Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cardiovasc Diabetol ; 23(1): 107, 2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38553774

RESUMEN

BACKGROUND: Diabetes-induced trained immunity contributes to the development of atherosclerosis and its complications. This study aimed to investigate in humans whether epigenetic signals involved in immune cell activation and inflammation are initiated in hematopoietic stem/progenitor cells (HSPCs) and transferred to differentiated progeny. METHODS AND RESULTS: High glucose (HG)-exposure of cord blood (CB)-derived HSPCs induced a senescent-associated secretory phenotype (SASP) characterized by cell proliferation lowering, ROS production, telomere shortening, up-regulation of p21 and p27genes, upregulation of NFkB-p65 transcription factor and increased secretion of the inflammatory cytokines TNFα and IL6. Chromatin immunoprecipitation assay (ChIP) of p65 promoter revealed that H3K4me1 histone mark accumulation and methyltransferase SetD7 recruitment, along with the reduction of repressive H3K9me3 histone modification, were involved in NFkB-p65 upregulation of HG-HSPCs, as confirmed by increased RNA polymerase II engagement at gene level. The differentiation of HG-HSPCs into myeloid cells generated highly responsive monocytes, mainly composed of intermediate subsets (CD14hiCD16+), that like the cells from which they derive, were characterized by SASP features and similar epigenetic patterns at the p65 promoter. The clinical relevance of our findings was confirmed in sternal BM-derived HSPCs of T2DM patients. In line with our in vitro model, T2DM HSPCs were characterized by SASP profile and SETD7 upregulation. Additionally, they generated, after myeloid differentiation, senescent monocytes mainly composed of proinflammatory intermediates (CD14hiCD16+) characterized by H3K4me1 accumulation at NFkB-p65 promoter. CONCLUSIONS: Hyperglycemia induces marked chromatin modifications in HSPCs, which, once transmitted to the cell progeny, contributes to persistent and pathogenic changes in immune cell function and composition.


Asunto(s)
Diabetes Mellitus Tipo 2 , Inmunidad Entrenada , Humanos , Fenotipo Secretor Asociado a la Senescencia , Células Madre Hematopoyéticas/metabolismo , Antígenos CD34/metabolismo , Epigénesis Genética , Diabetes Mellitus Tipo 2/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo
2.
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.

3.
J Mol Cell Cardiol ; 174: 56-62, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36414111

RESUMEN

OBJECTIVE: Arterial thrombosis may be initiated by endothelial inflammation or denudation, activation of blood-borne elements or the coagulation system. Tissue factor (TF), a central trigger of the coagulation cascade, is regulated by the pro-inflammatory NF-κB-dependent pathways. Sirtuin 6 (SIRT6) is a nuclear member of the sirtuin family of NAD+-dependent deacetylases and is known to inhibit NF-κB signaling. Its constitutive deletion in mice shows early lethality with hypoglycemia and accelerated aging. Of note, the role of SIRT6 in arterial thrombosis remains unknown. Thus, we hypothesized that endothelial SIRT6 protects from arterial thrombosis by modulating inhibition of NF-κB-associated pathways. APPROACH AND RESULTS: Using a laser-induced carotid thrombosis model, in vivo arterial occlusion occurred 45% faster in 12-week-old male endothelial-specific Sirt6-/- mice as compared to Sirt6fl/fl controls (n ≥ 9 per group; p = 0.0012). Levels of procoagulant TF were increased in animals lacking endothelial SIRT6 as compared to control littermates. Similarly, in cultured human aortic endothelial cells, SIRT6 knockdown increased TF mRNA, protein and activity. Moreover, SIRT6 knockdown increased mRNA levels of NF-κB-associated genes tumor necrosis factor alpha (TNF-α), poly [ADP-ribose] polymerase 1 (PARP-1), vascular cell adhesion molecule 1 (VCAM-1), and cyclooxygenase-2 (COX-2); at the protein level, COX-2, VCAM-1, TNF-α, and cleaved PARP-1 remained increased after Sirt6 knockdown. CONCLUSIONS: Endothelium-specific Sirt6 deletion promotes arterial thrombosis in mice. In cultured human aortic endothelial cells, SIRT6 silencing enhances TF expression and activates pro-inflammatory pathways including TNF-α, cleaved PARP-1, VCAM-1 and COX-2. Hence, endogenous endothelial SIRT6 exerts a protective role in experimental arterial thrombosis.


Asunto(s)
Sirtuinas , Trombosis , Animales , Humanos , Masculino , Ratones , Células Cultivadas , Ciclooxigenasa 2 , Células Endoteliales , FN-kappa B , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Sirtuinas/genética , Trombosis/genética , Factor de Necrosis Tumoral alfa , Molécula 1 de Adhesión Celular Vascular/genética
4.
Circ Res ; 131(6): 476-491, 2022 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-35968712

RESUMEN

BACKGROUND: Experimental evidence suggests a key role of SIRT1 (silent information regulator 1) in age- and metabolic-related vascular dysfunction. Whether these effects hold true in the human microvasculature is unknown. We aimed to investigate the SIRT1 role in very early stages of age- and obesity-related microvascular dysfunction in humans. METHODS: Ninety-five subjects undergoing elective laparoscopic surgery were recruited and stratified based on their body mass index status (above or below 30 kg/m2) and age (above or below 40 years) in 4 groups: Young Nonobese, Young Obese, Old Nonobese, and Old Obese. We measured small resistance arteries' endothelial function by pressurized micromyography before and after incubation with a SIRT1 agonist (SRT1720) and a mitochondria reactive oxygen species (mtROS) scavenger (MitoTEMPO). We assessed vascular levels of mtROS and nitric oxide availability by confocal microscopy and vascular gene expression of SIRT1 and mitochondrial proteins by qPCR. Chromatin immunoprecipitation assay was employed to investigate SIRT1-dependent epigenetic regulation of mitochondrial proteins. RESULTS: Compared with Young Nonobese, obese and older patients showed lower vascular expression of SIRT1 and antioxidant proteins (FOXO3 [forkhead box protein O3] and SOD2) and higher expression of pro-oxidant and aging mitochondria proteins p66Shc and Arginase II. Old Obese, Young Obese and Old Nonobese groups endothelial dysfunction was rescued by SRT1720. The restoration was comparable to the one obtained with mitoTEMPO. These effects were explained by SIRT1-dependent chromatin changes leading to reduced p66Shc expression and upregulation of proteins involved in mitochondria respiratory chain. CONCLUSIONS: SIRT1 is a novel central modulator of the earliest microvascular damage induced by age and obesity. Through a complex epigenetic control mainly involving p66Shc and Arginase II, it influences mtROS levels, NO availability, and the expression of proteins of the mitochondria respiratory chain. Therapeutic modulation of SIRT1 restores obesity- and age-related endothelial dysfunction. Early targeting of SIRT1 might represent a crucial strategy to prevent age- and obesity-related microvascular dysfunction.


Asunto(s)
Arginasa , Obesidad , Sirtuina 1 , Enfermedades Vasculares , Adulto , Arginasa/metabolismo , Epigénesis Genética , Humanos , Proteínas Mitocondriales/metabolismo , Óxido Nítrico/metabolismo , Obesidad/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sirtuina 1/genética , Sirtuina 1/metabolismo , Enfermedades Vasculares/etiología
5.
Handb Exp Pharmacol ; 270: 511-535, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33474673

RESUMEN

Emerging evidence suggests the growing importance of "nongenetic factors" in the pathogenesis of atherosclerotic vascular disease. Indeed, the inherited genome determines only part of the risk profile as genomic approaches do not take into account additional layers of biological regulation by "epi"-genetic changes. Epigenetic modifications are defined as plastic chemical changes of DNA/histone complexes which critically affect gene activity without altering the DNA sequence. These modifications include DNA methylation, histone posttranslational modifications, and non-coding RNAs and have the ability to modulate gene expression at both transcriptional and posttranscriptional level. Notably, epigenetic signals are mainly induced by environmental factors (i.e., pollution, smoking, noise) and, once acquired, may be transmitted to the offspring. The inheritance of adverse epigenetic changes may lead to premature deregulation of pathways involved in vascular damage and endothelial dysfunction. Here, we describe the emerging role of epigenetic modifications as fine-tuners of gene transcription in atherosclerosis. Specifically, the following aspects are described in detail: (1) discovery and impact of the epigenome in cardiovascular disease, (2) the epigenetic landscape in atherosclerosis; (3) inheritance of epigenetic signals and premature vascular disease; (4) epigenetic control of lipid metabolism, vascular oxidative stress, inflammation, autophagy, and apoptosis; (5) epigenetic biomarkers in patients with atherosclerosis; (6) novel therapeutic strategies to modulate epigenetic marks. Understanding the individual epigenetic profile may pave the way for new approaches to determine cardiovascular risk and to develop personalized therapies to treat atherosclerosis and its complications.


Asunto(s)
Aterosclerosis , Epigenoma , Aterosclerosis/genética , Metilación de ADN , Epigénesis Genética , Genoma , Humanos
6.
Antioxid Redox Signal ; 36(10-12): 667-684, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34913726

RESUMEN

Aims: Therapeutic modulation of blood vessel growth holds promise for the prevention of limb ischemia in diabetic (DM) patients with peripheral artery disease (PAD). Epigenetic changes, namely, posttranslational histone modifications, participate in angiogenic response suggesting that chromatin-modifying drugs could be beneficial in this setting. Apabetalone (APA), a selective inhibitor of bromodomain (BRD) and bromodomain and extraterminal containing protein family (BET) proteins, prevents bromodomain-containing protein 4 (BRD4) interactions with chromatin thus modulating transcriptional programs in different organs. We sought to investigate whether APA affects angiogenic response in diabetes. Results: Compared with vehicle, APA restored tube formation and migration in human aortic endothelial cells (HAECs) exposed to high-glucose (HG) levels. Expression profiling of angiogenesis genes showed that APA prevents HG-induced upregulation of the antiangiogenic molecule thrombospondin-1 (THBS1). ChIP-seq and chromatin immunoprecipitation (ChIP) assays in HG-treated HAECs showed the enrichment of both BRD4 and active marks (H3K27ac) on THBS1 promoter, whereas BRD4 inhibition by APA prevented chromatin accessibility and THBS1 transcription. Mechanistically, we show that THBS1 inhibits angiogenesis by suppressing vascular endothelial growth factor A (VEGFA) signaling, while APA prevents these detrimental changes. In diabetic mice with hind limb ischemia, epigenetic editing by APA restored the THBS1/VEGFA axis, thus improving limb vascularization and perfusion, compared with vehicle-treated animals. Finally, epigenetic regulation of THBS1 by BRD4/H3K27ac was also reported in DM patients with PAD compared with nondiabetic controls. Innovation: This is the first study showing that BET protein inhibition by APA restores angiogenic response in experimental diabetes. Conclusions: Our findings set the stage for preclinical studies and exploratory clinical trials testing APA in diabetic PAD. Antioxid. Redox Signal. 36, 667-684.


Asunto(s)
Diabetes Mellitus Experimental , Factores de Transcripción , Animales , Proteínas de Ciclo Celular/genética , Cromatina , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/genética , Células Endoteliales/metabolismo , Epigénesis Genética , Humanos , Isquemia , Ratones , Proteínas Nucleares/genética , Quinazolinonas , Factores de Transcripción/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
Minerva Cardiol Angiol ; 69(3): 331-345, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32996305

RESUMEN

Despite significant advances in our understanding of cardiovascular disease (CVD) we are still far from having developed breakthrough strategies to combat coronary atherosclerosis and heart failure, which account for most of CV deaths worldwide. Available cardiovascular therapies have failed to show to be equally effective in all patients, suggesting that inter-individual diversity is an important factor when it comes to conceive and deliver effective personalized treatments. Genome mapping has proved useful in identifying patients who could benefit more from specific drugs depending on genetic variances; however, our genetic make-up determines only a limited part of an individual's risk profile. Recent studies have demonstrated that epigenetic changes - defined as dynamic changes of DNA and histones which do not affect DNA sequence - are key players in the pathophysiology of cardiovascular disease and may participate to delineate cardiovascular risk trajectories over the lifetime. Epigenetic modifications include changes in DNA methylation, histone modifications and non-coding RNAs and these epigenetic signals have shown to cooperate in modulating chromatin accessibility to transcription factors and gene expression. Environmental factors such as air pollution, smoking, psychosocial context, and unhealthy diet regimens have shown to significantly modify the epigenome thus leading to altered transcriptional programs and CVD phenotypes. Therefore, the integration of genetic and epigenetic information might be invaluable to build individual maps of cardiovascular risk and hence, could be employed for the design of customized diagnostic and therapeutic strategies. In the present review, we discuss the growing importance of epigenetic information and its putative implications in cardiovascular precision medicine.


Asunto(s)
Sistema Cardiovascular , Medicina de Precisión , Sistema Cardiovascular/metabolismo , Metilación de ADN , Epigénesis Genética , Histonas/genética , Humanos
8.
Circ Res ; 127(10): 1261-1273, 2020 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-32815777

RESUMEN

RATIONALE: Hyperglycemia -induced reactive oxygen species are key mediators of cardiac dysfunction. JunD (Jund proto-oncogene subunit), a member of the AP-1 (activator protein-1) family of transcription factors, is emerging as a major gatekeeper against oxidative stress. However, its contribution to redox state and inflammation in the diabetic heart remains to be elucidated. OBJECTIVE: The present study investigates the role of JunD in hyperglycemia-induced and reactive oxygen species-driven myocardial dysfunction. METHODS AND RESULTS: JunD mRNA and protein expression were reduced in the myocardium of mice with streptozotocin-induced diabetes mellitus as compared to controls. JunD downregulation was associated with oxidative stress and left ventricular dysfunction assessed by electron spin resonance spectroscopy as well as conventional and 2-dimensional speckle-tracking echocardiography. Furthermore, myocardial expression of free radical scavenger superoxide dismutase 1 and aldehyde dehydrogenase 2 was reduced, whereas the NOX2 (NADPH [nicotinamide adenine dinucleotide phosphatase] oxidase subunit 2) and NOX4 (NADPH [nicotinamide adenine dinucleotide phosphatase] oxidase subunit 4) were upregulated. The redox changes were associated with increased NF-κB (nuclear factor kappa B) binding activity and expression of inflammatory mediators. Interestingly, mice with cardiac-specific overexpression of JunD via the α MHC (α- myosin heavy chain) promoter (α MHC JunDtg) were protected against hyperglycemia-induced cardiac dysfunction. We also showed that JunD was epigenetically regulated by promoter hypermethylation, post-translational modification of histone marks, and translational repression by miRNA (microRNA)-673/menin. Reduced JunD mRNA and protein expression were confirmed in left ventricular specimens obtained from patients with type 2 diabetes mellitus as compared to nondiabetic subjects. CONCLUSIONS: Here, we show that a complex epigenetic machinery involving DNA methylation, histone modifications, and microRNAs mediates hyperglycemia-induced JunD downregulation and myocardial dysfunction in experimental and human diabetes mellitus. Our results pave the way for tissue-specific therapeutic modulation of JunD to prevent diabetic cardiomyopathy.


Asunto(s)
Cardiomiopatías Diabéticas/genética , Epigénesis Genética , Hiperglucemia/complicaciones , Proteínas Proto-Oncogénicas c-jun/genética , Animales , Metilación de ADN , Cardiomiopatías Diabéticas/etiología , Cardiomiopatías Diabéticas/metabolismo , Código de Histonas , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Miocardio/metabolismo , NADPH Oxidasa 2/genética , NADPH Oxidasa 2/metabolismo , NADPH Oxidasa 4/genética , NADPH Oxidasa 4/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo
9.
High Blood Press Cardiovasc Prev ; 27(5): 363-371, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32740853

RESUMEN

Epigenetic processing takes centre stage in cardiometabolic diseases (obesity, metabolic syndrome, type 2 diabetes, hypertension), where it participates in adiposity, inflammation, endothelial dysfunction, vascular insulin resistance and atherosclerosis. Epigenetic modifications, defined as heritable changes in gene expression that do not entail mutation in the DNA sequence, are mainly induced by environmental stimuli (stress, pollution, cigarette smoking) and are gaining considerable interest due to their causal role in cardiovascular disease, and their amenability to pharmacological intervention. Importantly, epigenetic modifications acquired during life can be transmitted to the offspring and exert their biological effects across multiple generations. Indeed, such transgenerational transmission of epigenetic signals may contribute to anticipating cardiovascular and metabolic disease phenotypes already in children and young adults. A deeper understanding of environmental factors and their effects on the epigenetic machinery and transcriptional programs is warranted to develop effective mechanism-based therapeutic strategies. The clinical application of epigenetic drugs-also known as "epi-drugs"-is currently exploding in the field of cardiovascular disease. The present review describes the main epigenetic networks underlying cardiometabolic alterations and sheds light on specific points of intervention for pharmacological reprogramming in this setting.


Asunto(s)
Enfermedades Cardiovasculares/genética , Endotelio Vascular/metabolismo , Epigénesis Genética , Síndrome Metabólico/genética , Animales , Fármacos Cardiovasculares/uso terapéutico , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiopatología , Epigénesis Genética/efectos de los fármacos , Interacción Gen-Ambiente , Humanos , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/epidemiología , Síndrome Metabólico/metabolismo , Factores de Riesgo , Transducción de Señal
10.
J Mol Cell Cardiol ; 139: 24-32, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31972266

RESUMEN

AIMS: Sirtuin 6 (Sirt6) is a NAD+-dependent deacetylase that plays a key role in DNA repair, inflammation and lipid regulation. Sirt6-null mice show severe metabolic defects and accelerated aging. Macrophage-foam cell formation via scavenger receptors is a key step in atherogenesis. We determined the effects of bone marrow-restricted Sirt6 deletion on foam cell formation and atherogenesis using a mouse model. METHODS AND RESULTS: Sirt6 deletion in bone marrow-derived cells increased aortic plaques, lipid content and macrophage numbers in recipient Apoe-/- mice fed a high-cholesterol diet for 12 weeks (n = 12-14, p < .001). In RAW macrophages, Sirt6 overexpression reduced oxidized low-density lipoprotein (oxLDL) uptake, Sirt6 knockdown enhanced it and increased mRNA and protein levels of macrophage scavenger receptor 1 (Msr1), whereas levels of other oxLDL uptake and efflux transporters remained unchanged. Similarly, in human primary macrophages, Sirt6 knockdown increased MSR1 protein levels and oxLDL uptake. Double knockdown of Sirt6 and Msr1 abolished the increase in oxLDL uptake observed upon Sirt6 single knockdown. FACS analyses of macrophages from aortic plaques of Sirt6-deficient bone marrow-transplanted mice showed increased MSR1 protein expression. Double knockdown of Sirt6 and the transcription factor c-Myc in RAW cells abolished the increase in Msr1 mRNA and protein levels; c-Myc overexpression increased Msr1 mRNA and protein levels. CONCLUSIONS: Loss of Sirt6 in bone marrow-derived cells is proatherogenic; hereby macrophages play an important role given a c-Myc-dependent increase in MSR1 protein expression and an enhanced oxLDL uptake in human and murine macrophages. These findings assign endogenous SIRT6 in macrophages an important atheroprotective role.


Asunto(s)
Aterosclerosis/metabolismo , Aterosclerosis/patología , Médula Ósea/patología , Eliminación de Gen , Receptores Depuradores de Clase A/metabolismo , Sirtuinas/genética , Sirtuinas/metabolismo , Animales , Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/metabolismo , Trasplante de Médula Ósea , Regulación hacia Abajo , Técnicas de Silenciamiento del Gen , Hematopoyesis , Homocigoto , Humanos , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Modelos Biológicos , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Células RAW 264.7
11.
Thromb Haemost ; 120(1): 168-180, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31858519

RESUMEN

Ischemia/reperfusion (I/R) injury in acute myocardial infarction activates several deleterious molecular mechanisms. The transcription factor JunD regulates pathways involved in oxidative stress as well as in cellular proliferation, differentiation, and death. The present study investigated the potential role of JunD as a modulator of myocardial injury pathways in a mouse model of cardiac I/R injury. Infarct size, systemic and local inflammation, and production of reactive oxygen species, as well as cytosolic and mitochondrial apoptotic pathways were investigated in adult males after myocardial I/R. In wild-type (WT) mice, 30 minutes after ischemia and up to 24 hours following reperfusion, cardiac JunD messenger ribonucleic acid expression was reduced while JunB increased. Cardiac-specific JunD overexpressing mice (JunDTg/0 ) displayed larger infarcts compared with WT. However, postischemic inflammatory or oxidative responses did not differ. JunD overexpression reduced Sirt3 transcription by binding to its promoter, thus leading to mitochondrial dysfunction, myocardial cell death, and increased infarct size. On the other hand, JunD silencing reduced, while Sirt3 silencing increased infarct size. In human myocardial autopsy specimens, JunD-positive areas within the infarcted left ventricle staining corresponded to undetectable Sirt3 areas in consecutive sections of the same heart. Cardiac-specific JunD overexpression increases myocardial infarct size following I/R. These effects are mediated via Sirt3 transcriptional repression, mitochondrial swelling, and increased apoptosis, suggesting that JunD is a key regulator of myocardial I/R injury. The present data set the stage for further investigation of the potential role of Sirt3 activation as a novel target for the treatment of acute myocardial infarction.


Asunto(s)
Mitocondrias/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/fisiología , Proteínas Proto-Oncogénicas c-jun/metabolismo , Daño por Reperfusión/metabolismo , Sirtuina 3/metabolismo , Animales , Apoptosis , Modelos Animales de Enfermedad , Regulación hacia Abajo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/patología , Miocardio/patología , Especificidad de Órganos , Proteínas Proto-Oncogénicas c-jun/genética , Daño por Reperfusión/patología , Sirtuina 3/genética , Regulación hacia Arriba
12.
J Hypertens ; 37(9): 1861-1870, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30950975

RESUMEN

BACKGROUND: Tenascin C (TN-C) is considered to play a pathophysiological role in maladaptive left ventricular remodeling. Yet, the mechanism underlying TN-C-dependent cardiac dysfunction remains elusive. METHOD: The present study was designed to investigate the effect of hypoxia and hypertrophic stimuli on TN-C expression in H9c2 cells and its putative regulation by epigenetic mechanisms, namely DNA promoter methylation and microRNAs. In addition, rats subjected to myocardial infarction (MI) were investigated. H9c2 cells were subjected to oxygen and glucose deprivation; incubated with angiotensin II (Ang II); or human TN-C (hTN-C) purified protein. Hypertrophic and fibrotic markers, TN-C promoter methylation as well as mir-335 expression were assessed by reverse transcription and quantitative polymerase chain reaction while TN-C protein levels were assessed by ELISA. RESULTS: Tn-C mRNA expression was markedly increased by both oxygen and glucose deprivation and Ang II (P < 0.01, respectively). In addition, Ang-II-dependent TN-C upregulation was explained by reduced promoter methylation (P < 0.05). Cells treated with hTN-C displayed upregulation of Bnp, Mmp2, ß-Mhc, integrin α6 and integrin ß1. Furthermore, hTN-C treated cells showed a significant reduction in adenosine monophosphate and adenosine triphosphate levels. In vivo, plasma and myocardial TN-C levels were increased 7 days post MI (P < 0.05, respectively). This increment in TN-C was accompanied by upregulation of mir-335 (P < 0.01). In conclusion, both hypoxic and hypertrophic stimuli lead to epigenetically driven TN-C upregulation and subsequent impairment of cellular energy metabolism in cardiomyoblasts. CONCLUSION: These findings might enlighten our understanding on maladaptive left ventricular remodeling and direct towards a strong involvement of TN-C.


Asunto(s)
Cardiomegalia/metabolismo , Metilación de ADN , Hipoxia/metabolismo , Infarto del Miocardio/metabolismo , Tenascina/metabolismo , Angiotensina II , Animales , Enfermedad de la Arteria Coronaria , Metabolismo Energético , Epigénesis Genética , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular , Fibrosis , Cardiopatías/metabolismo , Humanos , Hipertrofia , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , MicroARNs/metabolismo , Miocardio/metabolismo , Proteínas del Tejido Nervioso , Ratas , Tenascina/genética , Remodelación Ventricular
13.
Vascul Pharmacol ; 115: 64-68, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30926561

RESUMEN

Obesity has many deleterious effects on the cardiovascular system, mediated by changes in insulin sensitivity, dyslipidaemia, oxidative stress and inflammation. Current therapies mainly focus on caloric intake suppression and bariatric surgery, however the efficacy of these approaches remains elusive as most patients regain their body weight within the next 5 years. A better understanding of the pathophysiology of obesity is of paramount importance for the development of new therapeutic strategies to prevent vascular complications. Autophagy has emerged as key self-degrading process responsible for the maintenance of cellular homeostasis. Defects in autophagy homeostasis are implicated in metabolic disorders, including obesity, insulin resistance, diabetes mellitus and atherosclerosis. Most importantly, autophagy regulates animal lifespan. Albeit ample preclinical evidence supports the therapeutic promise of autophagy modulators for the treatment of obesity and metabolic diseases, the clinical efficacy of pharmacological modulation of autophagy remains to be proven. Recent work has shown that GLP-1-based therapeutic approaches may positively affect autophagy in perivascular adipose tissue, thus improving obesity-related endothelial dysfunction. In the present review we discuss current evidence on the role of autophagy in obesity, with a specific focus on DPP-4 inhibitors (DPP-4i) and GLP-1 receptor agonists (GLP-1 RA) as modulators of this process. Experimental evidence on GLP-1-based approaches is critically discussed in light of recent clinical trials with DPP-4i and GLP-1 RA.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Fármacos Antiobesidad/uso terapéutico , Autofagia/efectos de los fármacos , Enfermedades Cardiovasculares/prevención & control , Ensayos Clínicos como Asunto , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/agonistas , Obesidad/tratamiento farmacológico , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Fármacos Antiobesidad/efectos adversos , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Inhibidores de la Dipeptidil-Peptidasa IV/efectos adversos , Medicina Basada en la Evidencia , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Humanos , Obesidad/epidemiología , Obesidad/metabolismo , Obesidad/patología , Factores de Riesgo , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento
14.
Eur Heart J ; 40(12): 997-1008, 2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30629164

RESUMEN

AIMS: Metabolic cardiomyopathy (MC)-characterized by intra-myocardial triglyceride (TG) accumulation and lipotoxic damage-is an emerging cause of heart failure in obese patients. Yet, its mechanisms remain poorly understood. The Activator Protein 1 (AP-1) member JunD was recently identified as a key modulator of hepatic lipid metabolism in obese mice. The present study investigates the role of JunD in obesity-induced MC. METHODS AND RESULTS: JunD transcriptional activity was increased in hearts from diet-induced obese (DIO) mice and was associated with myocardial TG accumulation and left ventricular (LV) dysfunction. Obese mice lacking JunD were protected against MC. In DIO hearts, JunD directly binds PPARγ promoter thus enabling transcription of genes involved in TG synthesis, uptake, hydrolysis, and storage (i.e. Fas, Cd36, Lpl, Plin5). Cardiac-specific overexpression of JunD in lean mice led to PPARγ activation, cardiac steatosis, and dysfunction, thereby mimicking the MC phenotype. In DIO hearts as well as in neonatal rat ventricular myocytes exposed to palmitic acid, Ago2 immunoprecipitation, and luciferase assays revealed JunD as a direct target of miR-494-3p. Indeed, miR-494-3p was down-regulated in hearts from obese mice, while its overexpression prevented lipotoxic damage by suppressing JunD/PPARγ signalling. JunD and miR-494-3p were also dysregulated in myocardial specimens from obese patients as compared with non-obese controls, and correlated with myocardial TG content, expression of PPARγ-dependent genes, and echocardiographic indices of LV dysfunction. CONCLUSION: miR-494-3p/JunD is a novel molecular axis involved in obesity-related MC. These results pave the way for approaches to prevent or treat LV dysfunction in obese patients.


Asunto(s)
Cardiomiopatías/metabolismo , Miocardio/metabolismo , Obesidad/complicaciones , Proteínas Proto-Oncogénicas c-jun/metabolismo , Animales , Cardiomiopatías/complicaciones , Cardiomiopatías/fisiopatología , Estudios de Casos y Controles , Dieta Alta en Grasa , Regulación hacia Abajo , Insuficiencia Cardíaca/etiología , Humanos , Metabolismo de los Lípidos , Ratones , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , PPAR gamma/metabolismo , Ratas , Factor de Transcripción AP-1/metabolismo , Activación Transcripcional , Triglicéridos/metabolismo , Disfunción Ventricular Izquierda/diagnóstico por imagen , Disfunción Ventricular Izquierda/fisiopatología , Disfunción Ventricular Izquierda/prevención & control
16.
J Physiol ; 594(8): 2061-73, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26391109

RESUMEN

Age is one of the major risk factors associated with cardiovascular disease (CVD). About one-fifth of the world population will be aged 65 or older by 2030, with an exponential increase in CVD prevalence. It is well established that environmental factors (overnutrition, smoking, pollution, sedentary lifestyles) may lead to premature defects in mitochondrial functionality, insulin signalling, endothelial homeostasis and redox balance, fostering early senescent features. Over the last few years, molecular investigations have unveiled common signalling networks which may link the ageing process with deterioration of cardiovascular homeostasis and metabolic disturbances, namely insulin resistance. These different processes seem to be highly interconnected and their interplay may favour adverse vascular and cardiac phenotypes responsible for myocardial infarction, stroke and heart failure. In the present review, we carefully describe novel molecular cues underpinning ageing, metabolism and CVD. In particular, we describe a dynamic interplay between emerging pathways such as FOXOs, AMPK, SIRT1, p66(Shc) , JunD and NF-kB. This overview will provide the background for attractive molecular targets to prevent age-driven pathology in the vasculature and the heart.


Asunto(s)
Envejecimiento/metabolismo , Enfermedades Cardiovasculares/metabolismo , Envejecimiento/patología , Animales , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/fisiopatología , Sistema Cardiovascular/crecimiento & desarrollo , Sistema Cardiovascular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Transducción de Señal
17.
Circ Cardiovasc Genet ; 8(1): 150-8, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25472959

RESUMEN

BACKGROUND: Cellular studies showed that histone methyltransferase Set7 mediates high glucose-induced inflammation via epigenetic regulation of the transcription factor NF-kB. However, the link between Set7 and vascular dysfunction in patients with diabetes mellitus remains unknown. This study was designed to investigate whether Set7 contributes to vascular dysfunction in patients with type 2 diabetes mellitus (T2DM). METHODS AND RESULTS: Set7-driven epigenetic changes on NF-kB p65 promoter and expression of NF-kB-dependent genes, cyclooxygenase 2 and inducible endothelial nitric oxide synthase, were assessed in peripheral blood mononuclear cells isolated from 68 subjects (44 patients with T2DM and 24 age-matched controls). Brachial artery flow-mediated dilation, 24-hour urinary levels of 8-isoprostaglandin F2α, and plasma adhesion molecules, intercellular cell adhesion molecule-1 and monocyte chemoattractant protein-1, were also determined. Experiments in human aortic endothelial cells exposed to high glucose were performed to elucidate the mechanisms of Set7-driven inflammation and oxidative stress. Set7 expression increased in peripheral blood mononuclear cells from patients with T2DM when compared with controls. Patients with T2DM showed Set7-dependent monomethylation of lysine 4 of histone 3 on NF-kB p65 promoter. This epigenetic signature was associated with upregulation of NF-kB, subsequent transcription of oxidant/inflammatory genes, and increased plasma levels of intercellular cell adhesion molecule-1 and monocyte chemoattractant protein-1. Interestingly, we found that Set7 expression significantly correlated with oxidative marker 8-isoprostaglandin F2α (r=0.38; P=0.01) and flow-mediated dilation (r=-0.34; P=0.04). In human aortic endothelial cells, silencing of Set7 prevented monomethylation of lysine 4 of histone 3 and abolished NF-kB-dependent oxidant and inflammatory signaling. CONCLUSIONS: Set7-induced epigenetic changes contribute to vascular dysfunction in patients with T2DM. Targeting this chromatin-modifying enzyme may represent a novel therapeutic approach to prevent atherosclerotic vascular disease in this setting.


Asunto(s)
Diabetes Mellitus Tipo 2/enzimología , Angiopatías Diabéticas/enzimología , Células Endoteliales/enzimología , Epigénesis Genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Adulto , Anciano , Células Cultivadas , Diabetes Mellitus Tipo 2/genética , Angiopatías Diabéticas/genética , Femenino , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Masculino , Persona de Mediana Edad , Regiones Promotoras Genéticas , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo
18.
Atherosclerosis ; 230(2): 191-7, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24075743

RESUMEN

Risk of diabetic complications continues to escalate overtime despite a multifactorial intervention with glucose-lowering drugs, anti-hypertensive agents and statins. In this perspective, a mechanisms-based therapeutic approach to vascular disease in diabetes represents a major challenge. Epigenetic signatures are emerging as important determinants of vascular disease in this setting. Methylation and acetylation of DNA and histones is a reversible process leading to dysregulation of oxidant and inflammatory genes such as mitochondrial adaptor p66(Shc) and transcription factor NF-kB p65. Epigenetic modifications associated with diabetes may contribute to the early identification of high risk individuals. Ongoing epigenomic analyses will be instrumental in identifying the epigenetic variations that are specifically associated with cardiovascular disease in patients with diabetes. Here, we describe a complex scenario of epigenetic changes and their putative link with diabetic vascular disease. Pharmacological reprogramming of diabetes-induced epigenetic signatures may be a promising option to dampen oxidative stress and inflammation, and thus prevent cardiovascular complications in this setting.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Epigénesis Genética , Enfermedades Vasculares/genética , Enfermedades Cardiovasculares/complicaciones , Enfermedades Cardiovasculares/prevención & control , Angiopatías Diabéticas/genética , Ambiente , Epigenómica , Humanos , Inflamación , Oxidantes/química , Estrés Oxidativo , Regiones Promotoras Genéticas , Especies Reactivas de Oxígeno , Riesgo , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Factor de Transcripción ReIA/metabolismo , Enfermedades Vasculares/prevención & control
19.
Int J Cancer ; 133(9): 2089-101, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23629727

RESUMEN

The RAS/RAF/MEK/MAPK and the PTEN/PI3K/AKT/mTOR pathways are key regulators of proliferation and survival in human cancer cells. Selective inhibitors of different transducer molecules in these pathways have been developed as molecular targeted anti-cancer therapies. The in vitro and in vivo anti-tumor activity of pimasertib, a selective MEK 1/2 inhibitor, alone or in combination with a PI3K inhibitor (PI3Ki), a mTOR inhibitor (everolimus), or with multi-targeted kinase inhibitors (sorafenib and regorafenib), that block also BRAF and CRAF, were tested in a panel of eight human lung and colon cancer cell lines. Following pimasertib treatment, cancer cell lines were classified as pimasertib-sensitive (IC50 for cell growth inhibition of 0.001 µM) or pimasertib-resistant. Evaluation of basal gene expression profiles by microarrays identified several genes that were up-regulated in pimasertib-resistant cancer cells and that were involved in both RAS/RAF/MEK/MAPK and PTEN/PI3K/AKT/mTOR pathways. Therefore, a series of combination experiments with pimasertib and either PI3Ki, everolimus, sorafenib or regorafenib were conducted, demonstrating a synergistic effect in cell growth inhibition and induction of apoptosis with sustained blockade in MAPK- and AKT-dependent signaling pathways in pimasertib-resistant human colon carcinoma (HCT15) and lung adenocarcinoma (H1975) cells. Finally, in nude mice bearing established HCT15 and H1975 subcutaneous tumor xenografts, the combined treatment with pimasertib and BEZ235 (a dual PI3K/mTOR inhibitor) or with sorafenib caused significant tumor growth delays and increase in mice survival as compared to single agent treatment. These results suggest that dual blockade of MAPK and PI3K pathways could overcome intrinsic resistance to MEK inhibition.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 2/antagonistas & inhibidores , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Femenino , Perfilación de la Expresión Génica , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Ratones , Ratones Endogámicos BALB C , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfohidrolasa PTEN/metabolismo , Compuestos de Fenilurea/farmacología , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piridinas/farmacología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Basic Res Cardiol ; 108(2): 334, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23411815

RESUMEN

The increasing population of cancer survivors faces considerable morbidity and mortality due to late effects of the antineoplastic therapy. Cardiotoxicity is a major limiting factor of therapy with doxorubicin (DOXO), the most effective anthracycline, and is characterized by a dilated cardiomyopathy that can develop even years after treatment. Studies in animals have proposed the cardiac progenitor cells (CPCs) as the cellular target responsible for DOXO-induced cardiomyopathy but the relevance of these observations to clinical settings is unknown. In this study, the analysis of the DOXO-induced cardiomyopathic human hearts showed that the majority of human CPCs (hCPCs) was senescent. In isolated hCPCs, DOXO triggered DNA damage response leading to apoptosis early after exposure, and telomere shortening and senescence at later time interval. Functional properties of hCPCs, such as migration and differentiation, were also negatively affected. Importantly, the differentiated progeny of DOXO-treated hCPCs prematurely expressed the senescence marker p16(INK4a). In conclusion, DOXO exposure severely affects the population of hCPCs and permanently impairs their function. Premature senescence of hCPCs and their progeny can be responsible for the decline in the regenerative capacity of the heart and may represent the cellular basis of DOXO-induced cardiomyopathy in humans.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Cardiomiopatía Dilatada/inducido químicamente , Senescencia Celular/efectos de los fármacos , Doxorrubicina/efectos adversos , Mioblastos Cardíacos/efectos de los fármacos , Células Madre/efectos de los fármacos , Adulto , Antibióticos Antineoplásicos/uso terapéutico , Western Blotting , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Muerte Celular/efectos de los fármacos , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Daño del ADN/efectos de los fármacos , Doxorrubicina/uso terapéutico , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-kit/metabolismo , Homeostasis del Telómero , beta-Galactosidasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA