Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
1.
Biopharm Drug Dispos ; 39(2): 99-115, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29243851

RESUMEN

The vitamin D-deficient model, established in the C57BL/6 mouse after 8 weeks of feeding vitamin D-deficient diets in the absence or presence of added calcium, was found associated with elevated levels of plasma parathyroid hormone (PTH) and plasma and liver cholesterol, and a reduction in cholesterol 7α-hydroxylase (Cyp7a1, rate-limiting enzyme for cholesterol metabolism) and renal Oat3 mRNA/protein expression levels. However, there was no change in plasma calcium and phosphate levels. Appraisal of the liver revealed an up-regulation of mRNA expressions of the small heterodimer partner (Shp) and attenuation of Cyp7a1, which contributed to hypercholesterolemia in vitamin D-deficiency. When vitamin D-sufficient or D-deficient mice were further rendered hypercholesterolemic with 3 weeks of feeding the respective, high fat/high cholesterol (HF/HC) diets, treatment with 1α,25-dihydroxyvitamin D3 [1,25(OH)2 D3 ], active vitamin D receptor (VDR) ligand, or vitamin D (cholecalciferol) to HF/HC vitamin D-deficient mice lowered the cholesterol back to baseline levels. Cholecalciferol treatment partially restored renal Oat3 mRNA/protein expression back to that of vitamin D-sufficient mice. When the protein expression of protein kinase C (PKC), a known, negative regulator of Oat3, was examined in murine kidney, no difference in PKC expression was observed for any of the diets with/without 1,25(OH)2 D3 /cholecalciferol treatment, inferring that VDR regulation of renal Oat3 did not involve PKC in mice. As expected, plasma calcium levels were not elevated by cholecalciferol treatment of vitamin D-deficient mice, while 1,25(OH)2 D3 treatment led to hypercalcemia. In conclusion, vitamin D-deficiency resulted in down-regulation of liver Cyp7a1 and renal Oat3, conditions that are alleviated upon replenishment of cholecalciferol.


Asunto(s)
Colesterol 7-alfa-Hidroxilasa/biosíntesis , Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Riñón/metabolismo , Hígado/metabolismo , Transportadores de Anión Orgánico Sodio-Independiente/biosíntesis , Deficiencia de Vitamina D/enzimología , Deficiencia de Vitamina D/genética , Animales , Ácidos y Sales Biliares/metabolismo , Calcifediol/sangre , Calcio/sangre , Calcio/farmacología , Colecalciferol/farmacología , Colesterol/sangre , Colesterol/metabolismo , Colesterol 7-alfa-Hidroxilasa/sangre , Colesterol 7-alfa-Hidroxilasa/genética , Dieta/métodos , Vesícula Biliar/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Transportadores de Anión Orgánico Sodio-Independiente/genética , Hormona Paratiroidea/sangre , Vitamina D/análogos & derivados , Vitamina D/sangre
2.
J Pharmacol Exp Ther ; 362(3): 385-394, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28645914

RESUMEN

The interference of bile acid secretion through bile salt export pump (BSEP) inhibition is one of the mechanisms for troglitazone (TGZ)-induced hepatotoxicity. Here, we investigated the impact of single or repeated oral doses of TGZ (200 mg/kg/day, 7 days) on bile acid homoeostasis in wild-type (WT) and Bsep knockout (KO) rats. Following oral doses, plasma exposures of TGZ were not different between WT and KO rats, and were similar on day 1 and day 7. However, plasma exposures of the major metabolite, troglitazone sulfate (TS), in KO rats were 7.6- and 9.3-fold lower than in WT on day 1 and day 7, respectively, due to increased TS biliary excretion. With Bsep KO, the mRNA levels of multidrug resistance-associated protein 2 (Mrp2), Mrp3, Mrp4, Mdr1, breast cancer resistance protein (Bcrp), sodium taurocholate cotransporting polypeptide, small heterodimer partner, and Sult2A1 were significantly altered in KO rats. Following seven daily TGZ treatments, Cyp7A1 was significantly increased in both WT and KO rats. In the vehicle groups, plasma exposures of individual bile acids demonstrated variable changes in KO rats as compared with WT. WT rats dosed with TGZ showed an increase of many bile acid species in plasma on day 1, suggesting the inhibition of Bsep. Conversely, these changes returned to base levels on day 7. In KO rats, alterations of most bile acids were observed after seven doses of TGZ. Collectively, bile acid homeostasis in rats was regulated through bile acid synthesis and transport in response to Bsep deficiency and TGZ inhibition. Additionally, our study is the first to demonstrate that repeated TGZ doses can upregulate Cyp7A1 in rats.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Ácidos y Sales Biliares/metabolismo , Cromanos/farmacología , Homeostasis/efectos de los fármacos , Homeostasis/genética , Hipoglucemiantes/farmacología , Tiazolidinedionas/farmacología , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP , Animales , Bilis/metabolismo , Colesterol 7-alfa-Hidroxilasa/biosíntesis , Colesterol 7-alfa-Hidroxilasa/genética , Técnicas de Inactivación de Genes , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Troglitazona , Regulación hacia Arriba/efectos de los fármacos
3.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(12): 1469-1480, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28888833

RESUMEN

Iron depletion (ID) has been shown to induce the liver expression of Cyp7a1, the rate-limiting enzyme initiating conversion of cholesterol to bile acids (BA), although the effect on bile acids metabolism and bile production is unknown. Therefore, we investigated changes in bile secretion and BA synthesis during diet-induced iron depletion (ID) in rats. ID increased bile flow along with augmented biliary excretion of bile acids, glutathione, cholesterol and phospholipids. Accordingly, we found transcriptional upregulation of the Cyp7a1, Cyp8b1, and Cyp27a1 BA synthetic enzymes, as well as induction of the Abcg5/8 cholesterol transporters in ID rat livers. In contrast, intravenous infusion of 3H-taurocholate failed to elicit any difference in biliary secretion of this compound in the ID rats. This corresponded with unchanged expression of canalicular rate-limiting transporters for BA as well as glutathione. We also observed that ID substantially changed the spectrum of BA in bile and decreased plasma concentrations of BA and cholesterol. Experiments with differentiated human hepatic HepaRG cells confirmed human CYP7A1 orthologue upregulation resulting from reduced iron concentrations. Results employing a luciferase reporter gene assay suggest that the transcriptional activation of the CYP7A1 promoter under ID conditions works independent of farnesoid X (FXR), pregnane X (PXR) and liver X (LXRα) receptors activation. It can be concluded that this study characterizes the molecular mechanisms of modified bile production as well as cholesterol as along with BA homeostasis during ID. We propose complex upregulation of BA synthesis, and biliary cholesterol secretion as the key factors affected by ID.


Asunto(s)
Ácidos y Sales Biliares/biosíntesis , Colesterol/metabolismo , Glutatión/metabolismo , Deficiencias de Hierro , Animales , Línea Celular , Colestanotriol 26-Monooxigenasa/biosíntesis , Colesterol 7-alfa-Hidroxilasa/biosíntesis , Humanos , Masculino , Ratas , Ratas Wistar , Esteroide 12-alfa-Hidroxilasa/biosíntesis
4.
Biochem J ; 470(2): 181-93, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26348907

RESUMEN

Bile acids are primarily synthesized from cholesterol in the liver and have important roles in dietary lipid absorption and cholesterol homoeostasis. Detailed roles of the orphan nuclear receptors regulating cholesterol 7α-hydroxylase (CYP7A1), the rate-limiting enzyme in bile acid synthesis, have not yet been fully elucidated. In the present study, we report that oestrogen-related receptor γ (ERRγ) is a novel transcriptional regulator of CYP7A1 expression. Activation of cannabinoid receptor type 1 (CB1 receptor) signalling induced ERRγ-mediated transcription of the CYP7A1 gene. Overexpression of ERRγ increased CYP7A1 expression in vitro and in vivo, whereas knockdown of ERRγ attenuated CYP7A1 expression. Deletion analysis of the CYP7A1 gene promoter and a ChIP assay revealed an ERRγ-binding site on the CYP7A1 gene promoter. Small heterodimer partner (SHP) inhibited the transcriptional activity of ERRγ and thus regulated CYP7A1 expression. Overexpression of ERRγ led to increased bile acid levels, whereas an inverse agonist of ERRγ, GSK5182, reduced CYP7A1 expression and bile acid synthesis. Finally, GSK5182 significantly reduced hepatic CB1 receptor-mediated induction of CYP7A1 expression and bile acid synthesis in alcohol-treated mice. These results provide the molecular mechanism linking ERRγ and bile acid metabolism.


Asunto(s)
Colesterol 7-alfa-Hidroxilasa/biosíntesis , Hígado/metabolismo , Receptor Cannabinoide CB1/metabolismo , Receptores de Estrógenos/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Células Cultivadas , Colesterol 7-alfa-Hidroxilasa/genética , Agonismo Inverso de Drogas , Etanol/farmacología , Expresión Génica , Glicéridos/farmacología , Células HEK293 , Hepatocitos/metabolismo , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Estrógenos/genética , Transcripción Genética
5.
BMC Dev Biol ; 15: 47, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26627605

RESUMEN

BACKGROUND: Biliary atresia (BA) is a human infant disease with inflammatory fibrous obstructions in the bile ducts and is the most common cause for pediatric liver transplantation. In contrast, the sea lamprey undergoes developmental BA with transient cholestasis and fibrosis during metamorphosis, but emerges as a fecund adult. Therefore, sea lamprey liver metamorphosis may serve as an etiological model for human BA and provide pivotal information for hepatobiliary transformation and possible therapeutics. RESULTS: We hypothesized that liver metamorphosis in sea lamprey is due to transcriptional reprogramming that dictates cellular remodeling during metamorphosis. We determined global gene expressions in liver at several metamorphic landmark stages by integrating mRNA-Seq and gene ontology analyses, and validated the results with real-time quantitative PCR, histological and immunohistochemical staining. These analyses revealed that gene expressions of protein folding chaperones, membrane transporters and extracellular matrices were altered and shifted during liver metamorphosis. HSP90, important in protein folding and invertebrate metamorphosis, was identified as a candidate key factor during liver metamorphosis in sea lamprey. Blocking HSP90 with geldanamycin facilitated liver metamorphosis and decreased the gene expressions of the rate limiting enzyme for cholesterol biosynthesis, HMGCoA reductase (hmgcr), and bile acid biosynthesis, cyp7a1. Injection of hsp90 siRNA for 4 days altered gene expressions of met, hmgcr, cyp27a1, and slc10a1. Bile acid concentrations were increased while bile duct and gall bladder degeneration was facilitated and synchronized after hsp90 siRNA injection. CONCLUSIONS: HSP90 appears to play crucial roles in hepatobiliary transformation during sea lamprey metamorphosis. Sea lamprey is a useful animal model to study postembryonic development and mechanisms for hsp90-induced hepatobiliary transformation.


Asunto(s)
Conductos Biliares Intrahepáticos/embriología , Atresia Biliar/embriología , Colestasis/embriología , Proteínas HSP90 de Choque Térmico/genética , Metamorfosis Biológica/fisiología , Petromyzon/embriología , Animales , Benzoquinonas/farmacología , Ácidos y Sales Biliares/metabolismo , Conductos Biliares Intrahepáticos/patología , Atresia Biliar/patología , Colesterol 7-alfa-Hidroxilasa/biosíntesis , Colesterol 7-alfa-Hidroxilasa/genética , Inhibidores Enzimáticos/farmacología , Matriz Extracelular/metabolismo , Fibrosis/embriología , Vesícula Biliar/embriología , Vesícula Biliar/patología , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Hidroximetilglutaril-CoA Reductasas/biosíntesis , Hidroximetilglutaril-CoA Reductasas/genética , Lactamas Macrocíclicas/farmacología , Hígado/embriología , Transportadores de Anión Orgánico Sodio-Dependiente/biosíntesis , Proteínas Proto-Oncogénicas c-met/biosíntesis , Interferencia de ARN , ARN Interferente Pequeño/genética , Simportadores/biosíntesis
6.
J Lipid Res ; 55(6): 1165-72, 2014 06.
Artículo en Inglés | MEDLINE | ID: mdl-24771866

RESUMEN

A new mechanism for formation of 7-ketocholesterol was recently described involving cytochrome P-450 (CYP)7A1-catalyzed conversion of 7-dehydrocholesterol into 7-ketocholesterol with cholesterol-7,8-epoxide as a side product. Some patients with cerebrotendinous xanthomatosis (CTX) and all patients with Smith-Lemli-Opitz syndrome (SLO) have markedly increased levels of 7-dehydrocholesterol in plasma and tissues. In addition, the former patients have markedly upregulated CYP7A1. We hypothesized that these patients may produce 7-ketocholesterol from 7-dehydrocholesterol with formation of cholesterol-7,8-epoxide as a side product. In accord with this hypothesis, two patients with CTX were found to have increased levels of 7-ketocholesterol and 7-dehydrocholesterol, as well as a significant level of cholesterol-7,8-epoxide. The latter steroid was not detectable in plasma from healthy volunteers. Downregulation of CYP7A1 activity by treatment with chenodeoxycholic acid reduced the levels of 7-ketocholesterol in parallel with decreased levels of 7-dehydrocholesterol and cholesterol-7,8-epoxide. Three patients with SLO were found to have markedly elevated levels of 7-ketocholesterol as well as high levels of cholesterol-7,8-epoxide. The results support the hypothesis that 7-dehydrocholesterol is a precursor to 7-ketocholesterol in SLO and some patients with CTX.


Asunto(s)
Deshidrocolesteroles/sangre , Cetocolesteroles/sangre , Síndrome de Smith-Lemli-Opitz/sangre , Xantomatosis Cerebrotendinosa/sangre , Adolescente , Adulto , Niño , Colesterol 7-alfa-Hidroxilasa/biosíntesis , Colesterol 7-alfa-Hidroxilasa/genética , Regulación hacia Abajo , Femenino , Regulación Enzimológica de la Expresión Génica , Humanos , Cetocolesteroles/genética , Síndrome de Smith-Lemli-Opitz/genética , Síndrome de Smith-Lemli-Opitz/patología , Xantomatosis Cerebrotendinosa/genética , Xantomatosis Cerebrotendinosa/patología
7.
Am J Physiol Gastrointest Liver Physiol ; 307(4): G479-86, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24994853

RESUMEN

It was proposed that CYP7A1 expression is suppressed through the gut-hepatic signaling pathway fibroblast growth factor (FGF) 15/19-fibroblast growth factor receptor 4, which is initiated by activation of farnesoid X receptor in the intestine rather than in the liver. The present study tested whether portal bile acid flux alone without ileal FGF19 could downregulate CYP7A1 expression in rabbits. A rabbit model was developed by infusing glycodeoxycholic acid (GDCA) through the splenic vein to bypass ileal FGF19. Study was conducted in four groups of rabbits: control; bile fistula + bovine serum albumin solution perfusion (BF); BF + GDCA (by portal perfusion); and BF + GDCA-f (by femoral perfusion). Compared with only BF, BF + GDCA (6 h portal perfusion) suppressed CYP7A1 mRNA, whereas BF + GDCA-f (via femoral vein) with the same perfusion rate of GDCA did not show inhibitory effects. Meanwhile, there was a decrease in ileal FGF19 expression and portal FGF19 protein levels, but an equivalent increase in biliary bile acid outputs in both GDCA perfusion groups. This study demonstrated that portal bile acid flux alone downregulated CYP7A1 expression with diminished FGF19 expression and protein levels, whereas the same bile acid flux reaching the liver through the hepatic artery via femoral vein had no inhibitory effect on CYP7A1. We propose that bile acid flux through the portal venous system may be a kind of "intestinal factor" that suppresses CYP7A1 expression.


Asunto(s)
Colesterol 7-alfa-Hidroxilasa/biosíntesis , Ácido Glicodesoxicólico/farmacología , Íleon/metabolismo , Animales , Fístula Biliar , Regulación hacia Abajo , Factores de Crecimiento de Fibroblastos/metabolismo , Vena Porta , Conejos
9.
J Biol Chem ; 287(49): 41334-41, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23038264

RESUMEN

Fibroblast growth factor 19 (FGF19) is a postprandial enterokine induced by the nuclear bile acid receptor, FXR, in ileum. FGF19 inhibits bile acid synthesis in liver through transcriptional repression of cholesterol 7α-hydroxylase (CYP7A1) via a mechanism involving the nuclear receptor SHP. Here, in a series of loss-of-function studies, we show that the nuclear receptors HNF4α and LRH-1 have dual roles in regulating Cyp7a1 in vivo. First, they cooperate in maintaining basal Cyp7a1 expression. Second, they enable SHP binding to the Cyp7a1 promoter and facilitate FGF19-mediated repression of bile acid synthesis. HNF4α and LRH-1 promote active transcription histone marks on the Cyp7a1 promoter that are reversed by FGF19 in a SHP-dependent manner. These findings demonstrate that both HNF4α and LRH-1 are important regulators of Cyp7a1 transcription in vivo.


Asunto(s)
Colesterol 7-alfa-Hidroxilasa/biosíntesis , Regulación de la Expresión Génica , Factor Nuclear 4 del Hepatocito/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Ácidos y Sales Biliares/metabolismo , Sitios de Unión , Factores de Crecimiento de Fibroblastos/metabolismo , Células HEK293 , Histonas/metabolismo , Humanos , Hígado/metabolismo , Ratones , Regiones Promotoras Genéticas , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transcripción Genética
10.
Br J Nutr ; 110(11): 1958-67, 2013 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-23631850

RESUMEN

Replacing dietary fishmeal (FM) and fish oil (FO) with plant ingredients in Atlantic salmon (Salmo salar L.) diets decreases dietary cholesterol and introduces phytosterols. The aim of the present study was to assess the effect of dietary sterol composition on cholesterol metabolism in Atlantic salmon. For this purpose, two dietary trials were performed, in which Atlantic salmon were fed either 100 % FM and FO (FM-FO) diet or one of the three diets with either high (80 %) or medium (40 %) plant protein (PP) and a high (70 %) or medium (35 %) vegetable oil (VO) blend (trial 1); or 70 % PP with either 100 % FO or 80 % of the FO replaced with olive, rapeseed or soyabean oil (trial 2). Replacing ≥ 70 % of FM with PP and ≥ 70 % of FO with either a VO blend or rapeseed oil increased plasma and liver TAG concentrations. These diets contained high levels of phytosterols and low levels of cholesterol. Fish fed low-cholesterol diets, but with less phytosterols, exhibited an increased expression of genes encoding proteins involved in cholesterol uptake and synthesis. The expression of these genes was, however, partially inhibited in rapeseed oil-fed fish possibly due to the high dietary and tissue phytosterol:cholesterol ratio. Atlantic salmon tissue and plasma cholesterol concentrations were maintained stable independent of the dietary sterol content.


Asunto(s)
Colesterol/metabolismo , Dieta/veterinaria , Hígado/metabolismo , Fitosteroles/metabolismo , Salmo salar/metabolismo , Triglicéridos/metabolismo , Animales , Acuicultura , Colesterol/administración & dosificación , Colesterol/sangre , Colesterol 7-alfa-Hidroxilasa/biosíntesis , Colesterol 7-alfa-Hidroxilasa/genética , Colesterol 7-alfa-Hidroxilasa/metabolismo , Dieta/efectos adversos , Proteínas en la Dieta/administración & dosificación , Proteínas en la Dieta/efectos adversos , Proteínas en la Dieta/metabolismo , Ácidos Grasos Monoinsaturados , Proteínas de Peces/biosíntesis , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hígado/enzimología , Hígado/crecimiento & desarrollo , Receptores X del Hígado , Aceite de Oliva , Receptores Nucleares Huérfanos/biosíntesis , Receptores Nucleares Huérfanos/genética , Receptores Nucleares Huérfanos/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/biosíntesis , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Fitosteroles/administración & dosificación , Fitosteroles/efectos adversos , Aceites de Plantas/administración & dosificación , Aceites de Plantas/efectos adversos , Aceites de Plantas/metabolismo , Proteínas de Plantas/administración & dosificación , Proteínas de Plantas/efectos adversos , Proteínas de Plantas/metabolismo , Aceite de Brassica napus , Salmo salar/sangre , Salmo salar/crecimiento & desarrollo , Aceite de Soja/administración & dosificación , Aceite de Soja/efectos adversos , Aceite de Soja/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/biosíntesis , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Triglicéridos/administración & dosificación , Triglicéridos/sangre , Aumento de Peso
11.
Biosci Biotechnol Biochem ; 77(6): 1326-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23748774

RESUMEN

Dietary licorice fravonoid oil (LFO) significantly decreased hepatic cholesterol and plasma lipoprotein cholesterol levels in high-fat diet rats. It significantly suppressed hydroxymethylglutaryl-CoA synthase activity and increased cholesterol 7α-hydroxylase activity. The low density lipoprotein receptor mRNA level was significantly increased by LFO. These results suggest that dietary LFO improves cholesterol metabolism in obese animals.


Asunto(s)
Colesterol/metabolismo , Flavonoides/administración & dosificación , Lipoproteínas/sangre , Obesidad/sangre , Aceites de Plantas/administración & dosificación , Animales , Colesterol/sangre , Colesterol 7-alfa-Hidroxilasa/biosíntesis , Dietoterapia , Dieta Alta en Grasa , Regulación de la Expresión Génica/efectos de los fármacos , Glycyrrhiza/química , Hidroximetilglutaril-CoA Sintasa/biosíntesis , Metabolismo de los Lípidos , Masculino , Obesidad/tratamiento farmacológico , Ratas
12.
Endocr J ; 60(10): 1171-7, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23912975

RESUMEN

Small heterodimer partner (SHP) is involved in bile, lipid, and glucose metabolism. The aim of this study was to investigate the effect of SHP on the development of atherosclerosis. Apolipoprotein E knockout (ApoE-/-) mice were crossed with SHP knockout (SHP-/-) mice to generate double knockout (ApoE-/-SHP-/-) mice. ApoE-/- and ApoE-/-SHP-/- male mice were fed a western diet for 20 weeks. Body weight in ApoE-/-SHP-/) mice was significantly lower than that in ApoE-/- mice (37±1 g vs. 42±1 g, p<0.01). Loss of SHP in ApoE-/- mice decreased the size of adipocytes in white adipose tissue and reduced lipid accumulation in the liver. Glucose intolerance was improved in ApoE-/-SHP-/- mice as compared with ApoE-/- mice (p<0.01). There was no statistical difference in non-high density lipoprotein cholesterol levels between ApoE-/-SHP-/- mice and ApoE-/- mice despite an increase of cholesterol 7α-hydroxylase expression in the liver. The proportion of atherosclerotic lesions in the aorta was significantly lower in ApoE-/-SHP-/- mice than in ApoE-/- mice (2.8±2.0% vs. 9.1±1.9%, p<0.01). In conclusion, loss of SHP function can prevent atherosclerosis, and resistance to diet-induced obesity is the primary factor contributing to this protective effect.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/prevención & control , Receptores Citoplasmáticos y Nucleares/deficiencia , Animales , Aorta/patología , Aterosclerosis/patología , Colesterol 7-alfa-Hidroxilasa/biosíntesis , Dieta/efectos adversos , Resistencia a la Insulina , Hígado/enzimología , Masculino , Ratones , Ratones Noqueados , Obesidad/prevención & control , Aumento de Peso/efectos de los fármacos
13.
Hepatology ; 53(3): 996-1006, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21319191

RESUMEN

UNLABELLED: We reported previously that mice overexpressing cytochrome P450 7a1 (Cyp7a1; Cyp7a1-tg mice) are protected against high fat diet-induced hypercholesterolemia, obesity, and insulin resistance. Here, we investigated the underlying mechanism of bile acid signaling in maintaining cholesterol homeostasis in Cyp7a1-tg mice. Cyp7a1-tg mice had two-fold higher Cyp7a1 activity and bile acid pool than did wild-type mice. Gallbladder bile acid composition changed from predominantly cholic acid (57%) in wild-type to chenodeoxycholic acid (54%) in Cyp7a1-tg mice. Cyp7a1-tg mice had higher biliary and fecal cholesterol and bile acid secretion rates than did wild-type mice. Surprisingly, hepatic de novo cholesterol synthesis was markedly induced in Cyp7a1-tg mice but intestine fractional cholesterol absorption in Cyp7a1-tg mice remained the same as wild-type mice despite the presence of increased intestine bile acids. Interestingly, hepatic but not intestinal expression of several cholesterol (adenosine triphosphate-binding cassette G5/G8 [ABCG5/G8], scavenger receptor class B, member 1) and bile acid (ABCB11) transporters were significantly induced in Cyp7a1-tg mice. Treatment of mouse or human hepatocytes with a farnesoid X receptor (FXR) agonist GW4064 or bile acids induced hepatic Abcg5/g8 expression. A functional FXR binding site was identified in the Abcg5 gene promoter. Study of tissue-specific Fxr knockout mice demonstrated that loss of the Fxr gene in the liver attenuated bile acid induction of hepatic Abcg5/g8 and gallbladder cholesterol content, suggesting a role of FXR in the regulation of cholesterol transport. CONCLUSION: This study revealed a new mechanism by which increased Cyp7a1 activity expands the hydrophobic bile acid pool, stimulating hepatic cholesterol synthesis and biliary cholesterol secretion without increasing intestinal cholesterol absorption. This study demonstrated that Cyp7a1 plays a critical role in maintaining cholesterol homeostasis and underscores the importance of bile acid signaling in regulating overall cholesterol homeostasis.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Colesterol 7-alfa-Hidroxilasa/biosíntesis , Colesterol/metabolismo , Hígado/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 5 , Transportador de Casete de Unión a ATP, Subfamilia G, Miembro 8 , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Hepatocitos/efectos de los fármacos , Homeostasis , Humanos , Isoxazoles/farmacología , Lipoproteínas/metabolismo , Ratones , Ratones Noqueados , Receptores Citoplasmáticos y Nucleares/agonistas
14.
J Lipid Res ; 52(2): 289-98, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21097822

RESUMEN

Cholesterol 7α-hydroxylase (CYP7A1) encodes for the rate-limiting step in the conversion of cholesterol to bile acids in the liver. In response to acute cholesterol feeding, mice upregulate CYP7A1 via stimulation of the liver X receptor (LXR) α. However, the effect of a chronic high-cholesterol diet on hepatic CYP7A1 expression in mice is unknown. We demonstrate that chronic cholesterol feeding (0.2% or 1.25% w/w cholesterol for 12 weeks) in FVB/NJ mice results in a >60% suppression of hepatic CYP7A1 expression associated with a >2-fold increase in hepatic cholesterol content. In contrast, acute cholesterol feeding induces a >3-fold upregulation of hepatic CYP7A1 expression. We show that chronic, but not acute, cholesterol feeding increases the expression of hepatic inflammatory cytokines, tumor necrosis factor (TNF)α, and interleukin (IL)-1ß, which are known to suppress hepatic CYP7A1 expression. Chronic cholesterol feeding also results in activation of the mitogen activated protein (MAP) kinases, c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK). Furthermore, we demonstrate in vitro that suppression of CYP7A1 by TNFα and IL-1ß is dependent on JNK and ERK signaling. We conclude that chronic high-cholesterol feeding suppresses CYP7A1 expression in mice. We propose that chronic cholesterol feeding induces inflammatory cytokine activation and liver damage, which leads to suppression of CYP7A1 via activation of JNK and ERK signaling pathways.


Asunto(s)
Colesterol 7-alfa-Hidroxilasa/biosíntesis , Colesterol en la Dieta/administración & dosificación , Hígado/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Colesterol/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factores de Crecimiento de Fibroblastos/biosíntesis , Células Hep G2 , Humanos , Interleucina-1beta/biosíntesis , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Hígado/efectos de los fármacos , Masculino , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
15.
Hepatology ; 52(2): 678-90, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20623580

RESUMEN

UNLABELLED: Cholesterol 7alpha-hydroxylase (CYP7A1) is the rate-limiting enzyme in the bile acid biosynthetic pathway that converts cholesterol into bile acids in the liver. Recent studies have shown that bile acids may play an important role in maintaining lipid, glucose, and energy homeostasis. However, the role of CYP7A1 in the development of obesity and diabetes is currently unclear. In this study, we demonstrated that transgenic mice overexpressing Cyp7a1 in the liver [i.e., Cyp7a1 transgenic (Cyp7a1-tg) mice] were resistant to high-fat diet (HFD)-induced obesity, fatty liver, and insulin resistance. Cyp7a1-tg mice showed increased hepatic cholesterol catabolism and an increased bile acid pool. Cyp7a1-tg mice had increased secretion of hepatic very low density lipoprotein but maintained plasma triglyceride homeostasis. Gene expression analysis showed that the hepatic messenger RNA expression levels of several critical lipogenic and gluconeogenic genes were significantly decreased in HFD-fed Cyp7a1-tg mice. HFD-fed Cyp7a1-tg mice had increased whole body energy expenditure and induction of fatty acid oxidation genes in the brown adipose tissue. CONCLUSION: This study shows that Cyp7a1 plays a critical role in maintaining whole body lipid, glucose, and energy homeostasis. The induction of CYP7A1 expression with the expansion of the hydrophobic bile acid pool may be a potential therapeutic strategy for treating metabolic disorders such as fatty liver diseases, obesity, and diabetes in humans.


Asunto(s)
Colesterol 7-alfa-Hidroxilasa/biosíntesis , Grasas de la Dieta/administración & dosificación , Resistencia a la Insulina , Hígado/metabolismo , Obesidad/metabolismo , Animales , Homeostasis , Metabolismo de los Lípidos , Lipoproteínas VLDL/metabolismo , Ratones , Ratones Transgénicos
16.
Hepatology ; 51(1): 277-85, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19957370

RESUMEN

UNLABELLED: Cholesterol 7alpha-hydroxylase (Cyp7a1) and the bile acid pool size are increased 2 to 3-fold in lactating postpartum rats. We investigated the interaction of nuclear receptors with the Cyp7a1 proximal promoter and the expression of regulatory signaling pathways in postpartum rats at day 10 (PPd10) versus female controls to identify the mechanisms of increased expression of Cyp7a1, which is maximal at 16 hours. Liver X receptor (LXRalpha) and RNA polymerase II (RNA Pol II) recruitment to Cyp7a1 chromatin were increased 1.5- and 2.5-fold, respectively, at 16 hours on PPd10. Expression of nuclear receptors farnesoid X receptor (FXR), LXRalpha, liver receptor homolog (LRH-1), hepatocyte nuclear factor 4alpha (HNF4alpha), and short heterodimer partner (SHP) messenger RNA (mRNA) and coactivator peroxisome proliferators-activated receptor gamma coactivator-1alpha (PGC-1alpha) mRNA was unchanged in PPd10 versus controls at 16 hours, whereas chicken ovalbumin upstream transcription factor II (COUP-TFII) was decreased 40% at 16 hours. Investigation of a repressive signaling pathway, the c-Jun-N-terminal kinase (JNK) signaling pathway in PPd10 versus controls, showed decreased mRNA expression of hepatocyte growth factor (HGF; decreased 60% at 16 hours) and tyrosine kinase receptor c-Met (decreased 44%-50% at 16 hours), but these were not accompanied by decreased expression of phosphorylated c-Jun. Importantly, expression of fibroblast growth factor 15 (FGF15) mRNA in the ileum was decreased 70% in PPd10 versus controls, whereas phosphorylated mitogen-activated protein kinase/extracellular signal-regulated kinase 1/2 (Erk1/2) protein expression in liver was decreased 88% at 16 hours. CONCLUSION: The increased recruitment of LXRalpha, a Cyp7a1 stimulatory pathway, and decreased expression of FGF15 and phosphorylated Erk1/2, a Cyp7a1 repressive pathway, combined to increase Cyp7a1 expression during lactation.


Asunto(s)
Ácidos y Sales Biliares/fisiología , Colesterol 7-alfa-Hidroxilasa/biosíntesis , Regulación Enzimológica de la Expresión Génica , Lactancia/fisiología , Animales , Factor de Transcripción COUP II/metabolismo , Ritmo Circadiano , Femenino , Factor de Crecimiento de Hepatocito/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Receptores X del Hígado , MAP Quinasa Quinasa 2/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Receptores Nucleares Huérfanos/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Periodo Posparto , Regiones Promotoras Genéticas/fisiología , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas de Unión al ARN/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/metabolismo
17.
Biol Pharm Bull ; 34(10): 1644-7, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21963511

RESUMEN

Our previous study using interleukin-1α/ß-knockout (IL-1-KO) and wild-type (WT) mice demonstrated that IL-1 acts as a positive factor for constitutive gene expression of hepatic cytochrome P4507a1 (Cyp7a1). In this study, to clarify the role of IL-1 in the expression of the hepatic Cyp7a1 gene, we focused on Cyp7a1 transcriptional regulators such as α-fetoprotein transcription factor (FTF), liver X receptor α (LXRα), hepatocyte nuclear factor 4α (HNF4α) and small heterodimer partner (SHP) and examined the effects of IL-1 on their gene expression by real-time reverse-transcription polymerase chain reaction using IL-1-KO and WT mice. We observed no significant differences between sex-matched IL-1-KO and WT mice with regard to gene expression levels of FTF, LXRα, and HNF4α, all of which are positive transcriptional regulators for the Cyp7a1 gene. However, interindividual differences in hepatic FTF and LXRα expression were closely dependent on the gene expression level(s) of hepatic IL-1 and tumor necrosis factor-α (TNF-α), while interindividual differences in hepatic HNF4α were clearly correlated with the expression of IL-1, but not TNF-α. In contrast, the gene expression level of SHP, which is a negative transcriptional regulator of the Cyp7a1 gene through inhibition of FTF function, was higher in IL-1-KO mice than in sex-matched WT mice. These findings demonstrate that, like TNF-α, IL-1 positively controls the gene expression of Cyp7a1 transcriptional upregulators but, in contrast to the previously reported action of TNF-α, IL-1 also acts to downregulate SHP gene expression.


Asunto(s)
Colesterol 7-alfa-Hidroxilasa/genética , Regulación de la Expresión Génica/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Hígado/efectos de los fármacos , Animales , Colesterol 7-alfa-Hidroxilasa/biosíntesis , Proteínas de Unión al ADN/efectos de los fármacos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Expresión Génica/fisiología , Regulación de la Expresión Génica/fisiología , Factor Nuclear 4 del Hepatocito/efectos de los fármacos , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Interleucina-1alfa/genética , Interleucina-1alfa/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Hígado/metabolismo , Receptores X del Hígado , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptores Nucleares Huérfanos/efectos de los fármacos , Receptores Nucleares Huérfanos/genética , Receptores Nucleares Huérfanos/metabolismo , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
18.
Cells ; 10(6)2021 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-34198609

RESUMEN

Alcohol-related liver disease is associated with intestinal dysbiosis. Functional changes in the microbiota affect bile acid metabolism and result in elevated serum bile acids in patients with alcohol-related liver disease. The aim of this study was to identify the potential role of the bile acid sequestrant colesevelam in a humanized mouse model of ethanol-induced liver disease. We colonized germ-free (GF) C57BL/6 mice with feces from patients with alcoholic hepatitis and subjected humanized mice to the chronic-binge ethanol feeding model. Ethanol-fed gnotobiotic mice treated with colesevelam showed reduced hepatic levels of triglycerides and cholesterol, but liver injury and inflammation were not decreased as compared with non-treated mice. Colesevelam reduced hepatic cytochrome P450, family 7, subfamily a, polypeptide 1 (Cyp7a1) protein expression, although serum bile acids were not lowered. In conclusion, our findings indicate that colesevelam treatment mitigates ethanol-induced liver steatosis in mice.


Asunto(s)
Colesterol 7-alfa-Hidroxilasa/biosíntesis , Clorhidrato de Colesevelam/farmacología , Etanol/toxicidad , Hígado Graso , Vida Libre de Gérmenes , Animales , Hígado Graso/inducido químicamente , Hígado Graso/tratamiento farmacológico , Hígado Graso/enzimología , Femenino , Ratones
19.
Metabolism ; 118: 154728, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33581130

RESUMEN

BACKGROUND & AIMS: Cholesterol gallstone disease (CGD) is a common gastrointestinal disease. Liraglutide, an analogue of glucagon-like peptide 1, has been approved to treat type 2 diabetes. Clinical studies have suggested a potential role of liraglutide in CGD. METHODS: Mice were subcutaneously injected with liraglutide, then fed a lithogenic diet. Bile duct cannulation was performed to collect bile output in mice. Intestinal-specific ablation or pharmacological inhibition of farnesoid X receptor (FXR) was used to study its functions in CGD. RESULTS: Liraglutide could protect mice against CGD. Liraglutide treatment increased the biliary concentration of cholesterol, phospholipids and bile acids and thereby decreased the cholesterol saturation index. The resistance to CGD conferred by liraglutide is likely a result of increased bile acid synthesis and efficient bile acid transport. The expression of a key bile acid synthetic enzyme, Cyp7a1, was significantly increased in liraglutide-treated mice. The increased expression of Cyp7a1 resulted from a relieved suppression signal of Fgf15 from the ileum. Mechanistically, liraglutide treatment altered bile acid composition and suppressed FXR activity in the ileum. Genetic ablation or pharmacological inhibition of FXR in the intestine protected mice against CGD. More importantly, intestinal FXR was required for liraglutide-mediated regulation of hepatic expression of Cyp7a1. CONCLUSION: Liraglutide improved CGD by increasing bile acid secretion and decreasing cholesterol saturation index. Liraglutide attenuates the negative feedback inhibition of bile acids through inhibiting intestinal FXR activity. Our results suggest that liraglutide may represent a novel way for treating or preventing cholesterol gallstones in individuals with high risk of CGD.


Asunto(s)
Colesterol/metabolismo , Cálculos Biliares/prevención & control , Mucosa Intestinal/metabolismo , Liraglutida/farmacología , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Ácidos y Sales Biliares/biosíntesis , Colesterol 7-alfa-Hidroxilasa/biosíntesis , Colesterol 7-alfa-Hidroxilasa/metabolismo , Dieta , Inducción Enzimática , Factores de Crecimiento de Fibroblastos/metabolismo , Cálculos Biliares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microsomas Hepáticos/enzimología , Transducción de Señal
20.
Sci Rep ; 11(1): 21596, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34732821

RESUMEN

To investigate the effect of duodenal-jejunal bypass (DJB) surgery on postoperative blood glucose in type 2 diabetic rats, and further explore possible mechanisms for the effect of surgical treatment of type 2 diabetes. Forty rats with type 2 diabetes were randomly assigned to 4 groups (n = 10 rats per group), which subsequently underwent DJB, new biliopancreatic diversion (NBPD) or duodenal-jejunal exclusion (DJE) surgery or a sham operation (SHAM). Fasting glucose, 2-h postprandial glucose and blood lipids were measured, and the mRNA in liver and intestinal tissue for bile acid receptor (FXR), as well as the FXR protein expression in the liver tissues were determined. Postprandial blood glucose and fasting TG and FFA in the DJB and NBPD groups were significantly lower than those in the SHAM group and preoperative (p < 0.05) at 8 weeks postoperation. Liver FXR protein was expressed at significantly higher in the DJB and NBPD groups than in the other two (p < 0.05), and the intestinal FXR mRNA in the DJE group were highest. DJB up-regulates the expression of bile acid receptors in the liver and down-regulates those receptors in the intestinal tract via biliopancreatic diversion. This process reduces TG levels, and subsequently any lipotoxicity to islet cells to produce a hypoglycemic effect.


Asunto(s)
Desviación Biliopancreática/métodos , Diabetes Mellitus Tipo 2/complicaciones , Hipoglucemia/complicaciones , Intestinos/cirugía , Anastomosis Quirúrgica , Animales , Apoptosis , Ácidos y Sales Biliares/sangre , Colesterol/sangre , Colesterol 7-alfa-Hidroxilasa/biosíntesis , Duodeno/cirugía , Tracto Gastrointestinal , Regulación de la Expresión Génica , Insulina/sangre , Resistencia a la Insulina , Islotes Pancreáticos/metabolismo , Yeyuno/cirugía , Hígado/metabolismo , Masculino , Proteínas de Unión al ARN/biosíntesis , Ratas , Ratas Sprague-Dawley , Triglicéridos/sangre
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda