Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Pharmacol Res ; 109: 24-31, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26987940

RESUMEN

Thioredoxin is one of the most important cellular antioxidant systems known to date, and is responsible of maintaining the reduced state of the intracellular space. Trx-1 is a small cytosolic protein whose transcription is induced by stress. Therefore it is possible that this antioxidant plays a protective role against the oxidative stress caused by an increase of reactive oxygen species concentration, as occurs during the reperfusion after an ischemic episode. However, in addition to its antioxidant properties, it is able to activate other cytoplasmic and nuclear mediators that confer cardioprotection. It is remarkable that Trx-1 also participates in myocardial protection mechanisms such as ischemic preconditioning and postconditioning, activating proteins related to cellular survival. In this sense, it has been shown that Trx-1 inhibition abolished the preconditioning cardioprotective effect, evidenced through apoptosis and infarct size. Furthermore, ischemic postconditioning preserves Trx-1 content at reperfusion, after ischemia. However, comorbidities such as aging can modify this powerful cellular defense leading to decrease cardioprotection. Even ischemic preconditioning and postconditioning protocols performed in aged animal models failed to decrease infarct size. Therefore, the lack of success of antioxidants therapies to treat ischemic heart disease could be solved, at least in part, avoiding the damage of Trx system.


Asunto(s)
Poscondicionamiento Isquémico , Precondicionamiento Isquémico , Daño por Reperfusión Miocárdica/metabolismo , Tiorredoxinas/metabolismo , Envejecimiento/metabolismo , Envejecimiento/fisiología , Animales , Vasos Coronarios/fisiología , Corazón/fisiopatología , Humanos , Miocardio/metabolismo , Tiorredoxinas/fisiología
2.
Liver Int ; 35(3): 953-66, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24750664

RESUMEN

BACKGROUND & AIMS: Hepatocyte apoptosis, the hallmark of non-alcoholic steatohepatitis (NASH) contributes to liver injury and fibrosis. Although, both the intrinsic and extrinsic apoptotic pathways are involved in the pathogenesis of NASH, the final common step of apoptosis is executed by a family of cysteine-proteases termed caspases. Thus, our aim was to ascertain if administration of Emricasan, a pan-caspase inhibitor, ameliorates liver injury and fibrosis in a murine model of NASH. METHODS: C57/BL6J-mice were fed regular chow or high fat diet (HFD) for 20 weeks. All mice were treated with vehicle or Emricasan. RESULTS: Mice fed a HFD diet demonstrate a five-fold increase in hepatocyte apoptosis by the TUNEL assay and a 1.5-fold and 1.3-fold increase in caspase-3 and-8 activities respectively; this increase in apoptosis was substantially attenuated in mice fed a HFD treated with Emricasan (HFD-Em). Likewise, liver injury and inflammation were reduced in mice fed HFD-Em as compare to HFD by measuring serum aspartate aminotransferase and alanine aminotransferase levels, NAS histological score and IL 1-ß, TNF-α, monocyte chemoattractant protein (MCP-1) and C-X-C chemokine ligand-2 (CXCL2) quantitative reverse-transcription polymerase chain reaction (qPCR). These differences could not be attributed to differences in hepatic steatosis as liver triglycerides content were similar in both HFD groups. Hepatic fibrosis was reduced by Emricasan in HFD animals by decreasing αSMA (a marker for hepatic stellate cell activation), fibrosis score, Sirius red staining, hydroxyproline liver content and profibrogenic cytokines by qPCR. CONCLUSION: In conclusion, these data demonstrate that in a murine model of NASH, liver injury and fibrosis are suppressed by inhibiting hepatocytes apoptosis and suggests that Emricasan may be an attractive antifibrotic therapy in NASH.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores de Caspasas/uso terapéutico , Hepatocitos/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Ácidos Pentanoicos/uso terapéutico , Animales , Inhibidores de Caspasas/farmacología , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Fibrosis , Hepatitis/prevención & control , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/patología , Ácidos Pentanoicos/farmacología
3.
Mol Cell Biochem ; 359(1-2): 169-76, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21842376

RESUMEN

Hemorrhage (H) is associated with a left ventricular (LV) dysfunction. However, the diastolic function has not been studied in detail. The main goal was to assess the diastolic function both during and 120 min after bleeding, in the absence and in the presence of L-NAME. Also, the changes in mRNA and protein expression of nitric oxide synthase (NOS) isoforms were determined. New Zealand rabbits were divided into three groups: Sham group, H group (hemorrhage 20% blood volume), and H L-NAME group (hemorrhage treated with L-NAME). We evaluated systolic and diastolic ventricular functions in vivo and in vitro (Langendorff technique). Hemodynamic parameters and LV function were measured before, during, and at 120 min after bleeding. We analyzed the isovolumic relaxation using t ½ in vivo (closed chest). After that, hearts were excised and perfused in vitro to measure myocardial stiffness. Samples were frozen to measure NOS mRNA and protein expression. The t½ increased during bleeding and returned to basal values 120 min after bleeding. L-NAME blunted this effect. Data from the H group revealed a shift to the left in the LV end diastolic pressure-volume curve at 120 min after bleeding, which was blocked by L-NAME. iNOS and nNOS protein expression and mRNA levels increased at 120 min after the hemorrhage. Acute hemorrhage induces early and transient isovolumic relaxation impairment and an increase in myocardial stiffness 120 min after bleeding. L-NAME blunted the LV dysfunction, suggesting that NO modulates ventricular function through iNOS and nNOS isoforms.


Asunto(s)
Diástole , Choque Hemorrágico/fisiopatología , Disfunción Ventricular Izquierda/tratamiento farmacológico , Animales , Diástole/efectos de los fármacos , Diástole/fisiología , Corazón/fisiopatología , Hemorragia , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo I , Óxido Nítrico Sintasa de Tipo II , Óxidos de Nitrógeno , Conejos , Choque Hemorrágico/complicaciones , Disfunción Ventricular Izquierda/enzimología , Disfunción Ventricular Izquierda/etiología
4.
Trends Biochem Sci ; 31(11): 614-21, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16996273

RESUMEN

The past few years have witnessed intense research into the biological significance of carbon monoxide (CO) as an essential signaling mediator in cells and tissues. To transduce the signal properly, CO must react selectively with functional and structural proteins containing moieties that show preferred reactivity towards this gaseous molecule. This selectivity is exemplified by the interaction of CO with iron- and heme-dependent proteins, although systems containing other transition metals can potentially become a preferential target for CO. Notably, transition metal carbonyls, which carry and liberate CO, are also emerging as a pharmacological tool to mimic the bioactivity of endogenously generated CO. Thus, exploring how CO binding to metal complexes is translated into a cytoprotective function is a challenging task and might open up opportunities for therapeutic applications based on CO delivery.


Asunto(s)
Monóxido de Carbono/química , Gases/toxicidad , Metales/química , Transducción de Señal , Animales , Citoprotección , Mamíferos
5.
Medicina (B Aires) ; 70(3): 233-9, 2010.
Artículo en Español | MEDLINE | ID: mdl-20529772

RESUMEN

In order to evaluate the relationship between systemic inflammatory response and mortality in the older hospitalized patient, we developed a prospective cohort study in which we evaluated a nutritional score (SGA), years of instruction, functional status, organic failure (Marshall), presence of sepsis, comorbidities (Charlson), cognitive state (MMSE), albumin, erythrocyte sedimentation rate and mortality. Fifty two patients were included, 19 men (36.5%) and 33 women (63.5%), mean age was 80 (Interquartile Range 12.5) years. 29 (55.8%) patients were well-nourished and 23 (44.2%) malnourished, 53.8% of patients developed sepsis at admission or during hospitalization. Total nosocomial mortality was 7.7 % (n = 4) and one-year mortality was 31.8% (n = 14). Comparative analyses showed older age (80 vs. 78; p = 0.012), less years of instruction (7 vs. 8; p = 0.027), lower MMST (14 vs. 27; p = 0.017), lower previous functional status (21 vs. 32; p < 0.0001), lower albumin (3 vs. 3.35; p = 0.014) and higher organic failure score at admission (3 vs. 1; p = 0.01) with more number of affected organs (2 vs. 1; p = 0.003) in malnourished patients compared to well nourished ones. Higher incidence of sepsis -at admission or during hospitalization- (73.9% vs. 37.9%; p = 0.01) and more severe stages of sepsis were also observed in malnourished patients. One-year mortality was significantly higher in malnourished (52.2% vs. 9.5%, log rank test = 0.002). In conclusion, malnourished patients presented greater systemic inflammatory response.


Asunto(s)
Evaluación Geriátrica/estadística & datos numéricos , Hospitalización/estadística & datos numéricos , Desnutrición/mortalidad , Evaluación Nutricional , Estado Nutricional/fisiología , Síndrome de Respuesta Inflamatoria Sistémica/mortalidad , Anciano , Anciano de 80 o más Años , Argentina/epidemiología , Comorbilidad , Femenino , Anciano Frágil/estadística & datos numéricos , Humanos , Pacientes Internos/estadística & datos numéricos , Masculino , Estudios Prospectivos
6.
Arch Biochem Biophys ; 484(2): 214-20, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19159609

RESUMEN

Mitochondria require nitric oxide ((.)NO) to exert a delicate control of metabolic rate as well as to regulate life functions, cell cycle activation and arrest, and apoptosis. All activities depend on the matrical (.)NO steady state concentration as provided by mitochondrial (mtNOS) and cytosolic sources (eNOS) and reduced by forming superoxide anion and H2O2 and a low peroxynirite (ONOO(-)) yield. We review herein the biochemical pathways involved in the control of (.)NO mitochondrial level and its biological and physiological significance in hormone effects and aging. At high ()NO, the cost of this physiological regulation is that ONOO(-) excess will lead to nitrosation/nitration and oxidization of mitochondrial and cell proteins and lipids. The disruption of (.)NO modulation of mitochondrial respiration supports then, a platform for prevalent neurodegenerative and metabolic diseases.


Asunto(s)
Mitocondrias/fisiología , Óxido Nítrico/fisiología , Ácido Peroxinitroso/metabolismo , Envejecimiento/fisiología , Animales , Humanos , Insulina/fisiología , Leptina/fisiología , Mitocondrias/enzimología , Mitocondrias/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Consumo de Oxígeno , Ratas , Transducción de Señal , Superóxido Dismutasa/metabolismo , Glándula Tiroides/fisiología
7.
Medicina (B Aires) ; 69(4): 460-5, 2009.
Artículo en Español | MEDLINE | ID: mdl-19770103

RESUMEN

The main function of the immune system is to protect the individual against potentially dangerous pathogens. It comprises innate and adaptive cellular and soluble components, both with the capacity to discriminate between harmful and harmless. These processes are regulated by homeostatic mechanisms that constitute the so-called immunological tolerance, which aims to limit the prolonged action of immune mediators and to silence the generation of potentially autoaggressive components. Failure to silence self-reactive T and B cells results in the generation of autoimmune disease. Recent advances in our knowledge of these pathological entities have opened a new chapter in the pharmacology of the immune system. Its promising potential currently offers new therapeutic agents to control and attenuate pathological tissue damage. Nevertheless, further research regarding these biologic agents is required, since they are not free from inconveniences. It is without question that upcoming findings in this field will instill hope into the quest for the "magic bullet".


Asunto(s)
Enfermedades Autoinmunes/inmunología , Autoinmunidad/inmunología , Enfermedades Transmisibles/inmunología , Tolerancia Inmunológica/inmunología , Enfermedades Autoinmunes/tratamiento farmacológico , Autoinmunidad/efectos de los fármacos , Enfermedades Transmisibles/tratamiento farmacológico , Humanos , Tolerancia Inmunológica/efectos de los fármacos
8.
Oncotarget ; 10(41): 4169-4179, 2019 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-31289615

RESUMEN

SPARC, also known as osteonectin and BM-40, is a matricellular protein with a number of biological functions. Hepatic SPARC expression is induced in response to thioacetamide, bile-duct ligation, and acute injuries such as concanavalin A and lipopolysacharide (LPS)/D-galactosamine. We have previously demonstrated that the therapeutic inhibition of SPARC or SPARC gene deletion protected mice against liver injury. We investigated the mechanisms involved in the protective effect of SPARC inhibition in mice. We performed a proteome analysis of livers from SPARC+/+ and SPARC-/- mice chronically treated with thioacetamide. Catalase activity, carbonylation levels, oxidative stress response, and mitochondrial function were studied. Genomic analysis revealed that SPARC-/- mice had an increased expression of cell proliferation genes. Proteins involved in detoxification of reactive oxygen species such as catalase, peroxirredoxine-1, and glutathione-S-transferase P1 and Mu1 were highly expressed as evidenced by proteome analysis; hepatic catalase activity was increased in SPARC-/- mice. Oxidative stress response and carbonylation levels were lower in livers from SPARC-/- mice. Hepatic mitochondria showed lower levels of nitrogen reactive species in the SPARC-/- concanavalin A-treated mice. Mitochondrial morphology was preserved, and its complex activity reduced in SPARC-/- mice. In conclusion, our data suggest that the protection associated with SPARC gene deletion may be partially due to a higher proliferative capacity of hepatocytes and an enhanced oxidative stress defense in SPARC-/- mice after liver injury.

9.
Eur J Neurosci ; 27(1): 123-31, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18184317

RESUMEN

NO-mediated toxicity contributes to neuronal damage after hypoxia; however, the molecular mechanisms involved are still a matter of controversy. Since mitochondria play a key role in signalling neuronal death, we aimed to determine the role of nitrative stress in hypoxia-induced mitochondrial damage. Therefore, we analysed the biochemical and ultrastructural impairment of these organelles in the optic lobe of chick embryos after in vivo hypoxia-reoxygenation. Also, we studied the NO-dependence of damage and examined modulation of mitochondrial nitric oxide synthase (mtNOS) after the hypoxic event. A transient but substantial increase in mtNOS content and activity was observed at 0-2 h posthypoxia, resulting in accumulation of nitrated mitochondrial proteins measured by immunoblotting. However, no variations in nNOS content were observed in the homogenates, suggesting an increased translocation to mitochondria and not a general de novo synthesis. In parallel with mtNOS kinetics, mitochondria exhibited prolonged inhibition of maximal complex I activity and ultrastructural phenotypes associated with swelling, namely, fading of cristae, intracristal dilations and membrane disruption. Administration of the selective nNOS inhibitor 7-nitroindazole 20 min before hypoxia prevented complex I inhibition and most ultrastructural damage. In conclusion, we show here for the first time that hypoxia induces NO-dependent complex I inhibition and ultrastructural damage by increasing mitochondrial NO in the developing brain.


Asunto(s)
Sistema Nervioso Central/embriología , Sistema Nervioso Central/ultraestructura , Complejo I de Transporte de Electrón/metabolismo , Hipoxia , Mitocondrias/enzimología , Óxido Nítrico/metabolismo , Animales , Sistema Nervioso Central/metabolismo , Embrión de Pollo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Regulación del Desarrollo de la Expresión Génica/fisiología , Hipoxia/metabolismo , Hipoxia/patología , Hipoxia/fisiopatología , Indazoles/farmacología , Microscopía Electrónica de Transmisión , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/metabolismo , Factores de Tiempo
10.
Front Biosci ; 12: 1041-8, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17127359

RESUMEN

In the last years, nitric oxide synthases (NOS) have been localized in mitochondria. At this site, NO yield directly regulates the activity of cytochrome oxidase, O(2) uptake and the production of reactive oxygen species. Recent studies showed that translocated neuronal nitric oxide synthase (nNOS) is posttranslationally modified including phosphorylation at Ser 1412 (in mice) and myristoylation in an internal residue. Different studies confirm that modified nNOS alpha is the main modulable isoform in mitochondria. Modulation of mtNOS was observed in different situations, like adaptation to reduced O(2) availability and hypoxia, adaptation to low environmental temperature, and processes linked to life and death by effects on kinases and transcription factors. We present here evidence about the role of mtNOS in the analyzed conditions.


Asunto(s)
Mitocondrias/enzimología , Óxido Nítrico Sintasa de Tipo I/fisiología , Adaptación Fisiológica , Animales , Encéfalo/embriología , Encéfalo/enzimología , Encéfalo/crecimiento & desarrollo , Hígado/embriología , Hígado/enzimología , Hígado/crecimiento & desarrollo , Ratones , Plasticidad Neuronal , Oxígeno/metabolismo , Triyodotironina/fisiología
11.
Int J Biochem Cell Biol ; 81(Pt B): 323-334, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27592449

RESUMEN

Sepsis-induced myocardial dysfunction is associated with increased oxidative stress and mitochondrial dysfunction. Current evidence suggests a protective role of thioredoxin-1 (Trx1) in the pathogenesis of cardiovascular diseases. However, it is unknown yet a putative role of Trx1 in sepsis-induced myocardial dysfunction, in which oxidative stress is an underlying cause. Transgenic male mice with Trx1 cardiac-specific overexpression (Trx1-Tg) and its wild-type control (wt) were subjected to cecal ligation and puncture or sham surgery. After 6, 18, and 24h, cardiac contractility, antioxidant enzymes, protein oxidation, and mitochondrial function were evaluated. Trx1 overexpression improved the average life expectancy (Trx1-Tg: 36, wt: 28h; p=0.0204). Sepsis induced a decrease in left ventricular developed pressure in both groups, while the contractile reserve, estimated as the response to ß-adrenergic stimulus, was higher in Trx1-Tg in relation to wt, after 6h of the procedure. Trx1 overexpression attenuated complex I inhibition, protein carbonylation, and loss of membrane potential, and preserved Mn superoxide dismutase activity at 24h. Ultrastructural alterations in mitochondrial cristae were accompanied by reduced optic atrophy 1 (OPA1) fusion protein, and activation of dynamin-related protein 1 (Drp1) (fission protein) in wt mice at 24h, suggesting mitochondrial fusion/fission imbalance. PGC-1α gene expression showed a 2.5-fold increase in Trx1-Tg at 24h, suggesting mitochondrial biogenesis induction. Autophagy, demonstrated by electron microscopy and increased LC3-II/LC3-I ratio, was observed earlier in Trx1-Tg. In conclusion, Trx1 overexpression extends antioxidant protection, attenuates mitochondrial damage, and activates mitochondrial turnover (mitophagy and biogenesis), preserves contractile reserve and prolongs survival during sepsis.


Asunto(s)
Expresión Génica , Mitocondrias/genética , Miocardio/metabolismo , Sepsis , Tiorredoxinas/genética , Tiorredoxinas/metabolismo , Animales , Antioxidantes/metabolismo , Masculino , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Mitocondrias/patología , Enfermedades Mitocondriales/genética , Miocardio/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Sepsis/fisiopatología
12.
Mol Aspects Med ; 25(1-2): 125-39, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15051322

RESUMEN

Mitochondria are the specialized organelles for energy metabolism but also participate in the production of O(2) active species, cell cycle regulation, apoptosis and thermogenesis. Classically, regulation of mitochondrial energy functions was based on the ADP/ATP ratio, which dynamically stimulates the transition between resting and maximal O(2) uptake. However, in the last years, NO was identified as a physiologic regulator of electron transfer and ATP synthesis by inhibiting cytochrome oxidase. Additionally, NO stimulates the mitochondrial production of O(2) active species, primarily O(2)(-) and H(2)O(2), and, depending on NO matrix concentration, of ONOO(-), which is responsible for the nitrosylation and nitration of mitochondrial components. By this means, alteration in mitochondrial complexes restricts energy output, further increases O(2) active species and changes cell signaling for proliferation and apoptosis through redox effects on specific pathways. These mechanisms are prototypically operating in prevalent generalized diseases like sepsis with multiorgan failure or limited neurodegenerative disorders like Parkinson's disease. Complex I appears to be highly susceptible to ONOO(-) effects and nitration, which defines an acquired group of mitochondrial disorders, in addition to the genetically induced syndromes. Increase of mitochondrial NO may follow over-expression of nNOS, induction and translocation of iNOS, and activation and/or increased content of the newly described mtNOS. Likewise, mtNOS is important in the modulation of O(2) uptake and cell signaling, and in mitochondrial pathology, including the effects of aging, dystrophin deficiency, hypoxia, inflammation and cancer.


Asunto(s)
Complejo I de Transporte de Electrón/fisiología , Mitocondrias/fisiología , Óxido Nítrico Sintasa/fisiología , Óxido Nítrico/fisiología , Humanos , Estrés Oxidativo/fisiología , Enfermedad de Parkinson/fisiopatología
13.
Free Radic Biol Med ; 32(2): 115-21, 2002 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11796199

RESUMEN

Oxidation of catecholamines is suggested to contribute to oxidative stress in Parkinson's disease. Nitric oxide (*NO) is able to oxidize cyclic compounds like ubiquinol; moreover, recent lines of evidence proposed a direct role of *NO and its by-product peroxynitrite in the pathophysiology of Parkinson's disease. The aim of this study was to analyze the potential reaction between 6-hydroxydopamine, a classic inducer of Parkinson's disease, and *NO. The results showed that *NO reacts with the deprotonated form of 6-hydroxydopamine at pH 7 and 37 degrees C with a second-order rate constant of 1.5 x 10(3) M(-1) x s(-1) as calculated by the rate of *NO decay measured with an amperometric sensor. Accordingly, the rates of formation of 6-hydroxy-dopamine quinone were dependent on *NO concentration. The coincubation of *NO and 6-hydroxydopamine with either bovine serum albumin or alpha-synuclein led to tyrosine nitration of the protein, in a concentration dependent-manner and sensitive to superoxide dismutase. These findings suggest the formation of peroxynitrite during the redox reactions following the interaction of 6-hydroxydopamine with *NO. The implications of this reaction for in vivo models are discussed in terms of the generation of reactive nitrogen and oxygen species within a propagation process that may play a significant role in neurodegenerative diseases.


Asunto(s)
Óxido Nítrico/metabolismo , Oxidopamina/metabolismo , Enfermedad de Parkinson/metabolismo , Ácido Peroxinitroso/metabolismo , Tirosina/análogos & derivados , Tirosina/biosíntesis , Animales , Bovinos , Proteínas del Tejido Nervioso/metabolismo , Oxidación-Reducción , Oxidopamina/efectos adversos , Enfermedad de Parkinson Secundaria/inducido químicamente , Albúmina Sérica Bovina/metabolismo , Superóxido Dismutasa/farmacología , Sinucleínas , Tirosina/antagonistas & inhibidores , alfa-Sinucleína
14.
Intensive Care Med ; 29(1): 135-8, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12528035

RESUMEN

OBJECTIVE: To determine outcome and natural course of systemic inflammatory response syndrome (SIRS) stages in adult febrile neutropenic patients. DESIGN AND SETTING: Retrospective cohort study in the medicine department and intensive care unit of a university hospital. PATIENT: Adults with cancer-related neutropenia and community-acquired fever. MEASUREMENTS AND RESULTS: Patients were classified on admission according to SIRS parameters, tumor type, and degree of neutropenia. Records of clinical and laboratory data during hospitalization were reviewed. Univariate and logistic regression analyses were performed. Seventy-nine events in 62 patients were analyzed. Overall mortality rate was 20.2% (16/79). Mortality increased as SIRS stage worsened on admission. No patients with stage 2 SIRS died (neutropenia and fever alone) but 11.1% of patients with SIRS 3, 43.4% with SIRS 4, 66.6% with sepsis induced hypotension, and 90% with septic shock. SIRS stage on admission was an independent predictor of death and was related directly to rate of progression to shock, i.e., none of the patients with SIRS 2, 2.7%(1/36) of those with SIRS 3, and 30.4% (7/23) of those admitted with SIRS 4. CONCLUSIONS: Mortality and progression to septic shock increased as more SIRS criteria were met on admission. SIRS stages could serve as a risk-assessing model in febrile neutropenic patients.


Asunto(s)
Neutropenia/complicaciones , Síndrome de Respuesta Inflamatoria Sistémica/epidemiología , Análisis de Varianza , Argentina/epidemiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Prevalencia , Pronóstico , Estudios Retrospectivos , Riesgo , Estadísticas no Paramétricas , Tasa de Supervivencia , Síndrome de Respuesta Inflamatoria Sistémica/diagnóstico , Síndrome de Respuesta Inflamatoria Sistémica/mortalidad
15.
Antioxid Redox Signal ; 15(9): 2395-406, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21529143

RESUMEN

AIMS: Obesity arises on defective neuroendocrine pathways that increase energy intake and reduce mitochondrial metabolism. In the metabolic syndrome, mitochondrial dysfunction accomplishes defects in fatty acid oxidation and reciprocal increase in triglyceride content with insulin resistance and hyperglycemia. Mitochondrial inhibition is attributed to reduced biogenesis, excessive fission, and low adipokine-AMP-activated protein kinase (AMPK) level, but lateness of the respiratory chain contributes to perturbations. Considering that nitric oxide (NO) binds cytochrome oxidase and inhibits respiration, we explored NO as a direct effector of mitochondrial dysfunction in the leptin-deficient ob/ob mice. RESULTS: A remarkable three- to fourfold increase in neuronal nitric oxide synthase (nNOS) expression and activity was detected by western blot, citrulline assay, electronic and confocal microscopy, flow cytometry, and NO electrode sensor in mitochondria from ob/ob mice. High NO reduced oxygen uptake in ob/ob mitochondria by inhibition of complex IV and nitration of complex I. Low metabolic status restricted ß-oxidation in obese mitochondria and displaced acetyl-CoA to fat synthesis; instead, small interference RNA nNOS caused a phenotype change with fat reduction in ob/ob adipocytes. INNOVATION: We evidenced that leptin increases mitochondrial respiration and fat utilization by potentially inhibiting NO release. Accordingly, leptin administration to ob/ob mice prevented nNOS overexpression and mitochondrial dysfunction in vivo and rescued leptin-dependent effects by matrix NO reduction, whereas leptin-Ob-Rb disruption increased the formation of mitochondrial NO in control adipocytes. We demonstrated that in ob/ob, hypoleptinemia is associated with critically low mitochondrial p-AMPK and that, oppositely to p-Akt2, p-AMPK is a negative modulator of nNOS. CONCLUSION: Thereby, defective leptin-AMPK pathway links mitochondrial NO to obesity with complex I syndrome and dysfunctional mitochondria.


Asunto(s)
Adenilato Quinasa/metabolismo , Leptina/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Óxido Nítrico/metabolismo , Obesidad/metabolismo , Animales , Western Blotting , Ácidos Grasos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Microscopía Confocal , Microscopía Electrónica , Mitocondrias/ultraestructura , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo I/metabolismo , ARN Interferente Pequeño , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
16.
Methods Enzymol ; 457: 169-92, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19426868

RESUMEN

In order to achieve the goal of this article, as an example we will describe the strategies followed to analyze the presence of the multi-kinase complex at the mitochondria and the posttranslational modification of two key mitochondrial proteins, which participate in the regulation of cholesterol transport across the mitochondrial membranes and in the regulation of steroid biosynthesis. Hormones, ions or growth factors modulate steroid biosynthesis by the posttranslational phosphorylation of proteins. The question still remains on how phosphorylation events transmit a specific signal to its mitochondrial site of action. Cholesterol transport requires specific interactions in mitochondria between several proteins including a multi-kinase complex. The presence of this multi-kinase complex at the mitochondria reveals the importance of the posttranslational modification of mitochondrial proteins for its activity and functions. The activation of PKA triggers the posttranslational modification of the mitochondrial acyl-CoA thioesterase (Acot2), which releases arachidonic acid (AA) in the mitochondria, and the activation of a kinase cascade that leads to the phoshorylation of the steroidogenic acute regulatory (StAR) protein. The function of StAR is to facilitate the access of cholesterol to the first enzyme of the biosynthesis process and its induction is dependent on Acot2 and intramitochondrial AA release. Truncation of the StAR protein is associated with the steroid deficiency disease, congenital lipoid adrenal hyperplasia.


Asunto(s)
Mitocondrias/enzimología , Proteínas Quinasas/análisis , Proteínas Quinasas/metabolismo , Esteroides/biosíntesis , Tioléster Hidrolasas/metabolismo , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/análisis , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Mitocondrias/química , Quinasas de Proteína Quinasa Activadas por Mitógenos/análisis , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Mutación , Fosforilación , Proteínas Quinasas/genética , Procesamiento Proteico-Postraduccional , Tioléster Hidrolasas/análisis , Tioléster Hidrolasas/genética , Transfección
17.
Exp Biol Med (Maywood) ; 234(9): 1020-8, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19546350

RESUMEN

Mitochondria are specialized organelles that control energy metabolism and also activate a multiplicity of pathways that modulate cell proliferation and mitochondrial biogenesis or, conversely, promote cell arrest and programmed cell death by a limited number of oxidative or nitrative reactions. Nitric oxide (NO) regulates oxygen uptake by reversible inhibition of cytochrome oxidase and the production of superoxide anion from the mitochondrial electron transfer chain. In this sense, NO produced by mtNOS will set the oxygen uptake level and contribute to oxidation-reduction reaction (redox)-dependent cell signaling. Modulation of translocation and activation of neuronal nitric oxide synthase (mtNOS activity) under different physiologic or pathologic conditions represents an adaptive response properly modulated to adjust mitochondria to different cell challenges.


Asunto(s)
Metabolismo Energético , Mitocondrias/enzimología , Mitocondrias/fisiología , Óxido Nítrico Sintasa/metabolismo , Estrés Fisiológico , Óxido Nítrico/metabolismo
18.
PLoS One ; 3(1): e1443, 2008 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-18197253

RESUMEN

ERK1/2 is known to be involved in hormone-stimulated steroid synthesis, but its exact roles and the underlying mechanisms remain elusive. Both ERK1/2 phosphorylation and steroidogenesis may be triggered by cAMP/cAMP-dependent protein kinase (PKA)-dependent and-independent mechanisms; however, ERK1/2 activation by cAMP results in a maximal steroidogenic rate, whereas canonical activation by epidermal growth factor (EGF) does not. We demonstrate herein by Western blot analysis and confocal studies that temporal mitochondrial ERK1/2 activation is obligatory for PKA-mediated steroidogenesis in the Leydig-transformed MA-10 cell line. PKA activity leads to the phosphorylation of a constitutive mitochondrial MEK1/2 pool with a lower effect in cytosolic MEKs, while EGF allows predominant cytosolic MEK activation and nuclear pERK1/2 localization. These results would explain why PKA favors a more durable ERK1/2 activation in mitochondria than does EGF. By means of ex vivo experiments, we showed that mitochondrial maximal steroidogenesis occurred as a result of the mutual action of steroidogenic acute regulatory (StAR) protein -a key regulatory component in steroid biosynthesis-, active ERK1/2 and PKA. Our results indicate that there is an interaction between mitochondrial StAR and ERK1/2, involving a D domain with sequential basic-hydrophobic motifs similar to ERK substrates. As a result of this binding and only in the presence of cholesterol, ERK1/2 phosphorylates StAR at Ser(232). Directed mutagenesis of Ser(232) to a non-phosphorylable amino acid such as Ala (StAR S232A) inhibited in vitro StAR phosphorylation by active ERK1/2. Transient transfection of MA-10 cells with StAR S232A markedly reduced the yield of progesterone production. In summary, here we show that StAR is a novel substrate of ERK1/2, and that mitochondrial ERK1/2 is part of a multimeric protein kinase complex that regulates cholesterol transport. The role of MAPKs in mitochondrial function is underlined.


Asunto(s)
Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Progesterona/biosíntesis , Animales , Línea Celular , Colesterol/metabolismo , AMP Cíclico/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Activación Enzimática , Factor de Crecimiento Epidérmico/farmacología , Ratones , Mitocondrias/metabolismo , Fosfoproteínas/metabolismo , Fosforilación
19.
PLoS One ; 3(3): e1749, 2008 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-18335029

RESUMEN

BACKGROUND: In the metabolic syndrome with hyperinsulinemia, mitochondrial inhibition facilitates muscle fat and glycogen accumulation and accelerates its progression. In the last decade, nitric oxide (NO) emerged as a typical mitochondrial modulator by reversibly inhibiting citochrome oxidase and oxygen utilization. We wondered whether insulin-operated signaling pathways modulate mitochondrial respiration via NO, to alternatively release complete glucose oxidation to CO(2) and H(2)O or to drive glucose storage to glycogen. METHODOLOGY/PRINCIPAL FINDINGS: We illustrate here that NO produced by translocated nNOS (mtNOS) is the insulin-signaling molecule that controls mitochondrial oxygen utilization. We evoke a hyperinsulinemic-normoglycemic non-invasive clamp by subcutaneously injecting adult male rats with long-lasting human insulin glargine that remains stable in plasma by several hours. At a precise concentration, insulin increased phospho-Akt2 that translocates to mitochondria and determines in situ phosphorylation and substantial cooperative mtNOS activation (+4-8 fold, P<.05), high NO, and a lowering of mitochondrial oxygen uptake and resting metabolic rate (-25 to -60%, P<.05). Comparing in vivo insulin metabolic effects on gastrocnemius muscles by direct electroporation of siRNA nNOS or empty vector in the two legs of the same animal, confirmed that in the silenced muscles disrupted mtNOS allows higher oxygen uptake and complete (U-(14)C)-glucose utilization respect to normal mtNOS in the vector-treated ones (respectively 37+/-3 vs 10+/-1 micromolO(2)/h.g tissue and 13+/-1 vs 7.2+/-1 micromol (3)H(2)O/h.g tissue, P<.05), which reciprocally restricted glycogen-synthesis by a half. CONCLUSIONS/SIGNIFICANCE: These evidences show that after energy replenishment, insulin depresses mitochondrial respiration in skeletal muscle via NO which permits substrates to be deposited as macromolecules; at discrete hyperinsulinemia, persistent mtNOS activation could contribute to mitochondrial dysfunction with insulin resistance and obesity and therefore, to the progression of the metabolic syndrome.


Asunto(s)
Insulina/fisiología , Síndrome Metabólico/metabolismo , Mitocondrias Musculares/fisiología , Óxido Nítrico Sintasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Activación Enzimática , Humanos , Síndrome Metabólico/enzimología , Mitocondrias Musculares/enzimología , Músculo Esquelético/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Transducción de Señal
20.
J Biol Chem ; 281(8): 4779-86, 2006 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-16361261

RESUMEN

Although transcriptional effects of thyroid hormones have substantial influence on oxidative metabolism, how thyroid sets basal metabolic rate remains obscure. Compartmental localization of nitric-oxide synthases is important for nitric oxide signaling. We therefore examined liver neuronal nitric-oxide synthase-alpha (nNOS) subcellular distribution as a putative mechanism for thyroid effects on rat metabolic rate. At low 3,3',5-triiodo-L-thyronine levels, nNOS mRNA increased by 3-fold, protein expression by one-fold, and nNOS was selectively translocated to mitochondria without changes in other isoforms. In contrast, under thyroid hormone administration, mRNA level did not change and nNOS remained predominantly localized in cytosol. In hypothyroidism, nNOS translocation resulted in enhanced mitochondrial nitric-oxide synthase activity with low O2 uptake. In this context, NO utilization increased active O2 species and peroxynitrite yields and tyrosine nitration of complex I proteins that reduced complex activity. Hypothyroidism was also associated to high phospho-p38 mitogen-activated protein kinase and decreased phospho-extracellular signal-regulated kinase 1/2 and cyclin D1 levels. Similarly to thyroid hormones, but without changing thyroid status, nitric-oxide synthase inhibitor N(omega)-nitro-L-arginine methyl ester increased basal metabolic rate, prevented mitochondrial nitration and complex I derangement, and turned mitogen-activated protein kinase signaling and cyclin D1 expression back to control pattern. We surmise that nNOS spatial confinement in mitochondria is a significant downstream effector of thyroid hormone and hypothyroid phenotype.


Asunto(s)
Complejo I de Transporte de Electrón/metabolismo , Hipotiroidismo/patología , Hígado/enzimología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Animales , Ciclina D1/metabolismo , Citosol/metabolismo , Electrones , Electroforesis en Gel de Poliacrilamida , Proteínas HSP90 de Choque Térmico/metabolismo , Hipotiroidismo/metabolismo , Immunoblotting , Inmunoprecipitación , Hígado/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Microscopía Inmunoelectrónica , Mitocondrias/metabolismo , Mitocondrias Hepáticas/metabolismo , Modelos Químicos , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/metabolismo , Oxidantes/metabolismo , Oxígeno/metabolismo , Ácido Peroxinitroso/química , Fenotipo , Isoformas de Proteínas , Transporte de Proteínas , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Fracciones Subcelulares/metabolismo , Hormonas Tiroideas/metabolismo , Transcripción Genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA