Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 295(52): 18179-18188, 2020 12 25.
Artículo en Inglés | MEDLINE | ID: mdl-33097593

RESUMEN

Bone morphogenetic protein-9 (BMP-9) is a circulating cytokine that is known to play an essential role in the endothelial homeostasis and the binding of BMP-9 to the receptor activin-like kinase 1 (ALK-1) promotes endothelial cell quiescence. Previously, using an unbiased screen, we identified ALK-1 as a high-capacity receptor for low-density lipoprotein (LDL) in endothelial cells that mediates its transcytosis in a nondegradative manner. Here we examine the crosstalk between BMP-9 and LDL and how it influences their interactions with ALK-1. Treatment of endothelial cells with BMP-9 triggers the extensive endocytosis of ALK-1, and it is mediated by caveolin-1 (CAV-1) and dynamin-2 (DNM2) but not clathrin heavy chain. Knockdown of CAV-1 reduces BMP-9-mediated internalization of ALK-1, BMP-9-dependent signaling and gene expression. Similarly, treatment of endothelial cells with LDL reduces BMP-9-induced SMAD1/5 phosphorylation and gene expression and silencing of CAV-1 and DNM2 diminishes LDL-mediated ALK-1 internalization. Interestingly, BMP-9-mediated ALK-1 internalization strongly re-duces LDL transcytosis to levels seen with ALK-1 deficiency. Thus, BMP-9 levels can control cell surface levels of ALK-1, via CAV-1, to regulate both BMP-9 signaling and LDL transcytosis.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Caveolina 1/metabolismo , Membrana Celular/metabolismo , Endocitosis , Endotelio Vascular/fisiología , Factor 2 de Diferenciación de Crecimiento/metabolismo , Lipoproteínas LDL/metabolismo , Receptores de Activinas Tipo II/genética , Caveolina 1/genética , Células Cultivadas , Endotelio Vascular/citología , Factor 2 de Diferenciación de Crecimiento/genética , Humanos , Fosforilación , Transducción de Señal
2.
Arterioscler Thromb Vasc Biol ; 40(6): 1510-1522, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32349535

RESUMEN

OBJECTIVE: Endothelial Cav-1 (caveolin-1) expression plays a relevant role during atherogenesis by controlling NO production, vascular inflammation, LDL (low-density lipoprotein) transcytosis, and extracellular matrix remodeling. Additional studies have identified cholesterol-rich membrane domains as important regulators of autophagy by recruiting ATGs (autophagy-related proteins) to the plasma membrane. Here, we investigate how the expression of Cav-1 in the aortic endothelium influences autophagy and whether enhanced autophagy contributes to the atheroprotective phenotype observed in Cav-1-deficient mice. Approach and Results: To analyze the impact of Cav-1 deficiency on regulation of autophagy in the aortic endothelium during the progression of atherosclerosis, we fed Ldlr-/- and Cav-1-/-Ldlr-/- mice a Western diet and assessed autophagy in the vasculature. We observe that the absence of Cav-1 promotes autophagy activation in athero-prone areas of the aortic endothelium by enhancing autophagic flux. Mechanistically, we found that Cav-1 interacts with the ATG5-ATG12 complex and influences the cellular localization of autophagosome components in lipid rafts, which controls the autophagosome formation and autophagic flux. Pharmacological inhibition of autophagy attenuates the atheroprotection observed in Cav-1-/- mice by increasing endothelial inflammation and macrophage recruitment, identifying a novel molecular mechanism by which Cav-1 deficiency protects against the progression of atherosclerosis. CONCLUSIONS: These results identify Cav-1 as a relevant regulator of autophagy in the aortic endothelium and demonstrate that pharmacological suppression of autophagic flux in Cav-1-deficient mice attenuates the atheroprotection observed in Cav-1-/- mice. Additionally, these findings suggest that activation of endothelial autophagy by blocking Cav-1 might provide a potential therapeutic strategy for cardiovascular diseases including atherosclerosis.


Asunto(s)
Aterosclerosis/prevención & control , Autofagia/fisiología , Caveolina 1/deficiencia , Endotelio Vascular/fisiopatología , Vasculitis/prevención & control , Adenina/análogos & derivados , Adenina/farmacología , Animales , Aorta/patología , Aorta/fisiopatología , Aorta/ultraestructura , Aterosclerosis/etiología , Autofagia/efectos de los fármacos , Caveolina 1/análisis , Caveolina 1/fisiología , Dieta Occidental , Células Endoteliales/química , Células Endoteliales/fisiología , Células Endoteliales/ultraestructura , Endotelio Vascular/química , Endotelio Vascular/ultraestructura , Femenino , Humanos , Masculino , Microdominios de Membrana/química , Microdominios de Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células 3T3 NIH , Receptores de LDL/deficiencia
3.
Circulation ; 140(3): 225-239, 2019 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-31154825

RESUMEN

BACKGROUND: Atherosclerosis is driven by synergistic interactions between pathological, biomechanical, inflammatory, and lipid metabolic factors. Our previous studies demonstrated that absence of caveolin-1 (Cav1)/caveolae in hyperlipidemic mice strongly inhibits atherosclerosis, which was attributed to activation of endothelial nitric oxide (NO) synthase (eNOS) and increased production of NO and reduced inflammation and low-density lipoprotein trafficking. However, the contribution of eNOS activation and NO production in the athero-protection of Cav1 and the exact mechanisms by which Cav1/caveolae control the pathogenesis of diet-induced atherosclerosis are still not clear. METHODS: Triple-knockout mouse lacking expression of eNOS, Cav1, and Ldlr were generated to explore the role of NO production in Cav1-dependent athero-protective function. The effects of Cav1 on lipid trafficking, extracellular matrix remodeling, and vascular inflammation were studied both in vitro and in vivo with a mouse model of diet-induced atherosclerosis. The expression of Cav1 and distribution of caveolae regulated by flow were analyzed by immunofluorescence staining and transmission electron microscopy. RESULTS: We found that absence of Cav1 significantly suppressed atherogenesis in Ldlr-/-eNOS-/- mice, demonstrating that athero-suppression is independent of increased NO production. Instead, we find that the absence of Cav1/caveolae inhibited low-density lipoprotein transport across the endothelium and proatherogenic fibronectin deposition and disturbed flow-mediated endothelial cell inflammation. Consistent with the idea that Cav1/caveolae may play a role in early flow-dependent inflammatory priming, distinct patterns of Cav1 expression and caveolae distribution were observed in athero-prone and athero-resistant areas of the aortic arch even in wild-type mice. CONCLUSIONS: These findings support a role for Cav1/caveolae as a central regulator of atherosclerosis that links biomechanical, metabolic, and inflammatory pathways independently of endothelial eNOS activation and NO production.


Asunto(s)
Aterosclerosis/metabolismo , Caveolina 1/fisiología , Endotelio Vascular/metabolismo , Lipoproteínas LDL/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Transcitosis/fisiología , Animales , Aterosclerosis/patología , Aterosclerosis/prevención & control , Células Cultivadas , Perros , Endotelio Vascular/patología , Activación Enzimática/fisiología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
4.
Int J Mol Sci ; 21(22)2020 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-33212990

RESUMEN

Insulin resistance (IR) is one of the key contributing factors in the development of type 2 diabetes mellitus (T2DM). However, the molecular mechanisms leading to IR are still unclear. The implication of microRNAs (miRNAs) in the pathophysiology of multiple cardiometabolic pathologies, including obesity, atherosclerotic heart failure and IR, has emerged as a major focus of interest in recent years. Indeed, upregulation of several miRNAs has been associated with obesity and IR. Among them, miR-27b is overexpressed in the liver in patients with obesity, but its role in IR has not yet been thoroughly explored. In this study, we investigated the role of miR-27b in regulating insulin signaling in hepatocytes, both in vitro and in vivo. Therefore, assessment of the impact of miR-27b on insulin resistance through the hepatic tissue is of special importance due to the high expression of miR-27b in the liver together with its known role in regulating lipid metabolism. Notably, we found that miR-27b controls post-transcriptional expression of numerous components of the insulin signaling pathway including the insulin receptor (INSR) and insulin receptor substrate 1 (IRS1) in human hepatoma cells. These results were further confirmed in vivo showing that overexpression and inhibition of hepatic miR-27 enhances and suppresses hepatic INSR expression and insulin sensitivity, respectively. This study identified a novel role for miR-27 in regulating insulin signaling, and this finding suggests that elevated miR-27 levels may contribute to early development of hepatic insulin resistance.


Asunto(s)
Hepatocitos/metabolismo , Insulina/metabolismo , Receptor de Insulina/metabolismo , Transducción de Señal , Línea Celular , Hepatocitos/citología , Humanos , Insulina/genética , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Receptor de Insulina/genética
5.
J Neurosci ; 35(44): 14717-26, 2015 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-26538644

RESUMEN

Dysregulation of amyloid-ß (Aß) metabolism is critical for Alzheimer's disease (AD) pathogenesis. Mounting evidence suggests that apolipoprotein E (ApoE) is involved in Aß metabolism. ATP-binding cassette transporter A1 (ABCA1) is a key regulator of ApoE lipidation, which affects Aß levels. Therefore, identifying regulatory mechanisms of ABCA1 expression in the brain may provide new therapeutic targets for AD. Here, we demonstrate that microRNA-33 (miR-33) regulates ABCA1 and Aß levels in the brain. Overexpression of miR-33 impaired cellular cholesterol efflux and dramatically increased extracellular Aß levels by promoting Aß secretion and impairing Aß clearance in neural cells. In contrast, genetic deletion of mir-33 in mice dramatically increased ABCA1 levels and ApoE lipidation, but it decreased endogenous Aß levels in cortex. Most importantly, pharmacological inhibition of miR-33 via antisense oligonucleotide specifically in the brain markedly decreased Aß levels in cortex of APP/PS1 mice, representing a potential therapeutic strategy for AD. SIGNIFICANCE STATEMENT: Brain lipid metabolism, in particular Apolipoprotein E (ApoE) lipidation, is critical to Aß metabolism and Alzheimer's disease (AD). Brain lipid metabolism is largely separated from the periphery due to blood-brain barrier and different repertoire of lipoproteins. Therefore, identifying the novel regulatory mechanism of brain lipid metabolism may provide a new therapeutic strategy for AD. Although there have been studies on brain lipid metabolism, its regulation, in particular by microRNAs, is relatively unknown. Here, we demonstrate that inhibition of microRNA-33 increases lipidation of brain ApoE and reduces Aß levels by inducing ABCA1. We provide a unique approach for AD therapeutics to increase ApoE lipidation and reduce Aß levels via pharmacological inhibition of microRNA in vivo.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Apolipoproteínas E/metabolismo , Encéfalo/metabolismo , Metabolismo de los Lípidos/fisiología , MicroARNs/fisiología , Péptidos beta-Amiloides/genética , Animales , Secuencia de Bases , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular
6.
FASEB J ; 29(2): 597-610, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25392271

RESUMEN

Atherosclerosis is the major cause of death and disability in diabetic and obese subjects with insulin resistance. Akt2, a phosphoinositide-dependent serine-threonine protein kinase, is highly express in insulin-responsive tissues; however, its role during the progression of atherosclerosis remains unknown. Thus, we aimed to investigate the contribution of Akt2 during the progression of atherosclerosis. We found that germ-line Akt2-deficient mice develop similar atherosclerotic plaques as wild-type mice despite higher plasma lipids and glucose levels. It is noteworthy that transplantation of bone marrow cells isolated from Akt2(-/-) mice to Ldlr(-/-) mice results in marked reduction of the progression of atherosclerosis compared with Ldlr(-/-) mice transplanted with wild-type bone marrow cells. In vitro studies indicate that Akt2 is required for macrophage migration in response to proatherogenic cytokines (monocyte chemotactic protein-1 and macrophage colony-stimulating factor). Moreover, Akt2(-/-) macrophages accumulate less cholesterol and have an alternative activated or M2-type phenotype when stimulated with proinflammatory cytokines. Together, these results provide evidence that macrophage Akt2 regulates migration, the inflammatory response and cholesterol metabolism and suggest that targeting Akt2 in macrophages might be beneficial for treating atherosclerosis.


Asunto(s)
Aterosclerosis/fisiopatología , Proteínas Proto-Oncogénicas c-akt/deficiencia , Proteínas Proto-Oncogénicas c-akt/genética , Animales , Glucemia/metabolismo , Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Movimiento Celular , Colesterol/metabolismo , Citocinas/metabolismo , Progresión de la Enfermedad , Inflamación , Insulina/química , Leucocitos/citología , Lípidos/sangre , Lipoproteínas LDL/metabolismo , Macrófagos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Microscopía Fluorescente , Placa Aterosclerótica , Receptores de LDL/genética
7.
Circ Res ; 112(12): 1592-601, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23519695

RESUMEN

RATIONALE: Foam cell formation because of excessive accumulation of cholesterol by macrophages is a pathological hallmark of atherosclerosis, the major cause of morbidity and mortality in Western societies. Liver X nuclear receptors (LXRs) regulate the expression of the adenosine triphosphate-binding cassette (ABC) transporters, including adenosine triphosphate-binding cassette transporter A1 (ABCA1) and adenosine triphosphate-binding cassette transporter G1 (ABCG1). ABCA1 and ABCG1 facilitate the efflux of cholesterol from macrophages and regulate high-density lipoprotein (HDL) biogenesis. Increasing evidence supports the role of microRNA (miRNAs) in regulating cholesterol metabolism through ABC transporters. OBJECTIVE: We aimed to identify novel miRNAs that regulate cholesterol metabolism in macrophages stimulated with LXR agonists. METHODS AND RESULTS: To map the miRNA expression signature of macrophages stimulated with LXR agonists, we performed an miRNA profiling microarray analysis in primary mouse peritoneal macrophages stimulated with LXR ligands. We report that LXR ligands increase miR-144 expression in macrophages and mouse livers. Overexpression of miR-144 reduces ABCA1 expression and attenuates cholesterol efflux to apolipoproteinA1 in macrophages. Delivery of miR-144 oligonucleotides to mice attenuates ABCA1 expression in the liver, reducing HDL levels. Conversely, silencing of miR-144 in mice increases the expression of ABCA1 and plasma HDL levels. Thus, miR-144 seems to regulate both macrophage cholesterol efflux and HDL biogenesis in the liver. CONCLUSIONS: miR-144 regulates cholesterol metabolism via suppressing ABCA1 expression and modulation of miRNAs may represent a potential therapeutical intervention for treating dyslipidemia and atherosclerotic vascular disease.


Asunto(s)
HDL-Colesterol/sangre , Hepatocitos/metabolismo , Macrófagos/metabolismo , MicroARNs/metabolismo , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Anticolesterolemiantes/farmacología , Apolipoproteína A-I/metabolismo , Células COS , Chlorocebus aethiops , Dieta Alta en Grasa , Perfilación de la Expresión Génica/métodos , Células Hep G2 , Hepatocitos/efectos de los fármacos , Homeostasis , Humanos , Hidrocarburos Fluorados/farmacología , Receptores X del Hígado , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Oligonucleótidos/metabolismo , Receptores Nucleares Huérfanos/agonistas , Receptores Nucleares Huérfanos/genética , Receptores Nucleares Huérfanos/metabolismo , Sulfonamidas/farmacología
8.
J Lipid Res ; 55(6): 1066-76, 2014 06.
Artículo en Inglés | MEDLINE | ID: mdl-24729624

RESUMEN

ABCA1 is a major regulator of cellular cholesterol efflux and plasma HDL biogenesis. Even though the transcriptional activation of ABCA1 is well established, the posttranscriptional regulation of ABCA1 expression is poorly understood. Here, we investigate the potential contribution of the RNA binding protein (RBP) human antigen R (HuR) on the posttranscriptional regulation of ABCA1 expression. RNA immunoprecipitation assays demonstrate a direct interaction between HuR and ABCA1 mRNA. We found that HuR binds to the 3' untranslated region of ABCA1 and increases ABCA1 translation, while HuR silencing reduces ABCA1 expression and cholesterol efflux to ApoA1 in human hepatic (Huh-7) and monocytic (THP-1) cells. Interestingly, cellular cholesterol levels regulate the expression, intracellular localization, and interaction between HuR and ABCA1 mRNA. Finally, we found that HuR expression was significantly increased in macrophages from human atherosclerotic plaques, suggesting an important role for this RBP in controlling macrophage cholesterol metabolism in vivo. In summary, we have identified HuR as a novel posttranscriptional regulator of ABCA1 expression and cellular cholesterol homeostasis, thereby opening new avenues for increasing cholesterol efflux from atherosclerotic foam macrophages and raising circulat-ing HDL cholesterol levels.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/biosíntesis , Proteína 1 Similar a ELAV/metabolismo , Regulación de la Expresión Génica , Transportador 1 de Casete de Unión a ATP/genética , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Colesterol/genética , Colesterol/metabolismo , Proteína 1 Similar a ELAV/genética , Células Espumosas/metabolismo , Células Espumosas/patología , Homeostasis , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células THP-1
9.
Crit Rev Clin Lab Sci ; 51(6): 305-20, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25034902

RESUMEN

Metabolic syndrome is a complex metabolic condition caused by abnormal adipose deposition and function, dyslipidemia and hyperglycemia, which affects >47 million American adults and ∼1 million children. Individuals with the metabolic syndrome have essentially twice the risk for developing cardiovascular disease (CVD) and Type 2 diabetes mellitus (T2D), compared to those without the syndrome. In the search for improved and novel therapeutic strategies, microRNAs (miRNA) have been shown to be interesting targets due to their regulatory role on gene networks controlling different crucial aspects of metabolism, including lipid and glucose homeostasis. More recently, the discovery of circulating miRNAs suggest that miRNAs may be involved in facilitating metabolic crosstalk between organs as well as serving as novel biomarkers of diseases, including T2D and atherosclerosis. These findings highlight the importance of miRNAs for regulating pathways that underlie metabolic diseases, and their potential as therapeutic targets for the development of novel treatments.


Asunto(s)
Enfermedades Metabólicas , MicroARNs , Tejido Adiposo/metabolismo , Aterosclerosis , Humanos , Hígado/metabolismo , Síndrome Metabólico , Páncreas/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 33(2): 178-85, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23325474

RESUMEN

Alterations in the metabolic control of lipid and glucose homeostasis predispose an individual to develop cardiometabolic diseases, such as type 2-diabetes mellitus and atherosclerosis. Work over the last years has suggested that microRNAs (miRNAs) play an important role in regulating these physiological processes. The contribution of miRNAs in regulating metabolism is exemplified by miR-33, an intronic miRNA encoded in the Srebp genes. miR-33 controls cellular cholesterol export and fatty acid degradation, whereas its host genes stimulate cholesterol and fatty acid synthesis. Other miRNAs, such as miR-122, also play a critical role in regulating lipid homeostasis by controlling cholesterol synthesis and lipoprotein secretion in the liver. This review article summarizes the recent findings in the field, highlighting the contribution of miRNAs in regulating lipid and glucose metabolism. We will also discuss how the modulation of specific miRNAs may be a promising strategy to treat metabolic diseases.


Asunto(s)
Enfermedades Metabólicas/genética , MicroARNs/metabolismo , Animales , Metabolismo de los Hidratos de Carbono/genética , Regulación de la Expresión Génica , Terapia Genética , Glucosa/metabolismo , Homeostasis/genética , Humanos , Insulina/metabolismo , Metabolismo de los Lípidos/genética , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/terapia , Transducción de Señal/genética
11.
Arterioscler Thromb Vasc Biol ; 33(8): 1973-7, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23702658

RESUMEN

OBJECTIVE: To study the efficacy of anti-miRNA-33 therapy on the progression of atherosclerosis. APPROACH AND RESULTS: Ldlr(-/-) mice were injected subcutaneously with PBS, control, or anti-miR-33 oligonucleotides weekly and fed a Western diet for 12 weeks. At the end of treatment, the expression of miR-33 target genes was increased in the liver and aorta, demonstrating effective inhibition of miR-33 function. Interestingly, plasma high-density lipoprotein (HDL)-cholesterol was significantly increased in anti-miR-33-treated mice but only when they were fed a chow diet. However, HDL isolated from anti-miR-33-treated mice showed an increase cholesterol efflux capacity compared with HDL isolated from nontargeting oligonucleotide-treated mice. Analysis of atherosclerosis revealed a significant reduction of plaque size and macrophage content in mice receiving anti-miR-33. In contrast, no differences in collagen content and necrotic areas were observed among the 3 groups. CONCLUSIONS: Long-term anti-miR-33 therapy significantly reduces the progression of atherosclerosis and improves HDL functionality. The antiatherogenic effect is independent of plasma HDL-cholesterol levels.


Asunto(s)
Aterosclerosis/genética , Aterosclerosis/terapia , Terapia Genética/métodos , MicroARNs/genética , Receptores de LDL/genética , Alimentación Animal , Animales , Aterosclerosis/patología , HDL-Colesterol/sangre , Progresión de la Enfermedad , Silenciador del Gen , Inyecciones Subcutáneas , Ratones , Ratones Noqueados , Oligonucleótidos/genética , Oligonucleótidos/farmacología
12.
Proc Natl Acad Sci U S A ; 108(22): 9232-7, 2011 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-21576456

RESUMEN

Cellular imbalances of cholesterol and fatty acid metabolism result in pathological processes, including atherosclerosis and metabolic syndrome. Recent work from our group and others has shown that the intronic microRNAs hsa-miR-33a and hsa-miR-33b are located within the sterol regulatory element-binding protein-2 and -1 genes, respectively, and regulate cholesterol homeostasis in concert with their host genes. Here, we show that miR-33a and -b also regulate genes involved in fatty acid metabolism and insulin signaling. miR-33a and -b target key enzymes involved in the regulation of fatty acid oxidation, including carnitine O-octaniltransferase, carnitine palmitoyltransferase 1A, hydroxyacyl-CoA-dehydrogenase, Sirtuin 6 (SIRT6), and AMP kinase subunit-α. Moreover, miR-33a and -b also target the insulin receptor substrate 2, an essential component of the insulin-signaling pathway in the liver. Overexpression of miR-33a and -b reduces both fatty acid oxidation and insulin signaling in hepatic cell lines, whereas inhibition of endogenous miR-33a and -b increases these two metabolic pathways. Together, these data establish that miR-33a and -b regulate pathways controlling three of the risk factors of metabolic syndrome, namely levels of HDL, triglycerides, and insulin signaling, and suggest that inhibitors of miR-33a and -b may be useful in the treatment of this growing health concern.


Asunto(s)
Ácidos Grasos/metabolismo , Insulina/metabolismo , MicroARNs/biosíntesis , Animales , Enfermedades Cardiovasculares/metabolismo , Colesterol/metabolismo , Citoplasma/metabolismo , Drosophila melanogaster/metabolismo , Homeostasis , Humanos , Inmunohistoquímica/métodos , Lípidos/química , Fosforilación , Procesamiento Postranscripcional del ARN , Transducción de Señal
13.
Biochim Biophys Acta Gene Regul Mech ; 1866(2): 194938, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37086967

RESUMEN

Dysregulation of cholesterol homeostasis is associated with several pathologies including cardiovascular diseases and neurological disorders such as Alzheimer's disease (AD). MicroRNAs (miRNAs) have emerged as key post-transcriptional regulators of cholesterol metabolism. We previously established the role of miR-7 in regulating insulin resistance and amyloidosis, which represents a common pathological feature between type 2 diabetes and AD. We show here an additional metabolic function of miR-7 in cholesterol biosynthesis. We found that miR-7 blocks the last steps of the cholesterol biosynthetic pathway in vitro by targeting relevant genes including DHCR24 and SC5D posttranscriptionally. Intracranial infusion of miR-7 on an adeno-associated viral vector reduced the expression of DHCR24 in the brain of wild-type mice, supporting in vivo miR-7 targeting. We also found that cholesterol regulates endogenous levels of miR-7 in vitro, correlating with transcriptional regulation through SREBP2 binding to its promoter region. In parallel to SREBP2 inhibition, the levels of miR-7 and hnRNPK (the host gene of miR-7) were concomitantly reduced in brain in a mouse model of Niemann Pick type C1 disease and in murine fatty liver, which are both characterized by intracellular cholesterol accumulation. Taken together, the results establish a novel regulatory feedback loop by which miR-7 modulates cholesterol homeostasis at the posttranscriptional level, an effect that could be exploited for therapeutic interventions against prevalent human diseases.


Asunto(s)
Diabetes Mellitus Tipo 2 , MicroARNs , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Humanos , Ratones , Animales , MicroARNs/genética , MicroARNs/metabolismo , Regulación de la Expresión Génica , Colesterol/metabolismo , Homeostasis , Proteínas del Tejido Nervioso/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo
14.
J Biol Chem ; 286(20): 18066-78, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21402708

RESUMEN

MicroRNAs function as important regulators of gene expression and are commonly linked to development, differentiation, and diseases such as cancer. To better understand their roles in various biological processes, identification of genes targeted by microRNAs is necessary. Although prediction tools have significantly helped with this task, experimental approaches are ultimately required for extensive target search and validation. We employed two independent yet complementary high throughput approaches to map a large set of mRNAs regulated by miR-122, a liver-specific microRNA implicated in regulation of fatty acid and cholesterol metabolism, hepatitis C infection, and hepatocellular carcinoma. The combination of luciferase reporter-based screening and shotgun proteomics resulted in the identification of 260 proteins significantly down-regulated in response to miR-122 in at least one method, 113 of which contain predicted miR-122 target sites. These proteins are enriched for functions associated with the cell cycle, differentiation, proliferation, and apoptosis. Among these miR-122-sensitive proteins, we identified a large group with strong connections to liver metabolism, diseases, and hepatocellular carcinoma. Additional analyses, including examination of consensus binding motifs for both miR-122 and target sequences, provide further insight into miR-122 function.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Genes Relacionados con las Neoplasias , Neoplasias Hepáticas/metabolismo , MicroARNs/metabolismo , Proteínas de Neoplasias/biosíntesis , ARN Neoplásico/metabolismo , Carcinoma Hepatocelular/genética , Línea Celular , Humanos , Neoplasias Hepáticas/genética , MicroARNs/genética , Proteínas de Neoplasias/genética , ARN Neoplásico/genética
15.
Arterioscler Thromb Vasc Biol ; 31(11): 2707-14, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21885853

RESUMEN

OBJECTIVE: The ATP-binding cassette transporter A1 (ABCA1) is a major regulator of macrophage cholesterol efflux and protects cells from excess intracellular cholesterol accumulation; however, the mechanism involved in posttranscriptional regulation of ABCA1 is poorly understood. We previously showed that microRNA-33 (miR-33) is 1 regulator. Here, we investigated the potential contribution of other microRNAs (miRNAs) to posttranscriptional regulation of ABCA1 and macrophage cholesterol efflux. METHODS AND RESULTS: We performed a bioinformatic analysis for identifying miRNA target prediction sites in ABCA1 gene and an unbiased genome-wide screen to identify miRNAs modulated by cholesterol excess in mouse peritoneal macrophages. Quantitative real-time reverse transcription-polymerase chain reaction confirmed that miR-758 is repressed in cholesterol-loaded macrophages. Under physiological conditions, high dietary fat excess in mice repressed miR-758 both in peritoneal macrophages and, to a lesser extent, in the liver. In mouse and human cells in vitro, miR-758 repressed the expression of ABCA1, and conversely, the inhibition of this miRNA by using anti-miR-758 increased ABCA1 expression. In mouse cells, miR-758 reduced cellular cholesterol efflux to apolipoprotein A1 (apoA1), and anti-miR-758 increased it. miR-758 directly targets the 3'-untranslated region of Abca1 as assessed by 3'-untranslated region luciferase reporter assays. Interestingly, miR-758 is highly expressed in the brain, where it also targets several genes involved in neurological functions, including Slc38a1, Ntm, Epha7, and Mytl1. CONCLUSION: We identified miR-758 as a novel miRNA that posttranscriptionally controls ABCA1 levels in different cells and regulates macrophage cellular cholesterol efflux to apoA1, opening new avenues to increase apoA1 and raise high-density lipoprotein levels.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Colesterol/metabolismo , MicroARNs/metabolismo , Procesamiento Postranscripcional del ARN/fisiología , Regiones no Traducidas 3'/genética , Regiones no Traducidas 3'/fisiología , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Animales , Apolipoproteína A-I/metabolismo , Astrocitos/metabolismo , Astrocitos/patología , Línea Celular , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Metabolismo de los Lípidos/fisiología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Ratones , MicroARNs/farmacología , Modelos Animales
16.
Biomolecules ; 12(2)2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-35204710

RESUMEN

Insulin resistance defines an impairment in the biologic response to insulin action in target tissues, primarily the liver, muscle, adipose tissue, and brain. Insulin resistance affects physiology in many ways, causing hyperglycemia, hypertension, dyslipidemia, visceral adiposity, hyperinsulinemia, elevated inflammatory markers, and endothelial dysfunction, and its persistence leads to the development metabolic disease, including diabetes, obesity, cardiovascular disease, or nonalcoholic fatty liver disease (NAFLD), as well as neurological disorders such as Alzheimer's disease. In addition to classical transcriptional factors, posttranscriptional control of gene expression exerted by microRNAs and RNA-binding proteins constitutes a new level of regulation with important implications in metabolic homeostasis. In this review, we describe miRNAs and RBPs that control key genes involved in the insulin signaling pathway and related regulatory networks, and their impact on human metabolic diseases at the molecular level, as well as their potential use for diagnosis and future therapeutics.


Asunto(s)
Resistencia a la Insulina , Enfermedades Metabólicas , Enfermedad del Hígado Graso no Alcohólico , Regulación de la Expresión Génica , Humanos , Insulina/metabolismo , Resistencia a la Insulina/genética , Enfermedades Metabólicas/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo
17.
Biomed Pharmacother ; 153: 113419, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36076541

RESUMEN

OBJECTIVE: miR-148a-3p (miR-148a) is a hepatic and immune-enriched microRNA (miRNA) that regulates macrophage-related lipoprotein metabolism, cholesterol homeostasis, and inflammation. The contribution of miR-148a-3p to the progression of atherosclerosis is unknown. In this study, we determined whether miR-148a silencing mitigated atherogenesis in APOBTGApobec-/-Ldlr+/- mice. METHODS: APOBTGApobec-/-Ldlr+/- mice were fed a typical Western-style diet for 22 weeks and injected with a nontargeting locked nucleic acid (LNA; LNA control) or miR-148a LNA (LNA 148a) for the last 10 weeks. At the end of the treatment, the mice were sacrificed, and circulating lipids, hepatic gene expression, and atherosclerotic lesions were analyzed. RESULTS: Examination of atherosclerotic lesions revealed a significant reduction in plaque size, with marked remodeling of the lesions toward a more stable phenotype. Mechanistically, miR-148a levels influenced macrophage cholesterol efflux and the inflammatory response. Suppression of miR-148a in murine primary macrophages decreased mRNA levels of proinflammatory M1-like markers (Nos2, Il6, Cox2, and Tnf) and increased the expression of anti-inflammatory genes (Arg1, Retlna, and Mrc1). CONCLUSIONS: Therapeutic silencing of miR148a mitigated the progression of atherosclerosis and promoted plaque stability. The antiatherogenic effect of miR-148a antisense therapy is likely mediated by the anti-inflammatory effects observed in macrophages treated with miR-148 LNA and independent of significant changes in circulating low-density lipoprotein cholesterol (LDL-C) and high-density lipoprotein cholesterol (HDL-C).


Asunto(s)
Aterosclerosis , MicroARNs , Placa Aterosclerótica , Desaminasas APOBEC , Animales , Apolipoproteínas B , Aterosclerosis/patología , HDL-Colesterol , Ratones , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo
18.
Front Cardiovasc Med ; 9: 908070, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35711383

RESUMEN

Monocytes participate in the development of atherosclerosis through the action of cytokines and other inflammatory mediators. Among them, CCR2 and its ligands, CCL2 and CCL7 play an important role, so the main objective of this work was to determine whether genetic variants affecting their activity were associated with cardiovascular disease. A cohort of 519 patients that have suffered coronary events was analyzed under a propensity score-matching protocol selecting a homogeneous set of cases and controls, according to age, sex, smoking status, dyslipidemia, arterial hypertension and type 2 diabetes as risk factors. While dyslipidemia and arterial hypertension were more prevalent among patients with angina pectoris, current smoking status and elevated inflammatory markers, including total leukocyte and monocyte counts, were more likely associated with acute coronary events. Propensity score matching analysis, performed to eliminate the influence of these risk factors and highlight genetic modifiers, revealed that a single nucleotide variant, rs17735770 at the 3'untranslated region of the CCL7 gene transcript, was associated with decreased cardiovascular risk in a group represented mostly by men, with an average age of 57, and without significant differences in traditional risk factors. Furthermore, the presence of this variant altered the local mRNA structure encompassing a binding site for miR-23ab, resulting in increased translation of a reporter gene in a miR23 independent fashion. The rs17735770 genetic variant led to increased expression of CCL7, a potential antagonist of CCR2 at inflammatory sites, where it could play a meaningful role during the evolution of atherosclerosis.

19.
Cells ; 11(16)2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-36010613

RESUMEN

The evident implication of the insulin-degrading enzyme (IDE) in Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM), among its capacity to degrade insulin and amyloid-ß peptide (Aß), suggests that IDE could be an essential link in the relation between hyperinsulinemia, insulin resistance and AD. However, little is known about the cellular and molecular regulation of IDE expression, and even less has been explored regarding the post-transcriptional regulation of IDE, although it represents a great molecular target of interest for therapeutic treatments. We recently described that miR-7, a novel candidate for linking AD and T2DM at the molecular level, regulates IDE and other key genes in both pathologies, including some key genes involved in the insulin signaling pathway. Here, we explored whether other miRNAs as well as other post-transcriptional regulators, such as RNA binding proteins (RBP), could potentially participate in the regulation of IDE expression in vitro. Our data showed that in addition to miR-7, miR-125, miR-490 and miR-199 regulate IDE expression at the post-transcriptional level. Moreover, we also found that IDE contains multiple potential binding sites for several RBPs, and a narrow-down prediction analysis led us to speculate on a novel regulation of IDE by RALY and HuD. Taken together, these results demonstrate the novel players controlling IDE expression that could represent potential therapeutical targets to treat several metabolic diseases with a high impact on human health, including AD and T2DM.


Asunto(s)
Enfermedad de Alzheimer , Diabetes Mellitus Tipo 2 , Insulisina , MicroARNs , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo C , Humanos , Insulina/metabolismo , Insulisina/metabolismo , MicroARNs/genética , MicroARNs/uso terapéutico
20.
Front Cardiovasc Med ; 9: 994080, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36407436

RESUMEN

Hypoxia is a crucial factor contributing to maintenance of atherosclerotic lesions. The ability of ABCA1 to stimulate the efflux of cholesterol from cells in the periphery, particularly foam cells in atherosclerotic plaques, is an important anti-atherosclerotic mechanism. The posttranscriptional regulation by miRNAs represents a key regulatory mechanism of a number of signaling pathways involved in atherosclerosis. Previously, miR-199a-5p has been shown to be implicated in the endocytic and retrograde intracellular transport. Although the regulation of miR-199a-5p and ABCA1 by hypoxia has been already reported independently, the role of miR-199a-5p in macrophages and its possible role in atherogenic processes such us regulation of lipid homeostasis through ABCA1 has not been yet investigated. Here, we demonstrate that both ABCA1 and miR-199a-5p show an inverse regulation by hypoxia and Ac-LDL in primary macrophages. Moreover, we demonstrated that miR-199a-5p regulates ABCA1 mRNA and protein levels by directly binding to its 3'UTR. As a result, manipulation of cellular miR-199a-5p levels alters ABCA1 expression and cholesterol efflux in primary mouse macrophages. Taken together, these results indicate that the correlation between ABCA1-miR-199a-5p could be exploited to control macrophage cholesterol efflux during the onset of atherosclerosis, where cholesterol alterations and hypoxia play a pathogenic role.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA