Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Metab ; 6(1): 94-112, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38216738

RESUMEN

Adipose tissue lipolysis is mediated by cAMP-protein kinase A (PKA)-dependent intracellular signalling. Here, we show that PKA targets p21-activated kinase 4 (PAK4), leading to its protein degradation. Adipose tissue-specific overexpression of PAK4 in mice attenuates lipolysis and exacerbates diet-induced obesity. Conversely, adipose tissue-specific knockout of Pak4 or the administration of a PAK4 inhibitor in mice ameliorates diet-induced obesity and insulin resistance while enhancing lipolysis. Pak4 knockout also increases energy expenditure and adipose tissue browning activity. Mechanistically, PAK4 directly phosphorylates fatty acid-binding protein 4 (FABP4) at T126 and hormone-sensitive lipase (HSL) at S565, impairing their interaction and thereby inhibiting lipolysis. Levels of PAK4 and the phosphorylation of FABP4-T126 and HSL-S565 are enhanced in the visceral fat of individuals with obesity compared to their lean counterparts. In summary, we have uncovered an important role for FABP4 phosphorylation in regulating adipose tissue lipolysis, and PAK4 inhibition may offer a therapeutic strategy for the treatment of obesity.


Asunto(s)
Lipólisis , Esterol Esterasa , Animales , Ratones , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Lipólisis/fisiología , Obesidad/metabolismo , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo , Esterol Esterasa/genética , Esterol Esterasa/metabolismo
3.
Nutrients ; 15(17)2023 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-37686772

RESUMEN

Chronic liver injury due to various hepatotoxic stimuli commonly leads to fibrosis, which is a crucial factor contributing to liver disease-related mortality. Despite the potential benefits of Suaeda glauca (S. glauca) as a natural product, its biological and therapeutic effects are barely known. This study investigated the effects of S. glauca extract (SGE), obtained from a smart farming system utilizing LED lamps, on the activation of hepatic stellate cells (HSCs) and the development of liver fibrosis. C57BL/6 mice received oral administration of either vehicle or SGE (30 or 100 mg/kg) during CCl4 treatment for 6 weeks. The supplementation of SGE significantly reduced liver fibrosis induced by CCl4 in mice as evidenced by histological changes and a decrease in collagen accumulation. SGE treatment also led to a reduction in markers of HSC activation and inflammation as well as an improvement in blood biochemical parameters. Furthermore, SGE administration diminished fibrotic responses following acute liver injury. Mechanistically, SGE treatment prevented HSC activation and inhibited the phosphorylation and nuclear translocation of Smad2/3, which are induced by transforming growth factor (TGF)-ß1 in HSCs. Our findings indicate that SGE exhibits anti-fibrotic effects by inhibiting TGFß1-Smad2/3 signaling in HSCs.


Asunto(s)
Chenopodiaceae , Células Estrelladas Hepáticas , Animales , Ratones , Ratones Endogámicos C57BL , Cirrosis Hepática/tratamiento farmacológico
4.
Nat Commun ; 14(1): 4987, 2023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37591884

RESUMEN

PPARα corepressor NCoR1 is a key regulator of fatty acid ß-oxidation and ketogenesis. However, its regulatory mechanism is largely unknown. Here, we report that oncoprotein p21-activated kinase 4 (PAK4) is an NCoR1 kinase. Specifically, PAK4 phosphorylates NCoR1 at T1619/T2124, resulting in an increase in its nuclear localization and interaction with PPARα, thereby repressing the transcriptional activity of PPARα. We observe impaired ketogenesis and increases in PAK4 protein and NCoR1 phosphorylation levels in liver tissues of high fat diet-fed mice, NAFLD patients, and hepatocellular carcinoma patients. Forced overexpression of PAK4 in mice represses ketogenesis and thereby increases hepatic fat accumulation, whereas genetic ablation or pharmacological inhibition of PAK4 exhibites an opposite phenotype. Interestingly, PAK4 protein levels are significantly suppressed by fasting, largely through either cAMP/PKA- or Sirt1-mediated ubiquitination and proteasome degradation. In this way, our findings provide evidence for a PAK4-NCoR1/PPARα signaling pathway that regulates fatty acid ß-oxidation and ketogenesis.


Asunto(s)
Ácidos Grasos , PPAR alfa , Quinasas p21 Activadas , Animales , Ratones , Proteínas Co-Represoras , Ácidos Grasos/metabolismo , Quinasas p21 Activadas/genética , PPAR alfa/genética , Co-Represor 1 de Receptor Nuclear/genética , Humanos , Fosforilación , Transducción de Señal
5.
Nat Commun ; 13(1): 6080, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36241614

RESUMEN

Disturbed lipid metabolism precedes alcoholic liver injury. Whether and how AhR alters degradation of lipids, particularly phospho-/sphingo-lipids during alcohol exposure, was not explored. Here, we show that alcohol consumption in mice results in induction and activation of aryl hydrocarbon receptor (AhR) in the liver, and changes the hepatic phospho-/sphingo-lipids content. The levels of kynurenine, an endogenous AhR ligand, are elevated with increased hepatic tryptophan metabolic enzymes in alcohol-fed mice. Either alcohol or kynurenine treatment promotes AhR activation with autophagy dysregulation via AMPK. Protein Phosphatase 2 Regulatory Subunit-Bdelta (Ppp2r2d) is identified as a transcriptional target of AhR. Consequently, PPP2R2D-dependent AMPKα dephosphorylation causes autophagy inhibition and mitochondrial dysfunction. Hepatocyte-specific AhR ablation attenuates steatosis, which is associated with recovery of phospho-/sphingo-lipids content. Changes of AhR targets are corroborated using patient specimens. Overall, AhR induction by alcohol inhibits autophagy in hepatocytes through AMPKα, which is mediated by Ppp2r2d gene transactivation, revealing an AhR-dependent metabolism of phospho-/sphingo-lipids.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Receptores de Hidrocarburo de Aril , Animales , Ratones , Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia , Etanol/metabolismo , Etanol/toxicidad , Quinurenina/metabolismo , Ligandos , Metabolismo de los Lípidos , Hígado/metabolismo , Fosfolípidos/metabolismo , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Triptófano/metabolismo
6.
Nat Commun ; 13(1): 1808, 2022 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-35379817

RESUMEN

Expanding the exercise capacity of skeletal muscle is an emerging strategy to combat obesity-related metabolic diseases and this can be achieved by shifting skeletal muscle fibers toward slow-twitch oxidative type. Here, we report that Sirt6, an anti-aging histone deacetylase, is critical in regulating myofiber configuration toward oxidative type and that Sirt6 activator can be an exercise mimetic. Genetic inactivation of Sirt6 in skeletal muscle reduced while its transgenic overexpression increased mitochondrial oxidative capacity and exercise performance in mice. Mechanistically, we show that Sirt6 downregulated Sox6, a key repressor of slow fiber specific gene, by increasing the transcription of CREB. Sirt6 expression is elevated in chronically exercised humans, and mice treated with an activator of Sirt6 showed an increase in exercise endurance as compared to exercise-trained controls. Thus, the current study identifies Sirt6 as a molecular target for reprogramming myofiber composition toward the oxidative type and for improving muscle performance.


Asunto(s)
Músculo Esquelético , Sirtuinas , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Ratones , Mitocondrias/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Factores de Transcripción SOXD , Sirtuinas/genética , Sirtuinas/metabolismo
7.
Free Radic Biol Med ; 184: 42-52, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35390453

RESUMEN

Alcoholic liver disease is the major cause of chronic liver diseases. Excessive alcohol intake results in endoplasmic reticulum (ER) stress. ERdj5, a member of DNAJ family, is an ER-resident chaperone protein, whose role in alcoholic liver disease remains to be investigated. In this study, we aim to address the effect of ERdj5 on alcoholic liver disease and the underlying mechanism. Hepatic Dnajc10 (ERdj5) mRNA expression was elevated in both human and mouse alcoholic hepatitis. In mice subjected to chronic and binge ethanol feeding, ERdj5 levels were also markedly increased. Hepatic Dnajc10 correlated with Xbp1s mRNA. Tunicamycin, an ER stress inducer, increased ERdj5 levels. Dnajc10 knockout mice exhibited exacerbated alcohol-induced liver injury and hepatic steatosis. However, the macrophage numbers and chemokine levels were similar to those in wild-type mice. Depletion of Dnajc10 promoted oxidative stress. Ethanol feeding increased hepatic H2O2 levels, and these were further increased in Dnajc10 knockout mice. Additionally, Dnajc10-deficient hepatocytes produced large amounts of reactive oxygen species. Notably, Nrf2, a central regulator of oxidative stress, was decreased by depletion of Dnajc10 in the nuclear fraction of ethanol-treated mouse liver. Consistently, liver tissues from ethanol-fed Dnajc10 knockout mice had reduced expression of downstream antioxidant genes. Furthermore, hepatic glutathione content in the liver of knockout mice declined compared to wild-type mice. In conclusion, our results demonstrate that ethanol-induced ERdj5 may regulate the Nrf2 pathway and glutathione contents, and have protective effects on liver damage and alcohol-mediated oxidative stress in mice. These suggest that ERdj5 has the potential to protect against alcoholic liver disease.


Asunto(s)
Proteínas del Choque Térmico HSP40 , Hepatopatías Alcohólicas , Chaperonas Moleculares , Factor 2 Relacionado con NF-E2 , Animales , Ratones , Etanol/toxicidad , Glutatión/metabolismo , Proteínas del Choque Térmico HSP40/genética , Proteínas del Choque Térmico HSP40/metabolismo , Peróxido de Hidrógeno/metabolismo , Hígado/metabolismo , Hepatopatías Alcohólicas/genética , Ratones Noqueados , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , ARN Mensajero/metabolismo
8.
Hepatology ; 76(2): 345-356, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35108418

RESUMEN

BACKGROUND AND AIMS: p21-activated kinase 4 (PAK4), an oncogenic protein, has emerged as a promising target for anticancer drug development. Its role in oxidative stress conditions, however, remains elusive. We investigated the effects of PAK4 signaling on hepatic ischemia/reperfusion (I/R) injury. APPROACH AND RESULTS: Hepatocyte- and myeloid-specific Pak4 knockout (KO) mice and their littermate controls were subjected to a partial hepatic I/R (HIR) injury. We manipulated the catalytic activity of PAK4, either through genetic engineering (gene knockout, overexpression of wild-type [WT] or dominant-negative kinase) or pharmacological inhibitor, coupled with a readout of nuclear factor erythroid 2-related factor 2 (Nrf2) activity, to test the potential function of PAK4 on HIR injury. PAK4 expression was markedly up-regulated in liver during HIR injury in mice and humans. Deletion of PAK4 in hepatocytes, but not in myeloid cells, ameliorated liver damages, as demonstrated in the decrease in hepatocellular necrosis and inflammatory responses. Conversely, the forced expression of WT PAK4 aggravated the pathological changes. PAK4 directly phosphorylated Nrf2 at T369, and it led to its nuclear export and proteasomal degradation, all of which impaired antioxidant responses in hepatocytes. Nrf2 silencing in liver abolished the protective effects of PAK4 deficiency. A PAK4 inhibitor protected mice from HIR injury. CONCLUSIONS: PAK4 phosphorylates Nrf2 and suppresses its transcriptional activity. Genetic or pharmacological suppression of PAK4 alleviates HIR injury. Thus, PAK4 inhibition may represent a promising intervention against I/R-induced liver injury.


Asunto(s)
Hepatopatías , Daño por Reperfusión , Quinasas p21 Activadas , Animales , Apoptosis , Humanos , Isquemia/metabolismo , Isquemia/patología , Hígado/patología , Hepatopatías/etiología , Hepatopatías/metabolismo , Hepatopatías/prevención & control , Ratones , Ratones Noqueados , Factor 2 Relacionado con NF-E2/metabolismo , Fosforilación , Daño por Reperfusión/metabolismo , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo
9.
J Cachexia Sarcopenia Muscle ; 12(6): 1776-1788, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34431242

RESUMEN

BACKGROUND: Skeletal muscle regeneration is an adaptive response to injury that is crucial to the maintenance of muscle mass and function. A p21-activated kinase 4 (PAK4) serine/threonine kinase is critical to the regulation of cytoskeletal changes, cell proliferation, and growth. However, PAK4's role in myoblast differentiation and regenerative myogenesis remains to be determined. METHODS: We used a mouse model of myotoxin (notexin)-induced muscle regeneration. In vitro myogenesis was performed in the C2C12 myoblast cell line, primary myoblasts, and primary satellite cells. In vivo overexpression of PAK4 or kinase-inactive mutant PAK4S474A was conducted in skeletal muscle to examine PAK4's kinase-dependent effect on muscle regeneration. The regeneration process was evaluated by determining the number and size of multinucleated myofibres and expression patterns of myogenin and eMyHC. To explore whether PAK4 inhibition improves muscle regeneration, mice were injected intramuscularly with siRNA that targeted PAK4 or orally administered with a chemical inhibitor of PAK4. RESULTS: p21-activated kinase 4 was highly expressed during the myoblast stage, but expression gradually and substantially decreased as myoblasts differentiated into myotubes. PAK4 overexpression, but not kinase-inactive mutant PAK4S474A overexpression, significantly impeded myoblast fusion and MyHC-positive myotube formation in C2C12 cells, primary myoblasts, and satellite cells (P < 0.01). Conversely, PAK4 silencing led to an 8.7% and a 20.3% increase in the number of multinucleated larger myotubes in C2C12 cells and primary myoblasts. Further, in vivo overexpression of PAK4 by adenovirus injection to mice prior to and after myotoxin-induced injury led to a 52.6% decrease in the number of eMyHC-positive myofibres on Day 5 in tibialis anterior muscles as compared with those injected with control adenoviruses (P < 0.01), while Ad-PAK4S474A showed comparable muscle regeneration parameters. PAK4-induced repression of muscle regeneration coincided with an increase in phosphatase and tensin homologue (PTEN) expression and a decrease in phosphoinositide 3-kinase-Akt signalling. In contrast, PAK4 silencing reduced PTEN expression in mice. Consistent with these findings, prodrug of PAK4 inhibitor CZh-226 (30 mg/kg) orally administered to mice repressed PTEN expression and accelerated myotube formation. Subsequent mechanistic studies revealed that PAK4 directly phosphorylates PPARγ at S273 to increase its transcription activity, thereby up-regulating PTEN expression. Importantly, an analysis of the Genotype-Tissue Expression database showed a positive correlation between PAK4 and PTEN in human skeletal muscle tissues (P < 0.01). CONCLUSIONS: p1-activated kinase 4 is a new member of PPARγ kinase, and PAK4 inhibition may have a therapeutic role as an accelerant of muscle regeneration.


Asunto(s)
Receptores Activados del Proliferador del Peroxisoma , Quinasas p21 Activadas , Animales , Ratones , Músculo Esquelético , PPAR gamma , Fosfatidilinositol 3-Quinasas , Regeneración , Quinasas p21 Activadas/genética
10.
Exp Mol Med ; 53(7): 1159-1169, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34285335

RESUMEN

In this study, we hypothesized that deregulation in the maintenance of the pool of coenzyme A (CoA) may play a crucial role in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). Specific deletion of Acot12 (Acot12-/-), the major acyl-CoA thioesterase, induced the accumulation of acetyl-CoA and resulted in the stimulation of de novo lipogenesis (DNL) and cholesterol biosynthesis in the liver. KEGG pathway analysis suggested PPARα signaling as the most significantly enriched pathway in Acot12-/- livers. Surprisingly, the exposure of Acot12-/- hepatocytes to fenofibrate significantly increased the accumulation of acetyl-CoA and resulted in the stimulation of cholesterol biosynthesis and DNL. Interaction analysis, including proximity-dependent biotin identification (BioID) analysis, suggested that ACOT12 may directly interact with vacuolar protein sorting-associated protein 33A (VPS33A) and play a role in vesicle-mediated cholesterol trafficking and the process of lysosomal degradation of cholesterol in hepatocytes. In summary, in this study, we found that ACOT12 deficiency is responsible for the pathogenesis of NAFLD through the accumulation of acetyl-CoA and the stimulation of DNL and cholesterol via activation of PPARα and inhibition of cholesterol trafficking.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico/metabolismo , Tioléster Hidrolasas/metabolismo , Acetilcoenzima A/metabolismo , Animales , Colesterol/biosíntesis , Colesterol/genética , Dieta Alta en Grasa/efectos adversos , Femenino , Humanos , Lípidos/biosíntesis , Lípidos/genética , Lipogénesis/fisiología , Lipólisis/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Enfermedad del Hígado Graso no Alcohólico/genética , PPAR alfa/genética , PPAR alfa/metabolismo , Embarazo , Tioléster Hidrolasas/genética
11.
Biomolecules ; 11(2)2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33567666

RESUMEN

Excess and sustained endoplasmic reticulum (ER) stress, paired with a failure of initial adaptive responses, acts as a critical trigger of nonalcoholic fatty liver disease (NAFLD) progression. Unfortunately, there is no drug currently approved for treatment, and the molecular basis of pathogenesis by ER stress remains poorly understood. Classical ER stress pathway molecules have distinct but inter-connected functions and complicated effects at each phase of the disease. Identification of the specific molecular signal mediators of the ER stress-mediated pathogenesis is, therefore, a crucial step in the development of new treatments. These signaling nodes may be specific to the cell type and/or the phase of disease progression. In this review, we highlight the recent advancements in knowledge concerning signaling nodes associated with ER stress and NAFLD progression in various types of liver cells.


Asunto(s)
Estrés del Retículo Endoplásmico , Enfermedad del Hígado Graso no Alcohólico/patología , Transducción de Señal , Animales , Modelos Animales de Enfermedad , Humanos , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo
12.
Hepatology ; 73(4): 1307-1326, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32557804

RESUMEN

BACKGROUND AND AIMS: Fat accumulation results from increased fat absorption and/or defective fat metabolism. Currently, the lipid-sensing nuclear receptor that controls fat utilization in hepatocytes is elusive. Liver X receptor alpha (LXRα) promotes accumulation of lipids through the induction of several lipogenic genes. However, its effect on lipid degradation is open for study. Here, we investigated the inhibitory role of LXRα in autophagy/lipophagy in hepatocytes and the underlying basis. APPROACH AND RESULTS: In LXRα knockout mice fed a high-fat diet, or cell models, LXRα activation suppressed the function of mitochondria by inhibiting autophagy/lipophagy and induced hepatic steatosis. Gene sets associated with "autophagy" were enriched in hepatic transcriptome data. Autophagy flux was markedly augmented in the LXRα knockout mouse liver and primary hepatocytes. Mechanistically, LXRα suppressed autophagy-related 4B cysteine peptidase (ATG4B) and Rab-8B, responsible for autophagosome and -lysosome formation, by inducing let-7a and microRNA (miR)-34a. Chromatin immunoprecipitation assay enabled us to find LXRα as a transcription factor of let-7a and miR-34a. Moreover, 3' untranslated region luciferase assay substantiated the direct inhibitory effects of let-7a and miR-34a on ATG4B and Rab-8B. Consistently, either LXRα activation or the let-7a/miR-34a transfection lowered mitochondrial oxygen consumption rate and mitochondrial transmembrane potential and increased fat levels. In obese animals or nonalcoholic fatty liver disease (NAFLD) patients, let-7a and miR-34a levels were elevated with simultaneous decreases in ATG4B and Rab-8B levels. CONCLUSIONS: LXRα inhibits autophagy in hepatocytes through down-regulating ATG4B and Rab-8B by transcriptionally activating microRNA let-7a-2 and microRNA 34a genes and suppresses mitochondrial biogenesis and fuel consumption. This highlights a function of LXRα that culminates in the progression of liver steatosis and steatohepatitis, and the identified targets may be applied for a therapeutic strategy in the treatment of NAFLD.


Asunto(s)
Proteínas Relacionadas con la Autofagia/metabolismo , Autofagia/fisiología , Cisteína Endopeptidasas/metabolismo , Hepatocitos/metabolismo , Receptores X del Hígado/metabolismo , Mitocondrias/fisiología , Proteínas de Unión al GTP rab/metabolismo , Activación Metabólica , Animales , Autofagia/genética , Proteínas Relacionadas con la Autofagia/genética , Cisteína Endopeptidasas/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación hacia Abajo , Hígado Graso/etiología , Hígado Graso/genética , Hígado Graso/metabolismo , Hígado Graso/fisiopatología , Células Hep G2/metabolismo , Células Hep G2/fisiología , Hepatocitos/fisiología , Humanos , Metabolismo de los Lípidos/genética , Metabolismo de los Lípidos/fisiología , Hígado/metabolismo , Hígado/fisiología , Hígado/fisiopatología , Receptores X del Hígado/genética , Receptores X del Hígado/fisiología , Ratones , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , MicroARNs/fisiología , Mitocondrias/metabolismo , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Biogénesis de Organelos , Consumo de Oxígeno/genética , Consumo de Oxígeno/fisiología , Transcriptoma , Proteínas de Unión al GTP rab/genética
13.
Theranostics ; 10(25): 11416-11427, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33052223

RESUMEN

Rationale: Statin, the most widely used medication in lowering cholesterol, is also associated with increased risk of type 2 diabetes, but its molecular basis remains unclear. Methods: Mice were injected intraperitoneally with statins alone or in combination with sirtuin (Sirt) 6 activator, and blood glucose levels were measured. Liver tissues from patients with statin use were analyzed for the expression of Sirt6. Results: Statin treatment up-regulated the hepatic expression of phosphoenolpyruvate carboxykinase and glucose-6-phosphatase, which was prevented by Sirt6 overexpression. Mechanistically, statin directly repressed Sirt6 expression by induction of microRNA (miR)-495, a novel inhibitor of Sirt6. Pathway analysis for predicted target genes of miR-495 recognized forkhead box protein (Fox)O1 as a key downstream signaling of Sirt6. Statin treatment increased the acetylation and protein stability of FoxO1, which was suppressed by Sirt6 overexpression. Inhibiting miR-495 recovered Sirt6 levels, blocking the ability of statin to increase FoxO1 mediated gluconeogenesis, and thus confirming the role of the miR-495/Sirt6/FoxO1 pathway in controlling gluconeogenesis. Moreover, the Sirt6 activator MDL801 prevented gluconeogenesis and hyperglycemia induced by statin in mice. Equally noteworthy was that human liver tissues obtained from statin users showed a significant decrease in Sirt6 protein levels compared to those of non-users. Conclusion: Statin induces miR-495 to suppress Sirt6 expression, which leads to enhancement of FoxO1-mediated hepatic gluconeogenesis. Thus, Sirt6 activation may offer a promising strategy for preventing statin-induced hyperglycemia.


Asunto(s)
Diabetes Mellitus Tipo 2/inducido químicamente , Gluconeogénesis/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , MicroARNs/agonistas , Sirtuinas/antagonistas & inhibidores , Adulto , Animales , Células Cultivadas , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/prevención & control , Femenino , Proteína Forkhead Box O1/metabolismo , Gluconeogénesis/genética , Glucosa-6-Fosfatasa/metabolismo , Hepatocitos , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Inyecciones Intraperitoneales , Hígado/citología , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Ratones , MicroARNs/metabolismo , Persona de Mediana Edad , Cultivo Primario de Células , Sirtuinas/análisis , Sirtuinas/genética , Sirtuinas/metabolismo , Adulto Joven
14.
Korean J Physiol Pharmacol ; 23(6): 493-499, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31680771

RESUMEN

Macrophage-associated inflammation is crucial for the pathogenesis of diverse diseases including metabolic disorders. Rhodanthpyrone (Rho) is an active component of Gentiana rhodantha, which has been used in traditional Chinese medicine to treat inflammation. Although synthesis procedures of RhoA and RhoB were reported, the biological effects of the specific compounds have never been explored. In this study, the anti-inflammatory activity and mechanisms of action of RhoA and RhoB were studied in lipopolysaccharide (LPS)-stimulated macrophages. Pretreatment with RhoA and RhoB decreased inducible nitric oxide synthase and cyclooxygenase-2 expressions in RAW 264.7 cells and in thioglycollate-elicited mouse peritoneal macrophages. In addition, it downregulated transcript levels of several inflammatory genes in LPS-stimulated RAW 264.7 cells, including inflammatory cytokines/chemokines (Tnfa, Il6, and Ccl2) and inflammatory mediators (Nos2 and Ptgs2). Macrophage chemotaxis was also inhibited by treatment with the compounds. Mechanistic studies revealed that RhoA and RhoB suppressed the nuclear factor (NF)-κB pathway, but not the canonical mitogen activated protein kinase pathway, in LPS-stimulated condition. Moreover, the inhibitory effect of RhoA and RhoB on inflammatory gene expressions was attenuated by treatment with an NF-κB inhibitor. Our findings suggest that RhoA and RhoB play an anti-inflammatory role at least in part by suppressing the NF-κB pathway during macrophage-mediated inflammation.

15.
Sci Transl Med ; 11(513)2019 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-31597753

RESUMEN

The impact of liver disease on whole-body glucose homeostasis is largely attributed to dysregulated release of secretory proteins in response to metabolic stress. The molecular cues linking liver to whole-body glucose metabolism remain elusive. We found that expression of G protein α-13 (Gα13) was decreased in the liver of mice and humans with diabetes. Liver-specific deletion of the Gna13 gene in mice resulted in systemic glucose intolerance. Comparative secretome analysis identified inter-α-trypsin inhibitor heavy chain 1 (ITIH1) as a protein secreted by liver that was responsible for systemic insulin resistance in Gna13-deficient mice. Liver expression of ITIH1 positively correlated with surrogate markers for diabetes in patients with impaired glucose tolerance or overt diabetes. Mechanistically, a decrease in hepatic Gα13 caused ITIH1 oversecretion by liver through induction of O-GlcNAc transferase expression, facilitating ITIH1 deposition on the hyaluronan surrounding mouse adipose tissue and skeletal muscle. Neutralization of secreted ITIH1 ameliorated glucose intolerance in obese mice. Our findings demonstrate systemic insulin resistance in mice resulting from liver-secreted ITIH1 downstream of Gα13 and its reversal by ITIH1 neutralization.


Asunto(s)
alfa-Globulinas/metabolismo , Resistencia a la Insulina/fisiología , Hígado/metabolismo , alfa-Globulinas/genética , Animales , Anticuerpos Neutralizantes/metabolismo , Línea Celular , Células Cultivadas , Cromatografía Liquida , Intolerancia a la Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Células HEK293 , Hepatocitos/metabolismo , Humanos , Resistencia a la Insulina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Espectrometría de Masas en Tándem
16.
Toxicol Res ; 35(4): 319-330, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31636843

RESUMEN

Adverse drug reactions (ADRs) constitute key factors in determining successful medication therapy in clinical situations. Integrative analysis of electronic medical record (EMR) data and use of proper analytical tools are requisite to conduct retrospective surveillance of clinical decisions on medications. Thus, we suggest that electronic medical recording and human genetic databases are considered together in future directions of pharmacovigilance. We analyzed EMR-based ADR studies indexed on PubMed during the period from 2005 to 2017 and retrospectively acquired 1161 (29.6%) articles describing drug-induced adverse reactions (e.g., liver, kidney, nervous system, immune system, and inflammatory responses). Of them, only 102 (8.79%) articles contained useful information to detect or predict ADRs in the context of clinical medication alerts. Since insufficiency of EMR datasets and their improper analyses may provide false warnings on clinical decision, efforts should be made to overcome possible problems on data-mining, analysis, statistics, and standardization. Thus, we address the characteristics and limitations on retrospective EMR database studies in hospital settings. Since gene expression and genetic variations among individuals impact ADRs, pharmacokinetics, and pharmacodynamics, appropriate paths for pharmacovigilance may be optimized using suitable databases available in public domain (e.g., genome-wide association studies (GWAS), non-coding RNAs, microRNAs, proteomics, and genetic variations), novel targets, and biomarkers. These efforts with new validated biomarker analyses would be of help to repurpose clinical and translational research infrastructure and ultimately future personalized therapy considering ADRs.

17.
Int J Mol Sci ; 20(18)2019 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-31491992

RESUMEN

Hepatocyte death is critical for the pathogenesis of liver disease progression, which is closely associated with endoplasmic reticulum (ER) stress responses. However, the molecular basis for ER stress-mediated hepatocyte injury remains largely unknown. This study investigated the effect of ER stress on dual-specificity phosphatase 5 (DUSP5) expression and its role in hepatocyte death. Analysis of Gene Expression Omnibus (GEO) database showed that hepatic DUSP5 levels increased in the patients with liver fibrosis, which was verified in mouse models of liver diseases with ER stress. DUSP5 expression was elevated in both fibrotic and acutely injured liver of mice treated with liver toxicants. Treatment of ER stress inducers enhanced DUSP5 expression in hepatocytes, which was validated in vivo condition. The induction of DUSP5 by ER stress was blocked by either treatment with a chemical inhibitor of the protein kinase RNA-like endoplasmic reticulum kinase (PERK) pathway, or knockdown of C/EBP homologous protein (CHOP), whereas it was not affected by the silencing of IRE1 or ATF6. In addition, DUSP5 overexpression decreased extracellular-signal-regulated kinase (ERK) phosphorylation, but increased cleaved caspase-3 levels. Moreover, the reduction of cell viability under ER stress condition was attenuated by DUSP5 knockdown. In conclusion, DUSP5 expression is elevated in hepatocytes by ER stress through the PERK-CHOP pathway, contributing to hepatocyte death possibly through ERK inhibition.


Asunto(s)
Fosfatasas de Especificidad Dual/genética , Estrés del Retículo Endoplásmico , Hepatocitos/metabolismo , Transducción de Señal , Factor de Transcripción CHOP/metabolismo , eIF-2 Quinasa/metabolismo , Animales , Apoptosis/genética , Muerte Celular/genética , Expresión Génica , Hepatocitos/patología , Humanos , Hepatopatías/etiología , Hepatopatías/metabolismo , Ratones
18.
Toxicol Appl Pharmacol ; 379: 114665, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31323261

RESUMEN

Ferroptosis is the non-apoptotic form of cell death caused by small molecules or conditions that inhibit glutathione biosynthesis or resulting in iron-dependent accumulation of lipid peroxidation by lipid reactive oxygen species (ROS). Sestrin2 (Sesn2), a conserved antioxidant protein, is responsive to various stresses including genotoxic, metabolic, and oxidative stresses and acts to restore homeostatic balance. Sesn2 expression was reported to be regulated via stress-responsive transcription factors including p53, Nrf2, and HIF-1α. However, the role of Sesn2 in regulating ferroptosis is not known. In the current study, we investigated whether ferroptosis inducing compounds including erastin, sorafenib, and buthionine sulfoximine affect Sesn2 expression and the role of Sesn2 in cytoprotection against ferroptosis-mediated cell death. Our data demonstrate that ferroptosis inducers significantly increased Sesn2 in hepatocytes in a dose- and time-dependent manner. Treatment with erastin upregulated Sesn2 mRNA levels and luciferase reporter gene activity, and erastin-mediated Sesn2 induction was transcriptionally regulated by NF-E2-related factor 2 (Nrf2). Furthermore, deletion of the antioxidant response element (ARE) in the Sesn2 promoter or Nrf2 knockout or knockdown abolished erastin-induced Sesn2 expression. In cells expressing Sesn2, erastin-induced cell death, ROS formation, and glutathione depletion were almost completely inhibited compared to that in control cells. Treatment with phenylhydrazine in mice, well-reported iron overload liver injury model, increased ALT and AST levels and altered histological features, which were almost completely inhibited by adenoviral Sesn2 infection. Collectively, our results suggest that ferroptosis-mediated Sesn2 induction is dependent on Nrf2 and plays a protective role against iron overload and ferroptosis-induced liver injury.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Ferroptosis , Sobrecarga de Hierro/complicaciones , Proteínas Nucleares/fisiología , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Glutatión/metabolismo , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Sobrecarga de Hierro/metabolismo , Peroxidación de Lípido , Hígado/metabolismo , Hígado/patología , Masculino , Ratones Endogámicos ICR , Ratones Noqueados , Proteínas Nucleares/metabolismo , Reacción en Cadena de la Polimerasa , Especies Reactivas de Oxígeno/metabolismo
19.
J Clin Invest ; 128(12): 5587-5602, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30300140

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) arises from mitochondrial dysfunction under sustained imbalance between energy intake and expenditure, but the underlying mechanisms controlling mitochondrial respiration have not been entirely understood. Heterotrimeric G proteins converge with activated GPCRs to modulate cell-signaling pathways to maintain metabolic homeostasis. Here, we investigated the regulatory role of G protein α12 (Gα12) on hepatic lipid metabolism and whole-body energy expenditure in mice. Fasting increased Gα12 levels in mouse liver. Gα12 ablation markedly augmented fasting-induced hepatic fat accumulation. cDNA microarray analysis from Gna12-KO liver revealed that the Gα12-signaling pathway regulated sirtuin 1 (SIRT1) and PPARα, which are responsible for mitochondrial respiration. Defective induction of SIRT1 upon fasting was observed in the liver of Gna12-KO mice, which was reversed by lentivirus-mediated Gα12 overexpression in hepatocytes. Mechanistically, Gα12 stabilized SIRT1 protein through transcriptional induction of ubiquitin-specific peptidase 22 (USP22) via HIF-1α increase. Gα12 levels were markedly diminished in liver biopsies from NAFLD patients. Consistently, Gna12-KO mice fed a high-fat diet displayed greater susceptibility to diet-induced liver steatosis and obesity due to decrease in energy expenditure. Our results demonstrate that Gα12 regulates SIRT1-dependent mitochondrial respiration through HIF-1α-dependent USP22 induction, identifying Gα12 as an upstream molecule that contributes to the regulation of mitochondrial energy expenditure.


Asunto(s)
Endopeptidasas/metabolismo , Hígado Graso/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Mitocondrias Hepáticas/metabolismo , Obesidad/metabolismo , Consumo de Oxígeno , Transducción de Señal , Sirtuina 1/metabolismo , Animales , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/farmacología , Endopeptidasas/genética , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Hígado Graso/inducido químicamente , Hígado Graso/genética , Hígado Graso/patología , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Ratones , Ratones Noqueados , Mitocondrias Hepáticas/genética , Mitocondrias Hepáticas/patología , Obesidad/inducido químicamente , Obesidad/genética , Obesidad/patología , Sirtuina 1/genética , Ubiquitina Tiolesterasa
20.
Cell Rep ; 24(11): 2985-2999, 2018 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-30208322

RESUMEN

Endoplasmic reticulum (ER) stress is associated with liver injury and fibrosis, and yet the hepatic factors that regulate ER stress-mediated inflammasome activation remain unknown. Here, we report that farnesoid X receptor (FXR) activation inhibits ER stress-induced NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome in hepatocytes. In patients with hepatitis B virus (HBV)-associated hepatic failure or non-alcoholic fatty liver disease, and in mice with liver injury, FXR levels in the liver inversely correlated with the extent of NLRP3 inflammasome activation. Fxr deficiency in mice augmented the ability of ER stress to induce NLRP3 and thioredoxin-interacting protein (TXNIP), whereas FXR ligand activation prevented it, ameliorating liver injury. FXR attenuates CCAAT-enhancer-binding protein homologous protein (CHOP)-dependent NLRP3 overexpression by inhibiting ER stress-mediated protein kinase RNA-like endoplasmic reticulum kinase (PERK) activation. Our findings implicate miR-186 and its target, non-catalytic region of tyrosine kinase adaptor protein 1 (NCK1), in mediating the inhibition of ER stress by FXR. This study provides the insights on how FXR regulation of ER stress ameliorates hepatocyte death and liver injury and on the molecular basis of NLRP3 inflammasome activation.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Hepatocitos/metabolismo , Inflamasomas/metabolismo , Hígado/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Estrés del Retículo Endoplásmico/genética , Inflamasomas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tiorredoxinas/genética , Tiorredoxinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...