Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Infect Dis Ther ; 13(1): 189-205, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38233606

RESUMEN

INTRODUCTION: Molnupiravir is an orally available prodrug of N-hydroxycytidine that received special approval for emergency treatment of coronavirus disease 2019 (COVID-19) in Japan in December 2021 and full approval in April 2023. To assess the real-world safety and effectiveness of molnupiravir in Japanese patients with COVID-19, we conducted nationwide post-marketing surveillance to collect data at registered institutions in Japan. METHODS: The surveillance data were collected from December 27, 2021, to May 2, 2023. All reported adverse events were collected for safety analysis. Adverse drug reactions (ADRs) were assessed by the treating physicians. Effectiveness was assessed by the composite of hospitalization or all-cause death in outpatients and the composite of oxygen/mechanical ventilation initiation or all-cause death in inpatients. The observation period was from molnupiravir initiation through day 29. RESULTS: Of 3214 patients enrolled in the survey, 3179 were analyzed for safety. At baseline, 52.31% (1663/3179) of patients were male, the median (range) age was 69.0 (18-107) years, 82.38% (2619/3179) received COVID-19 vaccines, and 95.72% (3043/3179) had risk factors for severe COVID-19 illness. COVID-19 severity at baseline was mild in 86.44% (2748/3179) and moderate I in 10.22% (325/3179). A total of 205 ADRs occurred in 5.50% (175/3179) of patients; ADRs that occurred in > 0.5% of patients were diarrhea (1.86% [59/3179]) and rash (0.69% [22/3179]). Seven serious ADRs were reported in seven patients. In the effectiveness analysis population, the incidence of all-cause death through day 29 was 1.14% (34/2988), and the incidence of death through day 29 related to COVID-19 was 0.40% (12/2988). The cumulative incidence of the composite endpoint was 2.34% (47/2006) in outpatients and 4.60% (38/826) in inpatients. CONCLUSIONS: This large-scale survey showed that molnupiravir was safe and effective in real-world settings in highly vaccinated Japanese patients with COVID-19, including older patients and those with comorbidities.

2.
Am J Physiol Endocrinol Metab ; 320(1): E150-E159, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33284091

RESUMEN

Sepsis remains a leading cause of mortality in critically ill patients. Muscle wasting is a major complication of sepsis and negatively affects clinical outcomes. Despite intense investigation for many years, the molecular mechanisms underlying sepsis-related muscle wasting are not fully understood. In addition, a potential role of muscle wasting in disease development of sepsis has not been studied. Myostatin is a myokine that downregulates skeletal muscle mass. We studied the effects of myostatin deficiency on muscle wasting and other clinically relevant outcomes, including mortality and bacterial clearance, in mice. Myostatin deficiency prevented muscle atrophy along with inhibition of increases in muscle-specific RING finger protein 1 (MuRF-1) and atrogin-1 expression and phosphorylation of signal transducer and activator of transcription protein 3 (STAT3; major players of muscle wasting) in septic mice. Moreover, myostatin deficiency improved survival and bacterial clearance of septic mice. Sepsis-induced liver dysfunction, acute kidney injury, and neutrophil infiltration into the liver and kidney were consistently mitigated by myostatin deficiency, as indicated by plasma concentrations of aspartate aminotransferase (AST), alanine aminotransferase (ALT), and neutrophil gelatinase-associated lipocalin (NGAL) and myeloperoxidase activity in the organs. Myostatin deficiency also inhibited sepsis-induced increases in plasma high-mobility group protein B1 (HMGB1) and macrophage inhibitory cytokine (MIC)-1/growth differentiation factor (GDF)-15 concentrations. These results indicate that myostatin plays an important role not only in muscle wasting but also in other clinically relevant outcomes in septic mice. Furthermore, our data raise the possibility that muscle wasting may not be simply a complication, but myostatin-mediated muscle cachexia and related changes in muscle may actually drive the development of sepsis as well.NEW & NOTEWORTHY Muscle wasting is a major complication of sepsis, but its role in the disease development is not known. Myostatin deficiency improved bacterial clearance and survival and mitigated damage in the liver and kidney in septic mice, which paralleled prevention of muscle wasting. These results raise the possibility that muscle wasting may not simply be a complication of sepsis, but myostatin-mediated cachexic changes may have a role in impaired bacterial clearance and mortality in septic mice.


Asunto(s)
Atrofia Muscular/genética , Miostatina/deficiencia , Miostatina/genética , Sepsis/genética , Lesión Renal Aguda/genética , Animales , Caquexia/genética , Caquexia/prevención & control , Lipocalina 2/sangre , Hepatopatías/etiología , Hepatopatías/genética , Pruebas de Función Hepática , Masculino , Ratones , Proteínas Musculares/biosíntesis , Proteínas Musculares/genética , Atrofia Muscular/prevención & control , Infiltración Neutrófila/genética , Fosforilación , Factor de Transcripción STAT3/biosíntesis , Factor de Transcripción STAT3/genética , Sepsis/microbiología , Sepsis/mortalidad , Análisis de Supervivencia , Proteínas de Motivos Tripartitos/biosíntesis , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/biosíntesis , Ubiquitina-Proteína Ligasas/genética
3.
FEBS Open Bio ; 9(2): 348-363, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30761259

RESUMEN

Mitochondrial dysfunction is associated with metabolic alterations in various disease states, including major trauma (e.g., burn injury). Metabolic derangements, including muscle insulin resistance and hyperlactatemia, are a clinically significant complication of major trauma. Coenzyme Q10 (CoQ10) is an essential cofactor for mitochondrial electron transport, and its reduced form acts as a lipophilic antioxidant. Here, we report that burn injury induces impaired muscle insulin signaling, hyperlactatemia, mitochondrial dysfunction (as indicated by suppressed mitochondrial oxygen consumption rates), morphological alterations of the mitochondria (e. g., enlargement, and loss of cristae structure), mitochondrial oxidative stress, and disruption of mitochondrial integrity (as reflected by increased mitochondrial DNA levels in the cytosol and circulation). All of these alterations were significantly alleviated by CoQ10 treatment compared with vehicle alone. These findings indicate that CoQ10 treatment is efficacious in protecting against mitochondrial dysfunction and insulin resistance in skeletal muscle of burned mice. Our data highlight CoQ10 as a potential new strategy to prevent mitochondrial damage and metabolic dysfunction in burn patients.


Asunto(s)
Quemaduras/metabolismo , Insulina/metabolismo , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Transducción de Señal , Ubiquinona/análogos & derivados , Animales , Masculino , Ratones , Ubiquinona/metabolismo
4.
Biochem Biophys Res Commun ; 501(4): 893-897, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29763605

RESUMEN

Previous studies have shown that androgenic alopecia is associated with metabolic syndrome and diabetes. However, the detailed mechanism whereby diabetes causes alopecia still remains unclear. We focused on the inflammatory response that is caused by diabetes or obesity, given that inflammation is a risk factor for hair loss. Inducible nitric oxide synthase (iNOS) is known to be upregulated under conditions of acute or chronic inflammation. To clarify the potential role of iNOS in diabetes-related alopecia, we generated obese diabetic iNOS-deficient (ob/ob; iNOS-KO mice). We observed that ob/ob; iNOS-KO mice were potentiated for the transition from telogen (rest phase) to anagen (growth phase) in the hair cycle compared with iNOS-proficient ob/ob mice. To determine the effect of nitric oxide (NO) on the hair cycle, we administered an iNOS inhibitor intraperitoneally (compound 1400 W, 10 mg/kg) or topically (10% aminoguanidine) in ob/ob mice. We observed that iNOS inhibitors promoted anagen transition in ob/ob mice. Next, we administered an NO donor (S-nitrosoglutathione, GSNO), to test whether NO has the telogen elongation effects. The NO donor was sufficient to induce telogen elongation in wild-type mice. Together, our data indicate that iNOS-derived NO plays a role in telogen elongation under the inflammatory conditions associated with diabetes in mice.


Asunto(s)
Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/fisiopatología , Cabello/fisiopatología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Obesidad/fisiopatología , Regeneración , Administración Tópica , Animales , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Cabello/efectos de los fármacos , Cabello/enzimología , Cabello/crecimiento & desarrollo , Inyecciones Intraperitoneales , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regeneración/efectos de los fármacos , S-Nitrosoglutatión/metabolismo
5.
Sci Rep ; 7(1): 6618, 2017 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-28747716

RESUMEN

Metabolic derangements are a clinically significant complication of major trauma (e.g., burn injury) and include various aspects of metabolism, such as insulin resistance, muscle wasting, mitochondrial dysfunction and hyperlactatemia. Nonetheless, the molecular pathogenesis and the relation between these diverse metabolic alterations are poorly understood. We have previously shown that burn increases farnesyltransferase (FTase) expression and protein farnesylation and that FTase inhibitor (FTI) prevents burn-induced hyperlactatemia, insulin resistance, and increased proteolysis in mouse skeletal muscle. In this study, we found that burn injury activated mTORC1 and hypoxia-inducible factor (HIF)-1α, which paralleled dysfunction, morphological alterations (i.e., enlargement, partial loss of cristae structure) and impairment of respiratory supercomplex assembly of the mitochondria, and ER stress. FTI reversed or ameliorated all of these alterations in burned mice. These findings indicate that these burn-induced changes, which encompass various aspects of metabolism, may be linked to one another and require protein farnesylation. Our results provide evidence of involvement of the mTORC1-HIF-1α pathway in burn-induced metabolic derangements. Our study identifies protein farnesylation as a potential hub of the signaling network affecting multiple aspects of metabolic alterations after burn injury and as a novel potential molecular target to improve the clinical outcome of severely burned patients.


Asunto(s)
Quemaduras/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Mitocondrias/metabolismo , Músculos/patología , Prenilación de Proteína , Animales , Modelos Animales de Enfermedad , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Estrés del Retículo Endoplásmico , Redes y Vías Metabólicas , Ratones Endogámicos C57BL , Multimerización de Proteína
6.
PLoS One ; 12(1): e0170391, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28099528

RESUMEN

Inflammation and apoptosis develop in skeletal muscle after major trauma, including burn injury, and play a pivotal role in insulin resistance and muscle wasting. We and others have shown that inducible nitric oxide synthase (iNOS), a major mediator of inflammation, plays an important role in stress (e.g., burn)-induced insulin resistance. However, it remains to be determined how iNOS induces insulin resistance. Moreover, the interrelation between inflammatory response and apoptosis is poorly understood, although they often develop simultaneously. Nuclear factor (NF)-κB and p53 are key regulators of inflammation and apoptosis, respectively. Sirt1 inhibits p65 NF-κB and p53 by deacetylating these transcription factors. Recently, we have shown that iNOS induces S-nitrosylation of Sirt1, which inactivates Sirt1 and thereby increases acetylation and activity of p65 NF-κB and p53 in various cell types, including skeletal muscle cells. Here, we show that iNOS enhances burn-induced inflammatory response and apoptotic change in mouse skeletal muscle along with S-nitrosylation of Sirt1. Burn injury induced robust expression of iNOS in skeletal muscle and gene disruption of iNOS significantly inhibited burn-induced increases in inflammatory gene expression and apoptotic change. In parallel, burn increased Sirt1 S-nitrosylation and acetylation and DNA-binding capacity of p65 NF-κB and p53, all of which were reversed or ameliorated by iNOS deficiency. These results indicate that iNOS functions not only as a downstream effector but also as an upstream enhancer of burn-induced inflammatory response, at least in part, by Sirt1 S-nitrosylation-dependent activation (acetylation) of p65 NF-κB. Our data suggest that Sirt1 S-nitrosylation may play a role in iNOS-mediated enhanced inflammatory response and apoptotic change, which, in turn, contribute to muscle wasting and supposedly to insulin resistance after burn injury.


Asunto(s)
Apoptosis/fisiología , Quemaduras/patología , Inflamación/patología , Músculo Esquelético/patología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Sirtuina 1/metabolismo , Factor de Transcripción ReIA/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Animales , Proteínas de Unión al ADN/metabolismo , Activación Enzimática , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/genética
7.
J Pharm Pharmacol ; 68(12): 1567-1576, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27757967

RESUMEN

OBJECTIVES: Non-alcoholic fatty liver disease (NAFLD) is associated with impaired liver function, and resveratrol could suppress NAFLD progression. This study examined the effects of NAFLD on the expression of major cytochrome P450 (CYP) subtypes in the liver and whether the expression could be attenuated by resveratrol. METHODS: C57BL/6 mice (male, 10 weeks of age) were fed a high-fat and high-sucrose (HFHS) diet to induce NAFLD. Major Cyp subtype mRNA expression in the liver was measured by real-time RT-PCR. KEY FINDINGS: Body and liver weights at 4 and 12 weeks were significantly higher in mice fed the HFHS diet compared with control. The HFHS diet significantly increased the accumulation of cholesterol and triglycerides at 12 weeks. Under this condition, the HFHS diet increased the expression of Cyp1a2 and decreased that of Cyp3a11 at 1 week and thereafter. On the other hand, Cyp1a1, 2b10 and 2c29 mRNA expression levels in the liver were significantly increased at 12 weeks only. Resveratrol (0.05% (w/w) in diet) slightly suppressed lipid accumulation in the liver, but failed to recover impaired Cyp gene expression levels in NAFLD. CONCLUSIONS: Drug metabolism may be impaired in NAFLD, and each Cyp subtype is regulated in a different manner.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Dieta Alta en Grasa , Sacarosa en la Dieta , Hígado/enzimología , Enfermedad del Hígado Graso no Alcohólico/enzimología , Animales , Sistema Enzimático del Citocromo P-450/genética , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica , Isoenzimas , Lípidos/sangre , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/sangre , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Resveratrol , Estilbenos/farmacología , Factores de Tiempo
8.
Biochem Biophys Res Commun ; 472(1): 250-4, 2016 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-26923074

RESUMEN

Dihydrotestosterone (DHT) causes the regression of human hair follicles in the parietal scalp, leading to androgenic alopecia (AGA). Sulforaphane (SFN) increases the expression of DHT degrading enzymes, such as 3α-hydroxysteroid dehydrogenases (3α-HSDs), and, therefore, SFN treatment may improve AGA. To determine the effects of SFN on hair growth, we administered SFN (10 mg/kg BW, IP) or vehicle (DMSO) to ob/ob mice for six weeks and examined hair regeneration and the plasma levels of testosterone and DHT. We also tested the effects of SFN on the expression of two forms of 3α-HSD, aldo-keto reductase 1c21 and dehydrogenase/reductase (SDR family) member 9, both in vitro and in vivo. SNF significantly enhanced hair regeneration in ob/ob mice. The mice treated with SFN showed lower plasma levels of testosterone and DHT than those treated with vehicle. SFN increased the mRNA and protein levels of the two forms of 3α-HSD in the liver of the mice and in cultured murine hepatocyte Hepa1c1c7 cells. These results suggest that SFN treatment increases the amount of 3α-HSDs in the liver, accelerates the degradation of blood DHT, and subsequently blocks the suppression of hair growth by DHT.


Asunto(s)
Dihidrotestosterona/metabolismo , Cabello/efectos de los fármacos , Cabello/crecimiento & desarrollo , Isotiocianatos/farmacología , 3-Hidroxiesteroide Deshidrogenasas/genética , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Alopecia/tratamiento farmacológico , Alopecia/metabolismo , Alopecia/patología , Animales , Línea Celular , Dihidrotestosterona/sangre , Modelos Animales de Enfermedad , Humanos , Cinética , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Obesos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Sulfóxidos , Testosterona/sangre
9.
PLoS One ; 10(7): e0132029, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26172834

RESUMEN

Voluntary exercise can ameliorate insulin resistance. The underlying mechanism, however, remains to be elucidated. We previously demonstrated that inducible nitric oxide synthase (iNOS) in the liver plays an important role in hepatic insulin resistance in the setting of obesity. In this study, we tried to verify our hypothesis that voluntary exercise improves insulin resistance by reducing the expression of iNOS and subsequent S-nitrosylation of key molecules of glucose metabolism in the liver. Twenty-one Otsuka Long-Evans Tokushima Fatty (OLETF) rats, a model of type 2 diabetes mellitus, and 18 non-diabetic control Long-Evans Tokushima Otsuka (LETO) rats were randomly assigned to a sedentary group or exercise group subjected to voluntary wheel running for 20 weeks. The voluntary exercise significantly reduced the fasting blood glucose and HOMA-IR in the OLETF rats. In addition, the exercise decreased the amount of iNOS mRNA in the liver in the OLETF rats. Moreover, exercise reduced the levels of S-nitrosylated Akt in the liver, which were increased in the OLETF rats, to those observed in the LETO rats. These findings support our hypothesis that voluntary exercise improves insulin resistance, at least partly, by suppressing the iNOS expression and subsequent S-nitrosylation of Akt, a key molecule of the signal transduction pathways in glucose metabolism in the liver.


Asunto(s)
Resistencia a la Insulina , Hígado/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico/metabolismo , Obesidad/metabolismo , Condicionamiento Físico Animal , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Proteínas Sustrato del Receptor de Insulina/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Obesidad/enzimología , Obesidad/patología , Obesidad/fisiopatología , Fosforilación , Ratas , Transducción de Señal , Triglicéridos/metabolismo
10.
Sci Signal ; 7(351): ra106, 2014 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-25389371

RESUMEN

Inflammation increases the abundance of inducible nitric oxide synthase (iNOS), leading to enhanced production of nitric oxide (NO), which can modify proteins by S-nitrosylation. Enhanced NO production increases the activities of the transcription factors p53 and nuclear factor κB (NF-κB) in several models of disease-associated inflammation. S-nitrosylation inhibits the activity of the protein deacetylase SIRT1. SIRT1 limits apoptosis and inflammation by deacetylating p53 and p65 (also known as RelA), a subunit of NF-κB. We showed in multiple cultured mammalian cell lines that NO donors or inflammatory stimuli induced S-nitrosylation of SIRT1 within CXXC motifs, which inhibited SIRT1 by disrupting its ability to bind zinc. Inhibition of SIRT1 reduced deacetylation and promoted activation of p53 and p65, leading to apoptosis and increased expression of proinflammatory genes. In rodent models of systemic inflammation, Parkinson's disease, or aging-related muscular atrophy, S-nitrosylation of SIRT1 correlated with increased acetylation of p53 and p65 and activation of p53 and NF-κB target genes, suggesting that S-nitrosylation of SIRT1 may represent a proinflammatory switch common to many diseases and aging.


Asunto(s)
Inflamación , Nitrógeno/química , Sirtuina 1/metabolismo , Factor de Transcripción ReIA/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Apoptosis , Células COS , Chlorocebus aethiops , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Estrés Oxidativo , Ratas , Ratas Endogámicas F344 , Homología de Secuencia de Aminoácido
11.
PLoS One ; 8(10): e75249, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24204574

RESUMEN

Herp was originally identified as an endoplasmic reticulum (ER) stress protein in vascular endothelial cells. ER stress is induced in atherosclerotic lesions, but it is not known whether Herp plays any role in the development of atherosclerosis. To address this question, we generated Herp- and apolipoprotein E (apoE)-deficient mice (Herp(-/-); apoE(-/-) mice) by crossbreeding Herp(-/-) mice and apoE(-/-) mice. Herp was expressed in the endothelial cells and medial smooth muscle cells of the aorta, as well as in a subset of macrophages in the atherosclerotic lesions in apoE(-/-) mice, while there was no expression of Herp in the Herp(-/-); apoE(-/-) mice. The doubly deficient mice developed significantly fewer atherosclerotic lesions than the apoE(-/-) mice at 36 and 72 weeks of age, whereas the plasma levels of cholesterol and triglycerides were not significantly different between the strains. The plasma levels of non-esterified fatty acids were significantly lower in the Herp(-/-); apoE(-/-) mice when they were eight and 16 weeks old. The gene expression levels of ER stress response proteins (GRP78 and CHOP) and inflammatory cytokines (IL-1ß, IL-6, TNF-α and MCP-1) in the aorta were significantly lower in Herp(-/-); apoE(-/-) mice than in apoE(-/-) mice, suggesting that Herp mediated ER stress-induced inflammation. In fact, peritoneal macrophages isolated from Herp-deficient mice and RAW264.7 macrophages in which Herp was eliminated with a siRNA expressed lower levels of mRNA for inflammatory cytokines when they were treated with tunicamycin. Herp deficiency affected the major mediators of the unfolded protein response, including IRE1 and PERK, but not ATF6. These findings suggest that a deficiency of Herp suppressed the development of atherosclerosis by attenuating the ER stress-induced inflammatory reactions.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/genética , Proteínas de la Membrana/deficiencia , Animales , Aorta/metabolismo , Aorta/patología , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Arterias/metabolismo , Aterosclerosis/metabolismo , Aterosclerosis/patología , Glucemia , Línea Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico , Expresión Génica , Inflamación/genética , Inflamación/metabolismo , Lípidos/sangre , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Tunicamicina/farmacología , Respuesta de Proteína Desplegada/efectos de los fármacos , Respuesta de Proteína Desplegada/genética
12.
Am J Physiol Heart Circ Physiol ; 304(8): H1134-46, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23417863

RESUMEN

Myocardial depression is an important contributor to morbidity and mortality in septic patients. Nitric oxide (NO) plays an important role in the development of septic cardiomyopathy, but also has protective effects. Recent evidence has indicated that NO exerts many of its downstream effects on the cardiovascular system via protein S-nitrosylation, which is negatively regulated by S-nitrosoglutathione reductase (GSNOR), an enzyme promoting denitrosylation. We tested the hypothesis that reducing cardiomyocyte S-nitrosylation by increasing GSNOR activity can improve myocardial dysfunction during sepsis. Therefore, we generated mice with a cardiomyocyte-specific overexpression of GSNOR (GSNOR-CMTg mice) and subjected them to endotoxic shock. Measurements of cardiac function in vivo and ex vivo showed that GSNOR-CMTg mice had a significantly improved cardiac function after lipopolysaccharide challenge (LPS, 50 mg/kg) compared with wild-type (WT) mice. Cardiomyocytes isolated from septic GSNOR-CMTg mice showed a corresponding improvement in contractility compared with WT cells. However, systolic Ca(2+) release was similarly depressed in both genotypes after LPS, indicating that GSNOR-CMTg cardiomyocytes have increased Ca(2+) sensitivity during sepsis. Parameters of inflammation were equally increased in LPS-treated hearts of both genotypes, and no compensatory changes in NO synthase expression levels were found in GSNOR-overexpressing hearts before or after LPS challenge. GSNOR overexpression however significantly reduced total cardiac protein S-nitrosylation during sepsis. Taken together, our results indicate that increasing the denitrosylation capacity of cardiomyocytes protects against sepsis-induced myocardial depression. Our findings suggest that specifically reducing protein S-nitrosylation during sepsis improves cardiac function by increasing cardiac myofilament sensitivity to Ca(2+).


Asunto(s)
Cardiomiopatías/prevención & control , Glutatión Reductasa/metabolismo , Contracción Miocárdica/fisiología , Miocitos Cardíacos/metabolismo , Óxido Nítrico/metabolismo , Choque Séptico/metabolismo , Alcohol Deshidrogenasa , Animales , Calcio/metabolismo , Cardiomiopatías/etiología , Cardiomiopatías/fisiopatología , Glutatión Reductasa/genética , Lipopolisacáridos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocardio/metabolismo , Miocitos Cardíacos/fisiología , Óxido Nítrico Sintasa/metabolismo , Choque Séptico/complicaciones , Choque Séptico/fisiopatología
13.
Metabolism ; 62(3): 341-6, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22995863

RESUMEN

OBJECTIVES: Recent studies suggest that activation of glycogen synthase kinase (GSK)-3ß may be involved in burn injury-induced metabolic derangements and protein breakdown in skeletal muscle. However, the mechanism for GSK-3ß activation after burn injury is unknown. To investigate the role of inducible nitric oxide synthase (iNOS) in this scenario, a major mediator of inflammation, we examined the effects of a specific inhibitor for iNOS, L-NIL, on GSK-3ß activity in skeletal muscle of burned rats. MATERIALS/METHODS: Full-thickness third degree burn injury comprising 40% of total body surface area was produced under anesthesia in male Sprague-Dawley rats (160-190g) by immersing the back of the trunk for 15s and the abdomen for 8s in 80°C water. Burned and sham-burned rats were treated with L-NIL (60mg/kg BW, b.i.d., IP) or phosphate-buffered saline for three days. GSK-3ß activity in skeletal muscle was evaluated by immune complex kinase assay, and by phosphorylation status of GSK-3ß and its endogenous substrate, glycogen synthase. RESULTS: GSK-3ß activity was increased in a time-dependent manner in skeletal muscle after burn injury, concomitant with the induction of iNOS expression. iNOS inhibitor, L-NIL, reverted the elevated GSK-3ß activity in skeletal muscle of burned rats, although L-NIL did not alter GSK-3ß activity in sham-burned rats. CONCLUSIONS: Our results clearly indicate that iNOS plays an important role in burn injury-induced GSK-3ß activation in skeletal muscle. These findings suggest that iNOS may contribute to burn injury-induced metabolic derangements, in part, by activating GSK-3ß.


Asunto(s)
Quemaduras/enzimología , Inhibidores Enzimáticos/farmacología , Glucógeno Sintasa Quinasa 3/metabolismo , Lisina/análogos & derivados , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/enzimología , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Animales , Quemaduras/metabolismo , Activación Enzimática/efectos de los fármacos , Glucógeno Sintasa/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Lisina/farmacología , Masculino , Músculo Esquelético/lesiones , Músculo Esquelético/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosforilación , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
14.
Metabolism ; 61(1): 127-36, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21816442

RESUMEN

Burn injury is associated with inflammatory responses and metabolic alterations including insulin resistance. Impaired insulin receptor substrate-1 (IRS-1)-mediated insulin signal transduction is a major component of insulin resistance in skeletal muscle following burn injury. To further investigate molecular mechanisms that underlie burn injury-induced insulin resistance, we study a role of inducible nitric oxide synthase (iNOS), a major mediator of inflammation, on burn-induced muscle insulin resistance in iNOS-deficient mice. Full-thickness third-degree burn injury comprising 12% of total body surface area was produced in wild-type and iNOS-deficient C57BL/6 mice. Insulin-stimulated activation (phosphorylation) of IR, IRS-1, and Akt was assessed by immunoblotting and immunoprecipitation. Insulin-stimulated glucose uptake by skeletal muscle was evaluated ex vivo. Burn injury caused induction of iNOS in skeletal muscle of wild-type mice. The increase of iNOS expression paralleled the increase of insulin resistance, as evidenced by decreased tyrosine phosphorylation of IR and IRS-1, IRS-1 expression, insulin-stimulated activation of phosphatidylinositol 3-kinase and Akt/PKB, and insulin-stimulated glucose uptake in mouse skeletal muscle. The absence of iNOS in genetically engineered mice significantly lessened burn injury-induced insulin resistance in skeletal muscle. In wild-type mice, insulin tolerance test revealed whole-body insulin resistance in burned mice compared with sham-burned controls. This effect was reversed by iNOS deficiency. Unexpectedly, however, blood glucose levels were depressed in both wild-type and iNOS-deficient mice after burn injury. Gene disruption of iNOS ameliorated the effect of burn on IRS-1-mediated insulin signaling in skeletal muscle of mice. These findings indicate that iNOS plays a significant role in burn injury-induced skeletal muscle insulin resistance.


Asunto(s)
Glucemia/metabolismo , Quemaduras/metabolismo , Resistencia a la Insulina/fisiología , Músculo Esquelético/metabolismo , Óxido Nítrico Sintasa de Tipo II/deficiencia , Óxido Nítrico Sintasa de Tipo II/metabolismo , Animales , Quemaduras/sangre , Quemaduras/fisiopatología , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa/métodos , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/fisiología , Músculo Esquelético/fisiopatología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Insulina/metabolismo , Transducción de Señal , Tirosina/metabolismo
15.
FEBS Lett ; 585(19): 3058-64, 2011 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-21888904

RESUMEN

Nitric oxide (NO) has been implicated in pancreatic ß-cell death in the development of diabetes. The mechanisms underlying NO-induced ß-cell death have not been clearly defined. Recently, receptor-interacting protein-1 (RIP1)-dependent necrosis, which is inhibited by necrostatin-1, an inhibitor of RIP1, has emerged as a form of regulated necrosis. Here, we show that NO donor-induced ß-cell death was inhibited by necrostatin-1. Unexpectedly, however, RIP1 knockdown neither inhibited cell death nor altered the protective effects of necrostatin-1 in NO donor-treated ß-cells. These results indicate that NO donor induces necrostatin-1-inhibitable necrotic ß-cell death independent of RIP1. Our findings raise the possibility that NO-mediated ß-cell necrosis may be a novel form of signal-regulated necrosis, which play a role in the progression of diabetes.


Asunto(s)
Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Imidazoles/metabolismo , Indoles/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/patología , Donantes de Óxido Nítrico/farmacología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Línea Celular , Ciclofilina A/metabolismo , Técnicas de Silenciamiento del Gen , Proteína HMGB1/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , S-Nitrosoglutatión/farmacología , Transducción de Señal/efectos de los fármacos
16.
J Biol Chem ; 286(40): 34959-75, 2011 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-21846719

RESUMEN

Inducible nitric-oxide synthase (iNOS), a major mediator of inflammation, plays an important role in obesity-induced insulin resistance. Inhibition of iNOS by gene disruption or pharmacological inhibitors reverses or ameliorates obesity-induced insulin resistance in skeletal muscle and liver in mice. It is unknown, however, whether increased expression of iNOS is sufficient to cause insulin resistance in vivo. To address this issue, we generated liver-specific iNOS transgenic (L-iNOS-Tg) mice, where expression of the transgene, iNOS, is regulated under mouse albumin promoter. L-iNOS-Tg mice exhibited mild hyperglycemia, hyperinsulinemia, insulin resistance, and impaired insulin-induced suppression of hepatic glucose output, as compared with wild type (WT) littermates. Insulin-stimulated phosphorylation of insulin receptor substrate-1 (IRS-1) and -2, and Akt was significantly attenuated in liver, but not in skeletal muscle, of L-iNOS-Tg mice relative to WT mice without changes in insulin receptor phosphorylation. Moreover, liver-specific iNOS expression abrogated insulin-stimulated phosphorylation of glycogen synthase kinase-3ß, forkhead box O1, and mTOR (mammalian target of rapamycin), endogenous substrates of Akt, along with increased S-nitrosylation of Akt relative to WT mice. However, the expression of insulin receptor, IRS-1, IRS-2, Akt, glycogen synthase kinase-3ß, forkhead box O1, protein-tyrosine phosphatase-1B, PTEN (phosphatase and tensin homolog), and p85 phosphatidylinositol 3-kinase was not altered by iNOS transgene. Hyperglycemia was associated with elevated glycogen phosphorylase activity and decreased glycogen synthase activity in the liver of L-iNOS-Tg mice, whereas phosphoenolpyruvate carboxykinase, glucose-6-phosphatase, and proliferator-activated receptor γ coactivator-1α expression were not altered. These results clearly indicate that selective expression of iNOS in liver causes hepatic insulin resistance along with deranged insulin signaling, leading to hyperglycemia and hyperinsulinemia. Our data highlight a critical role for iNOS in the development of hepatic insulin resistance and hyperglycemia.


Asunto(s)
Hiperglucemia/metabolismo , Hígado/enzimología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Albúminas/genética , Animales , Femenino , Prueba de Tolerancia a la Glucosa , Hepatocitos/metabolismo , Resistencia a la Insulina , Masculino , Ratones , Ratones Transgénicos , Fenotipo , Fosforilación , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Triglicéridos/metabolismo
17.
J Biol Chem ; 286(33): 29388-29396, 2011 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-21700708

RESUMEN

Insulin receptor substrate-2 (IRS-2) plays a critical role in the survival and function of pancreatic ß-cells. Gene disruption of IRS-2 results in failure of the ß-cell compensatory mechanism and diabetes. Nonetheless, the regulation of IRS-2 protein expression in ß-cells remains largely unknown. Inducible nitric-oxide synthase (iNOS), a major mediator of inflammation, has been implicated in ß-cell damage in type 1 and type 2 diabetes. The effects of iNOS on IRS-2 expression have not yet been investigated in ß-cells. Here, we show that iNOS and NO donor decreased IRS-2 protein expression in INS-1/832 insulinoma cells and mouse islets, whereas IRS-2 mRNA levels were not altered. Interleukin-1ß (IL-1ß), alone or in combination with interferon-γ (IFN-γ), reduced IRS-2 protein expression in an iNOS-dependent manner without altering IRS-2 mRNA levels. Proteasome inhibitors, MG132 and lactacystin, blocked the NO donor-induced reduction in IRS-2 protein expression. Treatment with NO donor led to activation of glycogen synthase kinase-3ß (GSK-3ß) and c-Jun N-terminal kinase (JNK/SAPK) in ß-cells. Inhibition of GSK-3ß by pharmacological inhibitors or siRNA-mediated knockdown significantly prevented NO donor-induced reduction in IRS-2 expression in ß-cells. In contrast, a JNK inhibitor, SP600125, did not effectively block reduced IRS-2 expression in NO donor-treated ß-cells. These data indicate that iNOS-derived NO reduces IRS-2 expression by promoting protein degradation, at least in part, through a GSK-3ß-dependent mechanism. Our findings suggest that iNOS-mediated decreased IRS-2 expression may contribute to the progression and/or exacerbation of ß-cell failure in diabetes.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Células Secretoras de Insulina/metabolismo , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacología , Animales , Antracenos/farmacología , Línea Celular Tumoral , Inhibidores de Cisteína Proteinasa/farmacología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Humanos , Proteínas Sustrato del Receptor de Insulina/genética , Células Secretoras de Insulina/citología , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Leupeptinas/farmacología , Ratones , Óxido Nítrico Sintasa de Tipo II/genética , Complejo de la Endopetidasa Proteasomal/genética , Inhibidores de Proteasoma , Ratas
18.
Biochem Biophys Res Commun ; 391(3): 1459-64, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20034462

RESUMEN

Endotoxemia plays an important role in the pathogenesis of sepsis and is accompanied by dysregulated apoptosis of immune and non-immune cells. Treatment with statins reduces mortality in rodent models of sepsis and endotoxemia. Inhibition of protein isoprenylation, including farnesylation, has been proposed as a mechanism to mediate the lipid-lowering-independent effects of statins. Nonetheless, the effects of the inhibition of isoprenylation have not yet been studied. To investigate the role of farnesylation, we evaluated the effects of farnesyltransferase inhibitor and statin on survival following lipopolysaccharide (LPS) challenge in mice. Both simvastatin (2mg/kg BW) and FTI-277 (20mg/kg BW) treatment improved survival by twofold after LPS injection, as compared with vehicle alone (p<0.01). LPS-induced cleavage (activation) of caspase-3, an indicator of apoptotic change, and increased protein expression of proapoptotic molecules, Bax and Bim, and activation of c-Jun NH(2)-terminal kinase (JNK/SAPK) in the liver and spleen were attenuated by both simvastatin and FTI-277. These results demonstrate that farnesyltransferase inhibitor as well as statin significantly reduced LPS-induced mortality in mice. Our findings also suggest that inhibition of protein farnesylation may contribute to the lipid-lowering-independent protective effects of statins in endotoxemia, and that protein farnesylation may play a role in LPS-induced stress response, including JNK/SAPK activation, and apoptotic change. Our data argue that farnesyltransferase may be a potential molecular target for treating patients with endotoxemia.


Asunto(s)
Endotoxemia/tratamiento farmacológico , Inhibidores Enzimáticos/uso terapéutico , Farnesiltransferasa/antagonistas & inhibidores , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Metionina/análogos & derivados , Simvastatina/uso terapéutico , Animales , Apoptosis , Modelos Animales de Enfermedad , Endotoxemia/enzimología , Endotoxemia/patología , Lipopolisacáridos , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/patología , MAP Quinasa Quinasa 4/metabolismo , Masculino , Metionina/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Bazo/efectos de los fármacos , Bazo/enzimología , Bazo/patología
20.
Atherosclerosis ; 196(1): 68-75, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17362969

RESUMEN

Both experimental and epidemiological studies suggest that leptin is one of the molecules responsible for accelerated atherosclerosis in obese humans. To confirm the notion, we studied whether leptin accelerates atherosclerosis in apoE(-/-) mice. Leptin deficient hyperlipidemic mice (ob/ob;apoE(-/-) mice) developed significantly less atherosclerosis than apoE(-/-) mice, when fed an atherogenic diet for 16 weeks from 8 weeks of age. Histological analysis revealed that most of the atherosclerotic lesions in ob/ob;apoE(-/-) mice remained as fatty streaks, while those in apoE(-/-) mice were mainly fibrous plaques. The decrease in atherosclerosis was not due to changes in the serum levels of cholesterol, TNF-alpha, or adiponectin. Exogenous leptin significantly increased atherosclerotic areas in apoE(-/-) mice, even though it decreased food intake and body weight. Our findings support the notion that leptin accelerates atherosclerosis.


Asunto(s)
Apolipoproteínas E/genética , Aterosclerosis/fisiopatología , Leptina/deficiencia , Leptina/fisiología , Obesidad/fisiopatología , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Dieta Aterogénica , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...