Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Genes Cells ; 28(8): 585-594, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37249025

RESUMEN

The nuclear receptors peroxisome proliferator-activated receptor gamma (PPARγ) and adipogenin (ADIG) play vital roles in lipid metabolism. However, the interaction between PPARγ and ADIG during liver steatosis remains unclear. In this study, we aimed to investigate the role of PPARγ in the transcriptional regulation of hepatic ADIG expression. Adig was found to be highly expressed in various fatty liver mouse models. Although hepatic Adig was expressed at high levels in the fatty liver of type 2 diabetic ob/ob mice and was upregulated by PPARγ agonist treatment, it was expressed at significantly low levels in liver-specific Pparg-knockout mice. Moreover, hepatic Adig expression was observed in other mouse models of liver steatosis, such as the leptin receptor mutant db/db and alcohol-fed mice. Adig was also highly expressed in the white and brown adipose tissues, skeletal muscles, and heart of ob/ob mice. Reporter and electromobility shift assays showed that PPARγ positively regulates Adig transcriptional activity by directly binding to a functional PPARγ-responsive element in the promoter region. Our results indicate that Adig is a novel target gene of hepatic PPARγ in liver steatosis.


Asunto(s)
Hígado Graso , Proteínas Nucleares , PPAR gamma , Animales , Ratones , Etanol/metabolismo , Hígado Graso/genética , Hígado Graso/metabolismo , Regulación de la Expresión Génica , Hígado/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR gamma/genética , PPAR gamma/metabolismo
2.
Biol Pharm Bull ; 46(2): 334-337, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36724961

RESUMEN

Morphinone (MO) is an electrophilic metabolite of morphine that covalently binds to protein thiols, resulting in toxicity in vitro and in vivo. We have previously identified a variety of redox signaling pathways that are activated during electrophilic stress. However, the role of MO in such activation remains unknown. In this study, we examined whether MO could activate heat shock protein (HSP) 90/heat shock factor (HSF) 1 signaling in HepG2 cells. MO exposure caused S-modification of HSP90 (determined using biotin-PEAC5-maleimide labeling) and nuclear translocation of transcription factor HSF1, thereby up-regulating its downstream genes encoding B-cell lymphoma 2-associated anthanogene 3 and heat shock 70 kDa protein 1. However, dihydromorphinone, a non-electrophilic metabolite of morphine, had little effect on HSF1 activation or upregulation of these genes, suggesting that covalent modification plays a role in this process and that the HSP90/HSF1 pathway is a redox-signaled adaptive response to morphine metabolism.


Asunto(s)
Proteínas de Unión al ADN , Morfina , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción del Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico , Morfina/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Humanos , Células Hep G2
3.
Biol Pharm Bull ; 46(2): 338-342, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36724962

RESUMEN

Morphinone (MO) is an electrophilic metabolite of morphine that covalently binds to protein thiols via its α,ß-unsaturated carbonyl group, resulting in toxicity in vitro and in vivo. Our previous studies identified a variety of redox signaling pathways that are activated during electrophilic stress. Here, we examined in vitro activation of a signaling pathway involving Kelch-like ECH-associated protein 1 (Keap1) and nuclear factor erythroid 2-related factor 2 (Nrf2) in response to MO. Exposure of HepG2 cells to MO caused covalent modification of Keap1 thiols (evaluated using biotin-PEAC5-maleimide labeling) and nuclear translocation of Nrf2, thereby up-regulating downstream genes encoding ATP binding cassette subfamily C member 2, solute carrier family 7 member 11, glutamate-cysteine ligase catalytic subunit, glutamate-cysteine ligase modifier subunit, glutathione S-transferase alpha 1, and heme oxygenase 1. However, dihydromorphinone, a metabolite of morphine lacking the reactive C7-C8 double bond, had little effect on Nrf2 activation. These results suggest that covalent modification is crucial in the Keap1/Nrf2 pathway activation and that this pathway is a redox signaling-associated adaptive response to MO metabolism.


Asunto(s)
Glutamato-Cisteína Ligasa , Factor 2 Relacionado con NF-E2 , Glutamato-Cisteína Ligasa/genética , Glutamato-Cisteína Ligasa/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Morfina/farmacología , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Compuestos de Sulfhidrilo , Humanos , Células Hep G2
4.
Endocr J ; 69(5): 567-575, 2022 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-35082200

RESUMEN

CCAAT/enhancer-binding protein α (C/EBPα) is a transcription factor abundantly expressed in the liver and white adipose tissue (WAT). In this study, we investigated the mechanism by which C/EBPα regulates the lipase family member N (Lipn) gene in the mouse liver. Mouse Lipn consists of non-coding exon 1 and the translation start site located in exon 2. Lipn expression in the fatty liver of ob/ob mice was significantly higher than that in OB/OB mice and was significantly repressed by liver-specific C/EBPα deficiency. Lipn expression in ob/ob mice was detected in the liver, epididymal WAT (eWAT), subcutaneous WAT (sWAT), brown adipose tissue (BAT), and skeletal muscle, but not in the kidney, brain, and heart. Lipn expression in the liver, eWAT, and sWAT of wild-type mice was undetectable, although C/EBPα was highly expressed in these tissues. The database analysis revealed four putative C/EBP-responsive elements (CEBPREs), highly homologous with the typical CEBPRE consensus sequence at positions -2,686/-2,678, -1,364/-1,356, -106/-98, and -45/-37 from the transcription start site (+1) of Lipn. Reporter assays using reporter constructs with serial or internal deletions of the 5'-flanking regions of Lipn showed that two functional CEBPREs (-106/-98 and -45/-37) in the Lipn promoter region are essential for enhancing Lipn transcriptional activity by C/EBPα. Electrophoretic mobility shift assay showed that C/EBPα/ß binds to CEBPRE (-106/-98). These results suggest that C/EBPα and type 2 diabetic environment may be required for hepatic Lipn expression.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Diabetes Mellitus Tipo 2 , Lipasa/genética , Hígado/metabolismo , Animales , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Lipasa/metabolismo , Ratones , Regiones Promotoras Genéticas
5.
Endocr J ; 67(1): 9-14, 2020 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-31484851

RESUMEN

Visceral adipose tissue-derived serine protease inhibitor (vaspin), initially identified in the visceral adipose tissue, is an adipokine that improves endoplasmic reticulum stress in obesity or insulin sensitivity and glucose tolerance. However, the transcriptional regulation of the hepatic vaspin gene remains elusive. We have previously shown that CCAAT-enhancer-binding protein α, a transcription factor of the basic leucine zipper class, positively regulates the vaspin gene. The present study aimed to investigate the nutritional or hormonal regulators of vaspin expression in the liver. For the fasting and refeeding study, mice in the fasting group were subjected to fasting for 24 h and then sacrificed. Mice in the refeeding group were subjected to fasting for 24 h and then refed with a 50% (w/w) sucrose/MF diet for further 24 h and then sacrificed. For the streptozotocin (STZ) study, STZ (50 mg/kg) was intraperitoneally injected into C57BL/6JJc1 mice for 5 d. Hepatic vaspin was repressed due to fasting for 24 h and was induced upon refeeding with a high-sucrose diet. In studies on liver-specific C/EBPα-deficient mice, C/EBPα was not involved in the induction of hepatic vaspin upon refeeding. In addition, the depletion of insulin by streptozotocin treatment markedly decreased hepatic vaspin expression. Finally, fasting-repressed vaspin expression in the liver was significantly increased by direct injection of insulin into fasting mice. In conclusion, our results suggest that insulin is a positive regulator of hepatic vaspin expression.


Asunto(s)
Adipoquinas/genética , Diabetes Mellitus Experimental/genética , Ayuno/metabolismo , Insulina/metabolismo , Hígado/metabolismo , Serpinas/genética , Adipoquinas/metabolismo , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Diabetes Mellitus Experimental/metabolismo , Sacarosa en la Dieta , Regulación de la Expresión Génica , Hipoglucemiantes/farmacología , Insulina/farmacología , Grasa Intraabdominal/metabolismo , Hígado/efectos de los fármacos , Ratones , Ratones Noqueados , ARN Mensajero/metabolismo , Serpinas/efectos de los fármacos , Serpinas/metabolismo
6.
Endocr J ; 67(1): 37-44, 2020 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-31564684

RESUMEN

The fat-specific protein 27 gene (Fsp27) belongs to the cell death-inducing DNA fragmentation factor 45-like effector family. Fsp27 is highly expressed in adipose tissue and fatty liver. In adipocytes, FSP27 localizes to the membrane of lipid droplets and promotes lipid droplet hypertrophy. Recently, FSP27 was shown to consist of two isoforms, FSP27α and FSP27ß. Previously, we demonstrated that Fsp27a is directly regulated by peroxisome proliferator-activated receptor γ (PPARγ) in fatty livers of genetically obese leptin deficient ob/ob mice and that Fsp27b may potentially be regulated by different factors transcriptionally as they both have a different promoter region. Thus, the aim of the present study was to elucidate whether Fsp27b is regulated by PPARγ in fatty liver. Fsp27a and Fsp27b were markedly induced in fatty liver of ob/ob mice compared with those in the normal liver. However, both Fsp27a/b were expressed at markedly lower levels in liver-specific PPARγ knockout mice with an ob/ob background. Further, the PPAR response element (PPRE) for the PPARγ-dependent promotion of Fsp27b promotor activity was revealed at position -1,163/-1,151 from the transcriptional start site (+1). Interestingly, the cis-element responsible for the PPARγ-dependent induction of Fsp27b was the same as that responsible for PPARγ-dependent induction of Fsp27a. These results suggest that PPARγ regulates not only Fsp27a but also Fsp27b in fatty liver of ob/ob mice through a common PPRE.


Asunto(s)
Hígado Graso/genética , PPAR gamma/genética , Proteínas/genética , Animales , Hígado Graso/metabolismo , Regulación de la Expresión Génica , Ratones , Ratones Noqueados , Ratones Obesos , Isoformas de Proteínas , Proteínas/metabolismo , Elementos de Respuesta
7.
Biol Pharm Bull ; 40(6): 888-893, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28566630

RESUMEN

The fat-specific protein 27 (Fsp27) gene belongs to the cell death-inducing DNA fragmentation factor 45-like effector family. Fsp27 is highly expressed in adipose tissue as well as the fatty liver of ob/ob mice. Fsp27 is directly regulated by the peroxisome proliferator-activated receptor γ (PPARγ) in livers of genetically obese leptin deficient ob/ob mice. In the present study, Fsp27 was markedly induced by 24 h fasting in genetically normal mouse livers and repressed by refeeding a high sucrose diet. In contrast with the liver, Fsp27 expression was decreased in adipose tissue by fasting and increased by refeeding. Interestingly, fasting-induced Fsp27 liver expression was independent of PPARγ. Moreover, Fsp27 expression was induced in the insulin-depleted livers of streptozotocin-treated mice. Finally, Fsp27 expression was repressed by direct injection of glucose or insulin in fasting mice. These results suggest that insulin represses Fsp27 expression in the fasting liver.


Asunto(s)
Ayuno/metabolismo , Insulina/farmacología , Hígado/metabolismo , Proteínas/genética , Tejido Adiposo Blanco/metabolismo , Animales , Masculino , Ratones Noqueados , Ratones Obesos , PPAR gamma/genética , Estreptozocina
8.
Gastroenterology ; 149(4): 1030-41.e6, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26099526

RESUMEN

BACKGROUND & AIMS: Alcoholic steatohepatitis (ASH) is the progressive form of alcoholic liver disease and may lead to cirrhosis and hepatocellular carcinoma. We studied mouse models and human tissues to identify molecules associated with ASH progression and focused on the mouse fat-specific protein 27 (FSP-27)/human cell death-inducing DFF45-like effector C (CIDEC) protein, which is expressed in white adipose tissues and promotes formation of fat droplets. METHODS: C57BL/6N mice or mice with hepatocyte-specific disruption of Fsp27 (Fsp27(Hep-/-) mice) were fed the Lieber-Decarli ethanol liquid diet (5% ethanol) for 10 days to 12 weeks, followed by 1 or multiple binges of ethanol (5 or 6 g/kg) during the chronic feeding. Some mice were given an inhibitor (GW9662) of peroxisome proliferator-activated receptor γ (PPARG). Adenoviral vectors were used to express transgenes or small hairpin (sh) RNAs in cultured hepatocytes and in mice. Liver tissue samples were collected from ethanol-fed mice or from 31 patients with alcoholic hepatitis (AH) with biopsy-proved ASH and analyzed histologically and immunohistochemically and by transcriptome, immunoblotting, and real-time PCR analyses. RESULTS: Chronic-plus-binge ethanol feeding of mice, which mimics the drinking pattern of patients with AH, produced severe ASH and mild fibrosis. Microarray analyses revealed similar alterations in expression of many hepatic genes in ethanol-fed mice and humans with ASH, including up-regulation of mouse Fsp27 (also called Cidec) and human CIDEC. Fsp27(Hep-/-) mice and mice given injections of adenovirus-Fsp27shRNA had markedly reduced ASH following chronic-plus-binge ethanol feeding. Inhibition of PPARG and cyclic AMP-responsive element binding protein H (CREBH) prevented the increases in Fsp27α and FSP27ß mRNAs, respectively, and reduced liver injury in this chronic-plus-binge ethanol feeding model. Overexpression of FSP27 and ethanol exposure had synergistic effects in inducing production of mitochondrial reactive oxygen species and damage to hepatocytes in mice. Hepatic CIDEC mRNA expression was increased in patients with AH and correlated with the degree of hepatic steatosis and disease severity including mortality. CONCLUSIONS: In mice, chronic-plus-binge ethanol feeding induces ASH that mimics some histological and molecular features observed in patients with AH. Hepatic expression of FSP27/CIDEC is highly up-regulated in mice following chronic-plus-binge ethanol feeding and in patients with AH; this up-regulation contributes to alcohol-induced liver damage.


Asunto(s)
Hígado Graso Alcohólico/metabolismo , Hepatocitos/metabolismo , Hígado/metabolismo , Proteínas/metabolismo , Adulto , Animales , Proteínas Reguladoras de la Apoptosis , Consumo Excesivo de Bebidas Alcohólicas , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Hígado Graso Alcohólico/genética , Hígado Graso Alcohólico/patología , Hígado Graso Alcohólico/prevención & control , Femenino , Perfilación de la Expresión Génica/métodos , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Humanos , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Mitocondrias Hepáticas/metabolismo , PPAR gamma/antagonistas & inhibidores , PPAR gamma/metabolismo , Proteínas/genética , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Índice de Severidad de la Enfermedad , Transducción de Señal , Factores de Tiempo , Transfección , Regulación hacia Arriba
9.
Cell Metab ; 7(4): 302-11, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18396136

RESUMEN

Peroxisome proliferator-activated receptor gamma (PPARgamma) is induced in leptin-deficient (ob/ob) mouse liver and is critical for the development of hepatic steatosis. The present study shows that fat-specific protein 27 (Fsp27) in ob/ob liver is a direct target gene of PPARgamma and can elevate hepatic triglyceride levels. FSP27 belongs to the CIDE family, composed of CIDE A, CIDE B, and FSP27/CIDE C, all of which contain a conserved CIDE-N domain. FSP27 was recently reported to be a lipid droplet-binding protein and to promote lipid accumulation in adipocytes. The Fsp27 gene was expressed at high levels in ob/ob liver and at markedly lower levels in ob/ob livers lacking PPARgamma. Forced expression of FSP27 by adenovirus in hepatocytes in vitro or in vivo led to increased triglyceride levels. Knockdown by adenovirus expressing FSP27 shRNA resulted in lower accumulation of hepatic triglycerides compared to control adenovirus-infected liver. Taken together, these results indicate that FSP27 is a direct mediator of PPARgamma-dependent hepatic steatosis.


Asunto(s)
Hígado Graso/metabolismo , Leptina/deficiencia , PPAR gamma/metabolismo , Proteínas/metabolismo , Adenoviridae/genética , Adipocitos/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Células Cultivadas , Hígado Graso/genética , Regulación de la Expresión Génica , Hepatocitos/metabolismo , Leptina/metabolismo , Ratones , Regiones Promotoras Genéticas , Proteínas/genética , ARN/metabolismo , Triglicéridos/metabolismo
10.
Biol Pharm Bull ; 36(11): 1766-72, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24189421

RESUMEN

Fat-specific protein 27 gene (FSP27), isolated by screening for genes specifically expressed in fully differentiated mouse adipocytes, belongs to the cell death-inducing DNA fragmentation factor, alpha subunit-like effector family. FSP27 is induced in not only adipose tissue but also the liver of ob/ob mice, and it promotes the development of fatty liver. The FSP27 gene is expressed in a fatty liver-specific manner and is not detected in the normal mouse liver. FSP27 expression is directly regulated by the induction of the hepatic peroxisome proliferator-activated receptor γ (PPARγ) in ob/ob fatty liver. In the present study, expression of hepatic FSP27 mRNA was determined in non-genetic fatty liver models. The FSP27 gene was markedly induced in the high-fat- or methionine- and choline-deficient (MCD) diet-induced fatty liver, but it was not elevated in alcohol-induced fatty liver. Interestingly, the induction of FSP27 mRNA due to the MCD diet was independent of PPARγ levels and completely absent in the liver from PPARγ-null mice. These results suggest that FSP27 mRNA expression in the liver depends on the etiology of fatty liver.


Asunto(s)
Diabetes Mellitus/genética , Hígado Graso/genética , Obesidad/genética , Proteínas/genética , Animales , Deficiencia de Colina , Diabetes Mellitus/metabolismo , Dieta , Dieta Alta en Grasa , Hígado Graso/etiología , Hígado Graso/metabolismo , Femenino , Hígado/metabolismo , Metionina/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Obesidad/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Proteínas/metabolismo , ARN Mensajero/metabolismo
11.
Eur Heart J ; 33(10): 1279-89, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21825308

RESUMEN

AIMS: Obesity is commonly associated with hypertension. Increased sympathetic tonus in obese subjects contributes to the underlying mechanism. However, the precise mechanisms whereby obesity induces this sympathetic activation remain unclear. Hepatic peroxisome proliferator-activated receptor (PPAR)-γ2 expression, which is reportedly upregulated during obesity development, affects sympathetic activation via hepatic vagal afferents. Herein, we report involvement of this neuronal relay in obesity-related hypertension. METHODS AND RESULTS: Peroxisome proliferator-activated receptor-γ and a direct PPARγ target, fat-specific protein 27 (Fsp27), were adenovirally overexpressed or knocked down in the liver, in combination with surgical dissection or pharmacological deafferentation of the hepatic vagus. Adenoviral PPARγ2 expression in the liver raised blood pressure (BP) in wild-type but not in ß1/ß2/ß3 adrenergic receptor-deficient mice. In addition, knockdown of endogenous PPARγ in the liver lowered BP in murine obesity models. Either surgical dissection or pharmacological deafferentation of the hepatic vagus markedly blunted BP elevation in mice with diet-induced and genetically-induced obesity. In contrast, BP was not elevated in other models of hepatic steatosis, DGAT1 and DGAT2 overexpressions, in which PPARγ is not upregulated in the liver. Thus, hepatic PPARγ upregulation associated with obesity is involved in BP elevation during obesity development. Furthermore, hepatic expression of Fsp27 raised BP and the effect was blocked by hepatic vagotomy. Hepatic Fsp27 is actually upregulated in murine obesity models and its knockdown reversed BP elevation. CONCLUSION: The hepatic PPARγ-Fsp27 pathway plays important roles in the development of obesity-related hypertension via afferent vagal signals from the liver.


Asunto(s)
Hipertensión/etiología , Hígado/metabolismo , Obesidad/etiología , PPAR gamma/metabolismo , Proteínas/metabolismo , Nervio Vago/fisiología , Animales , Capsaicina/farmacología , Técnicas de Silenciamiento del Gen , Ratones , Ratones Endogámicos C57BL , Bloqueo Nervioso/métodos , Fármacos del Sistema Sensorial/farmacología , Transducción de Señal/fisiología , Transmisión Sináptica/fisiología , Regulación hacia Arriba , Nervio Vago/cirugía
12.
Mol Cell Endocrinol ; 565: 111887, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-36781118

RESUMEN

Oxysterol-binding protein-like 3 (OSBPL3) plays a key role in the development of fatty liver disease. Herein, we found that OSBPL3 is highly expressed in the fatty liver of humans and mice. Although high expression of Osbpl3 was observed in the fatty liver of type 2 diabetic ob/ob mice, liver-specific Pparg knockout ameliorated this increase in these mice. Moreover, high hepatic Osbpl3 expression was observed in other mice models of fatty liver disease, such as leptin receptor-mutant db/db and alcohol-fed mice. Analysis of the human liver transcriptome data revealed that hepatic OSBPL3 expression is higher in patients with advanced non-alcoholic fatty liver disease (NAFLD) when compared to those with mild NAFLD. Reporter and electrophoretic mobility shift assays showed that PPARγ positively regulates Osbpl3 transcription by binding to the two functional PPARγ-responsive elements present in the 5' upstream region. Overall, our results indicate that Osbpl3 is a novel PPARγ target in the fatty liver.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Oxiesteroles , Animales , Humanos , Ratones , Etanol/metabolismo , Hígado/metabolismo , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Oxiesteroles/metabolismo , PPAR gamma/metabolismo
13.
iScience ; 25(5): 104196, 2022 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-35479397

RESUMEN

Peroxisome proliferator-activated receptor α (PPARA) is a key mediator of lipid metabolism and inflammation. Activation of PPARA in rodents causes hepatocyte proliferation, but the underlying mechanism is poorly understood. This study focused on genes repressed by PPARA and analyzed the mechanism by which PPARA promotes hepatocyte proliferation in mice. Activation of PPARA by agonist treatment was autoregulated, and induced expression of the epigenetic regulator UHRF1 via activation of the newly described PPARA target gene E2f8, which, in turn, regulates Uhrf1. UHRF1 strongly repressed the expression of CDH1 via methylation of the Cdh1 promoter marked with H3K9me3. Repression of CDH1 by PPARA activation was reversed by PPARA deficiency or knockdown of E2F8 or UHRF1. Furthermore, a forced expression of CDH1 inhibited expression of the Wnt signaling target genes such as Myc after PPARA activation, and suppressed hepatocyte hyperproliferation. These results demonstrate that the PPARA-E2F8-UHRF1-CDH1 axis causes epigenetic regulation of hepatocyte proliferation.

14.
Pharmacol Res ; 61(5): 466-72, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20006706

RESUMEN

RhoA protein is involved in the Ca(2+) sensitization of bronchial smooth muscle (BSM) contraction, and an upregulation of RhoA in BSMs has been suggested in allergic bronchial asthma. However, the mechanism of upregulation of RhoA remains poorly understood. In the present study, the transcriptional regulation of human RhoA gene was investigated in cultured human BSM cells stimulated with IL-13 and TNF-alpha, both of which have an ability to upregulate RhoA protein. Luciferase-based assay showed that the RhoA promoter activity was augmented by both IL-13 and TNF-alpha. The deletion studies revealed a significant level of promoter activity between the 112 bp upstream and the transcription start site, which contains the STAT6 (78-70 bp upstream) and NF-kappaB (84-74 bp upstream) binding regions. The promoter activity was also decreased significantly by the mutations of these regions. Thus, the current study for the first time characterized the transcriptional regulation of the human RhoA gene. The findings also suggest that STAT6 and NF-kappaB are important for the upregulation of RhoA in human BSM induced by IL-13 and TNF-alpha, both of which are major cytokines in the pathogenesis of allergic bronchial asthma.


Asunto(s)
Bronquios/metabolismo , Interleucina-13/farmacología , Músculo Liso/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , FN-kappa B/genética , Factor de Transcripción STAT6/genética , Factor de Necrosis Tumoral alfa/farmacología , Proteína de Unión al GTP rhoA/biosíntesis , Western Blotting , Bronquios/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Humanos , Luciferasas/genética , Músculo Liso/citología , Músculo Liso/efectos de los fármacos , Plásmidos/genética , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Elementos de Respuesta/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba/efectos de los fármacos , Proteína de Unión al GTP rhoA/genética
15.
J Pharmacol Sci ; 114(3): 239-47, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20948164

RESUMEN

Airway hyperresponsiveness to nonspecific stimuli is one of the characteristic features of allergic bronchial asthma. An elevated contractility of bronchial smooth muscle has been considered as one of the causes of the airway hyperresponsiveness. The contraction of smooth muscles including airway smooth muscles is mediated by both Ca²+-dependent and Ca²+-independent pathways. The latter Ca²+-independent pathway, termed Ca²+ sensitization, is mainly regulated by a monomeric GTP-binding protein, RhoA, and its downstream target Rho-kinase. In animal models of allergic bronchial asthma, an augmented agonist-induced, RhoA-mediated contraction of bronchial smooth muscle has been suggested. The RhoA/Rho-kinase signaling is now proposed as a novel target for the treatment of airway hyperresponsiveness in asthma. Herein, we will discuss the mechanism of development of bronchial smooth muscle hyperresponsiveness, one of the causes of the airway hyperresponsiveness, based on the recent studies using animal models of allergic bronchial asthma and/or cultured airway smooth muscle cells. The possibility of RhoA as a therapeutic target in asthma, especially airway hyperresponsiveness, will also be described.


Asunto(s)
Asma/tratamiento farmacológico , Asma/metabolismo , Bronquios/fisiopatología , Hiperreactividad Bronquial/tratamiento farmacológico , Músculo Liso/fisiopatología , Proteína de Unión al GTP rhoA/metabolismo , Animales , Hiperreactividad Bronquial/etiología , Hiperreactividad Bronquial/metabolismo , Calcio/metabolismo , Humanos , Ratones , Terapia Molecular Dirigida , Contracción Muscular , Ratas , Sistema Respiratorio/metabolismo , Sistema Respiratorio/fisiopatología , Transducción de Señal , Activación Transcripcional , Regulación hacia Arriba , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/genética
16.
J Pharmacol Sci ; 112(4): 467-72, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20308800

RESUMEN

RhoA upregulation has been suggested in bronchial smooth muscles (BSMs) of asthmatic rats. Here, we cloned/characterized the 5'-promoter region of the rat rhoA. A transcription-initiation site was identified at 66-bp upstream of the reference sequence, GenBank-BC061732. Luciferase assay using interleukin-13 (IL-13)-stimulated cells revealed a significant promoter activity at 238- to 166-bp upstream of the transcription-initiation site, which contains a signal transducer and activation of transcription (STAT) 6-binding region. The IL-13-induced increase in luciferase activity was inhibited by a STAT6 inhibitor, AS1517499, or a Janus kinases (JAKs) inhibitor, JAK Inhibitor-I, but not by tyrphostin-AG490, WHI-P131, or tyrphostin-AG9 (selective JAK2, JAK3, and Tyk2 inhibitors, respectively). Thus, rat BSM rhoA expression may have causal relation to the IL-13-JAK1-STAT6 signaling.


Asunto(s)
Regiones Promotoras Genéticas , Quinasas Asociadas a rho/genética , Animales , Interleucina-13/farmacología , Ratas , Factor de Transcripción STAT6/antagonistas & inhibidores , Factor de Transcripción STAT6/metabolismo
17.
Biol Pharm Bull ; 33(3): 346-50, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20190390

RESUMEN

Fat specific protein 27 (FSP27) was originally isolated by screen for genes specifically expressed in fully differentiated mouse adipocytes. FSP27 and cell death-inducing DFF45-like effector C (CIDEC), the human homologue of FSP27, belong to the CIDE family. The FSP27 in adipocytes was recently reported to be a lipid droplet (LD)-associated protein, that promotes the formation of unilocular LDs. An FSP27 knockout mouse demonstrated lean phenotypes with atrophic adipose tissue as a result of high-energy expenditure; this mouse line was also resistant to diet-induced obesity and insulin resistance. Interestingly, FSP27 was also expressed in the steatoic liver of a type II diabetes model mouse. The expression of FSP27 was markedly decreased in livers lacking the nuclear receptor peroxisome proliferator-activated receptor gamma. Forced expression of FSP27 in hepatocytes in vitro or in vivo led to an increase of LD through increased triglyceride levels. The current status of the physiological roles of FSP27/CIDEC in adipose tissue and liver are discussed along with its significance as a factor involved in the development of metabolic disorders.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , Enfermedades Metabólicas/metabolismo , Proteínas/metabolismo , Animales , Diabetes Mellitus Experimental/metabolismo , Hígado Graso/metabolismo , Regulación de la Expresión Génica , Humanos , Ratones , Obesidad/genética , Obesidad/metabolismo , Orgánulos/metabolismo , PPAR gamma/metabolismo , Proteínas/genética , Triglicéridos/metabolismo
18.
Gene ; 721: 144113, 2019 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-31505214

RESUMEN

Vaspin, initially identified in visceral adipose tissue, is an adipokine, and administration of recombinant vaspin leads to lowering of the endoplasmic reticulum stress which is elevated in obesity or enhancement of insulin sensitivity. CCAAT/enhancer binding protein (C/EBP), as a basic leucine zipper transcription factor, plays a critical role in adipocyte development and glucose and lipid metabolisms in liver. The present study aimed to investigate the effect of C/EBPα on vaspin gene expression. The expression of hepatic vaspin was markedly decreased in liver-specific C/EBPα knockout mice. A reporter assay indicated that two C/EBP-responsive elements (CEBPREs) are necessary for C/EBPα-dependent induction of vaspin promoter activities. Furthermore, electrophoretic mobility shift assay showed that C/EBPα in mouse liver is capable of directly binding the two CEBPREs. These results suggest that C/EBPα positively regulates hepatic vaspin expression through two functional CEBPREs. Thus, vaspin is a novel C/EBPα target gene in the liver.


Asunto(s)
Adipoquinas/biosíntesis , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Regulación de la Expresión Génica/fisiología , Hígado/metabolismo , Elementos de Respuesta/fisiología , Serpinas/biosíntesis , Adipoquinas/genética , Animales , Ratones , Ratones Noqueados , Serpinas/genética
19.
Mol Cell Endocrinol ; 474: 48-56, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-29454584

RESUMEN

Fat-specific protein 27 (FSP27) is highly expressed in the fatty liver of genetically obese ob/ob mice and promotes hepatic triglyceride (TG) accumulation. The nuclear hormone receptor liver X receptor α (LXRα) also plays a critical role in the control of TG levels in the liver. The present study demonstrated transcriptional regulation of Fsp27a and Fsp27b genes by LXRα. Treatment with the LXR ligand T0901317 markedly increased Fsp27a and Fsp27b mRNAs in wild-type C57BL/6J and ob/ob mouse livers. A reporter assay indicated that two LXR-responsive elements (LXREs) are necessary for LXRα-dependent induction of Fsp27a and Fsp27b promoter activities. Furthermore, the LXRα/retinoid X receptor α complex is capable of directly binding to the two LXREs both in vitro and in vivo. These results suggest that LXRα positively regulates Fsp27a and Fsp27b expression through two functional LXREs. Fsp27a/b are novel LXR target genes in the ob/ob fatty liver.


Asunto(s)
Receptores X del Hígado/metabolismo , Proteínas/genética , Animales , Secuencia de Bases , Exones/genética , Hígado Graso/metabolismo , Hígado Graso/patología , Regulación de la Expresión Génica , Células HEK293 , Humanos , Hidrocarburos Fluorados , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones Endogámicos C57BL , Ratones Obesos , Regiones Promotoras Genéticas , Unión Proteica/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Proteínas/metabolismo , Elementos de Respuesta/genética , Sulfonamidas
20.
J Clin Invest ; 111(5): 737-47, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12618528

RESUMEN

To elucidate the function of PPARgamma in leptin-deficient mouse (ob/ob) liver, a PPARgamma liver-null mouse on an ob/ob background, ob/ob-PPARgamma(fl/fl)AlbCre(+), was produced using a floxed PPARgamma allele, PPARgamma(fl/fl), and Cre recombinase under control of the albumin promoter (AlbCre). The liver of ob/ob-PPARgamma(fl/fl)AlbCre(+) mice had a deletion of exon 2 and a corresponding loss of full-length PPARgamma mRNA and protein. The PPARgamma-deficient liver in ob/ob mice was smaller and had a dramatically decreased triglyceride (TG) content compared with equivalent mice lacking the AlbCre transgene (ob/ob-PPARgamma(fl/fl)AlbCre(-)). Messenger RNA levels of the hepatic lipogenic genes, fatty acid synthase, acetyl-CoA carboxylase, and stearoyl-CoA desaturase-1, were reduced in ob/ob-PPARgamma(fl/fl)AlbCre(+) mice, and the levels of serum TG and FFA in ob/ob-PPARgamma(fl/fl)AlbCre(+) mice were significantly higher than in the control ob/ob-PPARgamma(fl/fl)AlbCre(-) mice. Rosiglitazone treatment exacerbated the fatty liver in ob/ob-PPARgamma(fl/fl)AlbCre(-) mice compared with livers from nonobese Cre(-) mice; there was no effect of rosiglitazone in ob/ob-PPARgamma(fl/fl)AlbCre(+) mice. The deficiency of hepatic PPARgamma further aggravated the severity of diabetes in ob/ob mice due to decreased insulin sensitivity in muscle and fat. These data indicate that hepatic PPARgamma plays a critical role in the regulation of TG content and in the homeostasis of blood glucose and insulin resistance in steatotic diabetic mice.


Asunto(s)
Hígado Graso/etiología , Hiperglucemia/etiología , Resistencia a la Insulina , Leptina/deficiencia , Hígado/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Tiazolidinedionas , Factores de Transcripción/fisiología , Animales , Glucemia/análisis , Ácidos Grasos no Esterificados/sangre , Hígado Graso/terapia , Lipoproteína Lipasa/metabolismo , Lipoproteínas VLDL/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Receptores de LDL/metabolismo , Receptores de Leptina , Rosiglitazona , Tiazoles/uso terapéutico , Triglicéridos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA