Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 41(1): e106459, 2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34806773

RESUMEN

In mammals, histone 3 lysine 4 methylation (H3K4me) is mediated by six different lysine methyltransferases. Among these enzymes, SETD1B (SET domain containing 1b) has been linked to syndromic intellectual disability in human subjects, but its role in the mammalian postnatal brain has not been studied yet. Here, we employ mice deficient for Setd1b in excitatory neurons of the postnatal forebrain, and combine neuron-specific ChIP-seq and RNA-seq approaches to elucidate its role in neuronal gene expression. We observe that Setd1b controls the expression of a set of genes with a broad H3K4me3 peak at their promoters, enriched for neuron-specific genes linked to learning and memory function. Comparative analyses in mice with conditional deletion of Kmt2a and Kmt2b histone methyltransferases show that SETD1B plays a more pronounced and potent role in regulating such genes. Moreover, postnatal loss of Setd1b leads to severe learning impairment, suggesting that SETD1B-dependent regulation of H3K4me levels in postnatal neurons is critical for cognitive function.


Asunto(s)
Regulación de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/metabolismo , Aprendizaje/fisiología , Neuronas/metabolismo , Animales , Animales Recién Nacidos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Núcleo Celular/metabolismo , Epigénesis Genética , Hipocampo/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Integrasas/metabolismo , Memoria/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Sitio de Iniciación de la Transcripción , Transcriptoma/genética
2.
Circ Res ; 133(2): e19-e46, 2023 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-37313752

RESUMEN

BACKGROUND: Systemic defects in intestinal iron absorption, circulation, and retention cause iron deficiency in 50% of patients with heart failure. Defective subcellular iron uptake mechanisms that are independent of systemic absorption are incompletely understood. The main intracellular route for iron uptake in cardiomyocytes is clathrin-mediated endocytosis. METHODS: We investigated subcellular iron uptake mechanisms in patient-derived and CRISPR/Cas-edited induced pluripotent stem cell-derived cardiomyocytes as well as patient-derived heart tissue. We used an integrated platform of DIA-MA (mass spectrometry data-independent acquisition)-based proteomics and signaling pathway interrogation. We employed a genetic induced pluripotent stem cell model of 2 inherited mutations (TnT [troponin T]-R141W and TPM1 [tropomyosin 1]-L185F) that lead to dilated cardiomyopathy (DCM), a frequent cause of heart failure, to study the underlying molecular dysfunctions of DCM mutations. RESULTS: We identified a druggable molecular pathomechanism of impaired subcellular iron deficiency that is independent of systemic iron metabolism. Clathrin-mediated endocytosis defects as well as impaired endosome distribution and cargo transfer were identified as a basis for subcellular iron deficiency in DCM-induced pluripotent stem cell-derived cardiomyocytes. The clathrin-mediated endocytosis defects were also confirmed in the hearts of patients with DCM with end-stage heart failure. Correction of the TPM1-L185F mutation in DCM patient-derived induced pluripotent stem cells, treatment with a peptide, Rho activator II, or iron supplementation rescued the molecular disease pathway and recovered contractility. Phenocopying the effects of the TPM1-L185F mutation into WT induced pluripotent stem cell-derived cardiomyocytes could be ameliorated by iron supplementation. CONCLUSIONS: Our findings suggest that impaired endocytosis and cargo transport resulting in subcellular iron deficiency could be a relevant pathomechanism for patients with DCM carrying inherited mutations. Insight into this molecular mechanism may contribute to the development of treatment strategies and risk management in heart failure.


Asunto(s)
Cardiomiopatía Dilatada , Insuficiencia Cardíaca , Células Madre Pluripotentes Inducidas , Deficiencias de Hierro , Humanos , Miocitos Cardíacos/metabolismo , Mutación , Cardiomiopatía Dilatada/genética , Células Madre Pluripotentes Inducidas/metabolismo , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Hierro/metabolismo , Clatrina/genética , Clatrina/metabolismo , Clatrina/farmacología
3.
FASEB J ; 33(2): 2278-2289, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30260706

RESUMEN

Endothelial cells can acquire a mesenchymal phenotype upon irritation [endothelial-to-mesenchymal transition (EndMT)]. Macrophages accumulate in the atherosclerotic plaque. This study addressed whether macrophages modulate EndMT and delineated a reciprocal effect of EndMT on macrophage functions in atherosclerosis. In atherosclerotic murine and human aortas, endothelial cells with mesenchymal markers were elevated by confocal microscopy and flow cytometric analysis. Increased EndMT master transcription factor Snai1 expression and extracellular matrix are consistent with enhanced EndMT in this condition. Hypoxia was detected in individual aortic EndMT cells in vivo and rapidly induced a similar EndMT phenotype in vitro. As a novel inducer of EndMT, macrophages, which are abundant in the atherosclerotic lesions, enhance mesothelial marker expression during coculture in vitro. In the reverse relationship, EndMT altered endothelial colony-stimulating factor expression. Functionally, EndMT cell-conditioned media attenuated macrophage proliferation, antigen-presenting cell marker expression, and TNF-α production in response to oxidized LDL but increased oxidized LDL uptake and scavenger receptor expression. These experiments demonstrate that macrophages promote partial EndMT. In turn, EndMT cells modulate macrophage phenotype and lipid uptake. Our data suggest that EndMT shapes macrophage and endothelial cell phenotypes, thus affecting internal atherosclerotic plaque in addition to surface structure.-Helmke, A., Casper, J., Nordlohne, J., David, S., Haller, H., Zeisberg, E. M., von Vietinghoff, S. Endothelial-to-mesenchymal transition shapes the atherosclerotic plaque and modulates macrophage function.


Asunto(s)
Endotelio/patología , Macrófagos/citología , Mesodermo/patología , Placa Aterosclerótica/patología , Animales , Antígenos/inmunología , Aorta/metabolismo , Biomarcadores/metabolismo , Antígenos CD36/metabolismo , Hipoxia de la Célula , Proliferación Celular , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Endotelio/metabolismo , Matriz Extracelular/patología , Femenino , Humanos , Lipoproteínas LDL/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/inmunología , Masculino , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis , Factores de Transcripción de la Familia Snail/metabolismo
4.
J Cardiovasc Magn Reson ; 22(1): 46, 2020 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-32564773

RESUMEN

BACKGROUND: Myocardial fibrosis is a major determinant of outcome in aortic stenosis (AS). Novel fast real-time (RT) cardiovascular magnetic resonance (CMR) mapping techniques allow comprehensive quantification of fibrosis but have not yet been compared against standard techniques and histology. METHODS: Patients with severe AS underwent CMR before (n = 110) and left ventricular (LV) endomyocardial biopsy (n = 46) at transcatheter aortic valve replacement (TAVR). Midventricular short axis (SAX) native, post-contrast T1 and extracellular volume fraction (ECV) maps were generated using commercially available modified Look-Locker Inversion recovery (MOLLI) (native: 5(3)3, post-contrast: 4(1)3(1)2) and RT single-shot inversion recovery Fast Low-Angle Shot (FLASH) with radial undersampling. Focal late gadolinium enhancement was excluded from T1 and ECV regions of interest. ECV and LV mass were used to calculate LV matrix volumes. Variability and agreements were assessed between RT, MOLLI and histology using intraclass correlation coefficients, coefficients of variation and Bland Altman analyses. RESULTS: RT and MOLLI derived ECV were similar for midventricular SAX slice coverage (26.2 vs. 26.5, p = 0.073) and septal region of interest (26.2 vs. 26.5, p = 0.216). MOLLI native T1 time was in median 20 ms longer compared to RT (p < 0.001). Agreement between RT and MOLLI was best for ECV (ICC > 0.91), excellent for post-contrast T1 times (ICC > 0.81) and good for native T1 times (ICC > 0.62). Diffuse collagen volume fraction by biopsies was in median 7.8%. ECV (RT r = 0.345, p = 0.039; MOLLI r = 0.40, p = 0.010) and LV matrix volumes (RT r = 0.45, p = 0.005; MOLLI r = 0.43, p = 0.007) were the only parameters associated with histology. CONCLUSIONS: RT mapping offers fast and sufficient ECV and LV matrix volume calculation in AS patients. ECV and LV matrix volume represent robust and universally comparable parameters with associations to histologically assessed fibrosis and may emerge as potential targets for clinical decision making.


Asunto(s)
Estenosis de la Válvula Aórtica/diagnóstico por imagen , Válvula Aórtica/diagnóstico por imagen , Imagen por Resonancia Cinemagnética , Miocardio/patología , Función Ventricular Izquierda , Anciano , Anciano de 80 o más Años , Válvula Aórtica/patología , Válvula Aórtica/fisiopatología , Válvula Aórtica/cirugía , Estenosis de la Válvula Aórtica/patología , Estenosis de la Válvula Aórtica/fisiopatología , Estenosis de la Válvula Aórtica/cirugía , Biopsia , Femenino , Fibrosis , Humanos , Masculino , Variaciones Dependientes del Observador , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Reemplazo de la Válvula Aórtica Transcatéter , Remodelación Ventricular
5.
Int J Mol Sci ; 21(9)2020 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-32344896

RESUMEN

The field of genome editing started with the discovery of meganucleases (e.g., the LAGLIDADG family of homing endonucleases) in yeast. After the discovery of transcription activator-like effector nucleases and zinc finger nucleases, the recently discovered clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR associated proteins (Cas) system has opened a new window of applications in the field of gene editing. Here, we review different Cas proteins and their corresponding features including advantages and disadvantages, and we provide an overview of the different endonuclease-deficient Cas protein (dCas) derivatives. These dCas derivatives consist of an endonuclease-deficient Cas9 which can be fused to different effector domains to perform distinct in vitro applications such as tracking, transcriptional activation and repression, as well as base editing. Finally, we review the in vivo applications of these dCas derivatives and discuss their potential to perform gene activation and repression in vivo, as well as their potential future use in human therapy.


Asunto(s)
Proteínas Bacterianas/metabolismo , Sistemas CRISPR-Cas , Endodesoxirribonucleasas/metabolismo , Epigenómica/métodos , Edición Génica/métodos , Proteína 9 Asociada a CRISPR/metabolismo , Cromatina/ultraestructura , ADN/metabolismo , Endonucleasas/metabolismo , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Imagen Óptica , ARN Guía de Kinetoplastida/genética , Proteínas Recombinantes de Fusión/análisis , Especificidad por Sustrato , Telómero/ultraestructura , Nucleasas de los Efectores Tipo Activadores de la Transcripción/metabolismo , Efectores Tipo Activadores de la Transcripción/metabolismo , Transcripción Genética , Dedos de Zinc
6.
J Mol Cell Cardiol ; 136: 113-124, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31520610

RESUMEN

BACKGROUND: Cardiovascular fibrosis is a major contributor to cardiovascular disease, the primary cause of death in patients with chronic kidney disease (CKD). We previously reported expression of endogenous Klotho in human arteries, and that CKD is a state of Klotho deficiency, resulting in vascular calcification, but myocardial expression of Klotho is poorly understood. This study aimed to further clarify endogenous Klotho's functional roles in cardiac fibrosis in patients with underlying CKD. METHODS AND RESULTS: Human atrial appendage specimens were collected during cardiac surgery from individuals with or without CKD. Cardiac fibrosis was quantified using trichrome staining. For endogenous Klotho functional studies, primary human cardiomyocytes (HCMs) were treated with uremic serum from CKD patients or recombinant human TGF-ß1. The effects of endogenous Klotho in HCMs were studied using Klotho-siRNA and Klotho-plasmid transfection. Both gene and protein expression of endogenous Klotho are found in human heart, but decreased Klotho expression is clearly associated with the degree of cardiac fibrosis in CKD patients. Moreover, we show that endogenous Klotho is expressed by HCMs and cardiac fibroblasts (HCFs) but that HCM expression is suppressed by uremic serum or TGF-ß1. Klotho knockdown or overexpression aggravates or mitigates TGF-ß1-induced fibrosis and canonical Wnt signaling in HCMs, respectively. Furthermore, co-culture of HCMs with HCFs increases TGF-ß1-induced fibrogenic proteins in HCFs, but overexpression of endogenous Klotho in HCMs mitigates this effect, suggesting functional crosstalk between HCMs and HCFs. CONCLUSIONS: Our data from analysis of human hearts as well as functional in vitro studies strongly suggests that the loss of cardiac endogenous Klotho in CKD patients, specifically in cardiomyocytes, facilitates intensified TGF-ß1 signaling which enables more vigorous cardiac fibrosis through upregulated Wnt signaling. Upregulation of endogenous Klotho inhibits pathogenic Wnt/ß-catenin signaling and may offer a novel strategy for prevention and treatment of cardiac fibrosis in CKD patients.


Asunto(s)
Glucuronidasa/metabolismo , Miocardio/patología , Insuficiencia Renal Crónica/complicaciones , Factor de Crecimiento Transformador beta1/metabolismo , Vía de Señalización Wnt , Adulto , Anciano , Anciano de 80 o más Años , Células Cultivadas , Femenino , Fibrosis , Glucuronidasa/genética , Humanos , Proteínas Klotho , Masculino , Persona de Mediana Edad , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Insuficiencia Renal Crónica/metabolismo
7.
Arterioscler Thromb Vasc Biol ; 38(9): 1986-1996, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30354260

RESUMEN

Endothelial-to-mesenchymal transition (EndMT) is a process in which endothelial cells lose their properties and transform into fibroblast-like cells. This transition process contributes to cardiac fibrosis, a common feature of patients with chronic heart failure. To date, no specific therapies to halt or reverse cardiac fibrosis are available, so knowledge of the underlying mechanisms of cardiac fibrosis is urgently needed. In addition, EndMT contributes to other cardiovascular pathologies such as atherosclerosis and pulmonary hypertension, but also to cancer and organ fibrosis. Remarkably, the molecular mechanisms driving EndMT are largely unknown. Epigenetics play an important role in regulating gene transcription and translation and have been implicated in the EndMT process. Therefore, epigenetics might be the missing link in unraveling the underlying mechanisms of EndMT. Here, we review the involvement of epigenetic regulators during EndMT in the context of cardiac fibrosis. The role of DNA methylation, histone modifications (acetylation and methylation), and noncoding RNAs (microRNAs, long noncoding RNAs, and circular RNAs) in the facilitation and inhibition of EndMT are discussed, and potential therapeutic epigenetic targets will be highlighted.


Asunto(s)
Epigénesis Genética , Transición Epitelial-Mesenquimal , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Acetilación , Enfermedad Crónica , Metilación de ADN , Fibrosis , Insuficiencia Cardíaca/patología , Histonas/metabolismo , Humanos , Metilación , ARN no Traducido/fisiología , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
8.
Kidney Int ; 91(1): 157-176, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27692563

RESUMEN

Acute kidney injury (AKI) and progressive chronic kidney disease (CKD) are intrinsically tied syndromes. In this regard, the acutely injured kidney often does not achieve its full regenerative capacity and AKI directly transitions into progressive CKD associated with tubulointerstitial fibrosis. Underlying mechanisms of such AKI-to-CKD progression are still incompletely understood and specific therapeutic interventions are still elusive. Because epigenetic modifications play a role in maintaining tissue fibrosis, we used a murine model of ischemia-reperfusion injury to determine whether aberrant promoter methylation of RASAL1 contributes causally to the switch between physiological regeneration and tubulointerstitial fibrogenesis, a hallmark of AKI-to-CKD progression. It is known that the antihypertensive drug hydralazine has demethylating activity, and that its optimum demethylating activity occurs at concentrations below blood pressure-lowering doses. Administration of low-dose hydralazine effectively induced expression of hydroxylase TET3, which catalyzed RASAL1 hydroxymethylation and subsequent RASAL1 promoter demethylation. Hydralazine-induced CpG promoter demethylation subsequently attenuated renal fibrosis and preserved excretory renal function independent of its blood pressure-lowering effects. In comparison, RASAL1 demethylation and inhibition of tubulointerstitial fibrosis was not detected upon administration of the angiotensin-converting enzyme inhibitor Ramipril in this model. Thus, RASAL1 promoter methylation and subsequent transcriptional RASAL1 suppression plays a causal role in AKI-to-CKD progression.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Proteínas de Unión al ADN/metabolismo , Proteínas Activadoras de GTPasa/genética , Hidralazina/uso terapéutico , Riñón/patología , Proteínas Proto-Oncogénicas/metabolismo , Insuficiencia Renal Crónica/prevención & control , Vasodilatadores/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Islas de CpG , Metilación de ADN , Dioxigenasas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Epigénesis Genética , Fibroblastos/metabolismo , Fibrosis , Humanos , Hidralazina/administración & dosificación , Riñón/citología , Riñón/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Cultivo Primario de Células , Regiones Promotoras Genéticas , Ramipril/farmacología , Eliminación Renal/efectos de los fármacos , Daño por Reperfusión/complicaciones , Vasodilatadores/administración & dosificación
9.
Circ Res ; 116(5): 857-66, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25587097

RESUMEN

RATIONALE: Endocardial fibroelastosis (EFE) is a unique form of fibrosis, which forms a de novo subendocardial tissue layer encapsulating the myocardium and stunting its growth, and which is typically associated with congenital heart diseases of heterogeneous origin, such as hypoplastic left heart syndrome. Relevance of EFE was only recently highlighted through the establishment of staged biventricular repair surgery in infant patients with hypoplastic left heart syndrome, where surgical removal of EFE tissue has resulted in improvement in the restrictive physiology leading to the growth of the left ventricle in parallel with somatic growth. However, pathomechanisms underlying EFE formation are still scarce, and specific therapeutic targets are not yet known. OBJECTIVE: Here, we aimed to investigate the cellular origins of EFE tissue and to gain insights into the underlying molecular mechanisms to ultimately develop novel therapeutic strategies. METHODS AND RESULTS: By utilizing a novel EFE model of heterotopic transplantation of hearts from newborn reporter mice and by analyzing human EFE tissue, we demonstrate for the first time that fibrogenic cells within EFE tissue originate from endocardial endothelial cells via aberrant endothelial to mesenchymal transition. We further demonstrate that such aberrant endothelial to mesenchymal transition involving endocardial endothelial cells is caused by dysregulated transforming growth factor beta/bone morphogenetic proteins signaling and that this imbalance is at least in part caused by aberrant promoter methylation and subsequent transcriptional suppression of bone morphogenetic proteins 5 and 7. Finally, we provide evidence that supplementation of exogenous recombinant bone morphogenetic proteins 7 effectively ameliorates endothelial to mesenchymal transition and experimental EFE in rats. CONCLUSIONS: In summary, our data point to aberrant endothelial to mesenchymal transition as a common denominator of infant EFE development in heterogeneous, congenital heart diseases, and to bone morphogenetic proteins 7 as an effective treatment for EFE and its restriction of heart growth.


Asunto(s)
Transdiferenciación Celular/fisiología , Fibroelastosis Endocárdica/patología , Endocardio/patología , Epitelio/patología , Mesodermo/patología , Animales , Animales Recién Nacidos , Antígenos CD/genética , Biomarcadores , Proteína Morfogenética Ósea 7/genética , Proteína Morfogenética Ósea 7/fisiología , Proteína Morfogenética Ósea 7/uso terapéutico , Cadherinas/genética , Transdiferenciación Celular/genética , Células Cultivadas , Metilación de ADN , Fibroelastosis Endocárdica/tratamiento farmacológico , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Trasplante de Corazón , Humanos , Síndrome del Corazón Izquierdo Hipoplásico/patología , Síndrome del Corazón Izquierdo Hipoplásico/cirugía , Lactante , Recién Nacido , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Ratas , Ratas Endogámicas Lew , Proteínas Recombinantes/uso terapéutico , Transducción de Señal/fisiología , Proteínas Smad/genética , Proteínas Smad/fisiología , Factor de Crecimiento Transformador beta/fisiología , Trasplante Heterotópico
10.
Differentiation ; 92(4): 225-236, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27266810

RESUMEN

Endothelial cells derived from human induced pluripotent stem cells (hiPSC- EC) are of significant value for research on human vascular development, in vitro disease models and drug screening. Here we report an alternative, highly efficient and cost-effective simple three step method (mesoderm induction, endothelial cell differentiation and endothelial cell expansion) to differentiate hiPSC directly into endothelial cells. We demonstrate that efficiency of described method to derive CD31+ and VE-Cadherin+ double positive cells is higher than 80% in 12 days. Most notably we established that hiPSC-EC differentiation efficacy depends on optimization of both mesoderm differentiation and endothelial cell differentiation steps.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/genética , Células Endoteliales/citología , Células Madre Pluripotentes Inducidas/citología , Antígenos CD/metabolismo , Cadherinas/metabolismo , Linaje de la Célula/genética , Proliferación Celular , Humanos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Células Madre Pluripotentes
11.
J Biol Chem ; 290(27): 16653-64, 2015 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-25971970

RESUMEN

Endothelial to mesenchymal transition (EndMT) was originally described in heart development where the endocardial endothelial cells that line the atrioventricular canal undergo an EndMT to form the endocardial mesenchymal cushion that later gives rise to the septum and mitral and tricuspid valves. In the postnatal heart specifically, endothelial cells that originate from the endocardium maintain increased susceptibility to undergo EndMT as remnants from their embryonic origin. Such EndMT involving adult coronary endothelial cells contributes to microvascular rarefaction and subsequent chronification of hypoxia in the injured heart, ultimately leading to cardiac fibrosis. Although in most endothelial beds hypoxia induces tip cell formation and sprouting angiogenesis, here we demonstrate that hypoxia is a stimulus for human coronary endothelial cells to undergo phenotypic changes reminiscent of EndMT via a mechanism involving hypoxia-inducible factor 1α-induced activation of the EndMT master regulatory transcription factor SNAIL. Our study adds further evidence for the unique susceptibility of endocardium-derived endothelial cells to undergo EndMT and provides novel insights into how hypoxia contributes to progression of cardiac fibrosis. Additional studies may be required to discriminate between distinct sprouting angiogenesis and EndMT responses of different endothelial cells populations.


Asunto(s)
Endocardio/metabolismo , Células Endoteliales/metabolismo , Transición Epitelial-Mesenquimal , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Endocardio/citología , Células Endoteliales/citología , Humanos , Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética
12.
Biochem Biophys Res Commun ; 472(3): 459-64, 2016 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-26774342

RESUMEN

Phosphate is an essential constituent of critical cellular functions including energy metabolism, nucleic acid synthesis and phosphorylation-dependent cell signaling. Increased plasma phosphate levels are an independent risk factor for lowered life-expectancy as well as for heart and kidney failure. Nevertheless, direct cellular effects of elevated phosphate concentrations within the microenvironment are poorly understood and have been largely neglected in favor of phosphor-regulatory hormones. Because interstitial fibrosis is the common determinant of chronic progressive kidney disease, and because fibroblasts are major mediators of fibrogenesis, we here explored the effect of high extracellular phosphate levels on renal fibroblasts. We demonstrate that high inorganic phosphate directly induces fibrotic fibroblast activation associated with increased proliferative activity, increased expression of α-smooth muscle actin and increased synthesis of type I collagen. We further demonstrate that such fibroblast activation is dependent on phosphate influx, aberrant phosphorylation of DNA methyltransferase DNMT1 and aberrant CpG island promoter methylation. In summary, our studies demonstrate that elevated phosphate concentrations induce pro-fibrotic fibroblast activation independent of phospho-regulatory hormones.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Fosfatos/toxicidad , Línea Celular , Proliferación Celular/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasa 1 , Relación Dosis-Respuesta a Droga , Fibroblastos/efectos de los fármacos , Fibrosis , Humanos , Fosfatos/administración & dosificación , Fosforilación/efectos de los fármacos
13.
Kidney Int ; 88(3): 429-31, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26323066

RESUMEN

Among gliptins, linagliptin is unique, because decreased glomerular filtration rate does not require dose reduction. Linagliptin was originally developed to lower blood glucose by inhibiting dipeptidyl peptidase-4 (DPP-4). However, DPP-4 has numerous additional substrates, and thus gliptins possess a vast range of additional off-target effects. Shi et al. report that linagliptin directly targets interaction of DPP-4 with integrin ß1, preventing endothelial-mesenchymal transition and ultimately renal fibrosis, providing additional rationale for use of linagliptin in diabetic nephropathy.


Asunto(s)
Diabetes Mellitus Experimental/enzimología , Nefropatías Diabéticas/enzimología , Dipeptidil Peptidasa 4/metabolismo , Células Endoteliales/enzimología , Transición Epitelial-Mesenquimal , Integrina beta1/metabolismo , Riñón/enzimología , Animales , Masculino
14.
Am J Pathol ; 184(10): 2687-98, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25111226

RESUMEN

Thrombospondin-1 (TSP1) is a multifunctional matricellular protein known to promote progression of chronic kidney disease. To gain insight into the underlying mechanisms through which TSP1 accelerates chronic kidney disease, we compared disease progression in Col4a3 knockout (KO) mice, which develop spontaneous kidney failure, with that of Col4a3;Tsp1 double-knockout (DKO) mice. Decline of excretory renal function was significantly delayed in the absence of TSP1. Although Col4a3;Tsp1 DKO mice did progress toward end-stage renal failure, their kidneys exhibited distinct histopathological lesions, compared with creatinine level-matched Col4a3 KO mice. Although kidneys of both Col4a3 KO and Col4a3;Tsp1 DKO mice exhibited a widened tubulointerstitium, predominant lesions in Col4a3 KO kidneys were collagen deposition and fibroblast accumulation, whereas in Col4a3;Tsp1 DKO kidney inflammation was predominant, with less collagen deposition. Altered disease progression correlated with impaired activation of transforming growth factor-ß1 (TGF-ß1) in vivo and in vitro in the absence of TSP1. In summary, our findings suggest that TSP1 contributes to progression of chronic kidney disease by catalyzing activation of latent TGF-ß1, resulting in promotion of a fibroproliferative response over an inflammatory response. Furthermore, the findings suggest that fibroproliferative and inflammatory lesions are independent entities, both of which contribute to decline of renal function.


Asunto(s)
Fibrosis/patología , Fallo Renal Crónico/patología , Insuficiencia Renal/patología , Trombospondina 1/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Autoantígenos/metabolismo , Colágeno Tipo IV/metabolismo , Creatinina/sangre , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fibrosis/metabolismo , Riñón/patología , Fallo Renal Crónico/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nefritis , Insuficiencia Renal/metabolismo
15.
J Am Soc Nephrol ; 25(5): 905-12, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24480825

RESUMEN

Methylation of CpG island promoters is an epigenetic event that can effectively silence transcription over multiple cell generations. Hypermethylation of the Rasal1 promoter contributes to activation of fibroblasts and progression of kidney fibrosis. Here, we explored whether such causative hypermethylation could be reversed through endogenous mechanisms and whether such reversal of hypermethylation is a constituent of the antifibrotic activity of bone morphogenic protein 7 (BMP7). We show that successful inhibition of experimental kidney fibrosis through administration of BMP7 associates with normalization of Rasal1 promoter hypermethylation. Furthermore, this reversal of pathologic hypermethylation was achieved specifically through Tet3-mediated hydroxymethylation. Collectively, our findings reveal a new mechanism that may be exploited to facilitate therapeutic DNA demethylation to reverse kidney fibrosis.


Asunto(s)
Proteína Morfogenética Ósea 7/uso terapéutico , Metilación de ADN/efectos de los fármacos , Proteínas de Unión al ADN/fisiología , Proteínas Activadoras de GTPasa/genética , Silenciador del Gen , Nefroesclerosis/etiología , Nefroesclerosis/prevención & control , Proteínas Proto-Oncogénicas/fisiología , Animales , Biomarcadores/metabolismo , Proteína Morfogenética Ósea 7/metabolismo , Proteína Morfogenética Ósea 7/farmacología , Células Cultivadas , Metilación de ADN/genética , Proteínas de Unión al ADN/genética , Dioxigenasas , Epigénesis Genética , Ratones , Nefroesclerosis/genética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Obstrucción Ureteral/etiología , Obstrucción Ureteral/genética , Obstrucción Ureteral/prevención & control
16.
J Pathol ; 229(2): 264-73, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23097091

RESUMEN

The aberrant methylation of CpG island promoters of selected genes is the prominent epigenetic mechanism by which gene transcription can be effectively silenced. Aberrant hypermethylation of a few selected genes plays an important role in facilitating fibrotic fibroblast activation and in driving fibrogenesis. Here we review mechanisms of DNA methylation and demethylation and their implications for fibroblast activation and tissue fibrosis.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Fibroblastos/metabolismo , Enfermedades Renales/genética , Riñón/metabolismo , Regiones Promotoras Genéticas , Animales , Islas de CpG , Fibroblastos/patología , Fibrosis , Regulación de la Expresión Génica , Silenciador del Gen , Humanos , Riñón/patología , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Proteínas Activadoras de ras GTPasa/genética
17.
Nat Med ; 13(8): 952-61, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17660828

RESUMEN

Cardiac fibrosis, associated with a decreased extent of microvasculature and with disruption of normal myocardial structures, results from excessive deposition of extracellular matrix, which is mediated by the recruitment of fibroblasts. The source of these fibroblasts is unclear and specific anti-fibrotic therapies are not currently available. Here we show that cardiac fibrosis is associated with the emergence of fibroblasts originating from endothelial cells, suggesting an endothelial-mesenchymal transition (EndMT) similar to events that occur during formation of the atrioventricular cushion in the embryonic heart. Transforming growth factor-beta1 (TGF-beta1) induced endothelial cells to undergo EndMT, whereas bone morphogenic protein 7 (BMP-7) preserved the endothelial phenotype. The systemic administration of recombinant human BMP-7 (rhBMP-7) significantly inhibited EndMT and the progression of cardiac fibrosis in mouse models of pressure overload and chronic allograft rejection. Our findings show that EndMT contributes to the progression of cardiac fibrosis and that rhBMP-7 can be used to inhibit EndMT and to intervene in the progression of chronic heart disease associated with fibrosis.


Asunto(s)
Diferenciación Celular , Fibrosis Endomiocárdica/patología , Células Endoteliales/patología , Mesodermo/patología , Animales , Células de la Médula Ósea/patología , Proteína Morfogenética Ósea 7 , Proteínas Morfogenéticas Óseas/farmacología , Proteínas Morfogenéticas Óseas/uso terapéutico , Línea Celular , Células Cultivadas , Enfermedad Crónica , Fibrosis Endomiocárdica/tratamiento farmacológico , Células Endoteliales/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/patología , Mesodermo/efectos de los fármacos , Ratones , Ratones Transgénicos , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta/uso terapéutico , Factor de Crecimiento Transformador beta1/farmacología
18.
EuroIntervention ; 20(12): e760-e769, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38887885

RESUMEN

BACKGROUND: Cardiac fibrosis plays a major pathophysiological role in any form of chronic heart disease, and high levels are associated with poor outcome. Diffuse and focal cardiac fibrosis are different subtypes, which have different pathomechanisms and prognostic implications. The total fibrosis burden in endomyocardial biopsy tissue was recently proved to play an independent prognostic role in aortic stenosis patients after transcatheter aortic valve implantation (TAVI). AIMS: Here, for the first time, we aim to assess the specific impact of different fibrosis subtypes on sudden cardiac death (SCD) as a primary reason for cardiovascular mortality after TAVI. METHODS: The fibrosis pattern was assessed histologically in the left ventricular biopsies obtained during TAVI interventions in 161 patients, who received a structured follow-up thereafter. RESULTS: Receiver operating characteristic analyses, performed 6, 12, 24 and 48 months after TAVI, showed diffuse, but not focal, fibrosis as a significant predictor for SCD at all timepoints, with the highest area under the curve at the first time point and a decrease in its SCD predictivity over time. In both multivariate Cox proportional hazards and Fine-Gray competing risk models, including both fibrosis subtypes, as well as age, sex and ejection fraction, high diffuse fibrosis remained statistically significant. Accordingly, it represents an independent SCD predictor, most importantly for the occurrence of early events. CONCLUSIONS: The burden of diffuse cardiac fibrosis plays an important and independent prognostic role regarding SCD early after TAVI. Therefore, the histological evaluation of fibrosis topography has value as a prognostic tool for TAVI patients and may help to tailor individualised approaches to optimise their postinterventional management.


Asunto(s)
Estenosis de la Válvula Aórtica , Muerte Súbita Cardíaca , Fibrosis , Reemplazo de la Válvula Aórtica Transcatéter , Humanos , Reemplazo de la Válvula Aórtica Transcatéter/efectos adversos , Masculino , Femenino , Anciano , Muerte Súbita Cardíaca/etiología , Estenosis de la Válvula Aórtica/cirugía , Estenosis de la Válvula Aórtica/complicaciones , Estenosis de la Válvula Aórtica/mortalidad , Anciano de 80 o más Años , Factores de Riesgo , Miocardio/patología , Pronóstico
19.
Diabetol Metab Syndr ; 15(1): 11, 2023 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-36698180

RESUMEN

BACKGROUND: The prevalence of diabetes mellitus has risen considerably and currently affects more than 422 million people worldwide. Cardiovascular diseases including myocardial infarction and heart failure represent the major cause of death in type 2 diabetes (T2D). Diabetes patients exhibit accelerated aortic stiffening which is an independent predictor of cardiovascular disease and mortality. We recently showed that aortic stiffness precedes hypertension in a mouse model of diabetes (db/db mice), making aortic stiffness an early contributor to cardiovascular disease development. Elucidating how aortic stiffening develops is a pressing need in order to halt the pathophysiological process at an early time point. METHODS: To assess EndMT occurrence, we performed co-immunofluorescence staining of an endothelial marker (CD31) with mesenchymal markers (α-SMA/S100A4) in aortic sections from db/db mice. Moreover, we performed qRT-PCR to analyze mRNA expression of EndMT transcription factors in aortic sections of db/db mice and diabetic patients. To identify the underlying mechanism by which EndMT contributes to aortic stiffening, we used aortas from db/db mice and diabetic patients in combination with high glucose-treated human umbilical vein endothelial cells (HUVECs) as an in vitro model of diabetes-associated EndMT. RESULTS: We demonstrate robust CD31/α-SMA and CD31/S100A4 co-localization in aortic sections of db/db mice which was almost absent in control mice. Moreover, we demonstrate a significant upregulation of EndMT transcription factors in aortic sections of db/db mice and diabetic patients. As underlying regulator, we identified miR-132-3p as the most significantly downregulated miR in the micronome of db/db mice and high glucose-treated HUVECs. Indeed, miR-132-3p was also significantly downregulated in aortic tissue from diabetic patients. We identified Kruppel-like factor 7 (KLF7) as a target of miR-132-3p and show a significant upregulation of KLF7 in aortic sections of db/db mice and diabetic patients as well as in high glucose-treated HUVECs. We further demonstrate that miR-132-3p overexpression and KLF7 downregulation ameliorates EndMT in high glucose-treated HUVECs. CONCLUSIONS: We demonstrate for the first time that EndMT contributes to aortic stiffening in T2D. We identified miR-132-3p and KLF7 as novel EndMT regulators in this context. Altogether, this gives us new insights in the development of aortic stiffening in T2D.

20.
ESC Heart Fail ; 10(4): 2307-2318, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37060191

RESUMEN

AIMS: There is evidence to suggest that the subtype of aortic stenosis (AS), the degree of myocardial fibrosis (MF), and level of aortic valve calcification (AVC) are associated with adverse cardiac outcome in AS. Because little is known about their respective contribution, we sought to investigate their relative importance and interplay as well as their association with adverse cardiac events following transcatheter aortic valve replacement (TAVR). METHODS AND RESULTS: One hundred consecutive patients with severe AS and indication for TAVR were prospectively enrolled between January 2017 and October 2018. Patients underwent transthoracic echocardiography, multidetector computed tomography, and left ventricular endomyocardial biopsies at the time of TAVR. The final study cohort consisted of 92 patients with a completed study protocol, 39 (42.4%) of whom showed a normal ejection fraction (EF) high-gradient (NEFHG) AS, 13 (14.1%) a low EF high-gradient (LEFHG) AS, 25 (27.2%) a low EF low-gradient (LEFLG) AS, and 15 (16.3%) a paradoxical low-flow, low-gradient (PLFLG) AS. The high-gradient phenotypes (NEFHG and LEFHG) showed the largest amount of AVC (807 ± 421 and 813 ± 281 mm3 , respectively) as compared with the low-gradient phenotypes (LEFLG and PLFLG; 503 ± 326 and 555 ± 594 mm3 , respectively, P < 0.05). Conversely, MF was most prevalent in low-output phenotypes (LEFLG > LEFHG > PLFLG > NEFHG, P < 0.05). This was paralleled by a greater cardiovascular (CV) mortality within 600 days after TAVR (LEFLG 28% > PLFLG 26.7% > LEFHG 15.4% > NEFHG 2.5%; P = 0.023). In patients with a high MF burden, a higher AVC was associated with a lower mortality following TAVR (P = 0.045, hazard ratio 0.261, 95% confidence interval 0.07-0.97). CONCLUSIONS: MF is associated with adverse CV outcome following TAVR, which is most prevalent in low EF situations. In the presence of large MF burden, patients with large AVC have better outcome following TAVR. Conversely, worse outcome in large MF and relatively little AVC may be explained by a relative prominence of an underlying cardiomyopathy. The better survival rates in large AVC patients following TAVR indicate TAVR induced relief of severe AS-associated pressure overload with subsequently improved outcome.


Asunto(s)
Estenosis de la Válvula Aórtica , Cardiomiopatías , Reemplazo de la Válvula Aórtica Transcatéter , Humanos , Válvula Aórtica/cirugía , Resultado del Tratamiento , Estenosis de la Válvula Aórtica/diagnóstico , Estenosis de la Válvula Aórtica/cirugía , Estenosis de la Válvula Aórtica/complicaciones , Fibrosis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA