Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Adv Biol (Weinh) ; 8(1): e2300211, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37794610

RESUMEN

Although a large amount of data consistently shows that genes affect immunometabolic characteristics and outcomes, epigenetic mechanisms are also heavily implicated. Epigenetic changes, including DNA methylation, histone modification, and noncoding RNA, determine gene activity by altering the accessibility of chromatin to transcription factors. Various factors influence these alterations, including genetics, lifestyle, and environmental cues. Moreover, acquired epigenetic signals can be transmitted across generations, thus contributing to early disease traits in the offspring. A closer investigation is critical in this aspect as it can help to understand the underlying molecular mechanisms further and gain insights into potential therapeutic targets for preventing and treating diseases arising from immuno-metabolic dysregulation. In this review, the role of chromatin alterations in the transcriptional modulation of genes involved in insulin resistance, systemic inflammation, macrophage polarization, endothelial dysfunction, metabolic cardiomyopathy, and nonalcoholic fatty liver disease (NAFLD), is discussed. An overview of emerging chromatin-modifying drugs and the importance of the individual epigenetic profile for personalized therapeutic approaches in patients with immuno-metabolic disorders is also presented.


Asunto(s)
Metilación de ADN , Enfermedad del Hígado Graso no Alcohólico , Humanos , Metilación de ADN/genética , Epigénesis Genética , Enfermedad del Hígado Graso no Alcohólico/genética , Cromatina , Inflamación/genética
2.
Cardiovasc Diabetol ; 22(1): 312, 2023 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-37957697

RESUMEN

BACKGROUND: Metabolic cardiomyopathy (MCM), characterized by intramyocardial lipid accumulation, drives the progression to heart failure with preserved ejection fraction (HFpEF). Although evidence suggests that the mammalian silent information regulator 1 (Sirt1) orchestrates myocardial lipid metabolism, it is unknown whether its exogenous administration could avoid MCM onset. We investigated whether chronic treatment with recombinant Sirt1 (rSirt1) could halt MCM progression. METHODS: db/db mice, an established model of MCM, were supplemented with intraperitoneal rSirt1 or vehicle for 4 weeks and compared with their db/ + heterozygous littermates. At the end of treatment, cardiac function was assessed by cardiac ultrasound and left ventricular samples were collected and processed for molecular analysis. Transcriptional changes were evaluated using a custom PCR array. Lipidomic analysis was performed by mass spectrometry. H9c2 cardiomyocytes exposed to hyperglycaemia and treated with rSirt1 were used as in vitro model of MCM to investigate the ability of rSirt1 to directly target cardiomyocytes and modulate malondialdehyde levels and caspase 3 activity. Myocardial samples from diabetic and nondiabetic patients were analysed to explore Sirt1 expression levels and signaling pathways. RESULTS: rSirt1 treatment restored cardiac Sirt1 levels and preserved cardiac performance by improving left ventricular ejection fraction, fractional shortening and diastolic function (E/A ratio). In left ventricular samples from rSirt1-treated db/db mice, rSirt1 modulated the cardiac lipidome: medium and long-chain triacylglycerols, long-chain triacylglycerols, and triacylglycerols containing only saturated fatty acids were reduced, while those containing docosahexaenoic acid were increased. Mechanistically, several genes involved in lipid trafficking, metabolism and inflammation, such as Cd36, Acox3, Pparg, Ncoa3, and Ppara were downregulated by rSirt1 both in vitro and in vivo. In humans, reduced cardiac expression levels of Sirt1 were associated with higher intramyocardial triacylglycerols and PPARG-related genes. CONCLUSIONS: In the db/db mouse model of MCM, chronic exogenous rSirt1 supplementation rescued cardiac function. This was associated with a modulation of the myocardial lipidome and a downregulation of genes involved in lipid metabolism, trafficking, inflammation, and PPARG signaling. These findings were confirmed in the human diabetic myocardium. Treatments that increase Sirt1 levels may represent a promising strategy to prevent myocardial lipid abnormalities and MCM development.


Asunto(s)
Diabetes Mellitus , Cardiomiopatías Diabéticas , Insuficiencia Cardíaca , Animales , Humanos , Ratones , Diabetes Mellitus/metabolismo , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/prevención & control , Insuficiencia Cardíaca/metabolismo , Inflamación/metabolismo , Lipidómica , Lípidos , Miocitos Cardíacos/metabolismo , PPAR gamma/metabolismo , Sirtuina 1/genética , Sirtuina 1/metabolismo , Volumen Sistólico , Triglicéridos/metabolismo , Función Ventricular Izquierda
3.
Front Cardiovasc Med ; 10: 1204483, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37522089

RESUMEN

Mitochondria are cellular organelles which generate adenosine triphosphate (ATP) molecules for the maintenance of cellular energy through the oxidative phosphorylation. They also regulate a variety of cellular processes including apoptosis and metabolism. Of interest, the inner part of mitochondria-the mitochondrial matrix-contains a circular molecule of DNA (mtDNA) characterised by its own transcriptional machinery. As with genomic DNA, mtDNA may also undergo nucleotide mutations that have been shown to be responsible for mitochondrial dysfunction. During physiological aging, the mitochondrial membrane potential declines and associates with enhanced mitophagy to avoid the accumulation of damaged organelles. Moreover, if the dysfunctional mitochondria are not properly cleared, this could lead to cellular dysfunction and subsequent development of several comorbidities such as cardiovascular diseases (CVDs), diabetes, respiratory and cardiovascular diseases as well as inflammatory disorders and psychiatric diseases. As reported for genomic DNA, mtDNA is also amenable to chemical modifications, namely DNA methylation. Changes in mtDNA methylation have shown to be associated with altered transcriptional programs and mitochondrial dysfunction during aging. In addition, other epigenetic signals have been observed in mitochondria, in particular the interaction between mtDNA methylation and non-coding RNAs. Mitoepigenetic modifications are also involved in the pathogenesis of CVDs where oxygen chain disruption, mitochondrial fission, and ROS formation alter cardiac energy metabolism leading to hypertrophy, hypertension, heart failure and ischemia/reperfusion injury. In the present review, we summarize current evidence on the growing importance of epigenetic changes as modulator of mitochondrial function in aging. A better understanding of the mitochondrial epigenetic landscape may pave the way for personalized therapies to prevent age-related diseases.

4.
Cardiovasc Diabetol ; 22(1): 144, 2023 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-37349757

RESUMEN

BACKGROUND: The nuclear receptor corepressor 1 (NCOR1) plays an important role in the regulation of gene expression in immunometabolic conditions by connecting chromatin-modifying enzymes, coregulators and transcription factors. NCOR1 has been shown to be involved in cardiometabolic diseases. Recently, we demonstrated that the deletion of macrophage NCOR1 aggravates atherosclerosis by promoting CD36-triggered foam cell formation via PPARG derepression. PURPOSE: Since NCOR1 modulates the function of several key regulators involved in hepatic lipid and bile acid metabolism, we hypothesized that its deletion in hepatocytes alters lipid metabolism and atherogenesis. METHODS: To test this hypothesis, we generated hepatocyte-specific Ncor1 knockout mice on a Ldlr-/- background. Besides assessing the progression of the disease in thoracoabdominal aortae en face, we analyzed hepatic cholesterol and bile acid metabolism at expression and functional levels. RESULTS: Our data demonstrate that liver-specific Ncor1 knockout mice on an atherosclerosis-prone background develop less atherosclerotic lesions than controls. Interestingly, under chow diet, plasma cholesterol levels of liver-specific Ncor1 knockout mice were slightly higher compared to control, but strongly reduced compared to control mice after feeding them an atherogenic diet for 12 weeks. Moreover, the hepatic cholesterol content was decreased in liver-specific Ncor1 knockout compared to control mice. Our mechanistic data revealed that NCOR1 reprograms the synthesis of bile acids towards the alternative pathway, which in turn reduce bile hydrophobicity and enhances fecal cholesterol excretion. CONCLUSIONS: Our data suggest that hepatic Ncor1 deletion in mice decreases atherosclerosis development by reprograming bile acid metabolism and enhancing fecal cholesterol excretion.


Asunto(s)
Aterosclerosis , Esteroles , Ratones , Animales , Esteroles/metabolismo , Hígado/metabolismo , Colesterol , Aterosclerosis/genética , Aterosclerosis/prevención & control , Aterosclerosis/metabolismo , Ratones Noqueados , Ácidos y Sales Biliares/metabolismo , Metabolismo de los Lípidos , Ratones Endogámicos C57BL , Co-Represor 1 de Receptor Nuclear/genética , Co-Represor 1 de Receptor Nuclear/metabolismo
5.
Eur Heart J ; 40(4): 383-391, 2019 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-29077881

RESUMEN

Aims: Accumulation of reactive oxygen species (ROS) promotes vascular disease in obesity, but the underlying molecular mechanisms remain poorly understood. The adaptor p66Shc is emerging as a key molecule responsible for ROS generation and vascular damage. This study investigates whether epigenetic regulation of p66Shc contributes to obesity-related vascular disease. Methods and results: ROS-driven endothelial dysfunction was observed in visceral fat arteries (VFAs) isolated from obese subjects when compared with normal weight controls. Gene profiling of chromatin-modifying enzymes in VFA revealed a significant dysregulation of methyltransferase SUV39H1 (fold change, -6.9, P < 0.01), demethylase JMJD2C (fold change, 3.2, P < 0.01), and acetyltransferase SRC-1 (fold change, 5.8, P < 0.01) in obese vs. control VFA. These changes were associated with reduced di-(H3K9me2) and trimethylation (H3K9me3) as well as acetylation (H3K9ac) of histone 3 lysine 9 (H3K9) on p66Shc promoter. Reprogramming SUV39H1, JMJD2C, and SRC-1 in isolated endothelial cells as well as in aortas from obese mice (LepOb/Ob) suppressed p66Shc-derived ROS, restored nitric oxide levels, and rescued endothelial dysfunction. Consistently, in vivo editing of chromatin remodellers blunted obesity-related vascular p66Shc expression. We show that SUV39H1 is the upstream effector orchestrating JMJD2C/SRC-1 recruitment to p66Shc promoter. Indeed, SUV39H1 overexpression in obese mice erased H3K9-related changes on p66Shc promoter, while SUV39H1 genetic deletion in lean mice recapitulated obesity-induced H3K9 remodelling and p66Shc transcription. Conclusion: These results uncover a novel epigenetic mechanism underlying endothelial dysfunction in obesity. Targeting SUV39H1 may attenuate oxidative transcriptional programmes and thus prevent vascular disease in obese individuals.


Asunto(s)
Regulación de la Expresión Génica , Histona Demetilasas con Dominio de Jumonji/genética , Metiltransferasas/genética , Coactivador 1 de Receptor Nuclear/genética , Obesidad/genética , Estrés Oxidativo/fisiología , Proteínas Represoras/genética , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/genética , Animales , Western Blotting , Células Cultivadas , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Femenino , N-Metiltransferasa de Histona-Lisina , Humanos , Histona Demetilasas con Dominio de Jumonji/biosíntesis , Masculino , Metiltransferasas/biosíntesis , Ratones Endogámicos C57BL , Ratones Mutantes , Persona de Mediana Edad , Coactivador 1 de Receptor Nuclear/biosíntesis , Obesidad/metabolismo , Obesidad/patología , ARN/genética , Especies Reactivas de Oxígeno/metabolismo , Proteínas Represoras/biosíntesis , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/biosíntesis , Transcripción Genética , Vasodilatación
6.
Int J Cardiol ; 250: 207-214, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-28988828

RESUMEN

Cardiovascular disease (CVD) is a recognized age-dependent condition whose incidence is set to increase due to the gradual aging of the population. Moreover, ischemic cardiovascular diseases (i.e. stroke, myocardial infarction, critical limb ischemia) requiring blood vessel growth are associated with a worse outcome in elderly patients. Therefore, understanding the molecular cues regulating the vascular repair process is of paramount importance to prevent undesirable cardiovascular complications in this setting. A growing body of evidence suggests that epigenetic modifications - changes to the genome that do not involve changes in DNA sequence - may significantly derail gene expression trajectories during the life course, thus affecting molecular phenotype and functionality of angiogenic cells, namely mature endothelial cells (ECs), endothelial progenitor cells (EPCs), and bone-marrow (BM)-derived angiogenic cells. In the present review, we discuss the emerging role of epigenetics in age-related impairment of the angiogenic process. Specifically, the following aspects are critically addressed: i) defective angiogenic process in aging; ii) impact of epigenetics (DNA methylation, histone modifications, microRNAs, long noncoding RNAs) on phenotype and function of ECs and BM-derived angiogenic cells; iii) clinical perspectives on epigenetic biomarkers and reprogramming approaches for autologous transplantation. A scrutiny characterization of the "old epigenome" may provide unprecedented insights to develop preventive strategies and regenerative therapeutic interventions in the elderly.


Asunto(s)
Envejecimiento/genética , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/terapia , Epigénesis Genética/genética , Medicina Regenerativa/tendencias , Anciano , Envejecimiento/patología , Enfermedades Cardiovasculares/diagnóstico , Humanos , Fenotipo , Medicina Regenerativa/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...