Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Pathol ; 189(2): 339-353, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30448404

RESUMEN

Patients affected by Duchenne muscular dystrophy (DMD) develop a progressive dilated cardiomyopathy characterized by inflammatory cell infiltration, necrosis, and cardiac fibrosis. Standard treatments consider the use of ß-blockers and angiotensin-converting enzyme inhibitors that are symptomatic and unspecific toward DMD disease. Medications that target DMD cardiac fibrosis are in the early stages of development. We found immunoproteasome dysregulation in affected hearts of mdx mice (murine animal model of DMD) and cardiomyocytes derived from induced pluripotent stem cells of patients with DMD. Interestingly, immunoproteasome inhibition ameliorated cardiomyopathy in mdx mice and reduced the development of cardiac fibrosis. Establishing the immunoproteasome inhibition-dependent cardioprotective role suggests the possibility of modulating the immunoproteasome as new and clinically relevant treatment to rescue dilated cardiomyopathy in patients with DMD.


Asunto(s)
Cardiomiopatías , Distrofia Muscular de Duchenne , Miocitos Cardíacos , Complejo de la Endopetidasa Proteasomal/inmunología , Animales , Cardiomiopatías/inmunología , Cardiomiopatías/patología , Fibrosis , Humanos , Células Madre Pluripotentes Inducidas/inmunología , Células Madre Pluripotentes Inducidas/patología , Masculino , Ratones , Ratones Endogámicos mdx , Distrofia Muscular de Duchenne/inmunología , Distrofia Muscular de Duchenne/patología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/patología
2.
Int J Mol Sci ; 20(10)2019 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-31096574

RESUMEN

Arrhythmogenic cardiomyopathy (ACM) is a genetic disorder characterized by the progressive substitution of functional myocardium with noncontractile fibro-fatty tissue contributing to ventricular arrhythmias and sudden cardiac death. Cyclophilin A (CyPA) is a ubiquitous protein involved in several pathological mechanisms, which also characterize ACM (i.e., fibrosis, inflammation, and adipogenesis). Nevertheless, the involvement of CyPA in ACM cardiac remodeling has not been investigated yet. Thus, we first evaluated CyPA expression levels in the right ventricle (RV) tissue specimens obtained from ACM patients and healthy controls (HC) by immunohistochemistry. Then, we took advantage of ACM- and HC-derived cardiac mesenchymal stromal cells (C-MSC) to assess CyPA modulation during adipogenic differentiation. Interestingly, CyPA was more expressed in the RV sections obtained from ACM vs. HC subjects and positively correlated with the adipose replacement extent. Moreover, CyPA was upregulated at early stages of C-MSC adipogenic differentiation and was secreted at higher level over time in ACM- derived C-MSC. Our study provides novel ex vivo and in vitro information on CyPA expression in ACM remodeling paving the way for future C-MSC-based mechanistic and therapeutic investigations.


Asunto(s)
Arritmias Cardíacas/metabolismo , Cardiomiopatías/metabolismo , Ciclofilina A/metabolismo , Remodelación Ventricular , Adipogénesis/fisiología , Tejido Adiposo/patología , Arritmias Cardíacas/patología , Cardiomiopatías/patología , Diferenciación Celular , Ciclofilina A/genética , Muerte Súbita Cardíaca/patología , Fibrosis , Expresión Génica , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Humanos , Inflamación , Células Madre Mesenquimatosas/patología , Miocardio
3.
J Transl Med ; 16(1): 352, 2018 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-30541573

RESUMEN

BACKGROUND: To date the TGF-ß1 activation mediated by integrin ανß5 during fibrosis is well-known. This process has been shown also in the heart, where cardiac fibroblasts (CF) differentiate into α-smooth muscle actin (α-SMA)-positive myofibroblasts (MyoFB). Here, we studied the effects on CF, isolated by spontaneously hypertensive rats (SHR), of integrin ανß5 inhibition in MyoFB differentiation. METHODS: Staining and immunohistochemistry were performed on rat cardiac tissue. CF were isolated by enzymatic digestion from SHR (SHR-CF) and normotensive WKY (WKY-CF) rat hearts and then treated for in vitro evaluation. RESULTS: SHR heart tissues revealed a higher TGF-ß1 expression vs. WKY samples. SHR-CF showed an enhanced SMAD2/3 activation and an up-regulated expression of α-SMA, a typical MyoFB marker, especially after TGF-ß1 treatment. Immunostaining on cardiac tissues revealed a higher expression of integrin ανß5 in SHR vs. WKY rat hearts. In vitro results confirmed the up-regulation of integrin ανß5 expression in SHR-CF at basal condition and after TGF-ß1 treatment, in comparison with WKY-CF. Inhibition of integrin ανß5 by cilengitide treatment led a decreased expression of ανß5, collagen I, and α-SMA in SHR-CF vs. WKY-CF, resulting in a diminished differentiation of CF into MyoFB. Taking together, results suggested that SHR-CF are more susceptible to TGF-ß1, showing an up-regulated activation of SMAD2/3 signaling, and an increased ανß5, α-SMA, and collagen I expression. Hypertension stimulus promoted an up-regulation of integrin ανß5 on SHR cardiac tissue and its in vitro inhibition reverted pro-fibrotic events of SHR-CF. CONCLUSION: Inhibition of integrin ανß5 exerted by cilengitide strongly diminished SHR-CF differentiation into detrimental MyoFB. So, integrin ανß5 might be considered a novel therapeutic target and cilengitide an effective pharmacological tool to limit the progression of hypertension-induced cardiac fibrosis.


Asunto(s)
Fibroblastos/metabolismo , Fibroblastos/patología , Miocardio/metabolismo , Miocardio/patología , Receptores de Vitronectina/antagonistas & inhibidores , Actinas/metabolismo , Animales , Biomarcadores/metabolismo , Presión Sanguínea/efectos de los fármacos , Colágeno Tipo I/metabolismo , Diástole/efectos de los fármacos , Masculino , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Miofibroblastos/patología , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptores de Vitronectina/genética , Receptores de Vitronectina/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo , Venenos de Serpiente/farmacología , Sístole/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
4.
Pharmacol Res ; 127: 77-91, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28235633

RESUMEN

Over the past two decades cardiac cell therapy (CCT) has emerged as a promising new strategy to cure heart diseases at high unmet need. Thousands of patients have entered clinical trials for acute or chronic heart conditions testing different cell types, including autologous or allogeneic bone marrow (BM)-derived mononuclear or selected cells, BM- or adipose tissue-derived mesenchymal cells, or cardiac resident progenitors based on their potential ability to regenerate scarred or dysfunctional myocardium. Nowadays, the original enthusiasm surrounding the regenerative medicine field has been cushioned by a cumulative body of evidence indicating an inefficient or modest efficacy of CCT in improving cardiac function, along with the continued lack of indisputable proof for long-term prognostic benefit. In this review, we have firstly comprehensively outlined the positive and negative results of cell therapy studies in patients with acute myocardial infarction, refractory angina and chronic heart failure. Next, we have discussed cell therapy- and patient-related variables (e.g. cell intrinsic and extrinsic characteristics as well as criteria of patient selection and proposed methodologies) that might have dampened the efficacy of past cell therapy trials. Finally, we have addressed critical factors to be considered before embarking on further clinical trials.


Asunto(s)
Angina de Pecho/terapia , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Insuficiencia Cardíaca/terapia , Infarto del Miocardio/terapia , Humanos , Selección de Paciente
5.
Cell Mol Life Sci ; 74(2): 267-277, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27535662

RESUMEN

Marfan syndrome (MFS) is a connective tissue disorder with multiple organ manifestations. The genetic cause of this syndrome is the mutation of the FBN1 gene, encoding the extracellular matrix (ECM) protein fibrillin-1. This genetic alteration leads to the degeneration of microfibril structures and ECM integrity in the tunica media of the aorta. Indeed, thoracic aortic aneurysm and dissection represent the leading cause of death in MFS patients. To date, the most effective treatment option for this pathology is the surgical substitution of the damaged aorta. To highlight novel therapeutic targets, we review the molecular mechanisms related to MFS etiology in vascular smooth muscle cells, the foremost cellular type involved in MFS pathogenesis.


Asunto(s)
Aneurisma/complicaciones , Aneurisma/patología , Aorta/patología , Síndrome de Marfan/complicaciones , Síndrome de Marfan/patología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Animales , Humanos , Mecanotransducción Celular
6.
J Cell Mol Med ; 21(5): 871-880, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27860185

RESUMEN

The recruitment of bone marrow (BM)-derived progenitor cells to the lung is related to pulmonary remodelling and the pathogenesis of pulmonary hypertension (PH). Although sildenafil is a known target in PH treatment, the underlying molecular mechanism is still elusive. To test the hypothesis that the therapeutic effect of sildenafil is linked to the reduced recruitment of BM-derived progenitor cells, we induced pulmonary remodelling in rats by two-week exposure to chronic hypoxia (CH, 10% oxygen), a trigger of BM-derived progenitor cells. Rats were treated with either placebo (saline) or sildenafil (1.4 mg/kg/day ip) during CH. Control rats were kept in room air (21% oxygen) with no treatment. As expected, sildenafil attenuated the CH-induced increase in right ventricular systolic pressure and right ventricular hypertrophy. However, sildenafil suppressed the CH-induced increase in c-kit+ cells in the adventitia of pulmonary arteries. Moreover, sildenafil reduced the number of c-kit+ cells that colocalize with tyrosine kinase receptor 2 (VEGF-R2) and CD68 (a marker for macrophages), indicating a positive effect on moderating hypoxia-induced smooth muscle cell proliferation and inflammation without affecting the pulmonary levels of hypoxia-inducible factor (HIF)-1α. Furthermore, sildenafil depressed the number of CXCR4+ cells. Collectively, these findings indicate that the improvement in pulmonary haemodynamic by sildenafil is linked to decreased recruitment of BM-derived c-kit+ cells in the pulmonary tissue. The attenuation of the recruitment of BM-derived c-kit+ cells by sildenafil may provide novel therapeutic insights into the control of pulmonary remodelling.


Asunto(s)
Células de la Médula Ósea/patología , Pulmón/patología , Citrato de Sildenafil/farmacología , Células Madre/patología , Animales , Análisis de los Gases de la Sangre , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Hipoxia de la Célula/efectos de los fármacos , GMP Cíclico/metabolismo , Inflamación/patología , Masculino , Músculos/efectos de los fármacos , Músculos/patología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Ratas Sprague-Dawley , Receptores CXCR4/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
7.
Cell Mol Life Sci ; 72(9): 1725-40, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25575564

RESUMEN

Cardiovascular disease is the leading cause of morbidity and mortality in the developed world. Although ongoing therapeutic strategies ameliorate symptoms and prolong life for patients with cardiovascular diseases, they do not solve the critical issue related to the loss of cardiac tissue. Accordingly, stem/progenitor cell therapy has emerged as a paramount approach for cardiac repair and regeneration. In this regard, c-kit(+) cells have animated much interest and controversy. These cells are self-renewing, clonogenic, and multipotent and display a noteworthy potential to differentiate into all cardiovascular lineages. However, their functional contribution to cardiomyocyte turnover is one of the centrally debated issues concerning their regenerative potential. Regardless, plentiful preclinical and clinical studies have been conducted which provide evidence for the capacity of c-kit(+) cells to improve cardiac function. The purpose of this review is to give a comprehensive, impartial, critical description and evaluation of the literature on c-kit(+) cells from bench to bedside in order to address their true potential, benefits and controversies.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Corazón/fisiología , Miocitos Cardíacos/citología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Regeneración , Células Madre/citología , Animales , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Corazón/fisiopatología , Humanos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteínas Proto-Oncogénicas c-kit/análisis , Células Madre/metabolismo , Células Madre/patología
8.
J Mol Cell Cardiol ; 81: 62-70, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25640160

RESUMEN

Age-associated cardiovascular diseases are at least partially ascribable to vascular cell senescence. Replicative senescence (RS) and stress-induced premature senescence (SIPS) are provoked respectively by endogenous (telomere erosion) and exogenous (H2O2, UV) stimuli resulting in cell cycle arrest in G1 and G2 phases. In both scenarios, mitochondria-derived ROS are important players in senescence initiation. We aimed to define whether a mtDNA-transcribed long-non-coding-RNA (lncRNA), ASncmtRNA-2, has a role in vascular aging and senescence. Aortas of old mice, characterized by increased senescence, showed an increment in ASncmtRNA-2 expression. In vitro analysis of Endothelial Cells (EC) and Vascular Smooth Muscle Cells (VSMC) established that ASncmtRNA-2 is induced in EC, but not in VSMC, during RS. Surprisingly, ASncmtRNA-2 is not upregulated in two different EC SIPS scenarios, treated with H2O2 and UV. The p16 gene displayed similar ASncmtRNA-2 expression patterns, suggesting a possible co-regulation of the two genes. Interestingly, the expression of two miRNAs, hsa-miR-4485 and hsa-miR-1973, with perfect homology to the double strand region of ASncmtRNA-2 and originating at least in part from a mitochondrial transcript, was induced in RS, opening to the possibility that this lncRNA functions as a non-canonical precursor of these miRNAs. Cell cycle analysis of EC transiently over-expressing ASncmtRNA-2 revealed an accumulation of EC in the G2/M phase, but not in the G1 phase. We propose that ASncmtRNA-2 in EC might be involved in the RS establishment by participating in the cell cycle arrest in G2/M phase, possibly through the production of hsa-miR-4485 and hsa-miR-1973. This article is part of a Special Issue entitled: Mitochondria.


Asunto(s)
Envejecimiento/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Mitocondrias/metabolismo , Miocitos del Músculo Liso/metabolismo , ARN Largo no Codificante/genética , ARN/genética , Envejecimiento/genética , Animales , Aorta/citología , Aorta/metabolismo , Secuencia de Bases , Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de la radiación , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de la radiación , Humanos , Peróxido de Hidrógeno/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Mitocondrias/genética , Datos de Secuencia Molecular , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/efectos de la radiación , ARN/metabolismo , ARN Largo no Codificante/metabolismo , ARN Mitocondrial , Transducción de Señal , Rayos Ultravioleta
9.
Circ Res ; 110(4): 560-8, 2012 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-22267843

RESUMEN

RATIONALE: Endothelial cells (EC) at regions exposed to disturbed flow (d-flow) are predisposed to inflammation and the subsequent development of atherosclerosis. We previously showed that thioredoxin interacting protein (TXNIP) was required for tumor necrosis factor-mediated expression of vascular cell adhesion molecule-1. OBJECTIVE: We sought to investigate the role of TXNIP in d-flow-induced cell adhesion molecule expression and leukocyte interaction with vessels, and the mechanisms by which TXNIP suppresses athero-protective gene expression. METHODS AND RESULTS: Using en face staining of mouse aorta, we found a dramatic increase of TXNIP in EC at sites exposed to d-flow as compared to steady flow. EC-specific TXNIP (EC-TXNIP) knockout mice showed significant decreases in vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 mRNA expression in the d-flow regions of mouse aorta. Intravital microscopy of mesenteric venules showed that leukocyte rolling time was decreased, whereas rolling velocity was increased significantly in EC-TXNIP knockout mice. In vitro experiments using a cutout flow chamber to generate varying flow patterns showed that increased TXNIP was required for d-flow-induced EC-monocyte adhesion. Furthermore, we found that the expression of Kruppel-like factor 2, a key anti-inflammatory transcription factor in EC, was inhibited by TXNIP. Luciferase and chromatin immunoprecipitation assays showed that TXNIP was present within a repressing complex on the Kruppel-like factor 2 promoter. CONCLUSIONS: These data demonstrate the essential role for TXNIP in mediating EC-leukocyte adhesion under d-flow, as well as define a novel mechanism by which TXNIP acts as a transcriptional corepressor to regulate Kruppel-like factor 2-dependent gene expression.


Asunto(s)
Aterosclerosis/metabolismo , Proteínas Portadoras/metabolismo , Células Endoteliales/metabolismo , Inflamación/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Rodamiento de Leucocito , Tiorredoxinas/metabolismo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/inmunología , Proteínas Portadoras/genética , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Células Endoteliales/inmunología , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inflamación/genética , Inflamación/inmunología , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas , Interferencia de ARN , Flujo Sanguíneo Regional , Estrés Mecánico , Tiorredoxinas/genética , Transfección , Factor de Necrosis Tumoral alfa/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 33(9): 2147-53, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23846495

RESUMEN

OBJECTIVE: Angiotensin II (AngII) signal transduction in vascular smooth muscle cells (VSMC) is mediated by reactive oxygen species (ROS). Cyclophilin A (CyPA) is a ubiquitously expressed cytosolic protein that possesses peptidyl-prolyl cis-trans isomerase activity, scaffold function, and significantly enhances AngII-induced ROS production in VSMC. We hypothesized that CyPA regulates AngII-induced ROS generation by promoting translocation of NADPH oxidase cytosolic subunit p47phox to caveolae of the plasma membrane. APPROACH AND RESULTS: Overexpression of CyPA in CyPA-deficient VSMC (CyPA(-/-)VSMC) significantly increased AngII-stimulated ROS production. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitors (VAS2870 or diphenylene iodonium) significantly attenuated AngII-induced ROS production in CyPA and p47phox-overexpressing CyPA(-/-)VSMC. Cell fractionation and sucrose gradient analyses showed that AngII-induced p47phox plasma membrane translocation, specifically to the caveolae, was reduced in CyPA(-/-)VSMC compared with wild-type-VSMC. Immunofluorescence studies demonstrated that AngII increased p47phox and CyPA colocalization and translocation to the plasma membrane. In addition, immunoprecipitation of CyPA followed by immunoblotting of p47phox and actin showed that AngII increased CyPA and p47phox interaction. AngII-induced p47phox and actin cell cytoskeleton association was attenuated in CyPA(-/-)VSMC. Mechanistically, inhibition of p47phox phosphorylation and phox homology domain deletion attenuated CyPA and p47phox interaction. Finally, cyclosporine A and CyPA-peptidyl-prolyl cis-trans isomerase mutant, R55A, inhibited AngII-stimulated CyPA and p47phox association in VSMC, suggesting that peptidyl-prolyl cis-trans isomerase activity was required for their interaction. CONCLUSIONS: These findings provide the mechanism by which CyPA is an important regulator for AngII-induced ROS generation in VSMC through interaction with p47phox and cell cytoskeleton, which enhances the translocation of p47phox to caveolae.


Asunto(s)
Angiotensina II/farmacología , Caveolas/efectos de los fármacos , Ciclofilina A/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , NADPH Oxidasas/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/enzimología , Animales , Western Blotting , Caveolas/enzimología , Ciclofilina A/genética , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Técnica del Anticuerpo Fluorescente , Células HeLa , Humanos , Inmunoprecipitación , Ratones , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente , Músculo Liso Vascular/enzimología , Mutación , Miocitos del Músculo Liso/enzimología , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/genética , Oligopéptidos , Péptidos/genética , Péptidos/metabolismo , Fosforilación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Transporte de Proteínas , Ratas , Especies Reactivas de Oxígeno/metabolismo , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo
11.
Arterioscler Thromb Vasc Biol ; 32(12): 2974-80, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23023376

RESUMEN

OBJECTIVE: Protein kinase C (PKC) ζ is a key pathological mediator of endothelial cell apoptosis. p62 is a scaffold protein that regulates several cell signaling pathways by binding to target proteins. Because PKCζ and p62 contain Phox/Bem1p (PB1) modules that mediate protein-protein interactions, we hypothesized that an interaction between p62 and PKCζ is required for tumor necrosis factor α-induced PKCζ signaling in endothelial cells. METHODS AND RESULTS: In human umbilical vein endothelial cell, tumor necrosis factor α (10 ng/mL) enhanced the interaction between p62 and PKCζ. Transfection with p62 small interfering RNA reduced the activation of both PKCζ and its downstream targets JNK and caspase 3, suggesting that p62 is necessary for PKCζ signaling. Overexpression of only the PB1 domain of p62 inhibited p62-PKCζ interaction, showing that binding of these 2 proteins is mediated by their PB1 domains. Furthermore, overexpression of the p62 PB1 domain suppressed tumor necrosis factor α-induced PKCζ activation and subsequent activation of JNK and caspase 3. Finally, transfection of either p62 small interfering RNA or the PB1 domain of p62 inhibited human umbilical vein endothelial cell apoptosis. CONCLUSIONS: Our results suggest a novel function of p62 that regulates the activity of PKCζ by binding to PKCζ, thereby activating the PKCζ-JNK-caspase 3 apoptotic pathway in endothelial cells.


Asunto(s)
Apoptosis/fisiología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Caspasa 3/metabolismo , Línea Celular , Humanos , MAP Quinasa Quinasa 4/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-myc/genética , ARN Interferente Pequeño/genética , Transducción de Señal/fisiología , Transfección
12.
Blood ; 116(11): 1971-9, 2010 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-20538799

RESUMEN

PKCζ has emerged as a pathologic mediator of endothelial cell dysfunction, based on its essential role in tumor necrosis factor α (TNFα)-mediated inflammation. In contrast, extracellular signal-regulated kinase 5 (ERK5) function is required for endothelial cell homeostasis as shown by activation of Krüppel-like factor 2 (KLF2), increased endothelial nitric-oxide synthase (eNOS) expression, and inhibition of apoptosis. We hypothesized that protein kinase C ζ (PKCζ) activation by TNFα would inhibit the ERK5/KLF2/eNOS pathway. TNFα inhibited the steady laminar flow-induced eNOS expression, and this effect was reversed by the dominant-negative form of PKCζ (Ad.DN-PKCζ). In addition, ERK5 function was inhibited by either TNFα or the transfection of the catalytic domain of PKCζ. This inhibition was reversed by PKCζ small interfering RNA. PKCζ was found to bind to ERK5 under basal conditions with coimmunoprecipitation and the mammalian 2-hybrid assay. Furthermore, PKCζ phosphorylates ERK5, and mutation analysis showed that the preferred site is S486. Most importantly, we found that the predominant effect of TNFα stimulation of PKCζ was to decrease eNOS protein stability that was recapitulated by transfecting Ad.ERK5S486A mutant. Finally, aortic en face analysis of ERK5/PKCζ activity showed high PKCζ and ERK5 staining in the athero-prone region. Taken together our results show that PKCζ binds and phosphorylates ERK5, thereby decreasing eNOS protein stability and contributing to early events of atherosclerosis.


Asunto(s)
Células Endoteliales/metabolismo , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteína Quinasa C/metabolismo , Animales , Aorta/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Proteína Quinasa 7 Activada por Mitógenos/genética , Mutación , Óxido Nítrico Sintasa de Tipo III/genética , Fosforilación/efectos de los fármacos , Unión Proteica , Proteína Quinasa C/genética , Estabilidad Proteica , Interferencia de ARN , Factor de Necrosis Tumoral alfa/farmacología
14.
Arterioscler Thromb Vasc Biol ; 31(5): 1116-23, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21330604

RESUMEN

OBJECTIVE: Cyclophilin A (CyPA, encoded by Ppia) is a proinflammatory protein secreted in response to oxidative stress in mice and humans. We recently demonstrated that CyPA increased angiotensin II (Ang II)-induced reactive oxygen species (ROS) production in the aortas of apolipoprotein E (Apoe)-/- mice. In this study, we sought to evaluate the role of CyPA in Ang II-induced cardiac hypertrophy. METHODS AND RESULTS: Cardiac hypertrophy was not significantly different between Ppia+/+ and Ppia-/- mice infused with Ang II (1000 ng/min per kg for 4 weeks). Therefore, we investigated the effect of CyPA under conditions of high ROS and inflammation using the Apoe-/- mice. In contrast to Apoe-/- mice, Apoe-/-Ppia-/- mice exhibited significantly less Ang II-induced cardiac hypertrophy. Bone marrow cell transplantation showed that CyPA in cells intrinsic to the heart plays an important role in the cardiac hypertrophic response. Ang II-induced ROS production, cardiac fibroblast proliferation, and cardiac fibroblast migration were markedly decreased in Apoe-/-Ppia-/- cardiac fibroblasts. Furthermore, CyPA directly induced the hypertrophy of cultured neonatal cardiac myocytes. CONCLUSIONS: CyPA is required for Ang II-mediated cardiac hypertrophy by directly potentiating ROS production, stimulating the proliferation and migration of cardiac fibroblasts, and promoting cardiac myocyte hypertrophy.


Asunto(s)
Apolipoproteínas E/deficiencia , Cardiomegalia/enzimología , Ciclofilina A/metabolismo , Miocardio/enzimología , Angiotensina II , Animales , Animales Recién Nacidos , Apolipoproteínas E/genética , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Cardiomegalia/inducido químicamente , Cardiomegalia/genética , Cardiomegalia/inmunología , Cardiomegalia/patología , Cardiomegalia/prevención & control , Comunicación Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Ciclofilina A/deficiencia , Ciclofilina A/genética , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/inmunología , Miocardio/patología , Miocitos Cardíacos/metabolismo , Estrés Oxidativo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/metabolismo , Factores de Tiempo
15.
Int J Biol Sci ; 17(10): 2399-2416, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34326683

RESUMEN

Myocardial aging increases the cardiovascular risk in the elderly. The Receptor for Advanced Glycation End-products (RAGE) is involved in age-related disorders. The soluble isoform (sRAGE) acts as a scavenger blocking the membrane-bound receptor activation. This study aims at investigating RAGE contribution to age-related cardiac remodeling. We analyzed the cardiac function of three different age groups of female Rage-/- and C57BL/6N (WT) mice: 2.5- (Young), 12- (Middle-age, MA) and 21-months (Old) old. While aging, Rage-/- mice displayed an increase in left ventricle (LV) dimensions compared to age-matched WT animals, with the main differences observed in the MA groups. Rage-/- mice showed higher fibrosis and a larger number of α-Smooth Muscle Actin (SMA)+ cells with age, along with increased expression of pro-fibrotic Transforming Growth Factor (TGF)-ß1 pathway components. RAGE isoforms were undetectable in LV of WT mice, nevertheless, circulating sRAGE declined with aging and inversely associated with LV diastolic dimensions. Human cardiac fibroblasts stimulated with sRAGE exhibited a reduction in proliferation, pro-fibrotic proteins and TGF-beta Receptor 1 (TGFbR1) expression and Smad2-3 activation. Finally, sRAGE administration to MA WT animals reduced cardiac fibrosis. Hence, our work shows that RAGE associates with age-dependent myocardial changes and indicates sRAGE as an inhibitor of cardiac fibroblasts differentiation and age-dependent cardiac fibrosis.


Asunto(s)
Actinas/metabolismo , Envejecimiento , Miocardio/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Línea Celular , Femenino , Fibroblastos/metabolismo , Fibrosis , Humanos , Ratones , Ratones Endogámicos C57BL , Miocardio/patología , Isoformas de Proteínas/metabolismo
16.
Cells ; 9(1)2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31936351

RESUMEN

: Background: Marfan syndrome (MFS) is a genetic disease, characterized by thoracic aortic aneurysm (TAA), which treatment is to date purely surgical. Understanding of novel molecular targets is mandatory to unveil effective pharmacological approaches. Cyclophilin A (CyPA) and its receptor EMMPRIN are associated with several cardiovascular diseases, including abdominal aortic aneurysm. Here, we envisioned the contribution of CyPA/EMMPRIN axis in MFS-related TAA. METHODS: We obtained thoracic aortic samples from healthy controls (HC) and MFS patients' aortas and then isolated vascular smooth muscle cells (VSMC) from the aortic wall. RESULTS: our findings revealed that MFS aortic tissue samples isolated from the dilated zone of aorta showed higher expression levels of EMMPRIN vs. MFS non-dilated aorta and HC. Interestingly, angiotensin II significantly stimulated CyPA secretion in MFS-derived VSMC (MFS-VSMC). CyPA treatment on MFS-VSMC led to increased levels of EMMPRIN and other MFS-associated pro-fibrotic mediators, such as TGF-ß1 and collagen I. These molecules were downregulated by in vitro treatment with CyPA inhibitor MM284. Our results suggest that CyPA/EMMPRIN axis is involved in MFS-related TAA development, since EMMPRIN is upregulated in the dilated zone of MFS patients' TAA and the inhibition of its ligand, CyPA, downregulated EMMPRIN and MFS-related markers in MFS-VSMC. CONCLUSIONS: these insights suggest both a novel detrimental role for CyPA/EMMPRIN axis and its inhibition as a potential therapeutic strategy for MFS-related TAA treatment.


Asunto(s)
Aneurisma de la Aorta Torácica/patología , Basigina/metabolismo , Ciclofilina A/metabolismo , Fibrosis/patología , Síndrome de Marfan/fisiopatología , Factor de Crecimiento Transformador beta1/metabolismo , Aneurisma de la Aorta Torácica/metabolismo , Estudios de Casos y Controles , Fibrosis/metabolismo , Humanos
17.
Circulation ; 117(24): 3088-98, 2008 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-18541741

RESUMEN

BACKGROUND: Oxidative stress, generated by excessive reactive oxygen species, promotes cardiovascular disease. Cyclophilin A (CyPA) is a 20-kDa chaperone protein secreted from vascular smooth muscle cells (VSMCs) in response to reactive oxygen species that stimulates VSMC proliferation and inflammatory cell migration in vitro; however, the role CyPA plays in vascular function in vivo remains unknown. METHODS AND RESULTS: We tested the hypothesis that CyPA contributes to vascular remodeling by analyzing the response to complete carotid ligation in CyPA knockout mice, wild-type mice, and mice that overexpress CyPA in VSMC (VSMC-Tg). After carotid ligation, CyPA expression in vessels of wild-type mice increased dramatically and was significantly greater in VSMC-Tg mice. Reactive oxygen species-induced secretion of CyPA from mouse VSMCs correlated significantly with intracellular CyPA expression. Intimal and medial hyperplasia correlated significantly with CyPA expression after 2 weeks of carotid ligation, with marked decreases in CyPA knockout mice and increases in VSMC-Tg mice. Inflammatory cell migration into the intima was significantly reduced in CyPA knockout mice and increased in VSMC-Tg mice. Additionally, VSMC proliferation assessed by Ki67(+) cells was significantly less in CyPA knockout mice and was increased in VSMC-Tg mice. The importance of CyPA for intimal and medial thickening was shown by strong correlations between CyPA expression and the number of both inflammatory cells and proliferating VSMCs in vivo and in vitro. CONCLUSIONS: In response to low flow, CyPA plays a crucial role in VSMC migration and proliferation, as well as inflammatory cell accumulation, thereby regulating flow-mediated vascular remodeling and intima formation.


Asunto(s)
Ciclofilina A/deficiencia , Inflamación/fisiopatología , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiología , Animales , Aorta , Arterias Carótidas/fisiología , División Celular , Medios de Cultivo Condicionados , Ciclofilina A/genética , Ciclofilina A/fisiología , Regulación de la Expresión Génica , Ratones , Ratones Noqueados , Ratones Transgénicos , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiopatología , Especies Reactivas de Oxígeno/metabolismo , Túnica Íntima/citología , Túnica Íntima/fisiología
18.
Theranostics ; 9(8): 2224-2234, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31149040

RESUMEN

Marfan syndrome (MFS) is a rare genetic disease characterized by a matrix metalloproteases (MMPs) dysregulation that leads to extracellular matrix degradation. Consequently, MFS patients are prone to develop progressive thoracic aortic enlargement and detrimental aneurysm. Since MMPs are activated by the extracellular MMP inducer (EMMPRIN) protein, we determined whether its plasmatic soluble form (sEMMPRIN) may be considered a marker of thoracic aortic ectasia (AE). Methods: We compared plasma sEMMPRIN levels of 42 adult Caucasian MFS patients not previously subjected to aortic surgery with those of matched healthy controls (HC) by ELISA. In the MFS cohort we prospectively evaluated the relationship between plasma sEMMPRIN levels and the main MFS-related manifestations. Results: MFS patients had lower plasma sEMMPRIN levels (mean±SD: 2071±637 pg/ml) than HC (2441±642 pg/ml, p=0.009). Amongst all considered MFS-related clinical features, we found that only aortic root dilatation associated with circulating sEMMPRIN levels. Specifically, plasma sEMMPRIN levels negatively correlated with aortic Z-score (r=-0.431, p=0.004), and were significantly lower in patients with AE (Z-score≥2, 1788±510 pg/ml) compared to those without AE (Z-score<2, 2355±634 pg/ml; p=0.003). ROC curve analysis revealed that plasma sEMMPRIN levels discriminated patients with AE (AUC [95%CI]: 0.763 [0.610-0.916], p=0.003) with 85.7% sensitivity, 76.2% specificity, and 81% accuracy. We defined plasma sEMMPRIN levels ≤2246 pg/ml as the best threshold discriminating the presence of AE in MFS patients with an odds ratio [95%CI] of 19.2 [3.947-93.389] (p<0.001). Conclusions: MFS patients are characterized by lower sEMMPRIN levels than HC. Notably, plasma sEMMPRIN levels are strongly associated with thoracic AE.


Asunto(s)
Aorta/patología , Basigina/sangre , Síndrome de Marfan/diagnóstico , Adulto , Biomarcadores/sangre , Dilatación Patológica/sangre , Dilatación Patológica/patología , Femenino , Humanos , Masculino , Síndrome de Marfan/sangre , Sensibilidad y Especificidad
19.
Free Radic Biol Med ; 45(6): 875-84, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18638547

RESUMEN

The mammalian thioredoxin (Trx) system, composed of Trx, Trx reductase (TrxR), and NADPH, is the most important thiol system involved in the redox control of signaling and regulatory proteins in apoptosis and cell proliferation. Here we addressed the inhibition of the Trx system by 13-hydroxy-15-oxo-zoapatlin (OZ), a nor-kaurane diterpene previously shown to possess proapoptotic potential and to cause cell cycle arrest in leukemia cells. OZ was found, by both biochemical and mass spectrometry-based approaches, to target Trx1 and TrxR in a cell-free system. In particular, the formation of reversible OZ adducts to Trx1 Cys35, Cys62, and Cys73 was demonstrated. We next showed that OZ efficiently inhibited Trx and TrxR catalytic activity in Molt4 cells. The occurrence of oxidative modifications of Trx molecules was assessed by "redox Western blot" analyses. OZ-mediated Trx oxidation resulted in apoptosis signaling kinase-1 release and activation of downstream JNK and p38 pathways. By means of specific inhibitors of these two stress-activated protein kinases, we demonstrated that the JNK pathway plays a major role in determining the apoptotic fate of OZ-exposed cells, whereas p38 activation seems to be involved mainly in OZ-induced G2/M block.


Asunto(s)
Antineoplásicos/farmacología , Diterpenos/farmacología , Leucemia/patología , Tiorredoxinas/antagonistas & inhibidores , Western Blotting , Sistema Libre de Células , Activación Enzimática , Proteínas Quinasas/metabolismo , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masas en Tándem
20.
Prog Cardiovasc Dis ; 61(3-4): 328-335, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30041021

RESUMEN

Marfan Syndrome (MFS) is a rare connective tissue disorder, resulting from mutations in the fibrillin-1 gene, characterized by pathologic phenotypes in multiple organs, the most detrimental of which affects the thoracic aorta. Indeed, thoracic aortic aneurysms (TAA), leading to acute dissection and rupture, are today the major cause of morbidity and mortality in adult MFS patients. Therefore, there is a compelling need for novel therapeutic strategies to delay TAA progression and counteract aortic dissection occurrence. Unfortunately, the wide phenotypic variability of MFS patients, together with the lack of a complete genotype-phenotype correlation, have represented until now a barrier hampering the conduction of translational studies aimed to predict disease prognosis and drug discovery. In this review, we will illustrate available therapeutic strategies to improve the health of MFS patients. Starting from gold standard surgical overtures and the description of the main pharmacological approaches, we will comprehensively review the state-of-the-art of in vivo MFS models and discuss recent clinical pharmacogenetic results. Finally, we will focus on induced pluripotent stem cells (iPSC) as a technology that, if integrated with preclinical research and pharmacogenetics, could contribute in determining the best therapeutic approach for each MFS patient on the base of individual differences. Finally, we will suggest the integration of preclinical studies, pharmacogenetics and iPSC technology as the most likely strategy to help solve the composite puzzle of precise medicine in this condition.


Asunto(s)
Aneurisma de la Aorta Torácica , Disección Aórtica , Síndrome de Marfan , Adulto , Disección Aórtica/etiología , Disección Aórtica/mortalidad , Disección Aórtica/prevención & control , Aneurisma de la Aorta Torácica/etiología , Aneurisma de la Aorta Torácica/mortalidad , Humanos , Síndrome de Marfan/complicaciones , Síndrome de Marfan/genética , Síndrome de Marfan/terapia , Pruebas de Farmacogenómica , Medicina de Precisión/métodos , Pronóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA