Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Circ Res ; 132(7): 849-863, 2023 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-36876496

RESUMEN

BACKGROUND: Removal of circulating plasma low-density lipoprotein cholesterol (LDL-C) by the liver relies on efficient endocytosis and intracellular vesicle trafficking. Increasing the availability of hepatic LDL receptors (LDLRs) remains a major clinical target for reducing LDL-C levels. Here, we describe a novel role for RNF130 (ring finger containing protein 130) in regulating plasma membrane availability of LDLR. METHODS: We performed a combination of gain-of-function and loss-of-function experiments to determine the effect of RNF130 on LDL-C and LDLR recycling. We overexpressed RNF130 and a nonfunctional mutant RNF130 in vivo and measured plasma LDL-C and hepatic LDLR protein levels. We performed in vitro ubiquitination assays and immunohistochemical staining to measure levels and cellular distribution of LDLR. We supplement these experiments with 3 separate in vivo models of RNF130 loss-of-function where we disrupted Rnf130 using either ASO (antisense oligonucleotides), germline deletion, or AAV CRISPR (adeno-associated virus clustered regularly interspaced short palindromic repeats) and measured hepatic LDLR and plasma LDL-C. RESULTS: We demonstrate that RNF130 is an E3 ubiquitin ligase that ubiquitinates LDLR resulting in redistribution of the receptor away from the plasma membrane. Overexpression of RNF130 decreases hepatic LDLR and increases plasma LDL-C levels. Further, in vitro ubiquitination assays demonstrate RNF130-dependent regulation of LDLR abundance at the plasma membrane. Finally, in vivo disruption of Rnf130 using ASO, germline deletion, or AAV CRISPR results in increased hepatic LDLR abundance and availability and decreased plasma LDL-C levels. CONCLUSIONS: Our studies identify RNF130 as a novel posttranslational regulator of LDL-C levels via modulation of LDLR availability, thus providing important insight into the complex regulation of hepatic LDLR protein levels.


Asunto(s)
Hígado , Receptores de LDL , LDL-Colesterol/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Hígado/metabolismo , Proteínas Portadoras/metabolismo , Ubiquitinación , Lipoproteínas LDL/metabolismo
2.
J Lipid Res ; 58(11): 2127-2138, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28874443

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is the leading cause of chronic liver disease. NAFLD progresses from benign steatosis to steatohepatitis (NASH) to cirrhosis and is linked to hepatocellular carcinoma. No targeted treatment is currently approved for NAFLD/NASH. We previously showed that fat-specific protein 27 (FSP27), a lipid droplet-associated protein that controls triglyceride turnover in the hepatocyte, is required for fasting- and diet-induced triglyceride accumulation in the liver. However, silencing Fsp27 with antisense oligonucleotides (ASOs) did not improve hepatosteatosis in genetic nor nutritional mouse models of obesity. Herein, we tested the therapeutic potential of ASO-Fsp27 when used in combination with the PPARα agonist fenofibrate. C57BL/6 mice were fed a high-trans-fat, high-cholesterol, high-fructose diet for eight weeks to establish NASH, then kept on diet for six additional weeks while dosed with ASOs and fenofibrate, alone or in combination. Data show that ASO-Fsp27 and fenofibrate synergize to promote resistance to diet-induced obesity and hypertriglyceridemia and to reverse hepatic steatosis, inflammation, oxidative stress, and fibrosis. This multifactorial improvement of liver disease noted when combining both drugs suggests that a course of treatment that includes both reduced FSP27 activity and activation of PPARα could provide therapeutic benefit to patients with NAFLD/NASH.


Asunto(s)
Dieta/efectos adversos , Fenofibrato/farmacología , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/genética , Oligonucleótidos Antisentido/genética , Proteínas/genética , Animales , Sinergismo Farmacológico , Fenofibrato/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Obesidad/tratamiento farmacológico , Obesidad/genética , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Riesgo
3.
J Lipid Res ; 58(1): 81-91, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27884961

RESUMEN

Obesity is a component of the metabolic syndrome, mechanistically linked to diabetes, fatty liver disease, and cardiovascular disease. Proteins that regulate the metabolic fate of intracellular lipid droplets are potential therapeutic candidates to treat obesity and its related consequences. CIDEC (cell death-inducing DFFA-like effector C), also known in mice as Fsp27 (fat-specific protein 27), is a lipid droplet-associated protein that prevents lipid mobilization and promotes intracellular lipid storage. The consequences of complete loss of FSP27 on hepatic metabolism and on insulin resistance are controversial, as both healthy and deleterious lipodystrophic phenotypes have been reported in Fsp27-/- mice. To test whether therapeutic silencing of Fsp27 might be useful to improve obesity, fatty liver, and glycemic control, we used antisense oligonucleotides (ASOs) in both nutritional (high-fat diet) and genetic (leptin-deficient ob/ob) mouse models of obesity, hyperglycemia, and hepatosteatosis. We show that partial silencing Fsp27 in either model results in the robust decrease in visceral fat, improved insulin sensitivity and whole-body glycemic control, and tissue-specific changes in transcripts controlling lipid oxidation and synthesis. These data suggest that partial reduction of FSP27 activity (e.g., using ASOs) might be exploited therapeutically in insulin-resistant obese or overweight patients.


Asunto(s)
Diabetes Mellitus/terapia , Hígado Graso/terapia , Obesidad/terapia , Oligonucleótidos Antisentido/administración & dosificación , Proteínas/genética , Animales , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Hígado Graso/genética , Hígado Graso/metabolismo , Humanos , Resistencia a la Insulina/genética , Gotas Lipídicas/metabolismo , Gotas Lipídicas/patología , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Obesos , Obesidad/genética , Oligonucleótidos Antisentido/genética , Proteínas/antagonistas & inhibidores
4.
J Lipid Res ; 58(2): 317-324, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28007964

RESUMEN

α-Chlorofatty aldehydes are generated from myeloperoxidase-derived HOCl targeting plasmalogens, and are subsequently oxidized to α-chlorofatty acids (α-ClFAs). The catabolic pathway for α-ClFA is initiated by ω-oxidation. Here, we examine PPAR-α activation as a mechanism to increase α-ClFA catabolism. Pretreating both HepG2 cells and primary mouse hepatocytes with the PPAR-α agonist, pirinixic acid (Wy 14643), increased the production of α-chlorodicarboxylic acids (α-ClDCAs) in cells treated with exogenous α-ClFA. Additionally, α-ClDCA production in Wy 14643-pretreated wild-type mouse hepatocytes was accompanied by a reduction in cellular free α-ClFA. The dependence of PPAR-α-accelerated α-ClFA catabolism was further demonstrated by both impaired metabolism in mouse PPAR-α-/- hepatocytes and decreased clearance of plasma α-ClFA in PPAR-α-/- mice. Furthermore, Wy 14643 treatments decreased plasma 2-chlorohexadecanoic acid levels in wild-type mice. Additional studies showed that α-ClFA increases PPAR-α, PPAR-δ, and PPAR-γ activities, as well as mRNA expression of the PPAR-α target genes, CD36, CPT1a, Cyp4a10, and CIDEC. Collectively, these results indicate that PPAR-α accelerates important pathways for the clearance of α-ClFA, and α-ClFA may, in part, accelerate its catabolism by serving as a ligand for PPAR-α.


Asunto(s)
Hepatocitos/metabolismo , PPAR alfa/genética , Ácidos Palmíticos/metabolismo , Animales , Ácidos Grasos/metabolismo , Hepatocitos/efectos de los fármacos , Humanos , Metabolismo/genética , Ratones , Ratones Noqueados , Oxidación-Reducción , PPAR alfa/metabolismo , PPAR delta/biosíntesis , ARN Mensajero/biosíntesis
5.
J Lipid Res ; 58(5): 941-954, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28264879

RESUMEN

Idiopathic pulmonary alveolar proteinosis (PAP) is a rare lung disease characterized by accumulation of surfactant. Surfactant synthesis and secretion are restricted to epithelial type 2 (T2) pneumocytes (also called T2 cells). Clearance of surfactant is dependent upon T2 cells and macrophages. ABCG1 is highly expressed in both T2 cells and macrophages. ABCG1-deficient mice accumulate surfactant, lamellar body-loaded T2 cells, lipid-loaded macrophages, B-1 lymphocytes, and immunoglobulins, clearly demonstrating that ABCG1 has a critical role in pulmonary homeostasis. We identify a variant in the ABCG1 promoter in patients with PAP that results in impaired activation of ABCG1 by the liver X receptor α, suggesting that ABCG1 basal expression and/or induction in response to sterol/lipid loading is essential for normal lung function. We generated mice lacking ABCG1 specifically in either T2 cells or macrophages to determine the relative contribution of these cell types on surfactant lipid homeostasis. These results establish a critical role for T2 cell ABCG1 in controlling surfactant and overall lipid homeostasis in the lung and in the pathogenesis of human lung disease.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/metabolismo , Surfactantes Pulmonares/metabolismo , Células A549 , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/deficiencia , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/genética , Adulto , Células Epiteliales Alveolares/citología , Células Epiteliales Alveolares/metabolismo , Animales , Colesterol/biosíntesis , Colesterol/metabolismo , Femenino , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Homeostasis , Humanos , Inmunoglobulinas/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Proteinosis Alveolar Pulmonar/metabolismo , Proteinosis Alveolar Pulmonar/patología
6.
Biochim Biophys Acta ; 1861(12 Pt B): 2053-2061, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-26869447

RESUMEN

Altered lipoprotein metabolism plays a key role during atherogenesis. For over 50years, epidemiological data have fueled the proposal that HDL-cholesterol (HDL-c) in circulation is inversely correlated to cardiovascular risk. However, the atheroprotective role of HDL is currently the focus of much debate and remains an active field of research. The emerging picture from research in the past decade suggests that HDL function, rather than HDL-c content, is important in disease. Recent developments demonstrate that miRNAs play an important role in fine-tuning the expression of key genes involved in HDL biogenesis, lipidation, and clearance, as well as in determining the amounts of HDL-c in circulation. Thus, it has been proposed that miRNAs that affect HDL metabolism might be exploited therapeutically in patients. Whether HDL-based therapies, alone or in combination with LDL-based treatments (e.g. statins), provide superior outcomes in patients has been recently questioned by human genetics studies and clinical trials. The switch in focus from "HDL-cholesterol" to "HDL function" opens a new paradigm to understand the physiology and therapeutic potential of HDL, and to find novel modulators of cardiovascular risk. In this review we summarize the current knowledge on the regulation of HDL metabolism and function by miRNAs. This article is part of a Special Issue entitled: MicroRNAs and lipid/energy metabolism and related diseases edited by Carlos Fernández-Hernando and Yajaira Suárez.


Asunto(s)
Metabolismo de los Lípidos/genética , Lipoproteínas HDL/genética , Lipoproteínas HDL/metabolismo , MicroARNs/genética , Animales , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/metabolismo , LDL-Colesterol/genética , LDL-Colesterol/metabolismo , Humanos , Factores de Riesgo
7.
Hepatology ; 63(5): 1660-74, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26473496

RESUMEN

UNLABELLED: Understanding the hepatic regenerative process has clinical interest as the effectiveness of many treatments for chronic liver diseases is conditioned by efficient liver regeneration. Experimental evidence points to the need for a temporal coordination between cytokines, growth factors, and metabolic signaling pathways to enable successful liver regeneration. One intracellular mediator that acts as a signal integration node for these processes is the serine-threonine kinase Akt/protein kinase B (Akt). To investigate the contribution of Akt during hepatic regeneration, we performed partial hepatectomy in mice lacking Akt1, Akt2, or both isoforms. We found that absence of Akt1 or Akt2 does not influence liver regeneration after partial hepatectomy. However, hepatic-specific Akt1 and Akt2 null mice show impaired liver regeneration and increased mortality. The major abnormal cellular events observed in total Akt-deficient livers were a marked reduction in cell proliferation, cell hypertrophy, glycogenesis, and lipid droplet formation. Most importantly, liver-specific deletion of FoxO1, a transcription factor regulated by Akt, rescued the hepatic regenerative capability in Akt1-deficient and Akt2-deficient mice and normalized the cellular events associated with liver regeneration. CONCLUSION: The Akt-FoxO1 signaling pathway plays an essential role during liver regeneration.


Asunto(s)
Factores de Transcripción Forkhead/fisiología , Regeneración Hepática , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/fisiología , Animales , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/antagonistas & inhibidores , Hepatocitos/patología , Hiperplasia , Metabolismo de los Lípidos , Masculino , Ratones , Transducción de Señal/fisiología
8.
Curr Opin Lipidol ; 27(6): 623-629, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27755115

RESUMEN

PURPOSE OF REVIEW: Better tools are sorely needed for both the prevention and treatment of cardiovascular diseases, which account for more than one-third of the deaths in Western countries. MicroRNAs typically regulate the expression of several mRNAs involved in the same biological process. Therapeutic manipulation of miRNAs could restore the expression of multiple players within the same physiologic pathway, and ideally offer better curative outcomes than conventional approaches that target only one single player within the pathway. This review summarizes available studies on the prospective value of targeting miRNAs to prevent dyslipidemia and atherogenesis. RECENT FINDINGS: Silencing the expression of miRNAs that target key genes involved in lipoprotein metabolism in vivo with antisense oligonucleotides results in the expected de-repression of target mRNAs in liver and atherosclerotic plaques. However, the consequences of long-term antimiRNA treatment on both circulating lipoproteins and athero-protection are yet to be established. SUMMARY: A number of studies have demonstrated the efficacy of miRNA mimics and inhibitors as novel therapeutic tools for treating dyslipidemia and cardiovascular diseases. Nevertheless, concerns over unanticipated side-effects related to de-repression of additional targets should not be overlooked for miRNA-based therapies.


Asunto(s)
Aterosclerosis/genética , Aterosclerosis/metabolismo , Metabolismo de los Lípidos/genética , MicroARNs/genética , Animales , Transporte Biológico/genética , Colesterol/metabolismo , Humanos
9.
Hepatology ; 61(4): 1227-38, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25418138

RESUMEN

UNLABELLED: The cell death-inducing DNA fragmentation factor alpha-like effector c (CIDEC; also known in rodents as FSP27 or fat-specific protein 27) is a lipid droplet-associated protein that promotes intracellular triglyceride (TAG) storage. CIDEC/Fsp27 is highly expressed in adipose tissue, but undetectable in normal liver. However, its hepatic expression rises during fasting or under genetic or diet-induced hepatosteatosis in both mice and patients. Herein, we demonstrate that CIDEC/Fsp27 is a direct transcriptional target of the nuclear receptor PPARα (peroxisome proliferator-activated receptor alpha) in both mouse and human hepatocytes, and that preventing Fsp27 induction accelerates PPARα-stimulated fatty acid oxidation. We show that adenoviral-mediated silencing of hepatic Fsp27 abolishes fasting-induced liver steatosis in the absence of changes in plasma lipids. Finally, we report that anti-Fsp27 short hairpin RNA and PPARα agonists synergize to ameliorate hepatosteatosis in mice fed a high fat diet. CONCLUSIONS: Together, our data highlight the physiological importance of CIDEC/Fsp27 in TAG homeostasis under both physiological and pathological liver steatosis. Our results also suggest that patients taking fibrates likely have elevated levels of hepatic CIDEC, which may limit the efficient mobilization and catabolism of hepatic TAGs.


Asunto(s)
Hígado Graso/etiología , PPAR alfa/fisiología , Proteínas/fisiología , Animales , Células Cultivadas , Dieta , Ayuno , Hepatocitos , Humanos , Hígado , Ratones
10.
Circ Res ; 115(1): 10-22, 2014 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-24753547

RESUMEN

RATIONALE: Several reports suggest that antisense oligonucleotides against miR-33 might reduce cardiovascular risk in patients by accelerating the reverse cholesterol transport pathway. However, conflicting reports exist about the impact of anti-miR-33 therapy on the levels of very low-density lipoprotein-triglycerides (VLDL-TAG). OBJECTIVE: We test the hypothesis that miR-33 controls hepatic VLDL-TAG secretion. METHODS AND RESULTS: Using therapeutic silencing of miR-33 and adenoviral overexpression of miR-33, we show that miR-33 limits hepatic secretion of VLDL-TAG by targeting N-ethylmaleimide-sensitive factor (NSF), both in vivo and in primary hepatocytes. We identify conserved sequences in the 3'UTR of NSF as miR-33 responsive elements and show that Nsf is specifically recruited to the RNA-induced silencing complex following induction of miR-33. In pulse-chase experiments, either miR-33 overexpression or knock-down of Nsf lead to decreased secretion of apolipoproteins and TAG in primary hepatocytes, compared with control cells. Importantly, Nsf rescues miR-33-dependent reduced secretion. Finally, we show that overexpression of Nsf in vivo increases global hepatic secretion and raises plasma VLDL-TAG. CONCLUSIONS: Together, our data reveal key roles for the miR-33-NSF axis during hepatic secretion and suggest that caution should be taken with anti-miR-33-based therapies because they might raise proatherogenic VLDL-TAG levels.


Asunto(s)
Lipoproteínas VLDL/metabolismo , MicroARNs/fisiología , Proteínas Sensibles a N-Etilmaleimida/fisiología , Triglicéridos/metabolismo , Animales , Apolipoproteína B-100 , Apolipoproteínas B/metabolismo , Apolipoproteínas B/fisiología , Proteínas Portadoras/fisiología , Hepatocitos/metabolismo , Lipoproteínas VLDL/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de LDL/fisiología , Proteína 2 de Unión a Elementos Reguladores de Esteroles/fisiología , Triglicéridos/sangre
11.
J Immunol ; 193(11): 5637-48, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25339664

RESUMEN

Many metabolic diseases, including atherosclerosis, type 2 diabetes, pulmonary alveolar proteinosis, and obesity, have a chronic inflammatory component involving both innate and adaptive immunity. Mice lacking the ATP-binding cassette transporter G1 (ABCG1) develop chronic inflammation in the lungs, which is associated with the lipid accumulation (cholesterol, cholesterol ester, and phospholipid) and cholesterol crystal deposition that are characteristic of atherosclerotic lesions and pulmonary alveolar proteinosis. In this article, we demonstrate that specific lipids, likely oxidized phospholipids and/or sterols, elicit a lung-specific immune response in Abcg1(-/-) mice. Loss of ABCG1 results in increased levels of specific oxysterols, phosphatidylcholines, and oxidized phospholipids, including 1-palmitoyl-2-(5'-oxovaleroyl)-sn-glycero-3-phosphocholine, in the lungs. Further, we identify a niche-specific increase in natural Ab (NAb)-secreting B-1 B cells in response to this lipid accumulation that is paralleled by increased titers of IgM, IgA, and IgG against oxidation-specific epitopes, such as those on oxidized low-density lipoprotein and malondialdehyde-modified low-density lipoprotein. Finally, we identify a cytokine/chemokine signature that is reflective of increased B cell activation, Ab secretion, and homing. Collectively, these data demonstrate that the accumulation of lipids in Abcg1(-/-) mice induces the specific expansion and localization of B-1 B cells, which secrete NAbs that may help to protect against the development of atherosclerosis. Indeed, despite chronic lipid accumulation and inflammation, hyperlipidemic mice lacking ABCG1 develop smaller atherosclerotic lesions compared with controls. These data also suggest that Abcg1(-/-) mice may represent a new model in which to study the protective functions of B-1 B cells/NAbs and suggest novel targets for pharmacologic intervention and treatment of disease.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Aterosclerosis/inmunología , Subgrupos de Linfocitos B/inmunología , Linfocitos B/inmunología , Lipoproteínas/metabolismo , Pulmón/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/genética , Traslado Adoptivo , Animales , Anticuerpos/metabolismo , Proteínas Aviares/metabolismo , Subgrupos de Linfocitos B/trasplante , Linfocitos B/trasplante , Células Cultivadas , Citocinas/metabolismo , Perfilación de la Expresión Génica , Homeostasis/genética , Errores Innatos del Metabolismo Lipídico/genética , Lipoproteínas/genética , Pulmón/inmunología , Activación de Linfocitos , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Fosfolípidos/metabolismo
12.
Circ Res ; 112(12): 1602-12, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23519696

RESUMEN

RATIONALE: The bile acid receptor farnesoid X receptor (FXR) regulates many aspects of lipid metabolism by variouscomplex and incompletely understood molecular mechanisms. We set out to investigate the molecular mechanisms for FXR-dependent regulation of lipid and lipoprotein metabolism. OBJECTIVE: To identify FXR-regulated microRNAs that were subsequently involved in regulating lipid metabolism. METHODS AND RESULTS: ATP binding cassette transporter A1 (ABCA1) is a major determinant of plasma high-density lipoprotein (HDL)-cholesterol levels. Here, we show that activation of the nuclear receptor FXR in vivo increases hepatic levels of miR-144, which in turn lowers hepatic ABCA1 and plasma HDL levels. We identified 2 complementary sequences to miR-144 in the 3' untranslated region of ABCA1 mRNA that are necessary for miR-144-dependent regulation. Overexpression of miR-144 in vitro decreased both cellular ABCA1 protein and cholesterol efflux to lipid-poor apolipoprotein A-I protein, whereas overexpression in vivo reduced hepatic ABCA1 protein and plasma HDL-cholesterol. Conversely, silencing miR-144 in mice increased hepatic ABCA1 protein and HDL-cholesterol. In addition, we used tissue-specific FXR-deficient mice to show that induction of miR-144 and FXR-dependent hypolipidemia requires hepatic, but not intestinal, FXR. Finally, we identified functional FXR response elements upstream of the miR-144 locus, consistent with direct FXR regulation. CONCLUSIONS: We have identified a novel pathway involving FXR, miR-144, and ABCA1 that together regulate plasma HDL-cholesterol.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , HDL-Colesterol/sangre , Hepatocitos/efectos de los fármacos , Isoxazoles/farmacología , MicroARNs/metabolismo , Quinolinas/farmacología , Receptores Citoplasmáticos y Nucleares/agonistas , Regiones no Traducidas 3' , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Animales , Apolipoproteína A-I/metabolismo , Línea Celular Tumoral , Regulación de la Expresión Génica , Células HEK293 , Hepatocitos/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Interferencia de ARN , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Elementos de Respuesta , Factores de Tiempo , Transfección
13.
J Hepatol ; 61(2): 358-65, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24768901

RESUMEN

BACKGROUND & AIMS: Perilipin-5 (PLIN5) is a member of the perilipin family of lipid droplet (LD)-associated proteins. PLIN5 is expressed in oxidative tissues including the liver, and is critical during LD biogenesis. Studies showed that statins reduce hepatic triglyceride contents in some patients with non-alcoholic fatty liver disease and in rodent models of diet-induced hepatosteatosis. Whether statins alter triglyceride synthesis, storage, and/or utilization within the hepatocyte is unknown, though. Here we tested the hypothesis that statins alter the metabolism of LD in the hepatocyte during physiological conditions, such as fasting-induced steatosis. METHODS: Mice were gavaged with saline or atorvastatin, and the expression of LD-associated genes was determined in fed and fasted animals. The accumulation of triglycerides and LD was studied in mouse or human primary hepatocytes in response to statins, and following knock-down of SREBP2 or PLIN5. RESULTS: We show that statins decrease the levels of PLIN5, but not other LD-associated genes, in both mouse liver and mouse/human primary hepatocytes, which is paralleled by a significant reduction in both intracellular triglycerides and the number of LD. We identify an atypical negative sterol regulatory sequence in the proximal promoter of mouse/human PLIN5 that recruits the transcription factor SREBP2 and confers response to statins. Finally, we show that the statin-dependent reduction of hepatocyte triglyceride contents is mimicked by partial knock-down of PLIN5; conversely, ectopic overexpression of PLIN5 reverts the statin effect. CONCLUSIONS: PLIN5 is a physiological regulator of triglyceride metabolism in the liver, and likely contributes to the pleiotropic effects of statins.


Asunto(s)
Hepatocitos/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas Musculares/fisiología , Triglicéridos/metabolismo , Animales , Hepatocitos/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/análisis , Gotas Lipídicas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Musculares/análisis , Proteína 2 de Unión a Elementos Reguladores de Esteroles/fisiología
14.
Arterioscler Thromb Vasc Biol ; 33(3): 455-8, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23288159

RESUMEN

OBJECTIVE: To determine the efficacy of long-term anti-miR-33 therapy on the progression of atherosclerosis in high-fat, high-cholesterol-fed Ldlr(-/-) mice. METHODS AND RESULTS: Ldlr(-/-) mice received saline, or control or anti-miR-33 oligonucleotides once a week for 14 weeks. The treatment was effective, as measured by reduced levels of hepatic miR-33 and increased hepatic expression of miR-33 targets. Analysis of plasma samples revealed an initial elevation in high-density lipoprotein cholesterol after 2 weeks of treatment that was not sustained by the end of the experiment. Additionally, we found a significant increase in circulating triglycerides in anti-miR-33-treated mice, compared with controls. Finally, examination of atheromata revealed no significant changes in the size or composition of lesions between the 3 groups. CONCLUSIONS: Prolonged silencing of miR-33 fails to maintain elevated plasma high-density lipoprotein cholesterol and does not prevent the progression of atherosclerosis in Ldlr(-/-) mice.


Asunto(s)
Aorta/metabolismo , Enfermedades de la Aorta/terapia , Aterosclerosis/terapia , Hígado/metabolismo , MicroARNs/metabolismo , Oligonucleótidos Antisentido/metabolismo , Receptores de LDL/deficiencia , Animales , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Biomarcadores/sangre , Colesterol en la Dieta/sangre , HDL-Colesterol/sangre , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación hacia Abajo , Masculino , Ratones , Ratones Noqueados , MicroARNs/genética , Receptores de LDL/genética , Factores de Tiempo , Triglicéridos/sangre
15.
J Hepatol ; 59(4): 731-7, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23712050

RESUMEN

BACKGROUND & AIMS: It is widely recognized that in the early stages of liver regeneration after partial hepatectomy, the hepatocytes accumulate a significant amount of lipids. The functional meaning of this transient steatosis and its effect on hepatocellular proliferation are not well defined. In addition, the basic mechanisms of this lipid accumulation are not well understood although some studies suggest the participation of the Low Density Lipoprotein Receptor (Ldlr). METHODS: To address these questions, we studied the process of liver regeneration in Ldlr null mice and wild type mice following partial hepatectomy. RESULTS: Ldlr deficiency was associated with a significant decrease in serum albumin concentration, during early stages of liver regeneration, and a delayed hepatic regeneration. Remnant livers of Ldlr(-)(/)(-) showed a time-shifted expression of interleukin-6 (IL6) and a defective activation of tumor necrosis factor-α (TNFα) and hepatocyte growth factor (HGF) expression in early phases of liver regeneration. Unexpectedly, Ldlr(-)(/)(-) showed no significant differences in the content of lipid droplets after partial hepatectomy compared to wild type mice. However, lipidomic analysis of the regenerating liver from Ldlr(-)(/)(-) revealed a lipid profile compatible with liver quiescence: high content of cholesterol esters and ceramide, and low levels of phosphatidylcholine. CONCLUSIONS: Ldlr deficiency is associated with significant changes in the hepatic lipidome that affect cytokine-growth factor signaling and impair liver regeneration. These results suggest that the analysis of the hepatic lipidome may help predict the success of liver regeneration in the clinical environment, specifically in the context of pre-existing liver steatosis.


Asunto(s)
Metabolismo de los Lípidos , Regeneración Hepática/fisiología , Hígado/fisiopatología , Receptores de LDL/deficiencia , Animales , Puntos de Control del Ciclo Celular , Colesterol/sangre , Colesterol/metabolismo , Hígado Graso/etiología , Hígado Graso/metabolismo , Hígado Graso/patología , Hepatectomía/efectos adversos , Factor de Crecimiento de Hepatocito/metabolismo , Hipercolesterolemia/etiología , Hipercolesterolemia/metabolismo , Interleucina-6/metabolismo , Hígado/patología , Regeneración Hepática/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de LDL/genética , Receptores de LDL/fisiología , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
16.
Anal Biochem ; 433(1): 56-64, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23072980

RESUMEN

Cholesterol efflux from macrophages and the vascular wall is the initial step of the cardiovascular protective reverse cholesterol transport process. This study demonstrates a mass spectrometry based assay to measure the cellular and medium content of [d(7)]cholesterol and unlabeled cholesterol that can be used to measure cholesterol efflux from cell lines. Using a triple-quadrupole electrospray ionization-MS instrument in direct infusion mode, product ion scanning for m/z 83, neutral loss (NL) 375.5 scanning, and NL 368.5 scanning were used to detect cholesterol (as an acetylated derivative), [d(7)]cholesteryl ester (CE), and unlabeled CE, respectively. The same mass of [d(7)]cholesterol was substituted for [(3)H]cholesterol under standard efflux assay conditions. At the end of [d(7)]cholesterol loading, the intracellular mass of [d(7)]cholesterol was twofold greater than that of unlabeled cholesterol, and the intracellular [d(7)]CE profile was similar to that of unlabeled CE. Efflux of cholesterol to apolipoprotein A-I and high-density lipoproteins was similar comparing efflux of either [d(7)]cholesterol or [(3)H]cholesterol as measured by following efflux of the tracers only. This technique also can be used to assess the efflux of unlabeled cholesterol to acceptors in medium that are initially cholesterol-free (e.g., apolipoprotein A-I). Taken together, this mass spectrometry-based assay provides new molecular detail to assess cholesterol efflux.


Asunto(s)
Colesterol/metabolismo , Espectrometría de Masa por Ionización de Electrospray , Acetilación , Animales , Transporte Biológico , Línea Celular , Colesterol/química , Ésteres del Colesterol/química , Ésteres del Colesterol/metabolismo , Deuterio/química , Macrófagos/metabolismo , Ratones , Sodio/química
17.
Proc Natl Acad Sci U S A ; 107(27): 12228-32, 2010 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-20566875

RESUMEN

The sterol regulatory element binding protein 2 (SREBP-2) and the liver X receptor (LXR) control antagonistic transcriptional programs that stimulate cellular cholesterol uptake and synthesis, and cholesterol efflux, respectively. The clinical importance of SREBP-2 is revealed in patients with hypercholesterolemia treated with statins, which reduce low-density lipoprotein (LDL) cholesterol levels by increasing hepatic expression of SREBP-2 and its target, the LDL receptor. Here we show that miR-33 is encoded within SREBP-2 and that both mRNAs are coexpressed. We also identify sequences in the 3' UTR of ABCA1 and ABCG1, sterol transporter genes both previously shown to be regulated by LXR, as targets for miR-33-mediated silencing. Our data show that LXR-dependent cholesterol efflux to both ApoAI and serum is ameliorated by miR-33 overexpression and, conversely, stimulated by miR-33 silencing. Finally, we show that ABCA1 mRNA and protein and plasma HDL levels decline after hepatic overexpression of miR-33, whereas they increase after hepatic miR-33 silencing. These results suggest novel ways to manage hypercholesterolemic patients.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , MicroARNs/genética , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética , Regiones no Traducidas 5'/genética , Transportador 1 de Casete de Unión a ATP , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Secuencia de Bases , Línea Celular , Colesterol/sangre , Colesterol/metabolismo , HDL-Colesterol/sangre , Regulación de la Expresión Génica , Humanos , Lipoproteínas LDL/sangre , Lipoproteínas LDL/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Elementos de Respuesta/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Activación Transcripcional , Transfección
18.
Mo Med ; 110(4): 325-30, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24003651

RESUMEN

The Cho and Baldan labs focus their efforts on novel pathways that control atherogenesis. MIF (Macrophage migration inhibitory factor) recruits macrophages to atherosclerotic lesions and activates the production of matrix proteinases, which in turn destabilize atherosclerotic plaques. On the other hand, miR-33 coordinates the expression of several sterol transporters essential for high-density lipoprotein metabolism and bile secretion. Thus, both MIF and miR-33 are promising therapeutic targets to manage patients at risk of developing atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , MicroARNs/metabolismo , Aterosclerosis/genética , Biomarcadores/metabolismo , HDL-Colesterol/metabolismo , Humanos
19.
bioRxiv ; 2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-38948707

RESUMEN

Low nephron endowment at birth is a risk factor for chronic kidney disease. The prevalence of this condition is increasing due to higher survival rates of preterm infants and children with multi- organ birth defect syndromes that affect the kidney and urinary tract. We created a mouse model of congenital low nephron number due to deletion of Mta2 in nephron progenitor cells. Mta2 is a core component of the Nucleosome Remodeling and Deacetylase (NuRD) chromatin remodeling complex. These mice developed albuminuria at 4 weeks of age followed by focal segmental glomerulosclerosis (FSGS) at 8 weeks, with progressive kidney injury and fibrosis. Our studies reveal that altered mitochondrial metabolism in the post-natal period leads to accumulation of neutral lipids in glomeruli at 4 weeks of age followed by reduced mitochondrial oxygen consumption. We found that NuRD cooperated with Zbtb7a/7b to regulate a large number of metabolic genes required for fatty acid oxidation and oxidative phosphorylation. Analysis of human kidney tissue also supported a role for reduced mitochondrial lipid metabolism and ZBTB7A/7B in FSGS and CKD. We propose that an inability to meet the physiological and metabolic demands of post-natal somatic growth of the kidney promotes the transition to CKD in the setting of glomerular hypertrophy due to low nephron endowment.

20.
Cell Metab ; 1(2): 121-31, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16054053

RESUMEN

Here we demonstrate that the ABC transporter ABCG1 plays a critical role in lipid homeostasis by controlling both tissue lipid levels and the efflux of cellular cholesterol to HDL. Targeted disruption of Abcg1 in mice has no effect on plasma lipids but results in massive accumulation of both neutral lipids and phospholipids in hepatocytes and in macrophages within multiple tissues following administration of a high-fat and -cholesterol diet. In contrast, overexpression of human ABCG1 protects murine tissues from dietary fat-induced lipid accumulation. Finally, we show that cholesterol efflux to HDL specifically requires ABCG1, whereas efflux to apoA1 requires ABCA1. These studies identify Abcg1 as a key gene involved in both cholesterol efflux to HDL and in tissue lipid homeostasis.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Colesterol/metabolismo , Regulación de la Expresión Génica , Metabolismo de los Lípidos , Lipoproteínas HDL/metabolismo , Lipoproteínas/fisiología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Animales , Compuestos Azo/farmacología , Colorantes/farmacología , Femenino , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Hibridación in Situ , Operón Lac , Hígado/metabolismo , Pulmón/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN/metabolismo , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA