Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Cardiovasc Res ; 119(2): 571-586, 2023 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35704040

RESUMEN

AIMS: Brain-derived neurotrophic factor (BDNF) is markedly decreased in heart failure patients. Both BDNF and its receptor, tropomyosin-related kinase receptor (TrkB), are expressed in cardiomyocytes; however, the role of myocardial BDNF signalling in cardiac pathophysiology is poorly understood. Here, we investigated the role of BDNF/TrkB signalling in cardiac stress response to exercise and pathological stress. METHODS AND RESULTS: We found that myocardial BDNF expression was increased in mice with swimming exercise but decreased in a mouse heart failure model and human failing hearts. Cardiac-specific TrkB knockout (cTrkB KO) mice displayed a blunted adaptive cardiac response to exercise, with attenuated upregulation of transcription factor networks controlling mitochondrial biogenesis/metabolism, including peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α). In response to pathological stress (transaortic constriction, TAC), cTrkB KO mice showed an exacerbated heart failure progression. The downregulation of PGC-1α in cTrkB KO mice exposed to exercise or TAC resulted in decreased cardiac energetics. We further unravelled that BDNF induces PGC-1α upregulation and bioenergetics through a novel signalling pathway, the pleiotropic transcription factor Yin Yang 1. CONCLUSION: Taken together, our findings suggest that myocardial BDNF plays a critical role in regulating cellular energetics in the cardiac stress response.


Asunto(s)
Insuficiencia Cardíaca , Factores de Transcripción , Animales , Humanos , Ratones , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Metabolismo Energético , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Transcripción YY1/metabolismo
2.
J Appl Physiol (1985) ; 130(5): 1510-1521, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33764167

RESUMEN

Non-Hispanic black individuals suffer from an elevated prevalence of hypertension and cardiovascular disease (CVD) relative to other populations. This elevated disease risk is, in large part, related to impaired vascular function, secondary to reduced nitric oxide (NO) bioavailability. Emerging evidence suggests that dietary nitrate supplementation improves several cardiovascular parameters, including vascular function, in part by increased NO bioavailability. However, whether these findings extend to a population of black individuals is unknown. This study tested the hypothesis that forearm blood flow responses in young, non-Hispanic, black (BL) men during a mental stress challenge would be blunted relative to young, non-Hispanic, white (WH) men. We further hypothesized that acute dietary nitrate supplementation would improve this response in BL men. This study comprised two parts (phase 1 and phase 2). Phase 1 investigated the difference in blood flow responses between young, BL, and WH men. In contrast, phase 2 investigated the effect of acute nitrate supplementation on the responses in a subset of the BL men from phase 1. Eleven (nine for phase 2) BL and eight WH men (23 ± 3 vs. 24 ± 4 yr, respectively) participated in this double-blind, placebo-controlled, randomized, crossover study. During each visit, hemodynamic responses during 3 min of mental stress were assessed in the brachial artery using duplex Doppler ultrasound. Phase 1 was completed in one visit, whereas phase 2 was completed over two visits separated by ∼1 wk. During phase 2, data were collected before and 2-h postconsumption of a beverage either high in nitrate content or nitrate depleted. In phase 1, peak forearm blood flow (FBF; P < 0.001), total FBF (P < 0.01), and forearm vascular conductance (FVC; P < 0.001) were blunted in the BL. During phase 2, prebeverage responses were similar to phase 1 and were unaffected following beverage consumption (P > 0.05 vs. prebeverage for all variables). These data indicate that young, BL men have blunted microvascular vasodilatory responses to acute mental stress, which may not be altered following acute nitrate supplementation.NEW & NOTEWORTHY This study tested the hypothesis that non-Hispanic black (BL) men have a blunted forearm hyperemic response to mental stress, which would be augmented following acute nitrate supplementation. The increase in forearm blood flow during mental stress was attenuated in BL men and was not impacted by nitrate supplementation. This supports findings of altered vascular function in this population. This is especially important as BL experience a higher prevalence of stress, which contributes to CVD risk.


Asunto(s)
Hiperemia , Nitratos , Negro o Afroamericano , Estudios Cruzados , Suplementos Dietéticos , Humanos , Masculino , Flujo Sanguíneo Regional
3.
JCI Insight ; 52019 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-31120438

RESUMEN

BACKGROUND: Physical function decreases with age, and though bioenergetic alterations contribute to this decline, the mechanisms by which mitochondrial function changes with age remains unclear. This is partially because human mitochondrial studies require highly invasive procedures, such as muscle biopsies, to obtain live tissue with functional mitochondria. However, recent studies demonstrate that circulating blood cells are potentially informative in identifying systemic bioenergetic changes. Here, we hypothesize that human platelet bioenergetics reflect bioenergetics measured in muscle biopsies. METHODS & RESULTS: We demonstrate that maximal and ATP-linked respiratory rate measured in isolated platelets from older adults (86-93 years) correlates significantly with maximal respiration (r = 0.595; P = 0.003) measured by muscle biopsy respirometry and maximal ATP production (r = 0.643; P = 0.004) measured by 31P-MRS respectively, in the same individuals. Comparison of platelet bioenergetics in this aged cohort to platelets from younger adults (18-35 years) shows aged adults demonstrate lower basal and ATP-linked respiration. Platelets from older adults also show enhanced proton leak, which is likely due to increased protein levels of uncoupling protein 2, and correlates with increased gate speed in this cohort (r = 0.58; P = 0.0019). While no significant difference in glycolysis was observed in older adults compared to younger adults, platelet glycolytic rate correlated with fatigability (r = 0.44; P = 0.016). CONCLUSIONS: These data advance the mechanistic understanding of age-related changes in mitochondrial function. Further, they suggest that measuring platelet bioenergetics provides a potential supplement or surrogate for muscle biopsy measurement and may be a valuable tool to study mitochondrial involvement in age-related decline of physical function.


Asunto(s)
Plaquetas/metabolismo , Metabolismo Energético/fisiología , Músculo Esquelético/metabolismo , Adenosina Trifosfato/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Mitocondrias Musculares/metabolismo , Proteínas Desacopladoras Mitocondriales/metabolismo , Músculos , Proteína Desacopladora 2/metabolismo , Adulto Joven
4.
Mech Ageing Dev ; 180: 97-106, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31002926

RESUMEN

Cellular senescence is a phenotype characterized by irreversible growth arrest, chronic elevated secretion of proinflammatory cytokines and matrix proteases, a phenomenon known as senescence-associated secretory phenotype (SASP). Biomarkers of cellular senescence have been shown to increase with age and degeneration of human disc tissue. Senescent disc cells in culture recapitulate features associated with age-related disc degeneration, including increased secretion of proinflammatory cytokines, matrix proteases, and fragmentation of matrix proteins. However, little is known of the metabolic changes that underlie the senescent phenotype of disc cells. To assess the metabolic changes, we performed a bioenergetic analysis of in vitro oxidative stress-induced senescent (SIS) human disc cells. SIS disc cells acquire SASP and exhibit significantly elevated mitochondrial content and mitochondrial ATP-linked respiration. The metabolic changes appear to be driven by the upregulated protein secretion in SIS cells as abrogation of protein synthesis using cycloheximide decreased mitochondrial ATP-linked respiration. Taken together, the results of the study suggest that the increased energy generation state supports the secretion of senescent associated proteins in SIS disc cells.


Asunto(s)
Senescencia Celular , Metabolismo Energético , Disco Intervertebral/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo , Consumo de Oxígeno , Adulto , Femenino , Humanos , Disco Intervertebral/patología , Masculino , Persona de Mediana Edad , Mitocondrias/patología
5.
Circulation ; 139(19): 2238-2255, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30759996

RESUMEN

BACKGROUND: Deficiencies of iron-sulfur (Fe-S) clusters, metal complexes that control redox state and mitochondrial metabolism, have been linked to pulmonary hypertension (PH), a deadly vascular disease with poorly defined molecular origins. BOLA3 (BolA Family Member 3) regulates Fe-S biogenesis, and mutations in BOLA3 result in multiple mitochondrial dysfunction syndrome, a fatal disorder associated with PH. The mechanistic role of BOLA3 in PH remains undefined. METHODS: In vitro assessment of BOLA3 regulation and gain- and loss-of-function assays were performed in human pulmonary artery endothelial cells using siRNA and lentiviral vectors expressing the mitochondrial isoform of BOLA3. Polymeric nanoparticle 7C1 was used for lung endothelium-specific delivery of BOLA3 siRNA oligonucleotides in mice. Overexpression of pulmonary vascular BOLA3 was performed by orotracheal transgene delivery of adeno-associated virus in mouse models of PH. RESULTS: In cultured hypoxic pulmonary artery endothelial cells, lung from human patients with Group 1 and 3 PH, and multiple rodent models of PH, endothelial BOLA3 expression was downregulated, which involved hypoxia inducible factor-2α-dependent transcriptional repression via histone deacetylase 1-mediated histone deacetylation. In vitro gain- and loss-of-function studies demonstrated that BOLA3 regulated Fe-S integrity, thus modulating lipoate-containing 2-oxoacid dehydrogenases with consequent control over glycolysis and mitochondrial respiration. In contexts of siRNA knockdown and naturally occurring human genetic mutation, cellular BOLA3 deficiency downregulated the glycine cleavage system protein H, thus bolstering intracellular glycine content. In the setting of these alterations of oxidative metabolism and glycine levels, BOLA3 deficiency increased endothelial proliferation, survival, and vasoconstriction while decreasing angiogenic potential. In vivo, pharmacological knockdown of endothelial BOLA3 and targeted overexpression of BOLA3 in mice demonstrated that BOLA3 deficiency promotes histological and hemodynamic manifestations of PH. Notably, the therapeutic effects of BOLA3 expression were reversed by exogenous glycine supplementation. CONCLUSIONS: BOLA3 acts as a crucial lynchpin connecting Fe-S-dependent oxidative respiration and glycine homeostasis with endothelial metabolic reprogramming critical to PH pathogenesis. These results provide a molecular explanation for the clinical associations linking PH with hyperglycinemic syndromes and mitochondrial disorders. These findings also identify novel metabolic targets, including those involved in epigenetics, Fe-S biogenesis, and glycine biology, for diagnostic and therapeutic development.


Asunto(s)
Endotelio Vascular/fisiología , Glicina/metabolismo , Hipertensión Pulmonar/genética , Proteínas Mitocondriales/metabolismo , Adolescente , Adulto , Animales , Respiración de la Célula , Células Cultivadas , Niño , Preescolar , Modelos Animales de Enfermedad , Femenino , Humanos , Hipertensión Pulmonar/metabolismo , Lactante , Proteínas Hierro-Azufre/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Mitocondriales/genética , Mutación/genética , Oxidación-Reducción , ARN Interferente Pequeño/genética , Adulto Joven
6.
Antioxid Redox Signal ; 31(10): 673-686, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30608004

RESUMEN

Aims: Asthma, characterized by airway obstruction and hyper-responsiveness, is more severe and less responsive to treatment in obese subjects. While alterations in mitochondrial function and redox signaling have been implicated in asthma pathogenesis, it is unclear whether these mechanisms differ in lean versus obese asthmatics. In addition, we previously demonstrated that circulating platelets from asthmatic individuals have altered bioenergetics; however, it is unknown whether platelet mitochondrial changes reflect those observed in airway epithelial cells. Herein we hypothesized that lean and obese asthmatics show differential bioenergetics and redox signaling in airway cells and that these alterations could be measured in platelets from the same individual. Results: Using freshly isolated bronchial airway epithelial cells and platelets from lean and obese asthmatics and healthy individuals, we show that both cell types from obese asthmatics have significantly increased glycolysis, basal and maximal respiration, and oxidative stress compared with lean asthmatics and healthy controls. This increased respiration was associated with enhanced arginine metabolism by arginase, which has previously been shown to drive respiration. Inducible nitric oxide synthase (iNOS) was also upregulated in cells from all asthmatics. However, due to nitric oxide synthase uncoupling in obese asthmatics, overall nitric oxide (NO) bioavailability was decreased, preventing NO-dependent inhibition in obese asthmatic cells that was observed in lean asthmatics. Innovation and Conclusion: These data demonstrate bioenergetic differences between lean and obese asthmatics that are, in part, due to differences in NO signaling. They also suggest that the platelet may serve as a useful surrogate to understand redox, oxidative stress and bioenergetic changes in the asthmatic airway.


Asunto(s)
Asma/metabolismo , Plaquetas/metabolismo , Óxido Nítrico/metabolismo , Obesidad/metabolismo , Delgadez/metabolismo , Adulto , Células Cultivadas , Epitelio/metabolismo , Femenino , Humanos , Pulmón/metabolismo , Masculino , Persona de Mediana Edad , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Estrés Oxidativo/fisiología , Transducción de Señal/fisiología , Adulto Joven
7.
Redox Biol ; 15: 277-283, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29304478

RESUMEN

Nitrite represents an endocrine reserve of bioavailable nitric oxide (NO) that mediates a number of physiological responses including conferral of cytoprotection after ischemia/reperfusion (I/R). It has long been known that nitrite can react with non-heme iron to form dinitrosyliron complexes (DNIC). However, it remains unclear how quickly nitrite-dependent DNIC form in vivo, whether formation kinetics differ from that of NO-dependent DNIC, and whether DNIC play a role in the cytoprotective effects of nitrite. Here we demonstrate that chronic but not acute nitrite supplementation increases DNIC concentration in the liver and kidney of mice. Although DNIC have been purported to have antioxidant properties, we show that the accumulation of DNIC in vivo is not associated with nitrite-dependent cytoprotection after hepatic I/R. Further, our data in an isolated mitochondrial model of anoxia/reoxygenation show that while NO and nitrite demonstrate similar S-nitrosothiol formation kinetics, DNIC formation is significantly greater with NO and associated with mitochondrial dysfunction as well as inhibition of aconitase activity. These data are the first to directly compare mitochondrial DNIC formation by NO and nitrite. This study suggests that nitrite-dependent DNIC formation is a physiological consequence of dietary nitrite. The data presented herein implicate mitochondrial DNIC formation as a potential mechanism underlying the differential cytoprotective effects of nitrite and NO after I/R, and suggest that DNIC formation is potentially responsible for the cytotoxic effects observed at high NO concentrations.


Asunto(s)
Antioxidantes/metabolismo , Hierro/metabolismo , Hígado/metabolismo , Mitocondrias/metabolismo , Óxido Nítrico/biosíntesis , Óxidos de Nitrógeno/metabolismo , Aconitato Hidratasa/metabolismo , Animales , Antioxidantes/química , Citoprotección/efectos de los fármacos , Hipoxia/metabolismo , Hipoxia/patología , Hierro/química , Riñón/metabolismo , Riñón/patología , Hígado/patología , Ratones , Mitocondrias/patología , Óxido Nítrico/metabolismo , Nitritos/química , Nitritos/metabolismo , Óxidos de Nitrógeno/química , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , S-Nitrosotioles/metabolismo
8.
Antioxid Redox Signal ; 28(1): 44-61, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-28816057

RESUMEN

AIMS: This study was designed to explore the neuroprotective potential of inorganic nitrite as a new therapeutic avenue in Parkinson's disease (PD). RESULTS: Administration of inorganic nitrite ameliorates neuropathology in phylogenetically distinct animal models of PD. Beneficial effects are not confined to prophylactic treatment and also occur if nitrite is administered when the pathogenic cascade is already active. Mechanistically, the effect is mediated by both complex I S-nitrosation, which under nitrite administration is favored over formation of other forms of oxidation, and down-stream activation of the antioxidant Nrf2 pathway. Nitrite also rescues respiratory reserve capacity and increases proton leakage in LRRK2 PD patients' dermal fibroblasts. INNOVATION: The study proposes an unprecedented approach based on the administration of the nitrosonium donor nitrite to contrast complex I and redox anomalies in PD. Dysfunctional mitochondrial complex I propagates oxidative stress in PD, and treatments mitigating this defect may, therefore, limit disease progression. Therapeutic complex I targeting has been successfully achieved in ischemia/reperfusion by using nitrosonium donors such as nitrite to reversibly modify its subunits and protect from oxidative damage after reperfusion. This evidence led to the innovative hypothesis that nitrite could exert protective effects also in pathological conditions where complex I dysfunction occurs in normoxia, such as in PD. CONCLUSIONS: Overall, these results demonstrate that administration of inorganic nitrite improves mitochondrial function in PD, and it, therefore, represents an amenable intervention to hamper disease progression. Antioxid. Redox Signal. 28, 44-61.


Asunto(s)
Complejo I de Transporte de Electrón/metabolismo , Metabolismo Energético , Mitocondrias/metabolismo , Enfermedad de Parkinson/metabolismo , Animales , Antioxidantes/metabolismo , Conducta Animal , Respiración de la Célula/efectos de los fármacos , Supervivencia Celular , Citoprotección , Modelos Animales de Enfermedad , Metabolismo Energético/efectos de los fármacos , Fibroblastos , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Masculino , Mitocondrias/efectos de los fármacos , Actividad Motora , Mutación , Neuronas/metabolismo , Nitritos/administración & dosificación , Nitritos/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Sustancias Protectoras/administración & dosificación , Ratas , Tirosina 3-Monooxigenasa/metabolismo
9.
Hypertension ; 70(3): 634-644, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28739973

RESUMEN

Dietary NO3- (nitrate) and NO2- (nitrite) support ˙NO (nitric oxide) generation and downstream vascular signaling responses. These nitrogen oxides also generate secondary nitrosating and nitrating species that react with low molecular weight thiols, heme centers, proteins, and unsaturated fatty acids. To explore the kinetics of NO3-and NO2-metabolism and the impact of dietary lipid on nitrogen oxide metabolism and cardiovascular responses, the stable isotopes Na15NO3 and Na15NO2 were orally administered in the presence or absence of conjugated linoleic acid (cLA). The reduction of 15NO2- to 15NO was indicated by electron paramagnetic resonance spectroscopy detection of hyperfine splitting patterns reflecting 15NO-deoxyhemoglobin complexes. This formation of 15NO also translated to decreased systolic and mean arterial blood pressures and inhibition of platelet function. Upon concurrent administration of cLA, there was a significant increase in plasma cLA nitration products 9- and 12-15NO2-cLA. Coadministration of cLA with 15NO2- also impacted the pharmacokinetics and physiological effects of 15NO2-, with cLA administration suppressing plasma NO3-and NO2-levels, decreasing 15NO-deoxyhemoglobin formation, NO2-inhibition of platelet activation, and the vasodilatory actions of NO2-, while enhancing the formation of 9- and 12-15NO2-cLA. These results indicate that the biochemical reactions and physiological responses to oral 15NO3-and 15NO2-are significantly impacted by dietary constituents, such as unsaturated lipids. This can explain the variable responses to NO3-and NO2-supplementation in clinical trials and reveals dietary strategies for promoting the generation of pleiotropic nitrogen oxide-derived lipid signaling mediators. Clinical Trial Registration- URL: http://www.clinicaltrials.gov . Unique identifier: NCT01681836.


Asunto(s)
Plaquetas/efectos de los fármacos , Sistema Cardiovascular/efectos de los fármacos , Ácidos Linoleicos Conjugados/farmacología , Nitratos/farmacología , Nitritos/farmacología , Administración Oral , Humanos , Ácidos Linoleicos Conjugados/administración & dosificación , Nitratos/administración & dosificación , Nitritos/administración & dosificación
12.
Am J Respir Crit Care Med ; 195(5): 596-606, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27753502

RESUMEN

Carbon monoxide (CO) poisoning affects 50,000 people a year in the United States. The clinical presentation runs a spectrum, ranging from headache and dizziness to coma and death, with a mortality rate ranging from 1 to 3%. A significant number of patients who survive CO poisoning suffer from long-term neurological and affective sequelae. The neurologic deficits do not necessarily correlate with blood CO levels but likely result from the pleiotropic effects of CO on cellular mitochondrial respiration, cellular energy utilization, inflammation, and free radical generation, especially in the brain and heart. Long-term neurocognitive deficits occur in 15-40% of patients, whereas approximately one-third of moderate to severely poisoned patients exhibit cardiac dysfunction, including arrhythmia, left ventricular systolic dysfunction, and myocardial infarction. Imaging studies reveal cerebral white matter hyperintensities, with delayed posthypoxic leukoencephalopathy or diffuse brain atrophy. Management of these patients requires the identification of accompanying drug ingestions, especially in the setting of intentional poisoning, fire-related toxic gas exposures, and inhalational injuries. Conventional therapy is limited to normobaric and hyperbaric oxygen, with no available antidotal therapy. Although hyperbaric oxygen significantly reduces the permanent neurological and affective effects of CO poisoning, a portion of survivors still have substantial morbidity. There has been some early success in therapies targeting the downstream inflammatory and oxidative effects of CO poisoning. New methods to directly target the toxic effect of CO, such as CO scavenging agents, are currently under development.


Asunto(s)
Intoxicación por Monóxido de Carbono/patología , Intoxicación por Monóxido de Carbono/terapia , Intoxicación por Monóxido de Carbono/diagnóstico , Humanos , Oxigenoterapia Hiperbárica
13.
Toxicol Appl Pharmacol ; 289(3): 397-408, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26529668

RESUMEN

Chronic exposure to arsenic in drinking water causes cancer and non-cancer diseases. However, mechanisms for chronic arsenic-induced pathogenesis, especially in response to lower exposure levels, are unclear. In addition, the importance of health impacts from xeniobiotic-promoted microbiome changes is just being realized and effects of arsenic on the microbiome with relation to disease promotion are unknown. To investigate impact of arsenic exposure on both microbiome and host metabolism, the stucture and composition of colonic microbiota, their metabolic phenotype, and host tissue and plasma metabolite levels were compared in mice exposed for 2, 5, or 10weeks to 0, 10 (low) or 250 (high) ppb arsenite (As(III)). Genotyping of colonic bacteria revealed time and arsenic concentration dependent shifts in community composition, particularly the Bacteroidetes and Firmicutes, relative to those seen in the time-matched controls. Arsenic-induced erosion of bacterial biofilms adjacent to the mucosal lining and changes in the diversity and abundance of morphologically distinct species indicated changes in microbial community structure. Bacterical spores increased in abundance and intracellular inclusions decreased with high dose arsenic. Interestingly, expression of arsenate reductase (arsA) and the As(III) exporter arsB, remained unchanged, while the dissimilatory nitrite reductase (nrfA) gene expression increased. In keeping with the change in nitrogen metabolism, colonic and liver nitrite and nitrate levels and ratios changed with time. In addition, there was a concomitant increase in pathogenic arginine metabolites in the mouse circulation. These data suggest that arsenic exposure impacts the microbiome and microbiome/host nitrogen metabolism to support disease enhancing pathogenic phenotypes.


Asunto(s)
Aminoácidos/metabolismo , Arsénico/farmacología , Colon/efectos de los fármacos , Colon/microbiología , Microbiota/efectos de los fármacos , Nitrógeno/metabolismo , Animales , Arginina/metabolismo , Bacterias/efectos de los fármacos , Bacterias/genética , Biopelículas/efectos de los fármacos , Colon/metabolismo , Genotipo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Microbiota/genética , Nitratos/metabolismo , Nitrito Reductasas/metabolismo , Nitritos/metabolismo
14.
Free Radic Biol Med ; 89: 333-41, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26385079

RESUMEN

A gap in our understanding of the beneficial systemic responses to dietary constituents nitrate (NO3(-)), nitrite (NO2(-)) and conjugated linoleic acid (cLA) is the identification of the downstream metabolites that mediate their actions. To examine these reactions in a clinical context, investigational drug preparations of (15)N-labeled NO3(-) and NO2(-) were orally administered to healthy humans with and without cLA. Mass spectrometry analysis of plasma and urine indicated that the nitrating species nitrogen dioxide was formed and reacted with the olefinic carbons of unsaturated fatty acids to yield the electrophilic fatty acid, nitro-cLA (NO2-cLA). These species mediate the post-translational modification (PTM) of proteins via reversible Michael addition with nucleophilic amino acids. The PTM of critical target proteins by electrophilic lipids has been described as a sensing mechanism that regulates adaptive cellular responses, but little is known about the endogenous generation of fatty acid nitroalkenes and their metabolites. We report that healthy humans consuming (15)N-labeled NO3(-) or NO2(-), with and without cLA supplementation, produce (15)NO2-cLA and corresponding metabolites that are detected in plasma and urine. These data support that the dietary constituents NO3(-), NO2(-) and cLA promote the further generation of secondary electrophilic lipid products that are absorbed into the circulation at concentrations sufficient to exert systemic effects before being catabolized or excreted.


Asunto(s)
Alquenos/metabolismo , Antiinflamatorios/metabolismo , Ácido Linoleico/metabolismo , Nitratos/administración & dosificación , Nitritos/administración & dosificación , Nitrocompuestos/metabolismo , Adulto , Cromatografía Liquida , Suplementos Dietéticos , Femenino , Humanos , Masculino , Nitratos/metabolismo , Nitritos/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
15.
Food Funct ; 6(3): 824-33, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25598082

RESUMEN

(-)-Epicatechin ((-)-EPI), a naturally occurring flavanol, has emerged as a likely candidate for cocoa-based product reported reductions in cardiometabolic risk. The present study aimed to determine the safety, tolerability, pharmacokinetics and pharmacodynamics of purified (-)-EPI administered to healthy volunteers. In this phase I, open-label, two-part single- and multiple-dose study, subjects received either a single dose (n = 9) of 50, 100 or 200 mg or multiple doses (n = 8) of 50 mg daily (q.d.) or twice daily (b.i.d) for 5 days. Blood was collected at 0, 0.5, 1, 2, 4 and 6 h after (-)-EPI administration in the single and multiple dose groups (blood collection repeated in day 5). Samples were analyzed by HPLC-HR-ESI-MS for EPI and metabolite quantification. In the q.d. and b.i.d. groups, blood samples were analyzed for NO surrogates and follistatin levels as well as, platelet mitochondrial complexes I, V and citrate synthase activity levels. (-)-EPI was well tolerated and readily absorbed with further phase 2 metabolism. On day 5, in the q.d. and b.i.d. groups, there were significant increases in plasma nitrite of 30% and 17%, respectively. In the q.d. group on day 5 vs. day 1, platelet mitochondrial complexes I, IV and citrate synthase activities demonstrated a significant increase of ∼92, 62 and 8%, respectively. Average day 5 follistatin AUC levels were ∼2.5 fold higher vs. day 1 AUC levels in the b.i.d. group. (-)-EPI was safe to use, with no observed adverse effects, and our findings suggest that increases in NO metabolites, mitochondrial enzyme function and plasma follistatin levels may underlie some of the beneficial effects of cocoa products or (-)-EPI as reported in other studies.


Asunto(s)
Antiinflamatorios no Esteroideos/efectos adversos , Antiinflamatorios no Esteroideos/metabolismo , Enfermedades Cardiovasculares/prevención & control , Catequina/efectos adversos , Catequina/metabolismo , Suplementos Dietéticos/efectos adversos , Absorción Intestinal , Adulto , Anciano , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/sangre , Biomarcadores/sangre , Biomarcadores/metabolismo , Plaquetas/enzimología , Enfermedades Cardiovasculares/inmunología , Catequina/administración & dosificación , Catequina/sangre , Citrato (si)-Sintasa/química , Citrato (si)-Sintasa/metabolismo , Suplementos Dietéticos/análisis , Proteínas del Complejo de Cadena de Transporte de Electrón/agonistas , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Femenino , Folistatina/sangre , Folistatina/metabolismo , Humanos , Cinética , Masculino , Persona de Mediana Edad , Óxido Nítrico/agonistas , Óxido Nítrico/sangre , Óxido Nítrico/metabolismo , Pruebas de Toxicidad Subcrónica , Adulto Joven
16.
J Natl Cancer Inst ; 105(15): 1111-22, 2013 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-23821767

RESUMEN

BACKGROUND: Efficacy of withaferin A (WA), an Ayurvedic medicine constituent, for prevention of mammary cancer and its associated mechanisms were investigated using mouse mammary tumor virus-neu (MMTV-neu) transgenic model. METHODS: Incidence and burden of mammary cancer and pulmonary metastasis were scored in female MMTV-neu mice after 28 weeks of intraperitoneal administration with 100 µg WA (three times/week) (n = 32) or vehicle (n = 29). Mechanisms underlying mammary cancer prevention by WA were investigated by determination of tumor cell proliferation, apoptosis, metabolomics, and proteomics using plasma and/or tumor tissues. Spectrophotometric assays were performed to determine activities of complex III and complex IV. All statistical tests were two-sided. RESULTS: WA administration resulted in a statistically significant decrease in macroscopic mammary tumor size, microscopic mammary tumor area, and the incidence of pulmonary metastasis. For example, the mean area of invasive cancer was lower by 95.14% in the WA treatment group compared with the control group (mean = 3.10 vs 63.77 mm2, respectively; difference = -60.67 mm2; 95% confidence interval = -122.50 to 1.13 mm2; P = .0536). Mammary cancer prevention by WA treatment was associated with increased apoptosis, inhibition of complex III activity, and reduced levels of glycolysis intermediates. Proteomics confirmed downregulation of many glycolysis-related proteins in the tumor of WA-treated mice compared with control, including M2-type pyruvate kinase, phospho glycerate kinase, and fructose-bisphosphate aldolase A isoform 2. CONCLUSIONS: This study reveals suppression of glycolysis in WA-mediated mammary cancer prevention in a clinically relevant mouse model.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Pulmonares/prevención & control , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/prevención & control , Witanólidos/farmacología , Animales , Apoptosis/efectos de los fármacos , Análisis por Conglomerados , Electroforesis en Gel Bidimensional , Femenino , Glucólisis/efectos de los fármacos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/virología , Virus del Tumor Mamario del Ratón , Medicina Ayurvédica , Ratones , Ratones Transgénicos , Distribución Aleatoria
17.
PLoS One ; 6(8): e23354, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21853114

RESUMEN

Withaferin A (WA), a promising anticancer constituent of Ayurvedic medicinal plant Withania somnifera, inhibits growth of MDA-MB-231 and MCF-7 human breast cancer cells in culture and MDA-MB-231 xenografts in vivo in association with apoptosis induction, but the mechanism of cell death is not fully understood. We now demonstrate, for the first time, that WA-induced apoptosis is mediated by reactive oxygen species (ROS) production due to inhibition of mitochondrial respiration. WA treatment caused ROS production in MDA-MB-231 and MCF-7 cells, but not in a normal human mammary epithelial cell line (HMEC). The HMEC was also resistant to WA-induced apoptosis. WA-mediated ROS production as well as apoptotic histone-associated DNA fragment release into the cytosol was significantly attenuated by ectopic expression of Cu,Zn-superoxide dismutase in both MDA-MB-231 and MCF-7 cells. ROS production resulting from WA exposure was accompanied by inhibition of oxidative phosphorylation and inhibition of complex III activity. Mitochondrial DNA-deficient Rho-0 variants of MDA-MB-231 and MCF-7 cells were resistant to WA-induced ROS production, collapse of mitochondrial membrane potential, and apoptosis compared with respective wild-type cells. WA treatment resulted in activation of Bax and Bak in MDA-MB-231 and MCF-7 cells, and SV40 immortalized embryonic fibroblasts derived from Bax and Bak double knockout mouse were significantly more resistant to WA-induced apoptosis compared with fibroblasts derived from wild-type mouse. In conclusion, the present study provides novel insight into the molecular circuitry of WA-induced apoptosis involving ROS production and activation of Bax/Bak.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Especies Reactivas de Oxígeno/metabolismo , Witanólidos/farmacología , Adenosina Trifosfato/metabolismo , Animales , Neoplasias de la Mama/enzimología , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Complejo III de Transporte de Electrones/metabolismo , Femenino , Humanos , Ratones , Fosforilación Oxidativa/efectos de los fármacos , Sustancias Protectoras/farmacología , Superóxido Dismutasa/metabolismo , Witanólidos/química , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Proteínas de Unión al GTP rho/metabolismo
18.
J Clin Invest ; 121(4): 1646-56, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21436585

RESUMEN

Vascular disease, a significant cause of morbidity and mortality in the developed world, results from vascular injury. Following vascular injury, damaged or dysfunctional endothelial cells and activated SMCs engage in vasoproliferative remodeling and the formation of flow-limiting intimal hyperplasia (IH). We hypothesized that vascular injury results in decreased bioavailability of NO secondary to dysregulated arginine-dependent NO generation. Furthermore, we postulated that nitrite-dependent NO generation is augmented as an adaptive response to limit vascular injury/proliferation and can be harnessed for its protective effects. Here we report that sodium nitrite (intraperitoneal, inhaled, or oral) limited the development of IH in a rat model of vascular injury. Additionally, nitrite led to the generation of NO in vessels and SMCs, as well as limited SMC proliferation via p21Waf1/Cip1 signaling. These data demonstrate that IH is associated with increased arginase-1 levels, which leads to decreased NO production and bioavailability. Vascular injury also was associated with increased levels of xanthine oxidoreductase (XOR), a known nitrite reductase. Chronic inhibition of XOR and a diet deficient in nitrate/nitrite each exacerbated vascular injury. Moreover, established IH was reversed by dietary supplementation of nitrite. The vasoprotective effects of nitrite were counteracted by inhibition of XOR. These data illustrate the importance of nitrite-generated NO as an endogenous adaptive response and as a pathway that can be harnessed for therapeutic benefit.


Asunto(s)
Arginina/fisiología , Óxido Nítrico Sintasa de Tipo III/fisiología , Óxido Nítrico/fisiología , Nitrito de Sodio/administración & dosificación , Túnica Íntima/fisiología , Animales , Arginasa/metabolismo , Proliferación Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Hiperplasia/patología , Hiperplasia/fisiopatología , Hiperplasia/prevención & control , Masculino , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Óxido Nítrico/biosíntesis , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Túnica Íntima/efectos de los fármacos , Túnica Íntima/lesiones , Túnica Íntima/patología , Xantina Deshidrogenasa/metabolismo
19.
Nat Chem Biol ; 5(12): 865-9, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19915529

RESUMEN

Inorganic nitrate and nitrite from endogenous or dietary sources are metabolized in vivo to nitric oxide (NO) and other bioactive nitrogen oxides. The nitrate-nitrite-NO pathway is emerging as an important mediator of blood flow regulation, cell signaling, energetics and tissue responses to hypoxia. The latest advances in our understanding of the biochemistry, physiology and therapeutics of nitrate, nitrite and NO were discussed during a recent 2-day meeting at the Nobel Forum, Karolinska Institutet in Stockholm.


Asunto(s)
Nitratos/metabolismo , Nitratos/uso terapéutico , Óxido Nítrico/metabolismo , Nitritos/metabolismo , Nitritos/uso terapéutico , Animales , Dieta , Metabolismo Energético , Humanos , Mitocondrias/metabolismo , Nitratos/administración & dosificación , Nitritos/administración & dosificación , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA