Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Am J Physiol Endocrinol Metab ; 326(5): E735-E746, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38597830

RESUMEN

Most studies on fat appetite have focused on long-chain triglycerides (LCTs) due to their obesogenic properties. Medium-chain triglycerides (MCTs), conversely, exhibit antiobesogenic effects; however, the regulation of MCT intake remains elusive. Here, we demonstrate that mice can distinguish between MCTs and LCTs, and the specific appetite for MCTs is governed by hepatic ß-oxidation. We generated liver-specific medium-chain acyl-CoA dehydrogenase (MCAD)-deficient (MCADL-/-) mice and analyzed their preference for MCT and LCT solutions using glyceryl trioctanoate (C8-TG), glyceryl tridecanoate (C10-TG), corn oil, and lard oil in two-bottle choice tests conducted over 8 days. In addition, we used lick microstructure analyses to evaluate the palatability and appetite for MCT and LCT solutions. Finally, we measured the expression levels of genes associated with fat ingestion (Galanin, Qrfp, and Nmu) in the hypothalamus 2 h after oral gavage of fat. Compared with control mice, MCADL-/- mice exhibited a significantly reduced preference for MCT solutions, with no alteration in the preference for LCTs. Lick analysis revealed that MCADL-/- mice displayed a significantly decreased appetite for MCT solutions only while the palatability of both MCT and LCT solutions remained unaffected. Hypothalamic Galanin expression in control mice was elevated by oral gavage of C8-TG but not by LCTs, and this response was abrogated in MCADL-/- mice. In summary, our data suggest that hepatic ß-oxidation is required for MCT-specific appetite but not for LCT-specific appetite. The induction of hypothalamic galanin upon MCT ingestion, dependent on hepatic ß-oxidation, could be involved in the regulation of MCT-specific appetite.NEW & NOTEWORTHY Whether and how medium-chain triglyceride (MCT) intake is regulated remains unknown. Here, we showed that mice can discriminate between MCTs and LCTs. Hepatic ß-oxidation participates in MCT-specific appetite, and hypothalamic galanin may be one of the factors that regulate MCT intake. Because of the antiobesity effects of MCTs, studying MCT-specific appetite may help combat obesity by promoting the intake of MCTs instead of LCTs.


Asunto(s)
Acil-CoA Deshidrogenasa , Apetito , Ácidos Grasos , Hígado , Ratones Noqueados , Oxidación-Reducción , Triglicéridos , Animales , Triglicéridos/metabolismo , Ratones , Oxidación-Reducción/efectos de los fármacos , Hígado/metabolismo , Hígado/efectos de los fármacos , Masculino , Ácidos Grasos/metabolismo , Apetito/efectos de los fármacos , Apetito/fisiología , Acil-CoA Deshidrogenasa/metabolismo , Acil-CoA Deshidrogenasa/genética , Ratones Endogámicos C57BL , Hipotálamo/metabolismo , Hipotálamo/efectos de los fármacos
2.
EMBO Rep ; 21(4): e48389, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32147946

RESUMEN

The Hippo signaling pathway is involved in the pathophysiology of various cardiovascular diseases. Yes-associated protein (YAP) and transcriptional enhancer activator domain (TEAD) transcriptional factors, the main transcriptional complex of the Hippo pathway, were recently identified as modulators of phenotypic switching of vascular smooth muscle cells (VSMCs). However, the intrinsic regulator of YAP/TEAD-mediated gene expressions involved in vascular pathophysiology remains to be elucidated. Here, we identified Homeobox A4 (HOXA4) as a potent repressor of YAP/TEAD transcriptional activity using lentiviral shRNA screen. Mechanistically, HOXA4 interacts with TEADs and attenuates YAP/TEAD-mediated transcription by competing with YAP for TEAD binding. We also clarified that the expression of HOXA4 is relatively abundant in the vasculature, especially in VSMCs. In vitro experiments in human VSMCs showed HOXA4 maintains the differentiation state of VSMCs via inhibition of YAP/TEAD-induced phenotypic switching. We generated Hoxa4-deficient mice and confirmed the downregulation of smooth muscle-specific contractile genes and the exacerbation of vascular remodeling after carotid artery ligation in vivo. Our results demonstrate that HOXA4 is a repressor of VSMC phenotypic switching by inhibiting YAP/TEAD-mediated transcription.


Asunto(s)
Genes Homeobox , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Remodelación Vascular , Animales , Ratones , Miocitos del Músculo Liso , Transducción de Señal
3.
Circ J ; 85(3): 323-329, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33518695

RESUMEN

Due to the COVID-19 pandemic, the 84thAnnual Meeting of the Japanese Circulation Society (JCS) was held in a web-based format for the first time in its history as "The Week for JCS 2020" from Monday, July 27 to Sunday, August 2, 2020. All sessions, including general abstracts, were streamed live or on-demand. The main theme of the meeting was "Change Practice!" and the aim was to organize the latest findings in the field of cardiovascular medicine and discuss how to change practice. The total number of registered attendees was over 16,800, far exceeding our expectations, and many of the sessions were viewed by far more people than at conventional face-to-face scientific meetings. At this conference, the power of online information dissemination was fully demonstrated, and the evolution of online academic meetings will be a direction that cannot be reversed in the future. The meeting was completed with great success, and we express our heartfelt gratitude to all affiliates for their enormous amount of work, cooperation, and support.


Asunto(s)
Cardiología/organización & administración , Congresos como Asunto/organización & administración , Sociedades Científicas/organización & administración , Telecomunicaciones/organización & administración , Cardiología/tendencias , Enfermedades Cardiovasculares/diagnóstico por imagen , Enfermedades Cardiovasculares/terapia , Congresos como Asunto/estadística & datos numéricos , Congresos como Asunto/tendencias , Humanos , Japón , Investigación , Encuestas y Cuestionarios , Telecomunicaciones/estadística & datos numéricos , Telecomunicaciones/tendencias
4.
Clin Sci (Lond) ; 133(4): 583-595, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30777884

RESUMEN

Recent reports, including ours, have indicated that microRNA (miR)-33 located within the intron of sterol regulatory element binding protein (SREBP) 2 controls cholesterol homeostasis and can be a potential therapeutic target for the treatment of atherosclerosis. Here, we show that SPAST, which encodes a microtubule-severing protein called SPASTIN, was a novel target gene of miR-33 in human. Actually, the miR-33 binding site in the SPAST 3'-UTR is conserved not in mice but in mid to large mammals, and it is impossible to clarify the role of miR-33 on SPAST in mice. We demonstrated that inhibition of miR-33a, a major form of miR-33 in human neurons, via locked nucleic acid (LNA)-anti-miR ameliorated the pathological phenotype in hereditary spastic paraplegia (HSP)-SPG4 patient induced pluripotent stem cell (iPSC)-derived cortical neurons. Thus, miR-33a can be a potential therapeutic target for the treatment of HSP-SPG4.


Asunto(s)
Terapia Genética/métodos , Células Madre Pluripotentes Inducidas/metabolismo , MicroARNs/genética , Células-Madre Neurales/metabolismo , Neuritas/metabolismo , Oligonucleótidos/genética , Paraplejía Espástica Hereditaria/terapia , Espastina/genética , Regiones no Traducidas 3' , Sitios de Unión , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/patología , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Células-Madre Neurales/patología , Neuritas/patología , Neurogénesis , Oligonucleótidos/metabolismo , Fenotipo , Paraplejía Espástica Hereditaria/genética , Paraplejía Espástica Hereditaria/metabolismo , Paraplejía Espástica Hereditaria/patología , Espastina/metabolismo
5.
Circ Res ; 120(5): 835-847, 2017 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-27920122

RESUMEN

RATIONALE: Heart failure and atherosclerosis share the underlying mechanisms of chronic inflammation followed by fibrosis. A highly conserved microRNA (miR), miR-33, is considered as a potential therapeutic target for atherosclerosis because it regulates lipid metabolism and inflammation. However, the role of miR-33 in heart failure remains to be elucidated. OBJECTIVE: To clarify the role of miR-33 involved in heart failure. METHODS AND RESULTS: We first investigated the expression levels of miR-33a/b in human cardiac tissue samples with dilated cardiomyopathy. Increased expression of miR-33a was associated with improving hemodynamic parameters. To clarify the role of miR-33 in remodeling hearts, we investigated the responses to pressure overload by transverse aortic constriction in miR-33-deficient (knockout [KO]) mice. When mice were subjected to transverse aortic constriction, miR-33 expression levels were significantly upregulated in wild-type left ventricles. There was no difference in hypertrophic responses between wild-type and miR-33KO hearts, whereas cardiac fibrosis was ameliorated in miR-33KO hearts compared with wild-type hearts. Despite the ameliorated cardiac fibrosis, miR-33KO mice showed impaired systolic function after transverse aortic constriction. We also found that cardiac fibroblasts were mainly responsible for miR-33 expression in the heart. Deficiency of miR-33 impaired cardiac fibroblast proliferation, which was considered to be caused by altered lipid raft cholesterol content. Moreover, cardiac fibroblast-specific miR-33-deficient mice also showed decreased cardiac fibrosis induced by transverse aortic constriction as systemic miR-33KO mice. CONCLUSION: Our results demonstrate that miR-33 is involved in cardiac remodeling, and it preserves lipid raft cholesterol content in fibroblasts and maintains adaptive fibrotic responses in the remodeling heart.


Asunto(s)
Colesterol/metabolismo , Microdominios de Membrana/metabolismo , MicroARNs/metabolismo , Miocardio/metabolismo , Miocardio/patología , Remodelación Ventricular/fisiología , Adulto , Anciano , Animales , Células Cultivadas , Femenino , Fibroblastos/metabolismo , Fibroblastos/fisiología , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Ratas , Ratas Sprague-Dawley
6.
Arterioscler Thromb Vasc Biol ; 38(10): 2460-2473, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30354203

RESUMEN

Objective- Atherosclerosis is a common disease caused by a variety of metabolic and inflammatory disturbances. MicroRNA (miR)-33a within SREBF2 (sterol regulatory element-binding factor 2) is a potent target for treatment of atherosclerosis through regulating both aspects; however, the involvement of miR-33b within SREBF1 remains largely unknown. Although their host genes difference could lead to functional divergence of miR-33a/b, we cannot dissect the roles of miR-33a/b in vivo because of lack of miR-33b sequences in mice, unlike human. Approach and Results- Here, we analyzed the development of atherosclerosis using miR-33b knock-in humanized mice under apolipoprotein E-deficient background. MiR-33b is prominent both in human and mice on atheroprone condition. MiR-33b reduced serum high-density lipoprotein cholesterol levels and systemic reverse cholesterol transport. MiR-33b knock-in macrophages showed less cholesterol efflux capacity and higher inflammatory state via regulating lipid rafts. Thus, miR-33b promotes vulnerable atherosclerotic plaque formation. Furthermore, bone marrow transplantation experiments strengthen proatherogenic roles of macrophage miR-33b. Conclusions- Our data demonstrated critical roles of SREBF1-miR-33b axis on both lipid profiles and macrophage phenotype remodeling and indicate that miR-33b is a promising target for treating atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , MicroARNs/metabolismo , Placa Aterosclerótica , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Apoptosis , Aterosclerosis/genética , Aterosclerosis/patología , Trasplante de Médula Ósea , Estudios de Casos y Controles , HDL-Colesterol/sangre , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Humanos , Absorción Intestinal , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Microdominios de Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , MicroARNs/genética , Persona de Mediana Edad , Fenotipo , Transducción de Señal , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Triglicéridos/sangre
7.
Arterioscler Thromb Vasc Biol ; 37(11): 2161-2170, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28882868

RESUMEN

OBJECTIVE: Abdominal aortic aneurysm (AAA) is an increasingly prevalent and ultimately fatal disease with no effective pharmacological treatment. Because matrix degradation induced by vascular inflammation is the major pathophysiology of AAA, attenuation of this inflammation may improve its outcome. Previous studies suggested that miR-33 (microRNA-33) inhibition and genetic ablation of miR-33 increased serum high-density lipoprotein cholesterol and attenuated atherosclerosis. APPROACH AND RESULTS: MiR-33a-5p expression in central zone of human AAA was higher than marginal zone. MiR-33 deletion attenuated AAA formation in both mouse models of angiotensin II- and calcium chloride-induced AAA. Reduced macrophage accumulation and monocyte chemotactic protein-1 expression were observed in calcium chloride-induced AAA walls in miR-33-/- mice. In vitro experiments revealed that peritoneal macrophages from miR-33-/- mice showed reduced matrix metalloproteinase 9 expression levels via c-Jun N-terminal kinase inactivation. Primary aortic vascular smooth muscle cells from miR-33-/- mice showed reduced monocyte chemotactic protein-1 expression by p38 mitogen-activated protein kinase attenuation. Both of the inactivation of c-Jun N-terminal kinase and p38 mitogen-activated protein kinase were possibly because of the increase of ATP-binding cassette transporter A1 that is a well-known target of miR-33. Moreover, high-density lipoprotein cholesterol derived from miR-33-/- mice reduced expression of matrix metalloproteinase 9 in macrophages and monocyte chemotactic protein-1 in vascular smooth muscle cells. Bone marrow transplantation experiments indicated that miR-33-deficient bone marrow cells ameliorated AAA formation in wild-type recipients. MiR-33 deficiency in recipient mice was also shown to contribute the inhibition of AAA formation. CONCLUSIONS: These data strongly suggest that inhibition of miR-33 will be effective as a novel strategy for treating AAA.


Asunto(s)
Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/prevención & control , Aortitis/prevención & control , Mediadores de Inflamación/metabolismo , MicroARNs/metabolismo , Angiotensina II , Animales , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/metabolismo , Aortitis/inducido químicamente , Aortitis/genética , Aortitis/metabolismo , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Trasplante de Médula Ósea , Cloruro de Calcio , Línea Celular , Quimiocina CCL2/metabolismo , HDL-Colesterol/sangre , Dilatación Patológica , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Fenotipo , Transducción de Señal , Factores de Tiempo , Transfección , Remodelación Vascular , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Circ J ; 82(5): 1231-1236, 2018 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-29526985

RESUMEN

Protein-coding genes account for less than 2% of the whole genome. However, the advances in RNA sequencing and genome-wide analysis have demonstrated that most of the genome is capable of being transcribed. Moreover, recent studies have suggested that long non-coding RNAs (lncRNAs) are critical regulators of gene expression and epigenesis in both physiological and disease states. Several lncRNAs are functionally involved in cardiovascular diseases and may be potential therapeutic targets. Here, we review the current strategies for the discovery of functional lncRNAs and recently discovered lncRNAs in the cardiovascular field, focusing on cardiac development, hypertrophy, heart failure, and atherosclerosis. We also discuss the therapeutic potentials of synthetic RNAs to modulate these lncRNAs and future directions in this research field.


Asunto(s)
Aterosclerosis , Cardiomegalia , Insuficiencia Cardíaca , ARN Largo no Codificante , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
9.
Circ Res ; 116(2): 279-88, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25362209

RESUMEN

RATIONALE: In some patients with type 2 diabetes mellitus (DM) without hypertension, cardiac hypertrophy and attenuated cardiac function are observed, and this insult is termed diabetic cardiomyopathy. To date, microRNA (miRNAs or miR) functions in diabetic cardiomyopathy remain to be elucidated. OBJECTIVE: To clarify the functions of miRNAs involved in diabetic cardiomyopathy caused by type 2 DM. METHODS AND RESULTS: C57BL/6 mice were fed a high-fat diet (HFD) for 20 weeks, which induced obesity and type 2 DM. miRNA microarray analyses and real-time polymerase chain reaction revealed that miR-451 levels were significantly increased in the type 2 DM mouse hearts. Because excess supply of saturated fatty acids is a cause of diabetic cardiomyopathy, we stimulated neonatal rat cardiac myocytes with palmitic acid and confirmed that miR-451 expression was increased in a dose- and time-dependent manner. Loss of miR-451 function ameliorated palmitate-induced lipotoxicity in neonatal rat cardiac myocytes. Calcium-binding protein 39 (Cab39) is a scaffold protein of liver kinase B1 (LKB1), an upstream kinase of AMP-activated protein kinase (AMPK). Cab39 was a direct target of miR-451 in neonatal rat cardiac myocytes and Cab39 overexpression rescued the lipotoxicity. To clarify miR-451 functions in vivo, we generated cardiomyocyte-specific miR-451 knockout mice. HFD-induced cardiac hypertrophy and contractile reserves were ameliorated in cardiomyocyte-specific miR-451 knockout mice compared with control mice. Protein levels of Cab39 and phosphorylated AMPK were increased and phosphorylated mammalian target of rapamycin (mTOR) was reduced in cardiomyocyte-specific miR-451 knockout mouse hearts compared with control mouse hearts. CONCLUSIONS: Our results demonstrate that miR-451 is involved in diabetic cardiomyopathy through suppression of the LKB1/AMPK pathway.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Cardiomegalia/metabolismo , Dieta Alta en Grasa/efectos adversos , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Animales Recién Nacidos , Cardiomegalia/patología , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Miocitos Cardíacos/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología
10.
Circ J ; 81(12): 1783-1791, 2017 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-28637969

RESUMEN

BACKGROUND: TheSCN5Agene encodes the α subunit of the cardiac voltage-gated sodium channel, NaV1.5. The missense mutation, D1275N, has been associated with a range of unusual phenotypes associated with reduced NaV1.5 function, including cardiac conduction disease and dilated cardiomyopathy. Curiously, the reported biophysical properties ofSCN5A-D1275N channels vary with experimental system.Methods and Results:First, using a human embryonic kidney (HEK) 293 cell-based heterologous expression system, theSCN5A-D1275N channels showed similar maximum sodium conductance but a significantly depolarizing shift of activation gate (+10 mV) compared to wild type. Second, we generated human-induced pluripotent stem cells (hiPSCs) from a 24-year-old female who carried heterozygousSCN5A-D1275N and analyzed the differentiated cardiomyocytes (CMs). AlthoughSCN5Atranscript levels were equivalent between D1275N and control hiPSC-CMs, both the total amount of NaV1.5 and the membrane fractions were reduced approximately half in the D1275N cells, which were rescued by the proteasome inhibitor MG132 treatment. Electrophysiological assays revealed that maximum sodium conductance was reduced to approximately half of that in control hiPSC-CMs in the D1275N cells, and maximum upstroke velocity of action potential was lower in D1275N, which was consistent with the reduced protein level of NaV1.5. CONCLUSIONS: This study successfully demonstrated diminished sodium currents resulting from lower NaV1.5 protein levels, which is dependent on proteasomal degradation, using a hiPSC-based model forSCN5A-D1275N-related sodium channelopathy.


Asunto(s)
Canalopatías/genética , Células Madre Pluripotentes Inducidas/citología , Mutación Missense , Canal de Sodio Activado por Voltaje NAV1.5/genética , Electrofisiología Cardíaca , Células HEK293 , Humanos , Miocitos Cardíacos/citología , Canal de Sodio Activado por Voltaje NAV1.5/análisis , Complejo de la Endopetidasa Proteasomal/metabolismo , Sodio/metabolismo
11.
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
12.
Circ J ; 79(2): 278-84, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25744742

RESUMEN

MicroRNAs (miRNAs; miRs) are small non-protein-coding RNAs that negatively regulate gene expression. They bind to the 3' UTR of specific mRNAs and either inhibit translation or promote mRNA degradation. There is emerging evidence linking miR-33a/b to lipid homoeostasis, targeting ABCA1,SREBF1, etc and it would appear that they have acted as "thrifty genes" during evolution to maintain cholesterol levels both at the cellular and whole body level. As we are now living in a period of "satiation", miR-33a/b no longer seem to be useful and could be potential therapeutic targets for lipid disorders and/or atherosclerosis. In this review, we describe the current understanding of the function of miR-33a/b in lipid homeostasis, focusing on the "thrifty" aspect.


Asunto(s)
Regiones no Traducidas 3' , Aterosclerosis/metabolismo , Metabolismo de los Lípidos , MicroARNs/metabolismo , Estabilidad del ARN , Transportador 1 de Casete de Unión a ATP/biosíntesis , Transportador 1 de Casete de Unión a ATP/genética , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Regulación de la Expresión Génica , Humanos , MicroARNs/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/biosíntesis , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética
13.
Int Heart J ; 56(4): 365-71, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26084456

RESUMEN

MicroRNAs (miRNAs) are small non-protein-coding RNAs that negatively regulate gene expression. They bind to the 3'-untranslated region of specific mRNAs and inhibit translation or promote mRNA degradation. Dyslipidemia/hyperlipidemia is a well-accepted risk factor for the development of atherosclerosis. The pathogenesis factors involved in lipid abnormalities are being examined extensively, and there is emerging evidence linking miRNAs to lipid metabolism. Among them, recent studies, including ours, have demonstrated that miRNAs control the expression of genes associated with high-density lipoprotein (HDL) cholesterol (HDL-C) metabolism, including ABCA1, ABCG1, and scavenger receptor class B, type I. Moreover, HDL-C itself was proved to carry miRNAs and deliver them to several different types of cells. In this review, we describe the current understanding of the functions of miRNAs in HDL metabolism and their potential in therapy for treating cardiometabolic diseases.


Asunto(s)
HDL-Colesterol , Dislipidemias/genética , MicroARNs/genética , Transportador 1 de Casete de Unión a ATP/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Aterosclerosis/prevención & control , HDL-Colesterol/genética , HDL-Colesterol/metabolismo , Dislipidemias/metabolismo , Dislipidemias/prevención & control , Regulación de la Expresión Génica , Humanos , Biosíntesis de Proteínas/genética , Receptores Depuradores de Clase B/genética
14.
BMJ Case Rep ; 17(3)2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38453224

RESUMEN

Systemic thromboembolism associated with atrial fibrillation (AF) is usually caused by thrombi in the left atrial appendage and acute onset. We experienced an unusual case of a woman in her 60s who presented to the outpatient district having bilateral intermittent claudication for more than 1 month, which turned out to be multiple thromboembolism from asymptomatic AF with tachycardia. She was also complicated with non-ischaemic dilated cardiomyopathy with reduced ejection fraction, consistent with arrhythmia-induced cardiomyopathy (AiCM), along with left atrial and left ventricular thrombi and thromboembolism in multiple organs. Rate control with beta-blockers was not effective. With the administration of amiodarone after adequate anticoagulation therapy, she returned to sinus rhythm, and the ejection fraction was restored. This case is instructive in that AiCM with AF can cause thrombosis in the left ventricle, and the patient may present with worsening intermittent claudication as a result of systemic embolism.


Asunto(s)
Fibrilación Atrial , Cardiomiopatías , Cardiopatías , Tromboembolia , Trombosis , Femenino , Humanos , Fibrilación Atrial/complicaciones , Fibrilación Atrial/tratamiento farmacológico , Cardiomiopatías/complicaciones , Cardiopatías/etiología , Claudicación Intermitente/etiología , Tromboembolia/complicaciones , Trombosis/complicaciones , Trombosis/diagnóstico por imagen , Trombosis/tratamiento farmacológico , Persona de Mediana Edad , Anciano
15.
Thromb Res ; 237: 141-144, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38593524

RESUMEN

The use of direct oral anticoagulants (DOACs) in breastfeeding women is currently challenging due to limited safety data for breastfeeding infants, and there have been no previous studies on the drug concentration in breastfeeding infants. We treated 2 patients (one case was twin pregnancy) with venous thromboembolisms in breastfeeding women administered rivaroxaban at our institution. Blood samples from the mothers and breastmilk samples were collected at time 0 and 2 h after the rivaroxaban administration, breastfeeding was conducted 2 h after the rivaroxaban administration, and blood samples from the infants were collected 2 h after breastfeeding (4 h after maternal rivaroxaban administration). The milk-to-plasma (M:P) ratios were 0.27 in Case 1 and 0.32 in Case 2. The estimated relative infant dose (RID) was 0.82 % in Case 1 Children 1 and 2, and 1.27 % in Case 2. The rivaroxaban concentration in the infant plasma was below the lower limit of quantification in all infants. In addition, even in the high-exposure case simulation based on 5 days of breastfeeding in Case 2, the infant plasma concentration level was below the lower limit of quantification. At 3 months of follow-up, breastfeeding was continued, and all infants grew and developed without any health problems including bleeding events. The current case series showed that there were no pharmacokinetic or clinical concerns for breastfeeding women or breastfed infants, and provides support for rivaroxaban as a safe treatment option for these patients.


Asunto(s)
Lactancia Materna , Inhibidores del Factor Xa , Leche Humana , Rivaroxabán , Humanos , Rivaroxabán/uso terapéutico , Rivaroxabán/farmacocinética , Femenino , Adulto , Inhibidores del Factor Xa/uso terapéutico , Inhibidores del Factor Xa/farmacocinética , Leche Humana/química , Leche Humana/metabolismo , Lactante , Tromboembolia Venosa/tratamiento farmacológico , Recién Nacido , Embarazo
16.
PLoS One ; 19(3): e0297231, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38507394

RESUMEN

Familial hypercholesterolemia is an inherited disorder that remains underdiagnosed. Conventional genetic testing methods such as next-generation sequencing (NGS) or target PCR are based on the amplification process. Due to the efficiency limits of polymerase and ligase enzymes, these methods usually target short regions and do not detect large mutations straightforwardly. This study combined the long-read nanopore sequencing and CRISPR-Cas9 system to sequence the target DNA molecules without amplification. We originally designed and optimized the CRISPR-RNA panel to target the low-density lipoprotein receptor gene (LDLR) and proprotein convertase subtilisin/kexin type 9 gene (PCSK9) from human genomic DNA followed by nanopore sequencing. The average coverages for LDLR and PCSK9 were 106× and 420×, versus 1.2× for the background genome. Among them, continuous reads were 52x and 307x, respectively, and spanned the entire length of LDLR and PCSK9. We identified pathogenic mutations in both coding and splicing donor regions in LDLR. We also detected an 11,029 bp large deletion in another case. Furthermore, using continuous long reads generated from the benchmark experiment, we demonstrated how a false-positive 670 bp deletion caused by PCR amplification errors was easily eliminated.


Asunto(s)
Hiperlipoproteinemia Tipo II , Secuenciación de Nanoporos , Humanos , Proproteína Convertasa 9/genética , Sistemas CRISPR-Cas/genética , Receptores de LDL/genética , Receptores de LDL/metabolismo , Hiperlipoproteinemia Tipo II/diagnóstico , Hiperlipoproteinemia Tipo II/genética , Mutación , Genómica , ADN
17.
Biomed Pharmacother ; 170: 115850, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38091636

RESUMEN

AIMS: As heart failure (HF) progresses, ATP levels in myocardial cells decrease, and myocardial contractility also decreases. Inotropic drugs improve myocardial contractility but increase ATP consumption, leading to poor prognosis. Kyoto University Substance 121 (KUS121) is known to selectively inhibit the ATPase activity of valosin-containing protein, maintain cellular ATP levels, and manifest cytoprotective effects in several pathological conditions. The aim of this study is to determine the therapeutic effect of KUS121 on HF models. METHODS AND RESULTS: Cultured cell, mouse, and canine models of HF were used to examine the therapeutic effects of KUS121. The mechanism of action of KUS121 was also examined. Administration of KUS121 to a transverse aortic constriction (TAC)-induced mouse model of HF rapidly improved the left ventricular ejection fraction and improved the creatine phosphate/ATP ratio. In a canine model of high frequency-paced HF, administration of KUS121 also improved left ventricular contractility and decreased left ventricular end-diastolic pressure without increasing the heart rate. Long-term administration of KUS121 to a TAC-induced mouse model of HF suppressed cardiac hypertrophy and fibrosis. In H9C2 cells, KUS121 reduced ER stress. Finally, in experiments using primary cultured cardiomyocytes, KUS121 improved contractility and diastolic capacity without changing peak Ca2+ levels or contraction time. These effects were not accompanied by an increase in cyclic adenosine monophosphate or phosphorylation of phospholamban and ryanodine receptors. CONCLUSIONS: KUS121 ameliorated HF by a mechanism totally different from that of conventional catecholamines. We propose that KUS121 is a promising new option for the treatment of HF.


Asunto(s)
Calcio , Insuficiencia Cardíaca , Humanos , Ratones , Animales , Perros , Calcio/metabolismo , Proteína que Contiene Valosina/metabolismo , Volumen Sistólico , Universidades , Función Ventricular Izquierda , Insuficiencia Cardíaca/metabolismo , Miocitos Cardíacos/metabolismo , Enfermedad Crónica , Adenosina Trifosfato/metabolismo , Modelos Animales de Enfermedad
18.
J Mol Cell Cardiol ; 57: 72-81, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23352991

RESUMEN

Metastasis-associated protein, S100A4 is suggested as a marker for fibrosis in several organs. It also modulates DNA binding of p53 and affects its function. However, the functional role of S100A4 in the myocardium has remained unclear. Therefore, we investigated the role of S100A4 and its relationship with p53 in cardiac fibrosis. In Dahl-rat hypertensive heart disease model, S100A4 was upregulated in the hypertrophic myocardium and further activated during transition to heart failure (HF). It was expressed in various cells including fibroblasts. In in vitro cardiac fibroblasts, the knockdown of S100A4 significantly suppressed both cell proliferation and collagen expressions. S100A4 co-localized and interacted with p53 in the nucleus. S100A4 knockdown increased the expression of p53-downstream genes, p21 and mdm2, and concomitant knockdown of p53 recovered cell proliferation and collagen expression. Transverse aortic constriction (TAC) was performed in S100A4 knockout (KO) mice, which showed a similar baseline-phenotype to wild type (WT) mice. Although there was no difference in hypertrophic response, KO mice showed reduced interstitial fibrosis, decreased myofibroblasts, and suppressed expressions of collagens and profibrotic cytokines in the left ventricle. Also, DNA microarray analysis showed that S100A4 knockout in vivo had a significant impact on expressions of p53-associated genes. These findings suggest that S100A4 modulates p53 function in fibroblasts and thereby mediates myocardial interstitial fibrosis through two distinct mechanisms; cell proliferation and collagen expression. Blockade of S100A4 may have therapeutic potential in cardiac hypertrophy and HF by attenuating cardiac fibrosis.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Ventrículos Cardíacos/patología , Miofibroblastos/metabolismo , Proteínas S100/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Angiotensina II/fisiología , Animales , Proliferación Celular , Colágeno/genética , Colágeno/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Fibrosis , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Insuficiencia Cardíaca/patología , Ventrículos Cardíacos/metabolismo , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/patología , Masculino , Ratones , Ratones Noqueados , Miofibroblastos/fisiología , Células 3T3 NIH , Péptido Natriurético Encefálico/sangre , Ratas , Ratas Endogámicas Dahl , Proteína de Unión al Calcio S100A4 , Transcriptoma
19.
Proc Natl Acad Sci U S A ; 107(40): 17321-6, 2010 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-20855588

RESUMEN

Sterol regulatory element-binding protein 2 (SREBP-2) transcription factor has been identified as a key protein in cholesterol metabolism through the transactivation of the LDL receptor and cholesterol biosynthesis genes. Here, we generated mice lacking microRNA (miR)-33, encoded by an intron of the Srebp2, and showed that miR-33 repressed the expression of ATP-binding cassette transporter A1 (ABCA1) protein, a key regulator of HDL synthesis by mediating cholesterol efflux from cells to apolipoprotein A (apoA)-I. In fact, peritoneal macrophages derived from miR-33-deficient mice showed a marked increase in ABCA1 levels and higher apoA-I-dependent cholesterol efflux than those from WT mice. ABCA1 protein levels in liver were also higher in miR-33-deficient mice than in WT mice. Moreover, miR-33-deficient mice had significantly higher serum HDL cholesterol levels than WT mice. These data establish a critical role for miR-33 in the regulation of ABCA1 expression and HDL biogenesis in vivo.


Asunto(s)
HDL-Colesterol/metabolismo , Intrones , MicroARNs/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética , 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 , Secuencia de Bases , Bovinos , Línea Celular , Pollos , HDL-Colesterol/genética , Femenino , Humanos , Macrófagos/metabolismo , Ratones , MicroARNs/genética , Datos de Secuencia Molecular , Alineación de Secuencia , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo
20.
Life Sci Alliance ; 6(8)2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37263777

RESUMEN

Nonalcoholic steatohepatitis (NASH) can lead to cirrhosis and hepatocellular carcinoma in their advanced stages; however, there are currently no approved therapies. Here, we show that microRNA (miR)-33b in hepatocytes is critical for the development of NASH. miR-33b is located in the intron of sterol regulatory element-binding transcription factor 1 and is abundantly expressed in humans, but absent in rodents. miR-33b knock-in (KI) mice, which have a miR-33b sequence in the same intron of sterol regulatory element-binding transcription factor 1 as humans and express miR-33b similar to humans, exhibit NASH under high-fat diet feeding. This condition is ameliorated by hepatocyte-specific miR-33b deficiency but unaffected by macrophage-specific miR-33b deficiency. Anti-miR-33b oligonucleotide improves the phenotype of NASH in miR-33b KI mice fed a Gubra Amylin NASH diet, which induces miR-33b and worsens NASH more than a high-fat diet. Anti-miR-33b treatment reduces hepatic free cholesterol and triglyceride accumulation through up-regulation of the lipid metabolism-related target genes. Furthermore, it decreases the expression of fibrosis marker genes in cultured hepatic stellate cells. Thus, inhibition of miR-33b using nucleic acid medicine is a promising treatment for NASH.


Asunto(s)
Neoplasias Hepáticas , MicroARNs , Enfermedad del Hígado Graso no Alcohólico , Ratones , Humanos , Animales , Enfermedad del Hígado Graso no Alcohólico/genética , Antagomirs , MicroARNs/genética , MicroARNs/metabolismo , Colesterol , Neoplasias Hepáticas/patología , Factores de Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA