Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cardiovasc Res ; 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38289873

RESUMEN

AIMS: Vascular smooth muscle cells (SMCs) and their derivatives are key contributors to the development of atherosclerosis. However, studying changes in SMC gene expression in heterogeneous vascular tissues is challenging due to the technical limitations and high cost associated with current approaches. In this paper, we apply Translating Ribosome Affinity Purification sequencing (TRAP-Seq) to profile SMC-specific gene expression directly from tissue. METHODS AND RESULTS: To facilitate SMC-specific translatome analysis, we generated SMCTRAP mice, a transgenic mouse line expressing EGFP-tagged ribosomal protein L10a (EGFP-L10a) under the control of the SMC-specific αSMA promoter. These mice were further crossed with the atherosclerosis model Ldlr-/-, ApoB100/100 to generate SMCTRAP-AS mice and used to profile atherosclerosis-associated SMCs in thoracic aorta samples of 15-month-old SMCTRAP and SMCTRAP-AS mice. Our analysis of SMCTRAP-AS mice showed that EGFP-L10a expression was localized to SMCs in various tissues, including the aortic wall and plaque. The TRAP fraction demonstrated high enrichment of known SMC-specific genes, confirming the specificity of our approach. We identified several genes, including Cemip, Lum, Mfge8, Spp1, and Serpina3, that are known to be involved in atherosclerosis-induced gene expression. Moreover, we identified several novel genes not previously linked to SMCs in atherosclerosis, such as Anxa4, Cd276, Itih4, Myof, Pcdh11x, Rab31, Serpinb6b, Slc35e4, Slc8a3, and Spink5. Among them, we confirmed the SMC-specific expression of Itih4 in atherosclerotic lesions using immunofluorescence staining of mouse aortic roots and spatial transcriptomics of human carotid arteries. Furthermore, our more detailed analysis of Itih4 showed its link to coronary artery disease (CAD) through the colocalization of GWAS, splice-QTL, and protein-QTL signals. CONCLUSIONS: We generated a SMC-specific TRAP mouse line to study atherosclerosis and identified Itih4 as a novel SMC-expressed gene in atherosclerotic plaques, warranting further investigation of its putative function in extracellular matrix stability and genetic evidence of causality.

2.
J Biol Chem ; 299(11): 105291, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37748649

RESUMEN

Impaired oxygen homeostasis is a frequently encountered pathophysiological factor in multiple complex diseases, including cardiovascular disease and cancer. While the canonical hypoxia response pathway is well characterized, less is known about the role of noncoding RNAs in this process. Here, we investigated the nascent and steady-state noncoding transcriptional responses in endothelial cells and their potential roles in regulating the hypoxic response. Notably, we identify a novel antisense long noncoding RNA that convergently overlaps the majority of the hypoxia inducible factor 1 alpha (HIF1A) locus, which is expressed across several cell types and elevated in atherosclerotic lesions. The antisense (HIF1A-AS) is produced as a stable, unspliced, and polyadenylated nuclear retained transcript. HIF1A-AS is highly induced in hypoxia by both HIF1A and HIF2A and exhibits anticorrelation with the coding HIF1A transcript and protein expression. We further characterized this functional relationship by CRISPR-mediated bimodal perturbation of the HIF1A-AS promoter. We provide evidence that HIF1A-AS represses the expression of HIF1a in cis by repressing transcriptional elongation and deposition of H3K4me3, and that this mechanism is dependent on the act of antisense transcription itself. Overall, our results indicate a critical regulatory role of antisense mediated transcription in regulation of HIF1A expression and cellular response to hypoxia.


Asunto(s)
Células Endoteliales , ARN Largo no Codificante , Humanos , Hipoxia de la Célula , Células Endoteliales/metabolismo , Hipoxia/genética , Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Regiones Promotoras Genéticas , ARN Largo no Codificante/genética , Células Cultivadas
3.
Elife ; 122023 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-37199488

RESUMEN

Background: Sporadic venous malformation (VM) and angiomatosis of soft tissue (AST) are benign, congenital vascular anomalies affecting venous vasculature. Depending on the size and location of the lesion, symptoms vary from motility disturbances to pain and disfigurement. Due to the high recurrence of the lesions, more effective therapies are needed. Methods: As targeting stromal cells has been an emerging concept in anti-angiogenic therapies, here, by using VM/AST patient samples, RNA-sequencing, cell culture techniques, and a xenograft mouse model, we investigated the crosstalk of endothelial cells (EC) and fibroblasts and its effect on vascular lesion growth. Results: We report, for the first time, the expression and secretion of transforming growth factor A (TGFA) in ECs or intervascular stromal cells in AST and VM lesions. TGFA induced secretion of vascular endothelial growth factor (VEGF-A) in paracrine fashion, and regulated EC proliferation. Oncogenic PIK3CA variant in p.H1047R, a common somatic mutation found in these lesions, increased TGFA expression, enrichment of hallmark hypoxia, and in a mouse xenograft model, lesion size, and vascularization. Treatment with afatinib, a pan-ErbB tyrosine-kinase inhibitor, decreased vascularization and lesion size in a mouse xenograft model with ECs expressing oncogenic PIK3CA p.H1047R variant and fibroblasts. Conclusions: Based on the data, we suggest that targeting of both intervascular stromal cells and ECs is a potential treatment strategy for vascular lesions having a fibrous component. Funding: Academy of Finland, Ella and Georg Ehnrooth foundation, the ERC grants, Sigrid Jusélius Foundation, Finnish Foundation for Cardiovascular Research, Jane and Aatos Erkko Foundation, GeneCellNano Flagship program, and Department of Musculoskeletal and Plastic Surgery, Helsinki University Hospital.


Asunto(s)
Células Endoteliales , Malformaciones Vasculares , Humanos , Ratones , Animales , Células Endoteliales/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Transducción de Señal , Inhibidores de Proteínas Quinasas/farmacología , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Malformaciones Vasculares/tratamiento farmacológico , Malformaciones Vasculares/genética , Malformaciones Vasculares/patología
4.
Am J Hum Genet ; 110(5): 722-740, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-37060905

RESUMEN

Coronary artery disease (CAD) is a pandemic disease where up to half of the risk is explained by genetic factors. Advanced insights into the genetic basis of CAD require deeper understanding of the contributions of different cell types, molecular pathways, and genes to disease heritability. Here, we investigate the biological diversity of atherosclerosis-associated cell states and interrogate their contribution to the genetic risk of CAD by using single-cell and bulk RNA sequencing (RNA-seq) of mouse and human lesions. We identified 12 disease-associated cell states that we characterized further by gene set functional profiling, ligand-receptor prediction, and transcription factor inference. Importantly, Vcam1+ smooth muscle cell state genes contributed most to SNP-based heritability of CAD. In line with this, genetic variants near smooth muscle cell state genes and regulatory elements explained the largest fraction of CAD-risk variance between individuals. Using this information for variant prioritization, we derived a hybrid polygenic risk score (PRS) that demonstrated improved performance over a classical PRS. Our results provide insights into the biological mechanisms associated with CAD risk, which could make a promising contribution to precision medicine and tailored therapeutic interventions in the future.


Asunto(s)
Aterosclerosis , Enfermedad de la Arteria Coronaria , Humanos , Aterosclerosis/genética , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/patología , Factores de Riesgo , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo/métodos , Polimorfismo de Nucleótido Simple/genética
5.
Arterioscler Thromb Vasc Biol ; 43(1): e46-e61, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36384268

RESUMEN

BACKGROUND: Diabetes is a major risk factor for peripheral arterial disease. Clinical and preclinical studies suggest an impaired collateral remodeling and angiogenesis in response to atherosclerotic arterial occlusion in diabetic conditions, although the underlying mechanisms are poorly understood. OBJECTIVE: To clarify the cellular and molecular mechanisms underlying impaired postischemic adaptive vascular responses and to evaluate rHDL (reconstituted HDL)-ApoA-I nanotherapy to rescue the defect in type 2 diabetic mouse model of hindlimb ischemia. METHODS AND RESULTS: Hindlimb ischemia was induced by unilateral femoral artery ligation. Collateral and capillary parameters together with blood flow recovery were analyzed from normoxic adductor and ischemic gastrocnemius muscles, respectively, at day 3 and 7 post-ligation. In response to femoral artery ligation, collateral lumen area was significantly reduced in normoxic adductor muscles. Distally, ischemic gastrocnemius muscles displayed impaired perfusion recovery and angiogenesis paralleled with persistent inflammation. Muscle-specific mRNA sequencing revealed differential expression of genes critical for smooth muscle proliferation and sprouting angiogenesis in normoxic adductor and ischemic gastrocnemius, respectively, at day 7 post-ligation. Genes typical for macrophage (Mϕ) subsets were differentially expressed across both muscle types. Cell-specific gene expression, flow cytometry, and immunohistochemistry revealed persistent IFN-I response gene upregulation in arterial endothelial cells, ECs and Mϕs from T2DM mice associated with impaired collateral remodeling, angiogenesis and perfusion recovery. Furthermore, rHDL nanotherapy rescued impaired collateral remodeling and angiogenesis through dampening EC and Mϕ inflammation in T2DM mice. CONCLUSIONS: Our results suggest that an impaired collateral remodeling and sprouting angiogenesis in T2DM mice is associated with persistent IFN-I response in ECs and Mϕs. Dampening persistent inflammation and skewing ECs and Mϕ phenotype toward less inflammatory ones using rHDL nanotherapy may serve as a potential therapeutic target for T2DM peripheral arterial disease.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Enfermedad Arterial Periférica , Ratones , Animales , Neovascularización Fisiológica , Células Endoteliales/metabolismo , Apolipoproteína A-I/metabolismo , Macrófagos/metabolismo , Isquemia , Músculo Esquelético/irrigación sanguínea , Arteria Femoral/metabolismo , Diabetes Mellitus Tipo 2/genética , Inflamación/metabolismo , Enfermedad Arterial Periférica/metabolismo , Fenotipo , Miembro Posterior/irrigación sanguínea , Ratones Endogámicos C57BL , Circulación Colateral
6.
Front Genet ; 12: 719456, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34422021

RESUMEN

Atherosclerosis is a chronic inflammatory disease characterized by extensive remodeling of medium and large-sized arteries. Inward remodeling (=lumen shrinkage) of the vascular walls is the underlying cause for ischemia in target organs. Therefore, inward remodeling can be considered the predominant feature of atherosclerotic pathology. Outward remodeling (=lumen enlargement) is a physiological response compensating for lumen shrinkage caused by neointimal hyperplasia, but as a pathological response to changes in blood flow, outward remodeling leads to substantial arterial wall thinning. Thinned vascular walls are prone to rupture, and subsequent thrombus formation accounts for the majority of acute cardiovascular events. Pathological remodeling is driven by inflammatory cells which induce vascular smooth muscle cells to switch from quiescent to a proliferative and migratory phenotype. After decades of intensive research, the molecular mechanisms of arterial remodeling are starting to unfold. In this mini-review, we summarize the current knowledge of the epigenetic and transcriptional regulation of vascular smooth muscle cell phenotype switching from the contractile to the synthetic phenotype involved in arterial remodeling and discuss potential therapeutic options.

8.
Circ Res ; 127(11): 1437-1455, 2020 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-32981416

RESUMEN

RATIONALE: Atherosclerotic lesions are known for their cellular heterogeneity, yet the molecular complexity within the cells of human plaques has not been fully assessed. OBJECTIVE: Using single-cell transcriptomics and chromatin accessibility, we gained a better understanding of the pathophysiology underlying human atherosclerosis. METHODS AND RESULTS: We performed single-cell RNA and single-cell ATAC sequencing on human carotid atherosclerotic plaques to define the cells at play and determine their transcriptomic and epigenomic characteristics. We identified 14 distinct cell populations including endothelial cells, smooth muscle cells, mast cells, B cells, myeloid cells, and T cells and identified multiple cellular activation states and suggested cellular interconversions. Within the endothelial cell population, we defined subsets with angiogenic capacity plus clear signs of endothelial to mesenchymal transition. CD4+ and CD8+ T cells showed activation-based subclasses, each with a gradual decline from a cytotoxic to a more quiescent phenotype. Myeloid cells included 2 populations of proinflammatory macrophages showing IL (interleukin) 1B or TNF (tumor necrosis factor) expression as well as a foam cell-like population expressing TREM2 (triggering receptor expressed on myeloid cells 2) and displaying a fibrosis-promoting phenotype. ATACseq data identified specific transcription factors associated with the myeloid subpopulation and T cell cytokine profiles underlying mutual activation between both cell types. Finally, cardiovascular disease susceptibility genes identified using public genome-wide association studies data were particularly enriched in lesional macrophages, endothelial, and smooth muscle cells. CONCLUSIONS: This study provides a transcriptome-based cellular landscape of human atherosclerotic plaques and highlights cellular plasticity and intercellular communication at the site of disease. This detailed definition of cell communities at play in atherosclerosis will facilitate cell-based mapping of novel interventional targets with direct functional relevance for the treatment of human disease.


Asunto(s)
Enfermedades de las Arterias Carótidas/genética , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Linfocitos/metabolismo , Células Mieloides/metabolismo , Miocitos del Músculo Liso/metabolismo , Placa Aterosclerótica , Análisis de la Célula Individual , Transcriptoma , Anciano , Anciano de 80 o más Años , Animales , Enfermedades de las Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/patología , Transdiferenciación Celular , Secuenciación de Inmunoprecipitación de Cromatina , Bases de Datos Genéticas , Células Endoteliales/patología , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Linfocitos/patología , Masculino , Ratones , Persona de Mediana Edad , Células Mieloides/patología , Miocitos del Músculo Liso/patología , Fenotipo , RNA-Seq
9.
Biochim Biophys Acta Gen Subj ; 1863(2): 481-490, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30508567

RESUMEN

Statins are effective drugs used to prevent and treat cardiovascular diseases but their effects in the absence of low density lipoprotein receptor (LDLR) and on the risk of diabetes are not yet well characterized. The aim of this study was to clarify systemic and pleiotropic effects of rosuvastatin on cardiovascular and diabetic phenotypes. IGF-II/LDLR-/-ApoB100/100 hypercholesterolemic prediabetic mice were used to test the effects of rosuvastatin on plasma glucose, insulin, lipids, atherosclerosis and liver steatosis. To get a more comprehensive view about changes in gene expression RNA-sequencing was done from the liver. Rosuvastatin significantly reduced plasma cholesterol in hypercholesterolemic mice in the absence of LDLR but had no effects on atherosclerosis at aortic sinus level or in coronary arteries. Rosuvastatin also significantly reduced liver steatosis without any harmful effects on glucose or insulin metabolism. RNA-sequencing showed relatively specific effects of rosuvastatin on genes involved in cholesterol metabolism together with a significant anti-inflammatory gene expression profile in the liver. In addition, significant changes were found in the expression of Perilipin 4 and 5 which are involved in lipid droplet formation in the liver. For the first time it could be shown that Tribbles proteins are affected by rosuvastatin treatment in the hyperlipidemic mice. Rosuvastatin had several positive effects on hypercholesterolemic mice showing early signs of diabetes, many of which are unrelated to cholesterol and lipoprotein metabolism. These results increase our understanding about the systemic and pleiotropic effects of rosuvastatin in the absence of LDLR expression.


Asunto(s)
Anticolesterolemiantes/farmacología , Hipercolesterolemia/tratamiento farmacológico , Receptores de LDL/metabolismo , Rosuvastatina Cálcica/farmacología , Animales , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Modelos Animales de Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento , Hipercolesterolemia/metabolismo , Gotas Lipídicas/efectos de los fármacos , Gotas Lipídicas/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Receptores de LDL/deficiencia
10.
Atherosclerosis ; 281: 168-179, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30591183

RESUMEN

Underlying mechanisms of cardiovascular diseases (CVD) have been investigated for over 100 years and novel molecular level mechanisms in the pathophysiology are still continuously being discovered. Genetic polymorphisms (SNPs = single nucleotide polymorphisms) have explained about one tenth of the CVD risk, but polymorphisms fail to account for gene-environment interactions i.e. explain the dynamics of epigenome modifications in CVD. Accumulating evidence suggests that epigenetic modifications are actively reshaping pathological processes (e.g. dedifferentiation of smooth muscle cells, accumulation of senescent cells) in CVD. Senescence of vascular cells in ageing arteries not only counteracts regenerative processes but also exacerbates atherogenesis. Epigenome modifications include changes in DNA methylation, histone code and expression of non-coding RNAs. DNA methylation is a major epigenetic regulator modulating cell-type specific gene expression in mural cells, but there is some controversy regarding how to interpret the role of DNA hyper- and hypomethylation in CVD pathology. DNA hypomethylation (loss of methyl cytosines) appears to predominate in atherosclerosis, while a few genes become more methylated (i.e. hypermethylated) as the disease progresses in medium-sized and large arteries. The actual time-course of atherosclerosis-linked changes in genomic DNA methylation is still poorly studied. This review highlights recent novel findings which link alterations in DNA methylation to atherogenesis and points out new potential approaches for novel treatments.


Asunto(s)
Aterosclerosis/genética , Metilación de ADN , Epigénesis Genética , Placa Aterosclerótica , Animales , Aterosclerosis/epidemiología , Aterosclerosis/patología , Predisposición Genética a la Enfermedad , Humanos , Fenotipo , Pronóstico , Medición de Riesgo , Factores de Riesgo , Factores de Tiempo
11.
Front Cardiovasc Med ; 5: 159, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30456215

RESUMEN

Hypoxia occurs in human atherosclerotic lesions and has multiple adverse effects on endothelial cell metabolism. Recently, key roles of long non-coding RNAs (lncRNAs) in the development of atherosclerosis have begun to emerge. In this study, we investigate the lncRNA profiles of human umbilical vein endothelial cells subjected to hypoxia using global run-on sequencing (GRO-Seq). We demonstrate that hypoxia regulates the nascent transcription of ~1800 lncRNAs. Interestingly, we uncover evidence that promoter-associated lncRNAs are more likely to be induced by hypoxia compared to enhancer-associated lncRNAs, which exhibit an equal distribution of up- and downregulated transcripts. We also demonstrate that hypoxia leads to a significant induction in the activity of super-enhancers next to transcription factors and other genes implicated in angiogenesis, cell survival and adhesion, whereas super-enhancers near several negative regulators of angiogenesis were repressed. Despite the majority of lncRNAs exhibiting low detection in RNA-Seq, a subset of lncRNAs were expressed at comparable levels to mRNAs. Among these, MALAT1, HYMAI, LOC730101, KIAA1656, and LOC339803 were found differentially expressed in human atherosclerotic lesions, compared to normal vascular tissue, and may thus serve as potential biomarkers for lesion hypoxia.

12.
Circ Genom Precis Med ; 11(9): e002030, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30354327

RESUMEN

BACKGROUND: Tobacco smoking is a major risk factor for atherosclerotic disease and has been associated with DNA methylation (DNAm) changes in blood cells. However, whether smoking influences DNAm in the diseased vascular wall is unknown but may prove crucial in understanding the pathophysiology of atherosclerosis. In this study, we associated current tobacco smoking to epigenome-wide DNAm in atherosclerotic plaques from patients undergoing carotid endarterectomy. METHODS: DNAm at commonly methylated sites (cytosine-guanine nucleotide pairs separated by a phospho-group [CpGs]) was assessed in atherosclerotic plaque samples and peripheral blood samples from 485 carotid endarterectomy patients. We tested the association of current tobacco smoking with DNAm corrected for age and sex. To control for bias and inflation because of cellular heterogeneity, we applied a Bayesian method to estimate an empirical null distribution as implemented by the R package bacon. Replication of the smoking-associated methylated CpGs in atherosclerotic plaques was executed in the second sample of 190 carotid endarterectomy patients, and results were meta-analyzed using a fixed-effects model. RESULTS: Tobacco smoking was significantly associated to differential DNAm in atherosclerotic lesions of 4 CpGs (false discovery rate <0.05) mapped to 2 different genes ( AHRR, ITPK1) and 17 CpGs mapped to 8 genes and RNAs in blood. The strongest associations were found for CpGs mapped to the gene AHRR, a repressor of the aryl hydrocarbon receptor transcription factor involved in xenobiotic detoxification. One of these methylated CpGs were found to be regulated by local genetic variation. CONCLUSIONS: The risk factor tobacco smoking associates with DNAm at multiple loci in carotid atherosclerotic lesions. These observations support further investigation of the relationship between risk factors and epigenetic regulation in atherosclerotic disease.


Asunto(s)
Aterosclerosis/genética , Enfermedades de las Arterias Carótidas/genética , Metilación de ADN , Epigenómica/métodos , Estudio de Asociación del Genoma Completo/métodos , Fumar/efectos adversos , Anciano , Aterosclerosis/etiología , Enfermedades de las Arterias Carótidas/etiología , Islas de CpG/genética , Endarterectomía Carotidea/métodos , Endarterectomía Carotidea/estadística & datos numéricos , Epigénesis Genética , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Placa Aterosclerótica/etiología , Placa Aterosclerótica/genética
13.
Circ Cardiovasc Genet ; 10(3)2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28611032

RESUMEN

BACKGROUND: Microarrays and RNA sequencing are widely used to profile transcriptome remodeling during myocardial ischemia. However, the steady-state RNA analysis lacks in sensitivity to detect all noncoding RNA species and does not provide separation between transcriptional and post-transcriptional regulations. Here, we provide the first comprehensive analysis of nascent RNA profiles of mRNAs, primary micro-RNAs, long noncoding RNAs, and enhancer RNAs in a large animal model of acute infarction. METHODS AND RESULTS: Acute infarction was induced by cardiac catheterization of domestic swine. Nuclei isolated from healthy, border zone, and ischemic regions of the affected heart were subjected to global run-on sequencing. Global run-on sequencing analysis indicated that half of affected genes are regulated at the level of transcriptional pausing. A gradient of induction of inflammatory mediators and repression of peroxisome proliferator-activated receptor signaling and oxidative phosphorylation was detected when moving from healthy toward infarcted area. In addition, we interrogated the transcriptional regulation of primary micro-RNAs and provide evidence that several arrhythmia-related target genes exhibit repression at post-transcriptional level. We identified 450 long noncoding RNAs differently regulated by ischemia, including novel conserved long noncoding RNAs expressed in antisense orientation to myocardial transcription factors GATA-binding protein 4, GATA-binding protein 6, and Krüppel-like factor 6. Finally, characterization of enhancers exhibiting differential expression of enhancer RNAs pointed a central role for Krüppel-like factor, MEF2C, ETS, NFY, ATF, E2F2, and NRF1 transcription factors in determining transcriptional responses to ischemia. CONCLUSIONS: Global run-on sequencing allowed us to follow the gradient of gene expression occurring in the ischemic heart and identify novel noncoding RNAs regulated by oxygen deprivation. These findings highlight potential new targets for diagnosis and treatment of myocardial ischemia.


Asunto(s)
Infarto del Miocardio/patología , Miocardio/metabolismo , ARN no Traducido/metabolismo , Animales , Células Cultivadas , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA6/genética , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Genoma , Factor 6 Similar a Kruppel/genética , MicroARNs/genética , MicroARNs/metabolismo , Infarto del Miocardio/genética , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Factor Nuclear 1 de Respiración/genética , Receptores Activados del Proliferador del Peroxisoma/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , ARN Mensajero/metabolismo , ARN no Traducido/genética , Transducción de Señal/genética , Porcinos
14.
Eur Heart J ; 36(16): 993-1000, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25411193

RESUMEN

AIMS: We conducted a genome-wide analysis to identify differentially methylated genes in atherosclerotic lesions. METHODS: DNA methylation at promoters, exons and introns was identified by massive parallel sequencing. Gene expression was analysed by microarrays, qPCR, immunohistochemistry and western blots. RESULTS: Globally, hypomethylation of chromosomal DNA predominates in atherosclerotic plaques and two-thirds of genes showing over 2.5-fold differential in DNA methylation are up-regulated in comparison to healthy mammary arteries. The imprinted chromatin locus 14q32 was identified for the first time as an extensively hypomethylated area in atherosclerosis with highly induced expression of miR127, -136, -410, -431, -432, -433 and capillary formation-associated gene RTL1. The top 100 list of hypomethylated promoters exhibited over 1000-fold enrichment for miRNAs, many of which mapped to locus 14q32. Unexpectedly, also gene body hypermethylation was found to correlate with stimulated mRNA expression. CONCLUSION: Significant changes in genomic methylation were identified in atherosclerotic lesions. The most prominent gene cluster activated via hypomethylation was detected at imprinted chromosomal locus 14q32 with several clustered miRNAs that were up-regulated. These results suggest that epigenetic changes are involved in atherogenesis and may offer new potential therapeutic targets for vascular diseases.


Asunto(s)
Metilación de ADN/genética , MicroARNs/genética , Placa Aterosclerótica/genética , Anciano , Estudios de Casos y Controles , Cromosomas Humanos Par 14/genética , Regulación hacia Abajo/genética , Exones/genética , Femenino , Ontología de Genes , Sitios Genéticos/genética , Estudio de Asociación del Genoma Completo , Humanos , Masculino , MicroARNs/metabolismo , Proteínas Gestacionales/genética , Proteínas Gestacionales/metabolismo , Regiones Promotoras Genéticas/genética , ARN Mensajero/metabolismo , Regulación hacia Arriba/genética
16.
Curr Opin Lipidol ; 24(5): 438-43, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23872638

RESUMEN

PURPOSE OF REVIEW: This review summarizes recent findings in the epigenetics of vascular cells and discusses the new challenges for therapeutic strategies of cardiovascular diseases. RECENT FINDINGS: There is emerging optimism that epigenetic mechanisms can provide the missing link to connect (epi)genomes with the cause of complex diseases. Environmental factors like intrauterine conditions during fetal development appear to preprogram humans for complex diseases. The purpose of this review is to summarize the newest results about the inheritable epigenetic features of cardiovascular diseases. Also, the recently discovered role of small RNAs in epigenetic gene regulation is discussed. SUMMARY: Epigenetic mechanisms of gene regulation will likely become major determinants in the pathogenesis of complex diseases and may offer new opportunities for the treatment of these diseases.


Asunto(s)
Enfermedades Cardiovasculares , Epigénesis Genética , Animales , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Humanos
17.
Biochim Biophys Acta ; 1790(9): 886-91, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19233248

RESUMEN

The contribution of epigenetic mechanisms to cardiovascular diseases remains poorly understood. Hypomethylation of genomic DNA is present in human atherosclerotic lesions and methylation changes also occur at the promoter level of several genes involved in the pathogenesis of atherosclerosis, such as extracellular superoxide dismutase, estrogen receptor-alpha, endothelial nitric oxide synthase and 15-lipoxygenase. So far, no clear data is available about histone modification marks in atherosclerotic lesions. It remains unclear whether epigenetic changes are causally related to the pathogenetic features, such as clonal proliferation of lesion smooth muscle cells, lipid accumulation and modulation of immune responses in the lesions, or whether they merely represent a consequence of the ongoing pathological process. However, epigenetic changes could at least partly explain poorly understood environmental and dietary effects on atherogenesis and the rapid increases and decreases in the incidence of coronary heart disease observed in various populations. RNAi mechanisms may also contribute to the epigenetic regulation of vascular cells. Therapies directed towards modification of the epigenetic status of vascular cells might provide new tools to control atherosclerosis-related cardiovascular diseases.


Asunto(s)
Aterosclerosis/genética , Epigénesis Genética , Animales , Metilación de ADN , Histonas/metabolismo , Humanos , Hiperhomocisteinemia/genética
18.
J Vasc Res ; 46(3): 240-52, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-18984963

RESUMEN

BACKGROUND: We have shown that the combination of sirolimus and imatinib synergistically inhibits denudation-induced neointimal hyperplasia in rats. We have now dissected the mechanisms behind this synergy and evaluated its long-term efficacy. METHODS: After aortic denudation injury, rats received established submaximal doses of sirolimus (1.0 mg/kg/day), imatinib (10.0 mg/kg/day), the combination of these, or vehicle per os from 3 days before the operation until 14 days after injury. Vessel histology and complete blood counts were monitored until 90 days after injury. Neointimal cell outgrowth, migration and proliferation were evaluated in ex vivo vessel cultures. Quantitative real-time polymerase chain reaction and immunohistochemistry were used for gene and protein expression analysis. RESULTS: The combination therapy caused a synergistic decrease in the number of neointimal nuclei and area throughout the observation period. It also prevented postinjury thrombocytosis and leukocytosis, and almost abolished neointimal cell outgrowth and migration. Furthermore, the combination therapy resulted in upregulation of smooth muscle cell (SMC) markers SM22alpha and cysteine and glycine-rich protein 2, and of the anti-apoptotic BCL2 mRNA. CONCLUSIONS: Combination therapy confers superior long-term vasculoprotection, possibly by inhibition of postoperative thrombocytosis and leukocytosis, inhibition of neointimal cell migration to the injury site and maintenance of cell integrity by inhibition of apoptosis and SMC dedifferentiation.


Asunto(s)
Piperazinas/administración & dosificación , Pirimidinas/administración & dosificación , Sirolimus/administración & dosificación , Túnica Íntima/efectos de los fármacos , Animales , Aorta/efectos de los fármacos , Aorta/patología , Benzamidas , Recuento de Células Sanguíneas , Sinergismo Farmacológico , Hiperplasia , Mesilato de Imatinib , Proteínas con Dominio LIM , Masculino , Proteínas de Microfilamentos/genética , Proteínas Musculares/genética , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Proteínas Nucleares/genética , Piperazinas/toxicidad , Proteínas Proto-Oncogénicas c-bcl-2/genética , Pirimidinas/toxicidad , Ratas , Ratas Wistar , Sirolimus/toxicidad , Túnica Íntima/patología , Factor de von Willebrand/genética
19.
Mol Cell Endocrinol ; 279(1-2): 34-8, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17945410

RESUMEN

We have shown that somatostatin agonist peptide CH275, selective to somatostatin receptor (sst) subtypes 1,4, was more effective in preventing intimal hyperplasia than the sst2,3,5-selective octreotide, raising the question what are the separate roles of the sst1- and 4-subtypes. Here, we dissect this observation further with highly subtype-selective peptidomimetics and demonstrate that, after rat carotid denudation, both the sst1- and 4-selective analogs (300 microg/kg/day, s.c.) increased lumen size, while only the sst4-selective analog significantly reduced intimal nuclei number, intimal area, and intima/media ratio. The 2,3,5-selective compounds had no effect on these parameters. The observed in vivo effects were further investigated ex vivo with explant outgrowth from pieces of vascular wall. The sst4-selective analog was more effective than the sst1-selective one in inhibiting the percent of outgrowth and the migration of cells from the explants while neither compound affected proliferation. Thus, selective targeting to sst4 should be considered when developing orally active vasculoprotective therapies.


Asunto(s)
Péptidos , Receptores de Somatostatina/clasificación , Somatostatina/farmacología , Túnica Íntima/efectos de los fármacos , Animales , Aorta/efectos de los fármacos , Aorta/fisiología , Masculino , Proteínas de la Membrana/efectos de los fármacos , Técnicas de Cultivo de Órganos , Péptidos/genética , Péptidos/farmacología , Ratas , Ratas Wistar , Receptores de Somatostatina/efectos de los fármacos , Somatostatina/agonistas , Somatostatina/genética , Túnica Íntima/lesiones , Túnica Íntima/patología
20.
Transplantation ; 83(6): 671-6, 2007 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-17414693

RESUMEN

BACKGROUND: Progressive injury that is refractory to conventional immunosuppression remains the major hurdle to indefinite survival of transplanted organs. Several clinical risk factors of chronic renal allograft rejection have been identified; although some (e.g., acute rejection) are direct manifestations of immunological injury, others (e.g., donor age) have been more difficult to conceptually link with graft dysfunction. METHODS: We conducted formal multivariate statistical analyses to reveal associations between established clinical risk factors and allograft histopathology. In a multicenter protocol biopsy-controlled study, 17 clinical risk factors were studied in relation to either the composite Chronic Allograft Damage Index (CADI) score or, to each of eight individual histological indices, using multiple linear regression with forward selection. RESULTS: Nine clinical risk factors were not significantly associated with any histopathological index. Four (donor age, acute rejection, recipient age, and cold ischemia time) were associated both with the total CADI score and, to varying extents, with the individual histopathological indices. In our analysis, clinical risk factors accounted for, at best, only about 60% of the interindividual variation in histopathological score. CONCLUSIONS: Our study reveals a missing link between specific clinical risk factors and early histopathological findings that are known to presage accelerated failure of clinically healthy grafts. Given the complex relationship between clinical risk factors, early histopathological changes, and graft outcome, we conclude that composite, quantitative histological indices are best suited to for evaluation of the histological status of the transplant.


Asunto(s)
Rechazo de Injerto/patología , Trasplante de Riñón/patología , Patología/métodos , Índice de Severidad de la Enfermedad , Adulto , Biopsia , Rechazo de Injerto/clasificación , Humanos , Riñón/patología , Trasplante de Riñón/clasificación , Modelos Lineales , Persona de Mediana Edad , Análisis Multivariante , Valor Predictivo de las Pruebas , Pronóstico , Estudios Prospectivos , Factores de Riesgo , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...