Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Endocrinology ; 149(4): 1551-61, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18187556

RESUMEN

Androgens increase muscle mass, decrease fat mass, and reduce high-density lipoprotein cholesterol (HDL), but the relationship between body composition, lipoprotein metabolism, and androgens has not been explained. Here we treated ovariectomized cynomolgus monkeys with 5alpha-dihydrotestosterone (DHT) or vehicle for 14 d and measured lipoprotein and triglycerides. Nuclear magnetic resonance analysis revealed that DHT dose-dependently reduced the cholesterol content of large HDL particles and decreased mean HDL particle size (P < 0.01) and also tended to lower low-density lipoprotein cholesterol without altering other lipoprotein particles. Liver and visceral fat biopsies taken before and after DHT treatment for 1 or 14 d were analyzed by genome-wide microarrays. In liver, DHT did not alter the expression of most genes involved in cholesterol synthesis or uptake but rapidly increased small heterodimer partner (SHP) RNA, along with concomitant repression of CYP7A1, a target of SHP transcriptional repression and the rate-limiting enzyme in bile acid synthesis. DHT regulation of SHP and CYP7A1 also occurs in rats, indicating a conserved mechanism. In adipose tissue, pathway analyses suggested coordinate regulation of adipogenesis, tissue remodeling, and lipid homeostasis. Genes encoding IGF-I and beta-catenin were induced, as were extracellular matrix, cell adhesion, and cytoskeletal components, whereas there was consistent down-regulation of genes involved in triacylglycerol metabolism. Interestingly, cholesterol ester transfer protein RNA was induced rapidly in monkey adipose tissue, whereas its inhibitor apolipoprotein CI was repressed. These data provide insight into the androgenic regulation of lipoprotein homeostasis and suggest that changes in adipose lipoprotein metabolism could contribute to HDL cholesterol reduction.


Asunto(s)
Tejido Adiposo/metabolismo , HDL-Colesterol/sangre , Dihidrotestosterona/farmacología , Animales , Composición Corporal , Colesterol 7-alfa-Hidroxilasa/genética , Colesterol 7-alfa-Hidroxilasa/fisiología , Proteínas de Transferencia de Ésteres de Colesterol/genética , LDL-Colesterol/sangre , Relación Dosis-Respuesta a Droga , Femenino , Hígado/metabolismo , Macaca fascicularis , Análisis de Secuencia por Matrices de Oligonucleótidos , Tamaño de la Partícula , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/genética
2.
Pharmacol Ther ; 116(3): 449-72, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17959250

RESUMEN

Mammals dispose of cholesterol mainly through 7alpha-hydroxylated bile acids, and the enzyme catalyzing the 7alpha-hydroxylation, cholesterol 7alpha-hydroxylase (CYP7A1), has a deep impact on cholesterol homeostasis. In this review, we present the study of regulation of CYP7A1 as a good exemplification of the extraordinary contribution of molecular biology to the advancement of our understanding of metabolic pathways that has taken place in the last 2 decades. Since the cloning of the gene from different species, experimental evidence has accumulated, indicating that the enzyme is mainly regulated at the transcriptional level and that bile acids are the most important physiological inhibitors of CYP7A1 transcription. Multiple mechanisms are involved in the control of CYP7A1 transcription and a variety of transcription factors and nuclear receptors participate in sophisticated regulatory networks. A higher order of transcriptional regulation, stemming from the so-called histone code, also applies to CYP7A1, and recent findings clearly indicate that chromatin remodelling events have profound effects on its expression. CYP7A1 also acts as a sensor of signals coming from the gut, thus representing another line of defence against the toxic effects of bile acids and a downstream target of agents acting at the intestinal level. From the pharmacological point of view, bile acid binding resins were the first primitive approach targeting the negative feed-back regulation of CYP7A1 to reduce plasma cholesterol. In recent years, new drugs have been designed based on recent discoveries of the regulatory network, thus confirming the position of CYP7A1 as a focus for innovative pharmacological intervention.


Asunto(s)
Anticolesterolemiantes/farmacología , Ácidos y Sales Biliares/metabolismo , Colesterol 7-alfa-Hidroxilasa/fisiología , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Resinas de Intercambio Iónico/farmacología , Animales , Colesterol/metabolismo , Colesterol 7-alfa-Hidroxilasa/efectos de los fármacos , Colesterol 7-alfa-Hidroxilasa/genética , Ritmo Circadiano , Dieta , Regulación Enzimológica de la Expresión Génica , Homeostasis , Humanos , Receptores Citoplasmáticos y Nucleares/fisiología , Transducción de Señal
3.
Am J Physiol Gastrointest Liver Physiol ; 289(3): G456-61, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15845870

RESUMEN

Phosphatidylcholine transfer protein (PC-TP) is a member of the steroidogenic acute regulatory transfer protein-related domain superfamily and is enriched in liver. To explore a role for PC-TP in hepatic cholesterol metabolism, Pctp-/- and wild-type C57BL/6J mice were fed a standard chow diet or a high-fat, high-cholesterol lithogenic diet. In chow-fed Pctp-/- mice, acyl CoA:cholesterol acyltransferase (Acat) activity was markedly increased, 3-hydroxy-3-methylglutaryl-CoA reductase activity was unchanged, and cholesterol 7alpha-hydroxylase activity was reduced. Consistent with increased Acat activity, esterified cholesterol concentrations in livers of Pctp-/- mice were increased, whereas unesterified cholesterol concentrations were reduced. Hepatic phospholipid concentrations were also decreased in the absence of PC-TP and consequently, unesterified cholesterol-to-phospholipid ratios in liver remained unchanged. The lithogenic diet downregulated 3-hydroxy-3-methylglutaryl-CoA reductase in wild-type and Pctp-/- mice, whereas Acat was increased only in wild-type mice. In response to the lithogenic diet, a greater reduction in cholesterol 7alpha-hydroxylase activity in Pctp-/- mice could be attributed to increased size and hydrophobicity of the bile salt pool. Despite higher hepatic phospholipid concentrations, the unesterified cholesterol-to-phospholipid ratio increased. The lack of Acat upregulation suggests that, in the setting of the dietary challenge, the capacity for esterification to defend against hepatic accumulation of unesterified cholesterol was exceeded in the absence of PC-TP expression. We speculate that regulation of cholesterol homeostasis is a physiological function of PC-TP in liver, which can be overcome with a cholesterol-rich lithogenic diet.


Asunto(s)
Colesterol/metabolismo , Hígado/fisiología , Proteínas de Transferencia de Fosfolípidos/fisiología , Animales , Colesterol 7-alfa-Hidroxilasa/fisiología , Grasas de la Dieta , Homeostasis , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Transferencia de Fosfolípidos/biosíntesis , Regulación hacia Arriba
4.
Arterioscler Thromb Vasc Biol ; 24(4): 768-74, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14962946

RESUMEN

OBJECTIVE: Cholesterol 7alpha-hydroxylase (cyp7a1) catalyzes the rate-limiting step in conversion of cholesterol to bile acids. To study the relationship between bile acid biosynthesis and triglyceride metabolism, we cross-bred mice lacking cyp7a1 on a hyperlipidemic APOE*3-Leiden background. METHODS AND RESULTS: Female mice received a chow or lipogenic diet. On both diets, fecal bile acid excretion was 70% decreased concomitantly with a 2-fold increased neutral sterol output. The differences in bile acid biosynthesis did not change plasma cholesterol levels. However, plasma triglyceride levels decreased by 41% and 38% in the cyp7a1-/-. APOE*3-Leiden mice as compared with APOE*3-Leiden mice on chow and lipogenic diet, respectively. Mechanistic studies showed that very-low-density lipoprotein (VLDL)-apolipoprotein B and VLDL-triglyceride production rates were reduced in cyp7a1-/-. APOE*3-Leiden mice as compared with APOE*3-Leiden mice (-34% and -35%, respectively). Cyp7a1 deficiency also increased the hepatic cholesteryl ester and triglyceride content (2.8-fold and 2.5-fold, respectively). In addition, hepatic anti-oxidative vitamin content, which can influence VLDL-production, was lower. Hepatic mRNA analysis showed decreased expression of genes involved in lipogenesis including srebf1. CONCLUSIONS: Cyp7a1 deficiency in APOE*3-Leiden mice decreases the VLDL particle production rate, as a consequence of a strongly reduced bile acid biosynthesis, leading to a decrease in plasma triglycerides. These data underscore the close relationship between bile acid biosynthesis and triglyceride levels.


Asunto(s)
Apolipoproteínas E/genética , Ácidos y Sales Biliares/metabolismo , Colesterol 7-alfa-Hidroxilasa/deficiencia , Metabolismo de los Lípidos , Lipoproteínas VLDL/biosíntesis , Aciltransferasas/metabolismo , Animales , Apolipoproteína E3 , Apolipoproteínas B/metabolismo , Colesterol 7-alfa-Hidroxilasa/genética , Colesterol 7-alfa-Hidroxilasa/fisiología , Ésteres del Colesterol/metabolismo , Cruzamientos Genéticos , Diacilglicerol O-Acetiltransferasa , Dieta Aterogénica , Heces , Femenino , Hiperlipoproteinemia Tipo III/genética , Hiperlipoproteinemia Tipo III/metabolismo , Cuerpos Cetónicos/metabolismo , Lipólisis , Lipoproteínas VLDL/sangre , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/biosíntesis , Esteroles/metabolismo , Triglicéridos/metabolismo , Vitamina A/metabolismo , Vitamina E/metabolismo
5.
Gastroenterology ; 125(2): 544-55, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12891557

RESUMEN

BACKGROUND & AIMS: Increased serum triglyceride levels constitute a risk factor for coronary heart disease. Apolipoprotein CIII (Apo CIII) is a determinant of serum triglyceride metabolism. In this study, we investigated whether activators of the nuclear farnesoid X receptor (FXR) modulate Apo CIII gene expression. METHODS: The influence of bile acids and synthetic FXR activators on Apo CIII and triglyceride metabolism was studied in vivo by using FXR wild-type and FXR-deficient mice and in vitro by using human primary hepatocytes and HepG2 cells. RESULTS: In mice, treatment with the FXR agonist taurocholic acid strongly decreased serum triglyceride levels, an effect associated with reduced Apo CIII serum and liver messenger RNA levels. By contrast, no change was observed in FXR-deficient mice. Incubation of human primary hepatocytes and HepG2 cells with bile acids or the nonsteroidal synthetic FXR agonist GW4064 resulted in a dose-dependent down-regulation of Apo CIII gene expression. Promoter transfection experiments and mutation analysis showed that bile acid-activated FXR decrease human Apo CIII promoter activity via a negative FXR response element located in the I(4) footprint between nucleotides -739 and -704. Chromatin immunoprecipitation experiments showed that bile acid treatment led to binding of FXR/retinoid X receptor heterodimers to and displacement of HNF4alpha from this site. Bile acid treatment still repressed liver Apo CIII gene expression in hepatic HNF4alpha-deficient mice, suggesting an active rather than a competitive mechanism of Apo CIII repression by the FXR. CONCLUSIONS: We identified bile acid and synthetic activators of the nuclear FXR as negative regulators of Apo CIII expression, an effect that may contribute to the triglyceride-decreasing action of FXR agonists.


Asunto(s)
Apolipoproteínas C/genética , Proteínas de Unión al ADN , Regulación de la Expresión Génica , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Apolipoproteína C-III , Apolipoproteínas C/metabolismo , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Ácidos y Sales Biliares/farmacología , Células Cultivadas , Colesterol 7-alfa-Hidroxilasa/fisiología , Resina de Colestiramina/farmacología , Dimerización , Factor Nuclear 4 del Hepatocito , Hepatocitos , Humanos , Isoxazoles/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfoproteínas/deficiencia , Fosfoproteínas/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/análisis , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores de Ácido Retinoico/fisiología , Elementos de Respuesta , Receptores X Retinoide , Factores de Transcripción/deficiencia , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Triglicéridos/sangre
7.
Arterioscler Thromb Vasc Biol ; 21(6): 887-98, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11397693

RESUMEN

The production of apolipoprotein B (apoB)-containing lipoproteins by the liver is regulated by a complex series of processes involving apoB being cotranslationally translocated across the endoplasmic reticulum and assembled into a lipoprotein particle. The translocation of apoB across the endoplasmic reticulum is facilitated by the intraluminal chaperone, microsomal triglyceride transfer protein (MTP). MTP facilitates the translocation and folding of apoB, as well as the addition of lipid to lipid-binding domains (which consist of amphipathic beta sheets and alpha helices). In the absence of MTP or sufficient lipid, apoB exhibits translocation arrest. Thus, apoB translation, translocation, and assembly with lipids to form a core-containing lipoprotein particle occur as concerted processes. Abrogation of >/=1 of these processes diverts apoB into a degradation pathway that is dependent on conjugation with ubiquitin and proteolysis by the proteasome. The nascent core-containing lipoprotein particle that forms within the lumen of the endoplasmic reticulum can be "enlarged" to form a mature very low density lipoprotein particle. Additional studies show that the assembly and secretion of apoB-containing lipoproteins are linked to the cholesterol/bile acid synthetic pathway controlled by cholesterol 7alpha-hydroxylase. Studies in cultured cells and transgenic mice indicate that the expression of cholesterol 7alpha-hydroxylase indirectly regulates the expression of lipogenic enzymes through changes in the cellular content of mature sterol response element binding proteins. Oxysterols and bile acids may also act via the ligand-activated nuclear receptors LXR and FXR to link the metabolic pathways controlling energy balance and lipid metabolism to nutritional state.


Asunto(s)
Apolipoproteínas B/metabolismo , Colesterol 7-alfa-Hidroxilasa/fisiología , Enfermedad de la Arteria Coronaria/metabolismo , Hígado/metabolismo , Apolipoproteínas/metabolismo , Apolipoproteínas B/biosíntesis , Apolipoproteínas B/genética , Ácidos y Sales Biliares/biosíntesis , Proteínas Potenciadoras de Unión a CCAAT/fisiología , Proteínas Portadoras/metabolismo , Colesterol/metabolismo , Enfermedad de la Arteria Coronaria/terapia , Cisteína Endopeptidasas/metabolismo , Proteínas de Unión al ADN/fisiología , Retículo Endoplásmico/metabolismo , Humanos , Lipoproteínas VLDL/metabolismo , Complejos Multienzimáticos/metabolismo , Complejo de la Endopetidasa Proteasomal , Proteína 1 de Unión a los Elementos Reguladores de Esteroles , Factores de Transcripción/fisiología , Ubiquitinas/metabolismo
9.
Gastroenterology ; 120(7): 1801-9, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11375960

RESUMEN

BACKGROUND & AIMS: The rate of 12alpha-hydroxylation of bile acid intermediates is believed to determine the ratio of cholic acid (CA) to chenodeoxycholic acid (CDCA) biosynthesis and the overall hydrophobicity of the bile acid pool. The aim of this study was to determine the effects of the level of expression of sterol 12alpha-hydroxylase (CYP8b1) and cholesterol 7alpha-hydroxylase (CYP7a1) on rates of CA biosynthesis and bile acid pool composition. METHODS: Expression of CYP8b1 and CYP7a1 was accomplished through infection of primary rat hepatocytes (PRH) or intact male SD rats with replication-defective recombinant adenoviruses encoding either CYP8b1 or CYP7a1. RESULTS: Increased expression of CYP7a1 over basal levels in PRH dramatically increased bile acid biosynthesis (586% +/- 82%, P < 0.001) but did not alter the ratio of CA to CDCA. Conversely, increased expression of CYP8b1 in vitro had no significant effect on the rates of total bile acid synthesis but significantly increased (4.1-fold) the rates of CA biosynthesis, resulting in an increase in the CA-CDCA ratio from 1:6.6 to 2.8:1. In whole rats, increased CYP8b1 expression over basal levels markedly increased the CA in the bile acid pool from 36% +/- 3.4% to 50% +/- 2.9% in 5 days. CDCA and its muricholic acid derivatives decreased from 64% +/- 3.4% to 50% +/- 2.9%. CONCLUSIONS: Increased expression of CYP8b1 led to a marked increase in CA biosynthesis both in PRH and in whole animals. CYP8b1 is capable of 12alpha-hydroxylating bile acid intermediates from both the classic and acidic pathways.


Asunto(s)
Ácido Quenodesoxicólico/biosíntesis , Ácido Cólico/biosíntesis , Sistema Enzimático del Citocromo P-450/fisiología , Hepatocitos/metabolismo , Esteroide Hidroxilasas/fisiología , Animales , Células Cultivadas , Colesterol 7-alfa-Hidroxilasa/genética , Colesterol 7-alfa-Hidroxilasa/fisiología , Sistema Enzimático del Citocromo P-450/genética , Masculino , Ratas , Ratas Sprague-Dawley , Esteroide 12-alfa-Hidroxilasa , Esteroide Hidroxilasas/genética
10.
J Biol Chem ; 276(26): 23304-11, 2001 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-11323427

RESUMEN

The finding that expression of a cholesterol 7alpha-hydroxylase (CYP7A1) transgene in cultured rat hepatoma cells caused a coordinate increase in lipogenesis and secretion of apoB-containing lipoproteins led to the hypothesis that hepatic production of apoB-containing lipoproteins may be linked to the expression of CYP7A1 (Wang, S.-L., Du, E., Martin, T. D., and Davis, R. A. (1997) J. Biol. Chem. 272, 19351-19358). To examine this hypothesis in vivo, a transgene encoding CYP7A1 driven by the constitutive liver-specific enhancer of the human apoE gene was expressed in C56BL/6 mice. The expression of CYP7A1 mRNA (20-fold), protein ( approximately 10-fold), and enzyme activity (5-fold) was markedly increased in transgenic mice compared with non-transgenic littermates. The bile acid pool of CYP7A1 transgenic mice was doubled mainly due to increased hydrophobic dihydroxy bile acids. In CYP7A1 transgenic mice, livers contained approximately 3-fold more sterol response element-binding protein-2 mRNA. Hepatic expression of mRNAs encoding lipogenic enzymes (i.e. fatty-acid synthase, acetyl-CoA carboxylase, stearoyl-CoA desaturase, squalene synthase, farnesyl-pyrophosphate synthase, 3-hydroxy-3-methylglutaryl-CoA reductase, and low density lipoprotein receptor) as well as microsomal triglyceride transfer protein were elevated approximately 3-5-fold in transgenic mice. CYP7A1 transgenic mice also displayed a >2-fold increase in hepatic production and secretion of triglyceride-rich apoB-containing lipoproteins. Despite the increased hepatic secretion of apoB-containing lipoproteins in CYP7A1 mice, plasma levels of triglycerides and cholesterol were not significantly increased. These data suggest that the 5-fold increased expression of the low density lipoprotein receptor displayed by the livers of CYP7A1 transgenic mice was sufficient to compensate for the 2-fold increase production of apoB-containing lipoproteins. These findings emphasize the important homeostatic role that CYP7A1 plays in balancing the anabolic lipoprotein assembly/secretion pathway with the cholesterol catabolic bile acid synthetic pathway.


Asunto(s)
Apolipoproteínas B/biosíntesis , Colesterol 7-alfa-Hidroxilasa/fisiología , Hígado/enzimología , Animales , Apolipoproteína B-100 , Apolipoproteínas B/sangre , Apolipoproteínas B/metabolismo , Ácidos y Sales Biliares/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Portadoras/metabolismo , Colesterol/metabolismo , Colesterol 7-alfa-Hidroxilasa/genética , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Hiperlipidemias/sangre , Metabolismo de los Lípidos , Lípidos/sangre , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Mensajero/biosíntesis , Receptores de LDL/biosíntesis , Receptores de LDL/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles , Proteína 2 de Unión a Elementos Reguladores de Esteroles , Ácido Tauroquenodesoxicólico/metabolismo , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Triglicéridos/sangre
11.
Curr Pharm Des ; 7(4): 231-59, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11254888

RESUMEN

The orphan nuclear receptors FXR and LXRalpha have become challenging targets for the discovery of new therapeutic agents. Bile acids and hydroxysterol intermediates are the respective natural ligands of these two structurally and functionally closely related receptors. Both FXR and LXRalpha; are thought to play a major role in the control of cholesterol catabolism by regulating the expression of cholesterol 7alpha-hydroxylase, the rate limiting enzyme of bile acid synthesis. Reverse cholesterol transport might also be affected by FXR and LXR since they control the expression of PLTP and CETP, two proteins involved in the transfer of phospholipid, cholesterol and cholesteryl esters among plasma lipoproteins. A new class of potent synthetic activators of FXR, the 1,1-bisphosphonate esters, has been discovered which up regulate the Intestinal Bile Acid Binding Protein gene (I-BABP) as demonstrated for chenodeoxycholic acid, however there are no known synthetic activators yet identified for LXRalpha. The evaluation of FXR as a potential target for the development of drugs affecting plasma cholesterol can take advantage of the fact that the activators of FXR (farnesol, bile acids and the 1,1-bisphosphonate esters) have been studied in various in vitro and in vivo models. Administration of chenodeoxycholic acid to animals and man did not result in the increase in plasma cholesterol expected from a decrease in cholesterol 7alpha-hydroxylase expression. Like farnesol, the 1,1-bisphosphonate esters increase the rate of degradation of HMGCoA reductase and have the unexpected property of inducing hypocholesterolemia in normal animals. The natural and synthetic FXR agonists trigger differentiation, inhibit cell proliferation and are potent inducers of apoptosis. The 1,1-bisphosphonate ester SR-45023A (Apomine) is presently being developed as an antineoplastic drug.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Unión al ADN/efectos de los fármacos , Metabolismo de los Lípidos , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Factores de Transcripción/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Anticolesterolemiantes/farmacología , Ácidos y Sales Biliares/metabolismo , Colesterol/metabolismo , Colesterol 7-alfa-Hidroxilasa/fisiología , Proteínas de Unión al ADN/fisiología , Diseño de Fármacos , Humanos , Receptores X del Hígado , Datos de Secuencia Molecular , Receptores Nucleares Huérfanos , Receptores Citoplasmáticos y Nucleares/fisiología , Receptores de Esteroides/química , Factores de Transcripción/fisiología
12.
Biol Chem ; 381(12): 1149-53, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11209749

RESUMEN

Circadian rhythms of important enzymes involved in the conversion of cholesterol to bile acids [sterol 12alpha-hydroxylase (12alpha-hydroxylase) and cholesterol 7alpha-hydroxylase (7alpha-hydroxylase)] and an albumin site D-binding protein (DBP) were examined in rats. When the animals were fed freely, they usually ate in the dark and the circadian rhythms of activities of 12alpha-hydroxylase and 7alpha-hydroxylase showed the same peaks (at 10 p.m.) and lows (at 2 p.m.). Their mRNA levels were determined at four timepoints: 3 a.m., 10 a.m., 3 p.m. and 10 p.m. A maximum of the rhythm of 12alpha-hydroxylase was observed at 3 p.m. and the minimum at 3 a.m. These results are distinct from those of 7alpha-hydroxylase, whose maximum point was at 10 p.m. and minimum at 3 p.m. When the rats were fed only in the day-time (from 9 a.m. to 5 p.m.), a marked shift of the activity and mRNA rhythms was observed with both enzymes. The circadian rhythms of the activities of both enzymes showed the same peaks (at 3 p.m.), but the mRNA levels of 12alpha-hydroxylase were distinct from those of 7alpha-hydroxylase, whose maximum point was at 3 a.m. and minimum at 10 p.m. Differences between the maximum and the minimum points of each enzyme mRNA level were statistically significant (P < 0.01 for 12alpha-hydroxylase and 0.05 for 7alpha-hydroxylase). Moreover, circadian rhythms of DBP were also markedly shifted with the change of feeding period. The maximum mRNA level was observed at 10 p.m. instead of 10 a.m. and the minimum was at 10 a.m. instead of 10 p.m.


Asunto(s)
Colesterol/metabolismo , Ritmo Circadiano/fisiología , Proteínas de Unión al ADN , Ratas/metabolismo , Esteroide Hidroxilasas/metabolismo , Factores de Transcripción/metabolismo , Animales , Colesterol/genética , Colesterol/fisiología , Colesterol 7-alfa-Hidroxilasa/genética , Colesterol 7-alfa-Hidroxilasa/metabolismo , Colesterol 7-alfa-Hidroxilasa/fisiología , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Sistema Enzimático del Citocromo P-450/fisiología , Conducta Alimentaria/fisiología , Masculino , ARN Mensajero/metabolismo , Ratas/fisiología , Ratas Wistar , Esteroide 12-alfa-Hidroxilasa , Esteroide Hidroxilasas/genética , Esteroide Hidroxilasas/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología
14.
Nutr Rev ; 57(6): 196-8, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10439633

RESUMEN

Cholesterol biosynthesis is regulated by transcription factors named steroid regulatory element-binding proteins. A newly discovered transcription factor, LXR, regulates the catabolic degradation of cholesterol by activation of the gene controlling cholesterol 7 alpha-hydroxylase, the rate-limiting enzyme in the formation of bile acids. Excessive dietary cholesterol leads to increased oxysterol formation. Oxysterol binds to LXR and thereby induces transcription of cholesterol 7 alpha-hydroxylase, thus increasing the removal of cholesterol as bile acids.


Asunto(s)
Colesterol 7-alfa-Hidroxilasa/fisiología , Colesterol/biosíntesis , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Ácidos y Sales Biliares/biosíntesis , Colesterol 7-alfa-Hidroxilasa/genética , Colesterol en la Dieta/metabolismo , Proteínas de Unión al ADN , Homeostasis , Humanos , Receptores X del Hígado , Ratones , Ratones Noqueados , Receptores Nucleares Huérfanos , Esteroles/metabolismo , Factores de Transcripción/fisiología
16.
Biochem Cell Biol ; 75(3): 255-62, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9404645

RESUMEN

The effect of administering a synthetic transgene encoding cholesterol 7 alpha-hydroxylase (cyp7) on plasma cholesterol metabolism of intact mice was investigated. The synthetic cyp7 transgene (Tg1) was constructed by placing the cDNA sequence encoding the full-length cyp7 polypeptide under the control of a heavy metal inducible metallothionein promoter. The transgene was complexed with asialoorosomucoid-polylysine conjugate and introduced into mice via the tail vein. Cell marking experiments using a beta-galactosidase (lacZ) transgene as a tag showed that 5-10% of the liver can be transfected by this procedure. Administration of the Tg1 transgene to older hypercholesterolemic chow-fed mice resulted in about a 50% reduction of plasma cholesterol, regardless of whether or not transgene expression was induced by zinc treatment. In diet-induced hypercholesterolemic mice, the reduction (20%) in total plasma cholesterol was seen only when transgene expression was induced, and this reduction was due primarily to a decrease in non-high-density lipoprotein cholesterol. The maximum reduction was evident at 6 days after the introduction of the transgene and was no longer evident after 9 days. Introduction of the Tg1 transgene into young chow-fed mice had no effect on the already low levels of plasma cholesterol. However, compared with the no-transgene and lacZ transgene controls, the gallbladder bile acid content of Tg1-treated mice was increased. The results show that non-viral-mediated delivery of a synthetic transgene encoding cyp7 to a subpopulation of hepatocytes in the liver of intact hypercholesterolemic mice is sufficient to facilitate the temporary reduction of plasma cholesterol content.


Asunto(s)
Colesterol 7-alfa-Hidroxilasa/genética , Colesterol/sangre , Colesterol/genética , Hipercolesterolemia/tratamiento farmacológico , Hipercolesterolemia/genética , Hígado/metabolismo , Transfección/métodos , Transgenes , Administración Oral , Animales , Colesterol/metabolismo , Colesterol 7-alfa-Hidroxilasa/administración & dosificación , Colesterol 7-alfa-Hidroxilasa/fisiología , Grasas de la Dieta/administración & dosificación , Femenino , Hipercolesterolemia/sangre , Ratones , Ratas
17.
Biochem Biophys Res Commun ; 202(2): 896-901, 1994 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-8048962

RESUMEN

Transfection of COS-cells with a cDNA coding for human cholesterol 7 alpha-hydroxylase resulted in significant production of cholesterol 7 alpha-hydroxylase enzyme and intracellular accumulation of the product, 7 alpha-hydroxycholesterol. Presence of this enzyme activity was always associated with increased HMG CoA reductase activity. In five different independent transfection experiments resulting in a cholesterol 7 alpha-hydroxylase activity of 0.26 +/- 0.05 pmol/min/mg in the transfected cells, the HMG CoA reductase activity increased to 158 +/- 14% of that of the control cells (p < 0.01). This change was not associated with significant changes in the cholesterol content or LDL-receptor expression of the COS-cells. It is evident that the two key enzymes in cholesterol synthesis and degradation interact with each other also in extra-hepatic cells that are unable to degrade cholesterol into bile acids. Possible mechanisms for the finding is discussed.


Asunto(s)
Colesterol 7-alfa-Hidroxilasa/fisiología , Hidroximetilglutaril-CoA Reductasas/metabolismo , Animales , Línea Celular , Colesterol 7-alfa-Hidroxilasa/genética , ADN Complementario/genética , Expresión Génica , Humanos , Hidroxicolesteroles/metabolismo , Transfección
18.
J Lipid Res ; 35(2): 319-27, 1994 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-7513337

RESUMEN

Among nine strains of rat, two were found that responded to phenobarbital treatment with increased activity of hepatic cholesterol 7 alpha-hydroxylase. This effect was maximal after 2-3 days of treatment and was then reduced. Interestingly the increased cholesterol 7 alpha-hydroxylase activity was associated with increased activity of hepatic HMG-CoA reductase in the two responding strains but not in the non-responding strains. In tissues other than the liver, HMG-CoA reductase activity was unaffected in responding rats. Most of the above stimulation occurred at a pretranslatory level and the mRNA levels corresponding to the two enzymes paralleled the activities. The phenobarbital treatment resulted in decreased content of free cholesterol in liver microsomes in a strain of rat that responded with increased cholesterol 7 alpha-hydroxylase activity. It was shown that depletion of cholesterol in the responding strain of rats by lymph fistulation also was associated with a parallel increase in levels of HMG-CoA reductase activity and mRNA. The findings are discussed in relation to the link between HMG-CoA reductase and cholesterol 7 alpha-hydroxylase. A primary upregulation of the cholesterol 7 alpha-hydroxylase by the cytochrome P450 inducer phenobarbital can be expected to lead to increased consumption of cholesterol substrate. This consumption may result in a compensatory increase in the activity of the HMG-CoA reductase. It is suggested that such a mechanism is responsible for part of the covariation of the two enzyme systems under different conditions.


Asunto(s)
Colesterol 7-alfa-Hidroxilasa/fisiología , Hidroximetilglutaril-CoA Reductasas/fisiología , Hígado/enzimología , Fenobarbital/farmacología , Animales , Northern Blotting , Colesterol/metabolismo , Colesterol 7-alfa-Hidroxilasa/análisis , Hidroximetilglutaril-CoA Reductasas/análisis , Hígado/efectos de los fármacos , Masculino , Microsomas Hepáticos/metabolismo , ARN/análisis , Ratas , Ratas Endogámicas , Ratas Sprague-Dawley , Ratas Wistar , Fracciones Subcelulares
19.
Cardiovasc Drugs Ther ; 6(2): 103-10, 1992 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-1390320

RESUMEN

Cholesterol plays an essential role in cell membrane synthesis and in cell growth and differentiation. In mammalian cells, cholesterol can be synthesized from acetate precursors or taken up from dietary or exogenous sources. The major catabolic route for disposal of cholesterol involves conversion into excretable bile acids. The maintenance of cholesterol homeostasis is influenced and carefully controlled by multiple feedback mechanisms. The key regulatory targets of these feedback mechanisms are 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase in cholesterol biosynthesis, the low-density lipoprotein (LDL) receptor in cholesterol uptake, and cholesterol 7 alpha-hydroxylase in cholesterol catabolism. The elucidation of regulatory mechanisms in cholesterol metabolism has been greatly facilitated by the discovery of a new class of lipid-lowering drugs, the HMG-CoA reductase inhibitors. In addition to proving therapeutically useful in the treatment of hypercholesterolemia, these drugs have revealed novel regulatory steps in cholesterol metabolism and several new targets for future drug development. This manuscript reviews recent developments in the cholesterol biosynthetic pathway and the regulatory mechanisms that maintain cholesterol homeostasis.


Asunto(s)
Acilcoenzima A/fisiología , Colesterol/biosíntesis , Hígado/metabolismo , Acilcoenzima A/antagonistas & inhibidores , Colesterol/metabolismo , Colesterol 7-alfa-Hidroxilasa/fisiología , Homeostasis , Humanos , Hígado/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA