratos/uso terapéutico , Insuficiencia Renal Crónica/dietoterapia , Adulto , Anciano , Beta vulgaris/química , Estudios Cruzados , Suplementos Dietéticos , Método Doble Ciego , Prueba de Esfuerzo/efectos de los fármacos , Femenino , Jugos de Frutas y Vegetales , Humanos , Masculino , Persona de Mediana Edad , Músculo Esquelético/efectos de los fármacos , Proyectos Piloto
5.
Int J Mol Sci ; 21(3)2020 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-32033212

RESUMEN

The placenta is metabolically active and supports the growth of the fetus. We hypothesize that deficits in the capacity of the placenta to maintain bioenergetic and metabolic stability during pregnancy may result in spontaneous preterm birth (SPTB). To explore this hypothesis, we performed a nested cased control study of metabolomic signatures in placentas from women with SPTB (<36 weeks gestation) compared to normal pregnancies (≥38 weeks gestation). To control for the effects of gestational age on placenta metabolism, we also studied a subset of metabolites in non-laboring preterm and term Rhesus monkeys. Comprehensive quantification of metabolites demonstrated a significant elevation in the levels of amino acids, prostaglandins, sphingolipids, lysolipids, and acylcarnitines in SPTB placenta compared to term placenta. Additional quantification of placental acylcarnitines by tandem mass spectrometry confirmed the significant elevation in SPTB human, with no significant differences between midgestation and term placenta in Rhesus macaque. Fatty acid oxidation as measured by the flux of 3H-palmitate in SPTB placenta was lower than term. Collectively, significant and biologically relevant alterations in the placenta metabolome were identified in SPTB placenta. Altered acylcarnitine levels and fatty acid oxidation suggest that disruption in normal substrate metabolism is associated with SPTB.


Asunto(s)
Placenta/metabolismo , Nacimiento Prematuro/metabolismo , Adulto , Estudios de Casos y Controles , Femenino , Feto/metabolismo , Edad Gestacional , Humanos , Recién Nacido , Metabolómica/métodos , Embarazo
6.
Circ Res ; 120(7): 1151-1161, 2017 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-27927683

RESUMEN

RATIONALE: Nitrate-rich beetroot juice has been shown to improve exercise capacity in heart failure with preserved ejection fraction, but studies using pharmacological preparations of inorganic nitrate are lacking. OBJECTIVES: To determine (1) the dose-response effect of potassium nitrate (KNO3) on exercise capacity; (2) the population-specific pharmacokinetic and safety profile of KNO3 in heart failure with preserved ejection fraction. METHODS AND RESULTS: We randomized 12 subjects with heart failure with preserved ejection fraction to oral KNO3 (n=9) or potassium chloride (n=3). Subjects received 6 mmol twice daily during week 1, followed by 6 mmol thrice daily during week 2. Supine cycle ergometry was performed at baseline (visit 1) and after each week (visits 2 and 3). Quality of life was assessed with the Kansas City Cardiomyopathy Questionnaire. The primary efficacy outcome, peak O2-uptake, did not significantly improve (P=0.13). Exploratory outcomes included exercise duration and quality of life. Exercise duration increased significantly with KNO3 (visit 1: 9.87, 95% confidence interval [CI] 9.31-10.43 minutes; visit 2: 10.73, 95% CI 10.13-11.33 minute; visit 3: 11.61, 95% CI 11.05-12.17 minutes; P=0.002). Improvements in the Kansas City Cardiomyopathy Questionnaire total symptom (visit 1: 58.0, 95% CI 52.5-63.5; visit 2: 66.8, 95% CI 61.3-72.3; visit 3: 70.8, 95% CI 65.3-76.3; P=0.016) and functional status scores (visit 1: 62.2, 95% CI 58.5-66.0; visit 2: 68.6, 95% CI 64.9-72.3; visit 3: 71.1, 95% CI 67.3-74.8; P=0.01) were seen after KNO3. Pronounced elevations in trough levels of nitric oxide metabolites occurred with KNO3 (visit 2: 199.5, 95% CI 98.7-300.2 µmol/L; visit 3: 471.8, 95% CI 377.8-565.8 µmol/L) versus baseline (visit 1: 38.0, 95% CI 0.00-132.0 µmol/L; P<0.001). KNO3 did not lead to clinically significant hypotension or methemoglobinemia. After 6 mmol of KNO3, systolic blood pressure was reduced by a maximum of 17.9 (95% CI -28.3 to -7.6) mm Hg 3.75 hours later. Peak nitric oxide metabolites concentrations were 259.3 (95% CI 176.2-342.4) µmol/L 3.5 hours after ingestion, and the median half-life was 73.0 (interquartile range 33.4-232.0) minutes. CONCLUSIONS: KNO3 is potentially well tolerated and improves exercise duration and quality of life in heart failure with preserved ejection fraction. This study reinforces the efficacy of KNO3 and suggests that larger randomized trials are warranted. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT02256345.


