Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Oxid Med Cell Longev ; 2022: 9142798, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35198097

RESUMEN

Proinflammatory cytokines play a causal role in the development of hyperinsulinemia and T2MD. FOXO1, a transcription factor which is known to enhance proinflammation, was recently shown to be involved in obesity-induced ß cell dysfunction. However, molecular mechanisms for the association remained elusive. In this study, we first found that both leptin (10 nM) and TNF-α (20 ng/ml) significantly inhibited glucose-stimulated insulin secretion (GSIS) of INS-1E cells. When in combination, the GSIS function of INS-1E cells was significantly increased compared with that of the leptin alone treatment, indicating that TNF-α attenuated the inhibiting effect of leptin on GSIS of INS-1E cells. Similarly, we found that TNF-α has the same inhibitory effect on leptin in regulating insulin synthesis and secretion, and the survival and apoptosis of insulin cells. Further studies showed that TNF-α blocks leptin pathway by reducing the expression of leptin receptor (LepRb, also called OBRb) and inhibiting the activation of STAT3, a key molecule involved in the leptin signaling pathway in INS-1E cells. Besides, the downregulated expression of phosphorylated FOXO1 was found to be involved in the possible mechanism of TNF-α. Overexpression of constitutively active FOXO1 markedly aggravated the LepRb reduction by TNF-α treatment of INS-1E cells, and the endogenous FOXO1 knockdown abolished the effect of TNF-α on INS-1E cells. Furthermore, we have proved that FOXO1 could directly bind to the promoter of LepRb as a negative transcription regulator. Taken together, the results of this study reveal that TNF-α-induced LepRb downregulated in pancreatic ß cells and demonstrate that transcriptional reduction of FOXO1 might be the primary mechanism underlying TNF-α promoting INS-1E leptin resistance and ß cell dysfunction. Conclusions. Our current studies based on INS-1E cells in vitro indicate that the inflammatory factor TNF-α plays an important role in the development of INS-1E leptin resistance and glucose metabolism disorders, probably through FOXO1-induced transcription reduction of LepRb promoter in pancreatic ß cells, and FOXO1 may be a novel target for treating ß cell dysfunction in obesity-induced hyperinsulinemia and T2DM.


Asunto(s)
Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Leptina/farmacología , Proteínas del Tejido Nervioso/metabolismo , Receptores de Leptina/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Supervivencia Celular , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Insulina/genética , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Janus Quinasa 2/metabolismo , Proteínas del Tejido Nervioso/genética , Ratas , Receptores de Leptina/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos
2.
Nat Commun ; 9(1): 2574, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29968710

RESUMEN

Metabolic reprogramming greatly contributes to the regulation of macrophage activation. However, the mechanism of lipid accumulation and the corresponding function in tumor-associated macrophages (TAMs) remain unclear. With primary investigation in colon cancer and confirmation in other cancer models, here we determine that deficiency of monoacylglycerol lipase (MGLL) results in lipid overload in TAMs. Functionally, macrophage MGLL inhibits CB2 cannabinoid receptor-dependent tumor progression in inoculated and genetic cancer models. Mechanistically, MGLL deficiency promotes CB2/TLR4-dependent macrophage activation, which further suppresses the function of tumor-associated CD8+ T cells. Treatment with CB2 antagonists delays tumor progression in inoculated and genetic cancer models. Finally, we verify that expression of macrophage MGLL is decreased in cancer tissues and positively correlated with the survival of cancer patients. Taken together, our findings identify MGLL as a switch for CB2/TLR4-dependent macrophage activation and provide potential targets for cancer therapy.


Asunto(s)
Macrófagos/inmunología , Monoacilglicerol Lipasas/metabolismo , Neoplasias/patología , Receptor Cannabinoide CB2/metabolismo , Anciano , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Humanos , Metabolismo de los Lípidos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Monoacilglicerol Lipasas/genética , Neoplasias/inmunología , Cultivo Primario de Células , Células RAW 264.7 , Receptor Cannabinoide CB2/antagonistas & inhibidores , Receptor Cannabinoide CB2/genética , Receptor Cannabinoide CB2/inmunología , Receptor Toll-Like 4/metabolismo
4.
Sci Rep ; 6: 31143, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27503002

RESUMEN

Electron acceptor redox potential (EARP) was presumed to be a determining factor for microbial metabolism in many natural and engineered processes. However, little is known about the potentially global effects of EARP on bacteria. In this study, we compared the physiological and transcriptomic properties of Shewanella decolorationis S12 respiring with different EARPs in microbial electrochemical systems to avoid the effects caused by the other physicochemical properties of real electron acceptor. Results showed that the metabolic activities of strain S12 were nonlinear responses to EARP. The tricarboxylic acid cycle for central carbon metabolism was down-regulated while glyoxylate shunt was up-regulated at 0.8 V compared to 0.2 and -0.2 V, which suggested that EARP is an important but not the only determinant for metabolic pathways of strain S12. Moreover, few cytochrome c genes were differentially expressed at different EARPs. The energy intensive flagella assembly and assimilatory sulfur metabolism pathways were significantly enriched at 0.8 V, which suggested strain S12 had stronger electrokinesis behavior and oxidative stress-response at high EARP. This study provides the first global information of EARP regulations on microbial metabolism, which will be helpful for understanding microorganism respiration.


Asunto(s)
Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica/fisiología , Shewanella/metabolismo , Transcriptoma/fisiología , Oxidación-Reducción , Shewanella/genética
5.
J Endocrinol ; 230(2): 227-37, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27288055

RESUMEN

In addition to the ascertained efficacy as antidiabetic drug, metformin is increasingly being used as weight-loss agent in obesity, and as insulin sensitizer in nonalcoholic fatty liver disease (NAFLD). However, the mechanisms underlying these effects are still incompletely understood. Emerging evidence suggest metformin as leptin sensitizer to mediate the weight-loss effect in the brain. In this study, we investigated effects of metformin on expression of leptin receptors in liver and kidney in mice. C57BL/6 mice were fed with chow diet (CD) or high-fat diet (HF) for 5months. Afterward, mice were treated with metformin (50mg/kg or 200mg/kg) for 15days. Metabolic parameters and hepatic gene expression were analyzed at the end of the treatment. We also tested the effects of metformin on plasma-soluble leptin receptor (sOB-R) levels in newly diagnosed type 2 diabetes mellitus (T2DM) patients, and assessed its effect on hepatosteatosis in mice. Results showed that metformin upregulates the expression of leptin receptors (OB-Ra, -Rb, -Rc, and -Rd) in liver but not kidney. The stimulation effect is dose-dependent in both chow and HF mice. Upregulation of OB-Rb, long signaling isoform, needs a relatively higher dose of metformin. This effect was paralleled by increased sOBR levels in mice and T2DM patients, and decreased hepatic triglyceride (TG) content and lipogenic gene expression, including sterol regulatory element-binding protein 1c (SREBP-1c), fatty acid synthase (FAS) and acetyl-CoA carboxylase-1 (ACC-1). Taken together, these data identify hepatic leptin receptor as target gene being upregulated by metformin which may enhance leptin sensitivity in liver to alleviate steatosis.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Hígado Graso/prevención & control , Hipoglucemiantes/uso terapéutico , Metformina/uso terapéutico , Receptores de Leptina/metabolismo , Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Anciano , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Evaluación Preclínica de Medicamentos , Hígado Graso/etiología , Hígado Graso/metabolismo , Femenino , Humanos , Hipoglucemiantes/farmacología , Riñón/efectos de los fármacos , Riñón/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Metformina/farmacología , Ratones Endogámicos C57BL , Persona de Mediana Edad , Distribución Aleatoria , Triglicéridos/metabolismo
6.
Clin Sci (Lond) ; 130(14): 1257-68, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27129186

RESUMEN

Transcription factor 4 (TCF-4) was recently identified as a candidate gene for the cause of type 2 diabetes, although the mechanisms have not been fully elucidated. In the present study, we demonstrated that the TCF-4 transgene in macrophages aggravated high-fat diet (HFD)-induced insulin resistance and chronic inflammation, characterized by the elevation of proinflammatory cytokines in the blood, liver and white adipose tissue, as well as a proinflammatory profile of immune cells in visceral fats in mice. Mechanistically, TCF-4 functioned as a co-activator of p65 to amplify the saturated free fatty acid (FFA)-stimulated promoter activity, mRNA transcription and secretion of proinflammatory cytokines in primary macrophages. Blockage of p65 with a specific interfering RNA or inhibitor could prevent TCF-4-enhanced expression of proinflammatory cytokines in FFA/lipopolysaccharide-treated primary macrophages. The p65 inhibitor could abolish macrophage TCF-4 transgene-aggravated systemic inflammation, glucose intolerance and insulin resistance in HFD-treated mice. In addition, we demonstrated that the mRNA expression of TCF-4 in the peripheral blood monocytes from humans was positively correlated to the levels of interleukin (IL)-1ß, tumour necrosis factor α, IL-6 and fasting plasma glucose. In summary, we identified TCF-4 as a co-activator of p65 in the potentiation of proinflammatory cytokine production in macrophages and aggravation of HFD-induced chronic inflammation and insulin resistance in mice.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/fisiología , Inflamación/etiología , Resistencia a la Insulina , Macrófagos/fisiología , Factor de Transcripción ReIA/fisiología , Animales , Células Cultivadas , Enfermedad Crónica , Citocinas/biosíntesis , Dieta Alta en Grasa , Humanos , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción 4
7.
Front Med ; 9(3): 275-87, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26290284

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) covers a spectrum of liver disorders ranging from simple steatosis to advanced pathologies, including nonalcoholic steatohepatitis and cirrhosis. NAFLD significantly contributes to morbidity and mortality in developed societies. Insulin resistance associated with central obesity is the major cause of hepatic steatosis, which is characterized by excessive accumulation of triglyceride-rich lipid droplets in the liver. Accumulating evidence supports that dysregulation of adipose lipolysis and liver de novo lipogenesis (DNL) plays a key role in driving hepatic steatosis. In this work, we reviewed the molecular mechanisms responsible for enhanced adipose lipolysis and increased hepatic DNL that lead to hepatic lipid accumulation in the context of obesity. Delineation of these mechanisms holds promise for developing novel avenues against NAFLD.


Asunto(s)
Resistencia a la Insulina/genética , Hígado/fisiopatología , Enfermedad del Hígado Graso no Alcohólico/genética , Obesidad/metabolismo , Triglicéridos/metabolismo , Humanos , Enfermedad del Hígado Graso no Alcohólico/epidemiología
8.
Drug Des Devel Ther ; 9: 1773-83, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25848214

RESUMEN

Altered expression of ADAMTS5 is associated with human carcinogenesis and tumor progression. However, the role of ADAMTS5 in hepatocellular carcinoma (HCC) is unclear. This study analyzed ADAMTS5 expression in HCC tissues and tested for association with clinicopathological and survival data from HCC patients and then explored the role of ADAMTS5 in HCC cells in vitro. Paraffin blocks from 48 HCC patients were used to detect ADAMTS5 and vascular endothelial growth factor (VEGF) expression and microvessel density (MVD). A normal liver cell line and HCC cell lines were used to detect ADAMTS5 expression and for ADAMTS5 manipulation. ADAMTS5 cDNA was stably transfected into HCC cells and ADAMTS5 expression assessed by Western blot analysis. Tumor cell-conditioned growth medium was used to assess human umbilical vein endothelial cell migration and Matrigel tube formation. Xenograft assay was performed to determine the role of ADAMTS5 in vivo. The data showed that the expression of ADAMTS5 was reduced in HCC, which was inversely associated with VEGF expression, MVD, and tumor size and associated with poor overall survival of HCC patients. Lentivirus-mediated ADAMTS5 expression significantly inhibited tumor angiogenesis by downregulating in vitro expression of VEGF and inhibiting migration and tube formations, and also inhibited tumor growth and VEGF expression and reduced MVD in vivo in a mouse xenograft model. Taken together, these results suggest that ADAMTS5 plays a role in suppression of HCC progression, which could be further studied as a promising novel therapeutic target and a potential prognostic marker in HCC.


Asunto(s)
Proteínas ADAM/biosíntesis , Carcinoma Hepatocelular/diagnóstico , Progresión de la Enfermedad , Neoplasias Hepáticas/diagnóstico , Proteína ADAMTS5 , Adulto , Anciano , Animales , Células Cultivadas , Femenino , Células Hep G2 , Humanos , Masculino , Ratones , Microvasos/patología , Persona de Mediana Edad , Neoplasias Experimentales/diagnóstico , Pronóstico , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Adulto Joven
9.
Clin Sci (Lond) ; 128(8): 493-506, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25431838

RESUMEN

Over-nutrition induces low-grade inflammation that dampens insulin sensitivity, but the underlying molecular mediators are not fully understood. Comparative gene identification-58 (CGI-58) is an intracellular lipolytic activator. In the present study, we show that in mouse visceral fat-derived macrophages or human peripheral blood monocytes, CGI-58 negatively and interleukin (IL)-1ß positively correlate with obesity. Saturated non-esterified fatty acid (NEFA) suppresses CGI-58 expression in macrophages and this suppression activates FOXO1 (forkhead box-containing protein O subfamily-1) through inhibition of FOXO1 phosphorylation. Activated FOXO1 binds to an insulin-responsive element in IL-1ß promoter region to potentiate IL-1ß transcription. Gain- and loss-of-function studies demonstrate that NEFA-induced CGI-58 suppression activates FOXO1 to augment IL-1ß transcription by dampening insulin signalling through induction of SOCS3 (suppressor of cytokine signalling 3) expression. CGI-58 deficiency-induced SOCS3 expression is NLRP3 (nucleotide-binding oligomerization domain-like receptor family, pyrin domain containing 3) inflammasome-dependent. Our data thus identified a vicious cycle (IL-1ß-SOCS3-FOXO1-IL-1ß) that amplifies IL-1ß secretion and is initiated by CGI-58 deficiency-induced activation of the NLRP3 inflammasome in macrophages. We further show that blocking this cycle with a FOXO1 inhibitor, an antioxidant that inhibits FOXO1 or IL-1 receptor antagonist alleviates chronic inflammation and insulin resistance in high-fat diet (HFD)-fed mice. Collectively, our data suggest that obesity-associated factors such as NEFA and lipopolysaccharide (LPS) probably adopt this vicious cycle to promote inflammation and insulin resistance.


Asunto(s)
1-Acilglicerol-3-Fosfato O-Aciltransferasa/deficiencia , Factores de Transcripción Forkhead/metabolismo , Interleucina-1beta/genética , Macrófagos/metabolismo , Transducción de Señal , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Transcripción Genética , 1-Acilglicerol-3-Fosfato O-Aciltransferasa/metabolismo , Animales , Índice de Masa Corporal , Dieta Alta en Grasa , Ácidos Grasos/farmacología , Proteína Forkhead Box O1 , Humanos , Inflamasomas/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Resistencia a la Insulina , Interleucina-1beta/metabolismo , Macrófagos/efectos de los fármacos , Masculino , Ratones , Modelos Biológicos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína 3 Supresora de la Señalización de Citocinas , Transcripción Genética/efectos de los fármacos , Aumento de Peso
10.
Cell Biochem Biophys ; 72(1): 221-5, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25519309

RESUMEN

A disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5) is considered to be an important anti-angiogenic protein, in which the first TSR domain is crucial for its anti-angiogenic activity. Previous study showed that ADAMTS5 plays a role in suppression of hepatocellular carcinoma (HCC) progression through its anti-angiogenic activity. The rs2380585 G>A single-nucleotide polymorphism (SNP) is a missense mutation, located in the ADAMTS5 first TSR domain coding sequence (CDS). In this study, we investigated the impacts of ADAMTS5 rs2380585 polymorphism on the risk and progress of hepatocellular carcinoma. A total of 220 HCC patients and 220 controls in a Chinese Han population were enrolled and genotyped. The associations between SNPs and HCC incidence and progression were analyzed with logistic regression model. We found that individuals with the ADAMTS5 rs2380585 A allele was significantly associated with decreased HCC risk (OR = 0.348, 95 % CI 0.236-0.512; p = 0.000). Individuals having the ADAMTS5 rs2380585 polymorphic genotype (GA+AA) had an OR of 0.348 (95 % CI 0.201-0.600; p = 0.000) for developing HCC, compared with individuals having the ADAMTS5 rs2380585 ancestral genotype. However, stratified analyses did not find any evident gene-covariates interaction. The SNP of rs2380585 was irrelevant to the frequencies of clinicopathological characteristics. Our results for the first time indicate that ADAMTS5 rs2380585 polymorphism contributes to HCC susceptibility.


Asunto(s)
Proteína ADAMTS5/genética , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Proteína ADAMTS5/metabolismo , Adulto , Anciano , Alelos , Carcinoma Hepatocelular/etnología , Carcinoma Hepatocelular/metabolismo , Estudios de Casos y Controles , China , Progresión de la Enfermedad , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Genotipo , Heterocigoto , Humanos , Neoplasias Hepáticas/etnología , Neoplasias Hepáticas/metabolismo , Masculino , Persona de Mediana Edad , Mutación Missense , Oportunidad Relativa , Polimorfismo de Nucleótido Simple
11.
PLoS One ; 9(9): e107913, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25238230

RESUMEN

CYP2E1, one of the cytochrome P450 mixed-function oxidases located predominantly in liver, plays a key role in metabolism of xenobiotics including ethanol and procarcinogens. Recently, down-expression of CYP2E1 was found in hepatocellular carcinoma (HCC) with the majority to be chronic hepatitis B virus (HBV) carriers. In this study, we tested a hypothesis that HBx may inhibit CYP2E1 gene expression via hepatocyte nuclear factor 4α (HNF4α). By enforced HBx gene expression in cultured HepG2 cells, we determined the effect of HBx on CYP2E1 mRNA and protein expression. With a bioinformatics analysis, we found a consensus HNF-4α binding sequence located on -318 to -294 bp upstream of human CYP2E1 promoter. Using reporter gene assay and site-directed mutagenesis, we have shown that mutation of this site dramatically decreased CYP2E1 promoter activity. By silencing endogenous HNF-4α, we have further validated knockdown of HNF-4α significantly decreased CYP2E1 expression. Ectopic overexpression of HBx in HepG2 cells inhibits HNF-4α expression, and HNF-4α levels were inversely correlated with viral proteins both in HBV-infected HepG2215 cells and as well as HBV positive HCC liver tissues. Moreover, the HBx-induced CYP2E1 reduction could be rescued by ectopic supplement of HNF4α protein expression. Furthermore, human hepatoma cells C34, which do not express CYP2E1, shows enhanced cell growth rate compared to E47, which constitutively expresses CYP2E1. In addition, the significantly altered liver proteins in CYP2E1 knockout mice were detected with proteomics analysis. Together, HBx inhibits human CYP2E1 gene expression via downregulating HNF4α which contributes to promotion of human hepatoma cell growth. The elucidation of a HBx-HNF4α-CYP2E1 pathway provides novel insight into the molecular mechanism underlining chronic HBV infection associated hepatocarcinogenesis.


Asunto(s)
Carcinoma Hepatocelular/genética , Citocromo P-450 CYP2E1/genética , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 4 del Hepatocito/genética , Neoplasias Hepáticas/genética , Animales , Línea Celular Tumoral , Regulación hacia Abajo , Humanos , Ratones , Ratones Noqueados
12.
Cell Rep ; 7(1): 223-35, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24703845

RESUMEN

Overnutrition activates a proinflammatory program in macrophages to induce insulin resistance (IR), but its molecular mechanisms remain incompletely understood. Here, we show that saturated fatty acid and lipopolysaccharide, two factors implicated in high-fat diet (HFD)-induced IR, suppress macrophage CGI-58 expression. Macrophage-specific CGI-58 knockout (MaKO) in mice aggravates HFD-induced glucose intolerance and IR, which is associated with augmented systemic/tissue inflammation and proinflammatory activation of adipose tissue macrophages. CGI-58-deficient macrophages exhibit mitochondrial dysfunction due to defective peroxisome proliferator-activated receptor (PPAR)γ signaling. Consequently, they overproduce reactive oxygen species (ROS) to potentiate secretion of proinflammatory cytokines by activating NLRP3 inflammasome. Anti-ROS treatment or NLRP3 silencing prevents CGI-58-deficient macrophages from oversecreting proinflammatory cytokines and from inducing proinflammatory signaling and IR in the cocultured fat slices. Anti-ROS treatment also prevents exacerbation of inflammation and IR in HFD-fed MaKO mice. Our data thus establish CGI-58 as a suppressor of overnutrition-induced NLRP3 inflammasome activation in macrophages.


Asunto(s)
1-Acilglicerol-3-Fosfato O-Aciltransferasa/deficiencia , Inflamasomas/metabolismo , Resistencia a la Insulina/fisiología , Macrófagos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , 1-Acilglicerol-3-Fosfato O-Aciltransferasa/biosíntesis , Animales , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/metabolismo , Regulación hacia Abajo , Ácidos Grasos/farmacología , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR
13.
Free Radic Biol Med ; 63: 175-86, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23669278

RESUMEN

Ethanol induces hypoxia and elevates HIF-1α in the liver. CYP2E1 plays a role in the mechanisms by which ethanol generates oxidative stress, fatty liver, and liver injury. This study evaluated whether CYP2E1 contributes to ethanol-induced hypoxia and activation of HIF-1α in vivo and whether HIF-1α protects against or promotes CYP2E1-dependent toxicity in vitro. Wild-type (WT), CYP2E1-knock-in (KI), and CYP2E1 knockout (KO) mice were fed ethanol chronically; pair-fed controls received isocaloric dextrose. Ethanol produced liver injury in the KI mice to a much greater extent than in the WT and KO mice. Protein levels of HIF-1α and downstream targets of HIF-1α activation were elevated in the ethanol-fed KI mice compared to the WT and KO mice. Levels of HIF prolyl hydroxylase 2, which promotes HIF-1α degradation, were decreased in the ethanol-fed KI mice in association with the increases in HIF-1α. Hypoxia occurred in the ethanol-fed CYP2E1 KI mice as shown by an increased area of staining using the hypoxia-specific marker pimonidazole. Hypoxia was lower in the ethanol-fed WT mice and lowest in the ethanol-fed KO mice and all the dextrose-fed mice. In situ double staining showed that pimonidazole and CYP2E1 were colocalized to the same area of injury in the hepatic centrilobule. Increased protein levels of HIF-1α were also found after acute ethanol treatment of KI mice. Treatment of HepG2 E47 cells, which express CYP2E1, with ethanol plus arachidonic acid (AA) or ethanol plus buthionine sulfoximine (BSO), which depletes glutathione, caused loss of cell viability to a greater extent than in HepG2 C34 cells, which do not express CYP2E1. These treatments elevated protein levels of HIF-1α to a greater extent in E47 cells than in C34 cells. 2-Methoxyestradiol, an inhibitor of HIF-1α, blunted the toxic effects of ethanol plus AA and ethanol plus BSO in the E47 cells in association with inhibition of HIF-1α. The HIF-1α inhibitor also blocked the elevated oxidative stress produced by ethanol/AA or ethanol/BSO in the E47 cells. These results suggest that CYP2E1 plays a role in ethanol-induced hypoxia, oxidative stress, and activation of HIF-1α and that HIF-1α contributes to CYP2E1-dependent ethanol-induced toxicity. Blocking HIF-1α activation and actions may have therapeutic implications for protection against ethanol/CYP2E1-induced oxidative stress, steatosis, and liver injury.


Asunto(s)
Citocromo P-450 CYP2E1/metabolismo , Etanol/administración & dosificación , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Estrés Oxidativo , Animales , Supervivencia Celular/efectos de los fármacos , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/fisiopatología , Citocromo P-450 CYP2E1/genética , Glutatión/metabolismo , Células Hep G2 , Humanos , Hipoxia/inducido químicamente , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Hígado/efectos de los fármacos , Ratones , Ratones Noqueados , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Procolágeno-Prolina Dioxigenasa/metabolismo , Especies Reactivas de Oxígeno/metabolismo
14.
Endocrinology ; 153(12): 5821-33, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23070544

RESUMEN

Increasing evidence suggests that inflammation/cytokines may modulate hypothalamic responses to leptin, which is a key regulator of energy homeostasis and inflammatory/stress responses. We investigated a possible role of TNF-α, a key early mediator of inflammation, in regulating the expression and trafficking of the long-isoform leptin receptor (LEPRb), the primary mediator of leptin signaling, in cultured cells. We found that TNF-α in a wide range of concentrations up-regulated LEPRb protein level and soluble LEPR (sLEPR) release via ectodomain shedding of LEPRb in multiple cell types, including neuronal cells. TNF-α also acutely increased LEPRb cell surface expression and leptin-induced STAT3 phosphorylation. In contrast, TNF-α had no significant effects on the protein level or cell surface expression of several other transmembrane proteins, including the transferrin receptor and cadherin. The stimulatory effects of TNF-α on LEPRb cell surface expression and sLEPR release were not dependent on de novo protein synthesis or functional lysosomes but were blocked by brefeldin A, suggesting that an intact Golgi or continuous endoplasmic reticulum to Golgi transport of newly synthesized proteins is required for these effects. However, TNF-α did not increase the half-life of cell surface LEPRb. Protein kinase C (PKC) inhibitor GF109203X abrogated the effects of TNF-α, whereas the pan-PKC activator phorbol 12-myristate 13-acetate mimicked the TNF-α effects. Taken together, our results suggest that TNF-α, via activation of PKC, regulates anterograde trafficking and/or degradation of LEPRb in the biosynthetic pathway, leading to concomitant increases in LEPRb protein level, cell surface expression, and sLEPR production. The finding that LEPRb cell surface expression and sLEPR production, key modulators of leptin sensitivity and bioavailability, are direct targets of TNF-α signaling could have a potentially important implication in the regulation of leptin signaling activity in different pathophysiological conditions as diverse as obesity and sepsis.


Asunto(s)
Membrana Celular/metabolismo , Proteína Quinasa C/metabolismo , Receptores de Leptina/biosíntesis , Factor de Necrosis Tumoral alfa/biosíntesis , Regulación hacia Arriba , Animales , Cadherinas/metabolismo , Línea Celular , Inhibidores Enzimáticos/farmacología , Aparato de Golgi/metabolismo , Homeostasis , Humanos , Indoles/farmacología , Maleimidas/farmacología , Ratones , Neuronas/metabolismo , Fosforilación , Proteína Quinasa C/antagonistas & inhibidores , Transporte de Proteínas , Receptores de Transferrina/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
15.
Mol Endocrinol ; 26(3): 423-37, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22240809

RESUMEN

Hepatic insulin resistance (IR) is associated with liver inflammatory diseases, but molecular mechanisms for the association remained elusive. IR is known to increase activity of forkhead box-containing protein O subfamily-1 (FOXO1), a transcription factor that was recently shown to enhance proinflammatory cytokine production in macrophages and adipocytes. Here we report that overexpression of constitutively active FOXO1 markedly increased chemokine ligand 20 (CCL20) expression and secretion in HepG2 hepatoma cells treated with TNF-α. The opposite was seen when endogenous FOXO1 was silenced. FOXO1 did not bind CCL20 promoter directly; instead, it potentiated CCL20 transcription through increasing the binding of p65/p50 heterodimer to a functional nuclear factor-κB site in the human CCL20 promoter. The conditional medium from TNF-α-treated HepG2 cells stimulated migration of human peripheral blood mononuclear cells. This stimulation was significantly enhanced when FOXO1 was overexpressed, and attenuated when FOXO1 was silenced. CCL20 antibody partly blocked the synergistic effect of FOXO1 and TNF-α on peripheral blood mononuclear cells migration. Additionally, TNF-α antagonizes the insulin/Akt signal transduction, thus leading to activation of FOXO1, which is capable of mediating a transcriptional activation role in response to TNF-α on CCL20 gene expression in HepG2 cells and promotes lymphocyte chemotaxis. Furthermore, we found that FOXO1 and CCL20 were coordinately up-regulated in the insulin resistant and inflammatory cell-infiltrated liver of db/db mice, an animal model that displayed hepatic and systemic low-grade inflammation. In conclusion, our data suggest that FOXO1 links IR to lymphocyte chemotaxis in the insulin-resistant hepatocytes and livers by amplifying nuclear factor-κB-dependent hepatic CCL20 production.


Asunto(s)
Quimiocina CCL20/metabolismo , Quimiotaxis , Factores de Transcripción Forkhead/fisiología , Linfocitos/fisiología , Subunidad p50 de NF-kappa B/metabolismo , Factor de Transcripción ReIA/metabolismo , Animales , Secuencia de Bases , Quimiocina CCL20/genética , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Células Hep G2 , Humanos , Inflamación/metabolismo , Inflamación/patología , Insulina/fisiología , Hígado/metabolismo , Linfocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Unión Proteica , Elementos de Respuesta , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética , Factor de Necrosis Tumoral alfa/fisiología
16.
Inflamm Res ; 61(4): 349-58, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22223069

RESUMEN

OBJECTIVE: Low-grade inflammation from hepatocytes plays a causal role in hepatic and systemic insulin resistance (IR). We aimed to explore whether and how FOXO1 was involved in IR-related inflammation in hepatocytes. METHODS: We determined FOXO1 expression and activity, insulin and NF-κB signaling, and pro-inflammatory cytokine production in tumor necrosis factor-α (TNF-α)- or dexamethasone (DEX)-induced IR model in vitro and in high fat diet-induced obese or diabetic db/db mice in vivo with quantitative RT-PCR and Western blotting. RESULTS: We identified two different but physiologically relevant IR models characterized by attenuated insulin-induced phosphorylation of insulin receptor substrate-1 and AKT in TNF-α- or DEX-treated HepG2 cells. DEX largely increased FOXO1 expression in hepatocytes, while TNF-α did not. Notably, FOXO1 phosphorylation was attenuated in both models. TNF-α-stimulated nuclear translocation of NF-κB (p65) and mRNA levels of interleukin (IL)-1, IL-6 and monocyte attractant protein-1 were partly blocked, while the anti-inflammatory role of DEX was largely potentiated by insulin. FOXO1 knockdown by human-specific FOXO1 small interfering RNA exerted an identical role to insulin. Furthermore, augmented hepatic FOXO1 expression and decreased phosphorylation were found to be associated with elevated pro-inflammatory cytokine production in high fat diet-induced obese and db/db mice. CONCLUSION: FOXO1 potentiates pro-inflammatory cytokine production in insulin-resistant hepatocytes.


Asunto(s)
Citocinas/genética , Factores de Transcripción Forkhead/metabolismo , Hepatocitos/metabolismo , Resistencia a la Insulina/fisiología , Línea Celular , Dexametasona , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Técnicas de Silenciamiento del Gen , Humanos , ARN Mensajero/metabolismo
17.
Mycologia ; 103(5): 946-9, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21471295

RESUMEN

Eight peptide toxins were isolated and purified from basidiocarps of Amanita exitialis with high performance liquid chromatography and were subjected to ultraviolet, nuclear magnetic resonance and mass spectrometry. We identified seven peptide toxins, α-amanitin, ß-amanitin, amaninamide, phallacin, phallacidin, phallisacin and desoxoviroidin. The molecular weight (729.5 Da) of the eighth compound did not match that of any reported Amanita toxins and, although the UV absorption spectrum indicated it to be a phallotoxin, further studies are required to identify this component. This is the first report of amaninamide, phallacin, phallisacin and desoxoviroidin in this lethal mushroom species.


Asunto(s)
Amanita/química , Cuerpos Fructíferos de los Hongos/química , Micotoxinas/química , Micotoxinas/aislamiento & purificación , Cromatografía Líquida de Alta Presión , Espectrometría de Masas , Resonancia Magnética Nuclear Biomolecular , Péptidos/química , Péptidos/aislamiento & purificación , Espectrofotometría Ultravioleta
18.
Gynecol Endocrinol ; 25(7): 464-71, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19903045

RESUMEN

In this study, we examined the long-term influence of a high-fat (HF) diet compared with a standard chow (SC) diet on glucose metabolism in genetically normal Sprague-Dawley rats. As the diet was given for a period of 14 months it represents almost a life-long exposure of the rats to the diet. Results showed that (1) after exposure to 14 months of SC or HF feeding, the fasting glucose levels were below 5.1 mmol/l in both genders; (2) the 2 h plasma glucose concentration (2 h PG) of male rats after an intraperitoneal glucose tolerance test for the SC groups and for the HF groups was significantly higher than for females; (3) the liver triglycerides content was significantly enhanced by HF feeding as compared with SC-feeding, and histological staining showed that both genders of the HF group developed severe liver steatosis; (4) the fasting insulin concentration in blood of HF-fed males was significantly higher than that of females and homeostasis model assessment of insulin resistance was also significantly higher; (5) the insulin secretion capabilities in response to glucose stimulation were significantly higher in HF-fed female rats than that of males. Taken together, these data indicate that glucose intolerance produced by a HF diet in rats shows a gender difference, and females are protected against development of impaired glucose tolerance under HF condition.


Asunto(s)
Alimentación Animal , Grasas de la Dieta/farmacología , Intolerancia a la Glucosa/dietoterapia , Intolerancia a la Glucosa/prevención & control , Adaptación Biológica , Animales , Glucemia/metabolismo , Peso Corporal/fisiología , Ácidos Grasos no Esterificados/sangre , Hígado Graso/dietoterapia , Hígado Graso/prevención & control , Femenino , Homeostasis/fisiología , Insulina/sangre , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiología , Hígado/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Caracteres Sexuales , Triglicéridos/metabolismo
19.
Sheng Wu Gong Cheng Xue Bao ; 23(4): 602-6, 2007 Jul.
Artículo en Chino | MEDLINE | ID: mdl-17822030

RESUMEN

To carry out the secretive expression of human 67 kD laminin receptor (67LR), recombinant expression plasmid pPIC9K-67LR was constructed by inserting of 67LR cDNA into yeast expression vector pPIC9K. The 67LR protein was expressed in Pichia pastoris after induced by methanol, and about 12.56 mg electrophoresis purity 67LR could be obtained after the purification of 1L culture using affinity chromatograph column. In vitro competitive binding assay showed that target protein has an excellent biological activity. The successful expression of 67LR has placed a solid foundation for the research on structure and functions of 67LR.


Asunto(s)
Pichia/metabolismo , Receptores de Laminina/biosíntesis , Proteínas Recombinantes/biosíntesis , Vectores Genéticos , Humanos , Pichia/genética , Plásmidos/genética , Receptores de Laminina/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
20.
Biochem Biophys Res Commun ; 337(4): 1092-6, 2005 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-16236254

RESUMEN

Sirtuin 1 (SirT1), an NAD-dependent deacetylase that is important for promoting longevity during caloric restriction, can deacetylate and enhance the function of forkhead box transcription factors, O subfamily (FoxO). We examined the effect of SirT1 on the regulation of insulin-like growth factor-binding protein 1 (IGFBP-1), a known target of FoxO proteins that is increased in fasting. Co-transfection with a SirT1 expression vector dose-dependently stimulated IGFBP-1 promoter activity and a heterologous reporter gene construct containing three FoxO-binding sites linked to a minimal promoter. This effect is mimicked by 20muM resveratrol, a potent SirT1 activator, and immunoprecipitation and Western blotting confirm that SirT1 and FoxO1 interact in cells. Interestingly, mutation of FoxO-binding sites in the IGFBP-1 promoter reduces, but does not completely disrupt, the stimulatory effect of SirT1 on promoter activity. We found that overexpression of SirT1 is accompanied by enhanced mitogen-activated protein kinase (MAPK) activation. Treatment of SirT1-cotransfected cells with PD98059, which inhibits MAPK activation, decreased IGFBP-1 promoter activity by approximately 50%, in a FoxO-binding site-independent manner, and disrupts the residual effect of SirT1. These results indicate that SirT1 stimulates IGFBP-1 promoter activity through FoxO-dependent and -independent mechanisms, and provides the first evidence that activation of MAPK contributes to effects of SirT1 on gene expression.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica/genética , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Sirtuinas/metabolismo , Secuencia de Bases , Línea Celular , Factores de Transcripción Forkhead/clasificación , Factores de Transcripción Forkhead/genética , Humanos , Sistema de Señalización de MAP Quinasas , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética , Sirtuinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...