Asunto(s)
Insuficiencia Cardíaca/tratamiento farmacológico , Nitratos/farmacocinética , Compuestos de Potasio/farmacocinética , Volumen Sistólico , Anciano , Ejercicio Físico , Femenino , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/rehabilitación , Humanos , Masculino , Persona de Mediana Edad , Nitratos/efectos adversos , Compuestos de Potasio/efectos adversos , Calidad de Vida
7.
Circ Res ; 117(9): 793-803, 2015 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-26259881

RESUMEN

RATIONALE: The regulation of calcium (Ca(2+)) homeostasis by ß-adrenergic receptor (ßAR) activation provides the essential underpinnings of sympathetic regulation of myocardial function, as well as a basis for understanding molecular events that result in hypertrophic signaling and heart failure. Sympathetic stimulation of the ßAR not only induces protein phosphorylation but also activates nitric oxide-dependent signaling, which modulates cardiac contractility. Nonetheless, the role of nitric oxide in ßAR-dependent regulation of Ca(2+) handling has not yet been explicated fully. OBJECTIVE: To elucidate the role of protein S-nitrosylation, a major transducer of nitric oxide bioactivity, on ßAR-dependent alterations in cardiomyocyte Ca(2+) handling and hypertrophy. METHODS AND RESULTS: Using transgenic mice to titrate the levels of protein S-nitrosylation, we uncovered major roles for protein S-nitrosylation, in general, and for phospholamban and cardiac troponin C S-nitrosylation, in particular, in ßAR-dependent regulation of Ca(2+) homeostasis. Notably, S-nitrosylation of phospholamban consequent upon ßAR stimulation is necessary for the inhibitory pentamerization of phospholamban, which activates sarcoplasmic reticulum Ca(2+)-ATPase and increases cytosolic Ca(2+) transients. Coincident S-nitrosylation of cardiac troponin C decreases myocardial sensitivity to Ca(2+). During chronic adrenergic stimulation, global reductions in cellular S-nitrosylation mitigate hypertrophic signaling resulting from Ca(2+) overload. CONCLUSIONS: S-Nitrosylation operates in concert with phosphorylation to regulate many cardiac Ca(2+)-handling proteins, including phospholamban and cardiac troponin C, thereby playing an essential and previously unrecognized role in cardiac Ca(2+) homeostasis. Manipulation of the S-nitrosylation level may prove therapeutic in heart failure.


Asunto(s)
Calcio/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Óxido Nítrico/metabolismo , Receptores Adrenérgicos beta/metabolismo , Agonistas Adrenérgicos beta/farmacología , Aldehído Oxidorreductasas , Animales , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Células Cultivadas , Hipertrofia , Immunoblotting , Isoproterenol/farmacología , Ratones Noqueados , Ratones Transgénicos , Mutación , Miocardio/patología , Miocitos Cardíacos/citología , Fosforilación , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico , Transducción de Señal/efectos de los fármacos , Troponina I/genética , Troponina I/metabolismo
8.
J Biol Chem ; 290(16): 10486-94, 2015 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-25737446

RESUMEN

Very long acyl-CoA dehydrogenase (VLCAD) deficiency is a genetic pediatric disorder presenting with a spectrum of phenotypes that remains for the most part untreatable. Here, we present a novel strategy for the correction of VLCAD deficiency by increasing mutant VLCAD enzymatic activity. Treatment of VLCAD-deficient fibroblasts, which express distinct mutant VLCAD protein and exhibit deficient fatty acid ß-oxidation, with S-nitroso-N-acetylcysteine induced site-specific S-nitrosylation of VLCAD mutants at cysteine residue 237. Cysteine 237 S-nitrosylation was associated with an 8-17-fold increase in VLCAD-specific activity and concomitant correction of acylcarnitine profile and ß-oxidation capacity, two hallmarks of the disorder. Overall, this study provides biochemical evidence for a potential therapeutic modality to correct ß-oxidation deficiencies.


Asunto(s)
Acetilcisteína/análogos & derivados , Acil-CoA Deshidrogenasa/metabolismo , Carnitina/análogos & derivados , Fibroblastos/efectos de los fármacos , Acetilcisteína/farmacología , Acil-CoA Deshidrogenasa/química , Acil-CoA Deshidrogenasa/genética , Acil-CoA Deshidrogenasa de Cadena Larga/deficiencia , Acil-CoA Deshidrogenasa de Cadena Larga/genética , Secuencia de Aminoácidos , Carnitina/metabolismo , Síndromes Congénitos de Insuficiencia de la Médula Ósea , Cisteína/metabolismo , Relación Dosis-Respuesta a Droga , Ácidos Grasos/metabolismo , Fibroblastos/enzimología , Fibroblastos/patología , Terapia Genética/métodos , Humanos , Cinética , Errores Innatos del Metabolismo Lipídico/tratamiento farmacológico , Errores Innatos del Metabolismo Lipídico/enzimología , Errores Innatos del Metabolismo Lipídico/genética , Errores Innatos del Metabolismo Lipídico/patología , Enfermedades Mitocondriales/tratamiento farmacológico , Enfermedades Mitocondriales/enzimología , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/patología , Datos de Secuencia Molecular , Enfermedades Musculares/tratamiento farmacológico , Enfermedades Musculares/enzimología , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Mutación , Oxidación-Reducción , Cultivo Primario de Células , Piel/efectos de los fármacos , Piel/enzimología , Piel/patología
9.
Circulation ; 131(4): 371-80; discussion 380, 2015 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-25533966

RESUMEN

BACKGROUND: Inorganic nitrate (NO3(-)), abundant in certain vegetables, is converted to nitrite by bacteria in the oral cavity. Nitrite can be converted to nitric oxide in the setting of hypoxia. We tested the hypothesis that NO3(-) supplementation improves exercise capacity in heart failure with preserved ejection fraction via specific adaptations to exercise. METHODS AND RESULTS: Seventeen subjects participated in this randomized, double-blind, crossover study comparing a single dose of NO3-rich beetroot juice (NO3(-), 12.9 mmol) with an identical nitrate-depleted placebo. Subjects performed supine-cycle maximal-effort cardiopulmonary exercise tests, with measurements of cardiac output and skeletal muscle oxygenation. We also assessed skeletal muscle oxidative function. Study end points included exercise efficiency (total work/total oxygen consumed), peak VO2, total work performed, vasodilatory reserve, forearm mitochondrial oxidative function, and augmentation index (a marker of arterial wave reflections, measured via radial arterial tonometry). Supplementation increased plasma nitric oxide metabolites (median, 326 versus 10 µmol/L; P=0.0003), peak VO2 (12.6±3.7 versus 11.6±3.1 mL O2·min(-1)·kg(-1); P=0.005), and total work performed (55.6±35.3 versus 49.2±28.9 kJ; P=0.04). However, efficiency was unchanged. NO3(-) led to greater reductions in systemic vascular resistance (-42.4±16.6% versus -31.8±20.3%; P=0.03) and increases in cardiac output (121.2±59.9% versus 88.7±53.3%; P=0.006) with exercise. NO3(-) reduced aortic augmentation index (132.2±16.7% versus 141.4±21.9%; P=0.03) and tended to improve mitochondrial oxidative function. CONCLUSIONS: NO3(-) increased exercise capacity in heart failure with preserved ejection fraction by targeting peripheral abnormalities. Efficiency did not change as a result of parallel increases in total work and VO2. NO3(-) increased exercise vasodilatory and cardiac output reserves. NO3(-) also reduced arterial wave reflections, which are linked to left ventricular diastolic dysfunction and remodeling. CLINICAL TRIAL REGISTRATION URL: www.clinicaltrials.gov. Unique identifier: NCT01919177.


Asunto(s)
Prueba de Esfuerzo/métodos , Tolerancia al Ejercicio/fisiología , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/fisiopatología , Nitratos/administración & dosificación , Volumen Sistólico/fisiología , Anciano , Estudios Cruzados , Método Doble Ciego , Prueba de Esfuerzo/efectos de los fármacos , Tolerancia al Ejercicio/efectos de los fármacos , Femenino , Insuficiencia Cardíaca/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/fisiología , Volumen Sistólico/efectos de los fármacos , Resultado del Tratamiento
11.
Mol Cell Proteomics ; 13(1): 63-72, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24105792

RESUMEN

Nitric oxide (NO) mediates a substantial part of its physiologic functions via S-nitrosylation, however the cellular substrates for NO-mediated S-nitrosylation are largely unknown. Here we describe the S-nitrosoproteome using a high-density protein microarray chip containing 16,368 unique human proteins. We identified 834 potentially S-nitrosylated human proteins. Using a unique and highly specific labeling and affinity capture of S-nitrosylated proteins, 138 cysteine residues on 131 peptides in 95 proteins were determined, defining critical sites of NO's actions. Of these cysteine residues 113 are novel sites of S-nitrosylation. A consensus sequence motif from these 834 proteins for S-nitrosylation was identified, suggesting that the residues flanking the S-nitrosylated cysteine are likely to be the critical determinant of whether the cysteine is S-nitrosylated. We identify eight ubiquitin E3 ligases, RNF10, RNF11, RNF41, RNF141, RNF181, RNF208, WWP2, and UBE3A, whose activities are modulated by S-nitrosylation, providing a unique regulatory mechanism of the ubiquitin proteasome system. These results define a new and extensive set of proteins that are susceptible to NO regulation via S-nitrosylation. Similar approaches could be used to identify other post-translational modification proteomes.


Asunto(s)
Óxido Nítrico/metabolismo , Análisis por Matrices de Proteínas , Procesamiento Proteico-Postraduccional/genética , Proteoma , Humanos , Proteínas/metabolismo
12.
Mol Microbiol ; 91(4): 790-804, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24354846

RESUMEN

We show that thiols in the 4-cysteine zinc-finger motif of DksA, an RNA polymerase accessory protein known to regulate the stringent response, sense oxidative and nitrosative stress. Hydrogen peroxide- or nitric oxide (NO)-mediated modifications of thiols in the DksA 4-cysteine zinc-finger motif release the metal cofactor and drive reversible changes in the α-helicity of the protein. Wild-type and relA spoT mutant Salmonella, but not isogenic dksA-deficient bacteria, experience the downregulation of r-protein and amino acid transport expression after NO treatment, suggesting that DksA can regulate gene expression in response to NO congeners independently of the ppGpp alarmone. Oxidative stress enhances the DksA-dependent repression of rpsM, while preventing the activation of livJ and hisG gene transcription that is supported by reduced, zinc-bound DksA. The inhibitory effects of oxidized DksA on transcription are reversible with dithiothreitol. Our investigations indicate that sensing of reactive species by DksA redox active thiols fine-tunes the expression of translational machinery and amino acid assimilation and biosynthesis in accord with the metabolic stress imposed by oxidative and nitrosative stress. Given the conservation of Cys(114) , and neighbouring hydrophobic and charged amino acids in DksA orthologues, phylogenetically diverse microorganisms may use the DksA thiol switch to regulate transcriptional responses to oxidative and nitrosative stress.


Asunto(s)
Nitrosación , Estrés Oxidativo , Salmonella typhimurium/enzimología , Compuestos de Sulfhidrilo/metabolismo , Factores de Transcripción/metabolismo , Dedos de Zinc , Cisteína/metabolismo , ARN Polimerasas Dirigidas por ADN/metabolismo , Regulación de la Expresión Génica , Oxidación-Reducción , Salmonella typhimurium/genética , Estrés Fisiológico , Factores de Transcripción/genética
13.
J Biol Chem ; 288(37): 26473-9, 2013 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-23861393

RESUMEN

NO is a versatile free radical that mediates numerous biological functions within every major organ system. A molecular pathway by which NO accomplishes functional diversity is the selective modification of protein cysteine residues to form S-nitrosocysteine. This post-translational modification, S-nitrosylation, impacts protein function, stability, and location. Despite considerable advances with individual proteins, the in vivo biological chemistry, the structural elements that govern the selective S-nitrosylation of cysteine residues, and the potential overlap with other redox modifications are unknown. In this minireview, we explore the functional features of S-nitrosylation at the proteome level and the structural diversity of endogenously modified residues, and we discuss the potential overlap and complementation that may exist with other cysteine modifications.


Asunto(s)
Cisteína/análogos & derivados , Regulación de la Expresión Génica , Proteínas/química , S-Nitrosotioles/química , Animales , Cisteína/química , Humanos , Ratones , Miocardio/metabolismo , Óxido Nítrico/química , Óxido Nítrico Sintasa/metabolismo , Nitrógeno/química , Oxidación-Reducción , Procesamiento Proteico-Postraduccional , Proteómica/métodos , Transducción de Señal
14.
Methods ; 62(2): 165-70, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23116708

RESUMEN

Protein S-nitrosylation is considered as one of the molecular mechanisms by which nitric oxide regulates signaling events and protein function. The present review presents an updated method which allows for the site-specific detection of S-nitrosylated proteins in vivo. The method is based on enrichment of S-nitrosylated proteins or peptides using organomercury compounds followed by LC-MS/MS detection. Technical aspects for determining the reaction and binding efficiency of the mercury resin that assists enrichment of S-nitrosylated proteins are presented and discussed. In addition, emphasis is given to the specificity of the method by providing technical details for the generation of four chemically distinct negative controls. Finally it is provided an overview of the key steps for generation and evaluation of mass spectrometry derived data.


Asunto(s)
Cisteína/análogos & derivados , Proteoma/aislamiento & purificación , S-Nitrosotioles/aislamiento & purificación , Animales , Cromatografía de Afinidad , Cisteína/química , Cisteína/aislamiento & purificación , Cisteína/metabolismo , Humanos , Muramidasa/química , Fragmentos de Péptidos/química , Fragmentos de Péptidos/aislamiento & purificación , Unión Proteica , Procesamiento Proteico-Postraduccional , Proteoma/química , Proteoma/metabolismo , S-Nitrosotioles/química , S-Nitrosotioles/metabolismo , Espectrometría de Masas en Tándem
15.
Adv Sci (Weinh) ; 11(12): e2306469, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38235614

RESUMEN

In Alzheimer's disease (AD), dysfunctional mitochondrial metabolism is associated with synaptic loss, the major pathological correlate of cognitive decline. Mechanistic insight for this relationship, however, is still lacking. Here, comparing isogenic wild-type and AD mutant human induced pluripotent stem cell (hiPSC)-derived cerebrocortical neurons (hiN), evidence is found for compromised mitochondrial energy in AD using the Seahorse platform to analyze glycolysis and oxidative phosphorylation (OXPHOS). Isotope-labeled metabolic flux experiments revealed a major block in activity in the tricarboxylic acid (TCA) cycle at the α-ketoglutarate dehydrogenase (αKGDH)/succinyl coenzyme-A synthetase step, metabolizing α-ketoglutarate to succinate. Associated with this block, aberrant protein S-nitrosylation of αKGDH subunits inhibited their enzyme function. This aberrant S-nitrosylation is documented not only in AD-hiN but also in postmortem human AD brains versus controls, as assessed by two separate unbiased mass spectrometry platforms using both SNOTRAP identification of S-nitrosothiols and chemoselective-enrichment of S-nitrosoproteins. Treatment with dimethyl succinate, a cell-permeable derivative of a TCA substrate downstream to the block, resulted in partial rescue of mitochondrial bioenergetic function as well as reversal of synapse loss in AD-hiN. These findings have therapeutic implications that rescue of mitochondrial energy metabolism can ameliorate synaptic loss in hiPSC-based models of AD.


Asunto(s)
Enfermedad de Alzheimer , Células Madre Pluripotentes Inducidas , Humanos , Enfermedad de Alzheimer/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Metabolismo Energético/fisiología , Glucólisis , Neuronas/metabolismo
16.
Biochim Biophys Acta ; 1820(6): 684-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21651963

RESUMEN

BACKGROUND: A biochemical pathway by which nitric oxide accomplishes functional diversity is the specific modification of protein cysteine residues to form S-nitrosocysteine. This post-translational modification, S-nitrosylation, impacts protein function, interactions and location. However, comprehensive studies exploring protein signaling pathways or interrelated protein clusters that are regulated by S-nitrosylation have not been performed on a global scale. SCOPE OF REVIEW: To provide insights to these important biological questions, sensitive, validated and quantitative proteomic approaches are required. This review summarizes current approaches for the global identification of S-nitrosylated proteins. MAJOR CONCLUSIONS: The application of novel methods for identifying S-nitrosylated proteins, especially when combined with mass-spectrometry based proteomics to provide site-specific identification of the modified cysteine residues, promises to deliver critical clues for the regulatory role of this dynamic posttranslational modification in cellular processes. GENERAL SIGNIFICANCE: Though several studies have established S-nitrosylation as a regulator of protein function in individual proteins, the biological chemistry and the structural elements that govern the specificity of this modification in vivo are vastly unknown. Additionally, a gap in knowledge exists concerning the potential global regulatory role(s) this modification may play in cellular physiology. By further studying S-nitrosylation at a global scale, a greater appreciation of nitric oxide and protein S-nitrosylation in cellular function can be achieved. This article is part of a Special Issue entitled Regulation of Cellular Processes by S-nitrosylation.


Asunto(s)
Cisteína/análogos & derivados , Cisteína/metabolismo , Proteínas/metabolismo , Cisteína/biosíntesis , Espectrometría de Masas , Óxido Nítrico/metabolismo , Nitrosación/fisiología , Procesamiento Proteico-Postraduccional , Proteínas/química , Proteómica , S-Nitrosotioles , Transducción de Señal
17.
Circulation ; 126(20): 2392-401, 2012 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-23081989

RESUMEN

BACKGROUND: Several lines of evidence support a pathophysiological role of immunity in atherosclerosis. Tyrosine-nitrated proteins, a footprint of oxygen- and nitrogen-derived oxidants generated by cells of the immune system, are enriched in atheromatous lesions and in circulation of patients with coronary artery disease (CAD). However, the consequences of possible immune reactions triggered by the presence of nitrated proteins in subjects with clinically documented atherosclerosis have not been explored. METHODS AND RESULTS: Specific immunoglobulins that recognize 3-nitrotyrosine epitopes were identified in human lesions, as well as in circulation of patients with CAD. The levels of circulating immunoglobulins against 3-nitrotyrosine epitopes were quantified in patients with CAD (n=374) and subjects without CAD (non-CAD controls, n=313). A 10-fold increase in the mean level of circulating immunoglobulins against protein-bound 3-nitrotyrosine was documented in patients with CAD (3.75±1.8 µg antibody Eq/mL plasma versus 0.36±0.8 µg antibody Eq/mL plasma), and was strongly associated with angiographic evidence of significant CAD. CONCLUSIONS: The results of this cross-sectional study suggest that posttranslational modification of proteins via nitration within atherosclerotic plaque-laden arteries and in circulation serve as neo-epitopes for the elaboration of immunoglobulins, thereby providing an association between oxidant production and the activation of the immune system in CAD.


Asunto(s)
Enfermedad de la Arteria Coronaria/inmunología , Epítopos/inmunología , Inmunoglobulinas/sangre , Tirosina/análogos & derivados , Anciano , Estudios de Casos y Controles , Angiografía Coronaria , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/diagnóstico por imagen , Estudios Transversales , Femenino , Humanos , Sistema Inmunológico/fisiopatología , Masculino , Persona de Mediana Edad , Tirosina/inmunología
18.
Proc Natl Acad Sci U S A ; 107(39): 16958-63, 2010 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-20837516

RESUMEN

S-nitrosylation, the selective posttranslational modification of protein cysteine residues to form S-nitrosocysteine, is one of the molecular mechanisms by which nitric oxide influences diverse biological functions. In this study, unique MS-based proteomic approaches precisely pinpointed the site of S-nitrosylation in 328 peptides in 192 proteins endogenously modified in WT mouse liver. Structural analyses revealed that S-nitrosylated cysteine residues were equally distributed in hydrophobic and hydrophilic areas of proteins with an average predicted pK(a) of 10.01 ± 2.1. S-nitrosylation sites were over-represented in α-helices and under-represented in coils as compared with unmodified cysteine residues in the same proteins (χ(2) test, P < 0.02). A quantile-quantile probability plot indicated that the distribution of S-nitrosocysteine residues was skewed toward larger surface accessible areas compared with the unmodified cysteine residues in the same proteins. Seventy percent of the S-nitrosylated cysteine residues were surrounded by negatively or positively charged amino acids within a 6-Å distance. The location of cysteine residues in α-helices and coils in highly accessible surfaces bordered by charged amino acids implies site directed S-nitrosylation mediated by protein-protein or small molecule interactions. Moreover, 13 modified cysteine residues were coordinated with metals and 15 metalloproteins were endogenously modified supporting metal-catalyzed S-nitrosylation mechanisms. Collectively, the endogenous S-nitrosoproteome in the liver has structural features that accommodate multiple mechanisms for selective site-directed S-nitrosylation.


Asunto(s)
Cisteína/análogos & derivados , Hígado/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas/metabolismo , S-Nitrosotioles/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Cisteína/análisis , Cisteína/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Datos de Secuencia Molecular , Proteínas/química , Proteoma , Proteómica , S-Nitrosotioles/análisis
19.
Cell Chem Biol ; 30(8): 965-975.e6, 2023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37478858

RESUMEN

A causal relationship between mitochondrial metabolic dysfunction and neurodegeneration has been implicated in synucleinopathies, including Parkinson disease (PD) and Lewy body dementia (LBD), but underlying mechanisms are not fully understood. Here, using human induced pluripotent stem cell (hiPSC)-derived neurons with mutation in the gene encoding α-synuclein (αSyn), we report the presence of aberrantly S-nitrosylated proteins, including tricarboxylic acid (TCA) cycle enzymes, resulting in activity inhibition assessed by carbon-labeled metabolic flux experiments. This inhibition principally affects α-ketoglutarate dehydrogenase/succinyl coenzyme-A synthetase, metabolizing α-ketoglutarate to succinate. Notably, human LBD brain manifests a similar pattern of aberrantly S-nitrosylated TCA enzymes, indicating the pathophysiological relevance of these results. Inhibition of mitochondrial energy metabolism in neurons is known to compromise dendritic length and synaptic integrity, eventually leading to neuronal cell death. Our evidence indicates that aberrant S-nitrosylation of TCA cycle enzymes contributes to this bioenergetic failure.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedad de Parkinson , Sinucleinopatías , Humanos , Sinucleinopatías/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Enfermedad de Parkinson/metabolismo , Neuronas/metabolismo , Encéfalo/metabolismo
20.
J Immunol ; 185(11): 6664-9, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20980633

RESUMEN

NO is critical to immunity, but its role in the development of the immune system is unknown. In this study, we show that S-nitrosoglutathione reductase (GSNOR), a protein key to the control of protein S-nitrosylation, is important for the development of lymphocytes. Genetic deletion of GSNOR in mice results in significant decrease in both T and B lymphocytes in the periphery. In thymus, GSNOR deficiency causes excessive protein S-nitrosylation, increases apoptosis, and reduces the number of CD4 single-positive thymocytes. Lymphopenia and increase in S-nitrosylation and apoptosis in GSNOR-deficient mice are largely abolished by genetic deletion of inducible NO synthase. Furthermore, the protection of lymphocyte development by GSNOR is apparently intrinsic to hematopoietic cells. Thus, GSNOR, likely through regulation of S-nitrosylation and apoptosis, physiologically plays a protective role in the development of the immune system.


Asunto(s)
Aldehído Oxidorreductasas/fisiología , Diferenciación Celular/inmunología , Subgrupos Linfocitarios/enzimología , Subgrupos Linfocitarios/inmunología , Aldehído Oxidorreductasas/deficiencia , Aldehído Oxidorreductasas/metabolismo , Animales , Apoptosis/genética , Apoptosis/inmunología , Médula Ósea/inmunología , Médula Ósea/patología , Diferenciación Celular/genética , Predisposición Genética a la Enfermedad , Mediadores de Inflamación/fisiología , Subgrupos Linfocitarios/patología , Linfopenia/enzimología , Linfopenia/inmunología , Linfopenia/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/deficiencia , Óxido Nítrico Sintasa de Tipo II/genética , Infecciones por Pasteurella/enzimología , Infecciones por Pasteurella/inmunología , Infecciones por Pasteurella/patología , Quimera por Radiación/genética , Quimera por Radiación/inmunología , S-Nitrosotioles/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA