Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Basic Res Cardiol ; 119(2): 291-307, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38430261

RESUMEN

The coronary perivascular adipose tissue (cPVAT) has been associated to the burden of cardiovascular risk factors and to the underlying vessel atherosclerotic plaque severity. Although the "outside to inside" hypothesis of PVAT-derived-adipokine regulation of vessel function is currently accepted, whether the resident mesenchymal stem cells (ASCs) in PVAT have a regulatory role on the underlying vascular arterial smooth muscle cells (VSMCs) is not known. Here, we investigated the interactions between resident PVAT-ASCs and VSMCs. ASCs were obtained from PVAT overlying the left anterior descending (LAD) coronary artery of hearts removed at heart transplant operations. PVAT was obtained both from patients with non-ischemic and ischemic heart disease as the cause of heart transplant. ASCs were isolated from PVAT, phenotypically characterized by flow cytometry, functionally tested for proliferation, and differentiation. Crosstalk between ASCs and VSMCs was investigated by co-culture studies. ASCs were detected in the adventitia of the LAD-PVAT showing differentiation capacity and angiogenic potential. ASCs obtained from PVAT of non-ischemic and ischemic hearts showed different tissue factor (TF) expression levels, different VSMCs recruitment capacity through the axis ERK1/2-ETS1 signaling and different angiogenic potential. Induced upregulation of TF in ASCs isolated from ischemic PVAT rescued their angiogenic capacity in subcutaneously implanted plugs in mice, whereas silencing TF in ASCs decreased the proangiogenic capacity of non-ischemic ASCs. The results indicate for the first time a novel mechanism of regulation of VSMCs by PVAT-ASCs in angiogenesis, mediated by TF expression in ASCs. Regulation of TF in ASCs may become a therapeutic intervention to increase cardiac protection.


Asunto(s)
Adipocitos , Tromboplastina , Humanos , Ratones , Animales , Tromboplastina/metabolismo , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Corazón , Células Madre
2.
Circ Res ; 125(1): 74-89, 2019 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-31219744

RESUMEN

RATIONALE: Adipose-derived stem cells (ASCs) are a potential adult mesenchymal stem cell source for restoring endothelial function in ischemic tissues. However, the mechanism that promotes ASCs differentiation toward endothelial cells (ECs) is not known. OBJECTIVE: To investigate the mechanisms of ASCs differentiation into ECs. METHODS AND RESULTS: ASCs were isolated from clinical lipoaspirates and cultured with DMEM or endothelial cell-conditioned medium. Endothelial cell-conditioned medium induced downregulation of miR-145 in ASCs and promoted endothelial differentiation. We identified bFGF (basic fibroblast growth factor) released by ECs as inducer of ASCs differentiation through receptor-induced AKT (protein kinase B) signaling and phosphorylation of FOXO1 (forkhead box protein O1) suppressing its transcriptional activity and decreasing miR-145 expression. Blocking bFGF-receptor or PI3K/AKT signaling in ASCs increased miR-145 levels. Modulation of miR-145 in ASCs, using a miR-145 inhibitor, regulated their differentiation into ECs: increasing proliferation, migration, inducing expression of EC markers (VE-cadherin, VEGFR2 [vascular endothelial growth factor receptor 2], or VWF [von Willebrand Factor]), and tube-like formation. Furthermore, in vivo, downregulation of miR-145 in ASCs enhanced angiogenesis in subcutaneously implanted plugs in mice. In a murine hindlimb ischemia model injection of ASCs with downregulated miR-145 induced collateral flow and capillary formation evidenced by magnetic resonance angiography. Next, we identified ETS1 (v-ets avian erythroblastosis virus E26 oncogene homolog 1) as the target of miR-145. Upregulation of miR-145 in ASCs, by mimic miR-145, suppressed ETS1 expression and consequently abolished EC differentiation and the angiogenic properties of endothelial cell-conditioned medium-preconditioned ASCs; whereas, overexpression of ETS1 reversed the abrogated antiangiogenic capacity of miR-145. ETS1 overexpression induced similar results to those obtained with miR-145 knockdown. CONCLUSIONS: bFGF released by ECs induces ASCs differentiation toward ECs through miR-145-regulated expression of ETS1. Downregulation of miR-145 in ASCs induce vascular network formation in ischemic muscle.


Asunto(s)
Adipocitos/metabolismo , Diferenciación Celular/fisiología , Células Endoteliales/metabolismo , MicroARNs/metabolismo , Microvasos/metabolismo , Neovascularización Fisiológica/fisiología , Adipocitos/patología , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Células Cultivadas , Células Endoteliales/patología , Células HeLa , Humanos , Isquemia/metabolismo , Isquemia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , MicroARNs/antagonistas & inhibidores , Microvasos/patología
3.
Arterioscler Thromb Vasc Biol ; 40(10): 2481-2493, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32847390

RESUMEN

OBJECTIVE: HDL (high-density lipoprotein) role in atherosclerosis is controversial. Clinical trials with CETP (cholesterylester transfer protein)-inhibitors have not provided benefit. We have shown that HDL remodeling in hypercholesterolemia reduces HDL cardioprotective potential. We aimed to assess whether hypercholesterolemia affects HDL-induced atherosclerotic plaque regression. Approach and Results: Atherosclerosis was induced in New Zealand White rabbits for 3-months by combining a high-fat-diet and double-balloon aortic denudation. Then, animals underwent magnetic resonance imaging (basal plaque) and randomized to receive 4 IV infusions (1 infusion/wk) of HDL isolated from normocholesterolemic (NC-HDL; 75 mg/kg; n=10), hypercholesterolemic (HC-HDL; 75 mg/Kg; n=10), or vehicle (n=10) rabbits. Then, animals underwent a second magnetic resonance imaging (end plaque). Blood, aorta, and liver samples were obtained for analyses. Follow-up magnetic resonance imaging revealed that NC-HDL administration regressed atherosclerotic lesions by 4.3%, whereas, conversely, the administration of HC-HDLs induced a further 6.5% progression (P<0.05 versus basal). Plaque characterization showed that HC-HDL administered animals had a 2-fold higher lipid and cholesterol content versus those infused NC-HDL and vehicle (P<0.05). No differences were observed among groups in CD31 levels, nor in infiltrated macrophages or smooth muscle cells. Plaques from HC-HDL administered animals exhibited higher Casp3 (caspase 3) content (P<0.05 versus vehicle and NC-HDL) whereas plaques from NC-HDL infused animals showed lower expression of Casp3, Cox1 (cyclooxygenase 1), inducible nitric oxide synthase, and MMP (metalloproteinase) activity (P<0.05 versus HC-HDL and vehicle). HDLs isolated from animals administered HC-HDL displayed lower antioxidant potential and cholesterol efflux capacity as compared with HDLs isolated from NC-HDL-infused animal and vehicle or donor HDL (P<0.05). There were no differences in HDL-ApoA1 content, ABCA1 (ATP-binding cassette transporter A1) vascular expression, and SRB1 (scavenger receptor B1) and ABCA1 liver expression. CONCLUSIONS: HDL particles isolated from a hypercholesterolemic milieu lose their ability to regress and stabilize atherosclerotic lesions. Our data suggest that HDL remodeling in patients with co-morbidities may lead to the loss of HDL atheroprotective functions.


Asunto(s)
Anticolesterolemiantes/administración & dosificación , Aorta Abdominal/efectos de los fármacos , Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , HDL-Colesterol/administración & dosificación , Hipercolesterolemia/tratamiento farmacológico , Imagen por Resonancia Magnética , Placa Aterosclerótica , Animales , Anticolesterolemiantes/toxicidad , Aorta Abdominal/diagnóstico por imagen , Aorta Abdominal/metabolismo , Enfermedades de la Aorta/sangre , Enfermedades de la Aorta/diagnóstico por imagen , Enfermedades de la Aorta/etiología , Aterosclerosis/sangre , Aterosclerosis/diagnóstico por imagen , Aterosclerosis/etiología , Biomarcadores/sangre , HDL-Colesterol/sangre , HDL-Colesterol/toxicidad , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Hipercolesterolemia/sangre , Hipercolesterolemia/complicaciones , Infusiones Intravenosas , Masculino , Conejos
4.
Int J Mol Sci ; 22(10)2021 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-34066088

RESUMEN

Familial hypercholesterolemia (FH) is increasingly associated with inflammation, a phenotype that persists despite treatment with lipid lowering therapies. The alternative C3 complement system (C3), as a key inflammatory mediator, seems to be involved in the atherosclerotic process; however, the relationship between C3 and lipids during plaque progression remains unknown. The aim of the study was to investigate by a systems biology approach the role of C3 in relation to lipoprotein levels during atherosclerosis (AT) progression and to gain a better understanding on the effects of C3 products on the phenotype and function of human lipid-loaded vascular smooth muscle cells (VSMCs). By mass spectrometry and differential proteomics, we found the extracellular matrix (ECM) of human aortas to be enriched in active components of the C3 complement system, with a significantly different proteomic signature in AT segments. Thus, C3 products were more abundant in AT-ECM than in macroscopically normal segments. Furthermore, circulating C3 levels were significantly elevated in FH patients with subclinical coronary AT, evidenced by computed tomographic angiography. However, no correlation was identified between circulating C3 levels and the increase in plaque burden, indicating a local regulation of the C3 in AT arteries. In cell culture studies of human VSMCs, we evidenced the expression of C3, C3aR (anaphylatoxin receptor) and the integrin αMß2 receptor for C3b/iC3b (RT-PCR and Western blot). C3mRNA was up-regulated in lipid-loaded human VSMCs, and C3 protein significantly increased in cell culture supernatants, indicating that the C3 products in the AT-ECM have a local vessel-wall niche. Interestingly, C3a and iC3b (C3 active fragments) have functional effects on VSMCs, significantly reversing the inhibition of VSMC migration induced by aggregated LDL and stimulating cell spreading, organization of F-actin stress fibers and attachment during the adhesion of lipid-loaded human VSMCs. This study, by using a systems biology approach, identified molecular processes involving the C3 complement system in vascular remodeling and in the progression of advanced human atherosclerotic lesions.


Asunto(s)
Aterosclerosis/patología , Complemento C3/metabolismo , Hiperlipoproteinemia Tipo II/patología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Proteoma/metabolismo , Adulto , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Estudios de Casos y Controles , Adhesión Celular , Células Cultivadas , Femenino , Humanos , Hiperlipoproteinemia Tipo II/inmunología , Hiperlipoproteinemia Tipo II/metabolismo , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/inmunología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/metabolismo , Proteoma/análisis , Remodelación Vascular , Cicatrización de Heridas , Adulto Joven
5.
Br J Haematol ; 189(6): 1064-1073, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32130737

RESUMEN

Anti-cluster of differentiation 20 (CD20) monoclonal antibodies (mAbs) have shown promise in follicular lymphoma (FL) as post-induction therapy, by enhancing antibody-dependent cellular cytotoxicity (ADCC). However, cytotoxic cells are reduced after this treatment. We hypothesised that ex vivo expanded lymphokine-activated killer (LAK) cells administered to FL-remission patients are safe and improve anti-CD20 efficacy. This open, prospective, phase II, single-arm study assessed safety and efficacy of ex vivo expanded LAK cells in 20 FL-remission patients following rituximab maintenance. Mononuclear cells were obtained in odd rituximab cycles and stimulated with interleukin 2 (IL-2) for 8 weeks, after which >5 × 108 LAK cells were injected. Patients were followed-up for 5 years. At the end of maintenance, peripheral blood cells phenotype had not changed markedly. Natural killer, LAK and ADCC activities of mononuclear cells increased significantly after recombinant human IL-2 (rhIL-2) stimulation in all cycles. Rituximab significantly enhanced cytotoxic activity. No patients discontinued treatment. There were no treatment-related serious adverse events. Three patients had progressed by the end of follow-up. After a median (interquartile range) follow-up of 59.4 (43.8-70.9) months, 85% of patients remained progression free. No deaths occurred. Quality-of-life improved throughout the study. Post-induction LAK cells with rituximab seem safe in the long term. Larger studies are warranted to confirm efficacy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Células Asesinas Activadas por Linfocinas/trasplante , Linfoma Folicular/terapia , Quimioterapia de Mantención , Adolescente , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Ciclofosfamida/administración & dosificación , Ciclofosfamida/efectos adversos , Doxorrubicina/administración & dosificación , Doxorrubicina/efectos adversos , Femenino , Estudios de Seguimiento , Humanos , Linfoma Folicular/patología , Masculino , Persona de Mediana Edad , Prednisona/administración & dosificación , Prednisona/efectos adversos , Estudios Prospectivos , Rituximab/administración & dosificación , Rituximab/efectos adversos , Vincristina/administración & dosificación , Vincristina/efectos adversos
6.
Int J Mol Sci ; 21(6)2020 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-32188077

RESUMEN

Although the advent of combined antiretroviral therapy has substantially improved the survival of HIV-1-infected individuals, non-AIDS-related diseases are becoming increasingly prevalent in HIV-1-infected patients. Persistent abnormalities in coagulation appear to contribute to excess risk for a broad spectrum of non-AIDS defining complications. Alterations in coagulation biology in the context of HIV infection seem to be largely a consequence of a chronically inflammatory microenvironment leading to endothelial cell (EC) dysfunction. A possible direct role of HIV-1 proteins in sustaining EC dysfunction has been postulated but not yet investigated. The HIV-1 matrix protein p17 (p17) is secreted from HIV-1-infected cells and is known to sustain inflammatory processes by activating ECs. The aim of this study was to investigate the possibility that p17-driven stimulation of human ECs is associated with increased production of critical coagulation factors. Here we show the involvement of autophagy in the p17-induced accumulation and secretion of von Willebrand factor (vWF) by ECs. In vivo experiments confirmed the capability of p17 to exert a potent pro-coagulant activity soon after its intravenous administration.


Asunto(s)
Antitrombina III/metabolismo , Autofagia/fisiología , Células Endoteliales/metabolismo , Antígenos VIH/metabolismo , Péptido Hidrolasas/metabolismo , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo , Factor de von Willebrand/metabolismo , Animales , Antirretrovirales/uso terapéutico , Femenino , Infecciones por VIH/complicaciones , VIH-1/fisiología , Humanos , Ratones
7.
FASEB J ; 30(8): 2849-59, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27150622

RESUMEN

The epicardial adipose tissue (EAT) is a reservoir of adipose-derived stem cells (ASCs), with as yet unknown effects on myocardial and coronary arteries homeostasis. The purpose of this study was to investigate the angiogenic function of epicardial ASCs and their regulation by the common cardiovascular risk factors (CVRFs) affecting heart disease. Epicardial fat was obtained from a rodent model with clustering of CVRFs [Zucker diabetic fatty (ZDF)-Lepr(fa)] rats and from their lean control (ZDF-Crl) littermates without CVRFs, ASCs were isolated, and their function was assessed by proliferation and differentiation assays, flow cytometry, gene expression, and in vivo Matrigel angiogenesis analysis. Epicardial ASCs from both groups showed adipogenic and osteogenic differentiation capacity; however, epicardial ASCs from CVRF animals had a lesser ability to form tubular structures in vitro after endothelial differentiation, as well as a reduced angiogenic potential in vivo compared to control animals. Epicardial ASCs from CVRF rats showed up-regulation of the downstream Notch signaling genes Hes7, Hey1, and Heyl compared with control animals. The inhibition of Notch signaling by conditioning epicardial ASCs from CVRF animals with a γ-secretase inhibitor induced a reduction in Hes/Hey gene expression and rescued their angiogenic function in vivo We report for the first time the impact of CVRF burden on the ASCs of EAT and that the defective function is in part caused by increased Notch signaling. Conditioning ASCs by blocking Notch signaling rescues their angiogenic potential.-Bejar, M. T., Ferrer-Lorente, R., Peña, E., Badimon, L. Inhibition of Notch rescues the angiogenic potential impaired by cardiovascular risk factors in epicardial adipose stem cells.


Asunto(s)
Tejido Adiposo/citología , Enfermedades Cardiovasculares/etiología , Neovascularización Patológica/metabolismo , Receptores Notch/metabolismo , Células Madre/fisiología , Animales , Diabetes Mellitus , Regulación de la Expresión Génica/fisiología , Masculino , Miocitos Cardíacos/metabolismo , Obesidad , Ratas , Ratas Zucker , Receptores Notch/genética , Factores de Riesgo , Regulación hacia Arriba
8.
BMC Fam Pract ; 18(1): 74, 2017 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-28633627

RESUMEN

BACKGROUND: Overweight and obesity are common health problems which increase the risk of developing several serious health conditions. The main difficulty in the management of weight-loss lies in its maintenance, once it is achieved. The aim of this study was to investigate whether a motivational intervention, together with current clinical practice, was more efficient than a traditional intervention, in the treatment of overweight and obesity and whether this intervention reduces cardiovascular risk factors associated with overweight and obesity. METHODS: Multi-centre cluster randomized trial with a 24-month follow-up included 864 overweight/obese patients randomly assigned. Motivational intervention group (400 patients), delivered by a nurse trained by an expert psychologist, in 32 sessions, 1 to 12 fortnightly, and 13 to 32, monthly, on top of their standard programmed diet and exercise. The control group (446 patients), received the usual follow-up. RESULTS: Weight reduction was statistically significant in the second year with a mean reduction of 1.0 Kg in the control group and 2.5 Kg in the intervention group (p = 0. 02). While 18.1% of patients in the control group reduced their weight by more than 5%, this percentage rose to 26.9% in the intervention group, which is statistically significant (p = 0.04). Patients in the motivational intervention group had significantly greater improvements in triglycerides and APOB/APOA1ratio. CONCLUSIONS: The results highlight the importance of the group motivational interview in the treatment of overweight /obese patients in primary care, and in the improvement of their associated cardiovascular risks factors. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT01006213 October 30, 2009.


Asunto(s)
Entrevista Motivacional , Obesidad/terapia , Sobrepeso/terapia , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Obesidad/psicología , Sobrepeso/psicología , Atención Primaria de Salud/métodos , Programas de Reducción de Peso/métodos
9.
Arterioscler Thromb Vasc Biol ; 35(2): 348-57, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25425620

RESUMEN

OBJECTIVE: Therapeutic angiogenesis is a promising strategy for treating ischemia. Our previous work showed that endogenous endothelial tissue factor (TF) expression induces intracrine signaling and switches-on angiogenesis in microvascular endothelial cells (mECs). We have hypothesized that activated mECs could exert a further paracrine regulation through the release of TF-rich microvascular endothelial microparticles (mEMPs) and induce neovascularization of ischemic tissues. APPROACH AND RESULTS: Here, we describe for the first time that activated mECs are able to induce reparative neovascularization in ischemic zones by releasing TF-rich microparticles. We show in vitro and in vivo that mEMPs released by both wild-type and TF-upregulated-mECs induce angiogenesis and collateral vessel formation, whereas TF-poor mEMPs derived from TF-silenced mECs are not able to trigger angiogenesis. Isolated TF-bearing mEMPs delivered to nonperfused adductor muscles in a murine hindlimb ischemia model enhance collateral flow and capillary formation evidenced by MRI. TF-bearing mEMPs increase angiogenesis operating via paracrine regulation of neighboring endothelial cells, signaling through the ß1-integrin pathway Rac1-ERK1/2-ETS1 and triggering CCL2 (chemokine [C-C motif] ligand 2) production to form new and competent mature neovessels. CONCLUSIONS: These findings demonstrate that TF-rich mEMPs released by microvascular endothelial cells can overcome the consequences of arterial occlusion and tissue ischemia by promoting postischemic neovascularization and tissue reperfusion.


Asunto(s)
Micropartículas Derivadas de Células/trasplante , Circulación Colateral , Células Endoteliales/trasplante , Isquemia/cirugía , Microvasos/metabolismo , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/metabolismo , Neovascularización Fisiológica , Tromboplastina/metabolismo , Animales , Línea Celular , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/patología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Miembro Posterior , Humanos , Integrina beta1/metabolismo , Isquemia/genética , Isquemia/metabolismo , Isquemia/patología , Isquemia/fisiopatología , Angiografía por Resonancia Magnética , Masculino , Ratones Desnudos , Microvasos/patología , Microvasos/fisiopatología , Músculo Esquelético/patología , Comunicación Paracrina , Interferencia de ARN , Transducción de Señal , Tromboplastina/genética , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Transfección
10.
Eur Heart J ; 36(16): 965-75, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25217443

RESUMEN

AIM: Thrombus formation is a dynamic process regulated by flow, blood cells, and plasma proteins. The present study was performed to investigate the characteristics of human coronary thrombus in ST-segment elevation myocardial infarction (STEMI). METHODS AND RESULTS: Patients admitted with ST-elevation myocardial infarction, in which thrombectomy was performed, were included (n = 86). Intracoronary thrombi and blood from the culprit coronary site and the systemic circulation were obtained during percutaneous coronary intervention (PCI). Thrombi were categorized by onset-of-pain-to-PCI elapsed time in thrombus of <3 (T3) and more than 6 h of evolution (T6). Clinical, morphological, and proteomic variables were investigated. While T3 were mainly composed by platelets and fibrin(ogen), T6 were characterized by a reduced platelet content, increased leucocytes infiltration (including monocytes, neutrophils, T-cells, and B-cells), and appearance of undifferentiated progenitor cells. Significant differences between T3 and T6 were found in the cell cytoskeleton-associated proteome (beta-actin and tropomyosin 3 and 4). By discovery proteomics, we have identified profilin-1 (Pfn-1) in the coronary thrombi and detected higher levels in T3 than in T6. While plasma Pfn-1 levels were low in T3 patients, levels significantly increased in both coronary and peripheral circulation in T6 patients indicating release. In vitro platelet aggregation studies showed that platelets secrete Pfn-1 upon complete activation. CONCLUSION: Coronary thrombi show rapid dynamic changes both in structure and cell composition as a function of elapsed onset-of-pain-to-PCI time. Aged ischaemic thrombi were more likely to have reduced Pfn-1 content releasing Pfn-1 to the circulation. Onset-of-pain-to-PCI elapsed time in STEMI patients and hence age of occlusive thrombus can be profiled by Pfn-1 levels found in the peripheral circulation.


Asunto(s)
Trombosis Coronaria/patología , Infarto del Miocardio/patología , Profilinas/metabolismo , Análisis de Varianza , Plaquetas/patología , Clopidogrel , Trombosis Coronaria/metabolismo , Trombosis Coronaria/terapia , Femenino , Fibrinógeno/metabolismo , Humanos , Leucocitos/patología , Masculino , Persona de Mediana Edad , Infarto del Miocardio/metabolismo , Infarto del Miocardio/terapia , Intervención Coronaria Percutánea , Agregación Plaquetaria/fisiología , Inhibidores de Agregación Plaquetaria/uso terapéutico , Trombectomía/métodos , Trombina/fisiología , Ticlopidina/análogos & derivados , Ticlopidina/uso terapéutico , Tiempo de Tratamiento
11.
Arterioscler Thromb Vasc Biol ; 32(9): 2141-8, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22837468

RESUMEN

OBJECTIVE: Patients with diabetes mellitus have an increased risk of suffering atherothrombotic syndromes and are prone to clustering cardiovascular risk factors. However, despite their dysregulated glucose metabolism, intensive glycemic control has proven insufficient to reduce thrombotic complications. Therefore, we aimed to elucidate the determinants of thrombosis in a model of type 2 diabetes mellitus with cardiovascular risk factors clustering. METHODS AND RESULTS: Intravital microscopy was used to analyze thrombosis in vivo in Zucker diabetic fatty rats (ZD) and lean normoglycemic controls. Bone marrow (BM) transplants were performed to test the contribution of each compartment (blood or vessel wall) to thrombogenicity. ZD showed significantly increased thrombosis compared with lean normoglycemic controls. BM transplants demonstrated the key contribution of the hematopoietic compartment to increased thrombogenicity. Indeed, lean normoglycemic controls transplanted with ZD-BM showed increased thrombosis with normal glucose levels, whereas ZD transplanted with lean normoglycemic controls-BM showed reduced thrombosis despite presenting hyperglycemia. Significant alterations in megakaryopoiesis and platelet-endoplasmic reticulum stress proteins, protein disulfide isomerase and 78-kDa glucose-regulated protein, were detected in ZD, and increased tissue factor procoagulant activity was detected in plasma and whole blood of ZD. CONCLUSIONS: Our results indicate that diabetes mellitus with cardiovascular risk factor clustering favors BM production of hyperreactive platelets with altered protein disulfide isomerase and 78-kDa glucose-regulated protein expression that can contribute to increase thrombotic risk independently of blood glucose levels.


Asunto(s)
Coagulación Sanguínea , Plaquetas/metabolismo , Células de la Médula Ósea/metabolismo , Complicaciones de la Diabetes/etiología , Diabetes Mellitus Tipo 2/complicaciones , Estrés del Retículo Endoplásmico , Retículo Endoplásmico/metabolismo , Proteínas de Choque Térmico/metabolismo , Trombosis/etiología , Animales , Pruebas de Coagulación Sanguínea , Glucemia/metabolismo , Plaquetas/patología , Células de la Médula Ósea/patología , Trasplante de Médula Ósea , Complicaciones de la Diabetes/sangre , Complicaciones de la Diabetes/patología , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/patología , Modelos Animales de Enfermedad , Retículo Endoplásmico/patología , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Activación Plaquetaria , Pruebas de Función Plaquetaria , Proteína Disulfuro Isomerasas/metabolismo , Ratas , Ratas Transgénicas , Ratas Wistar , Ratas Zucker , Tromboplastina/metabolismo , Trombopoyesis , Trombosis/sangre , Trombosis/patología , Factores de Tiempo
12.
Angiogenesis ; 15(4): 657-69, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22869003

RESUMEN

Tissue factor (TF) has well-recognized roles as initiator of blood coagulation as well as an intracellular signaling receptor. TF signaling regulates gene transcription and protein translation. Recently, we have shown that TF-induced mature neovessel formation is ultimately driven by CCL2 expression. However, the signaling process induced by TF to promote microvessel formation remains to be determined. This study was designed with the objective to investigate the mechanisms involved in TF-induced neovessel formation. Here, we have identified that Ets-1 expression is a downstream effector of TF signaling. TF-siRNA induced a highly significant reduction in Ets-1 expression levels and in Ets-1/DNA binding while inducing abrogation of microvessel formation. Activation of Ets-1 rescued the effect of TF inhibition and restored microvessel formation confirming the critical role of Ets-1 in TF-induced angiogenesis. VE-cadherin expression, a key regulator of endothelial intercellular junctions, and an Ets-1 target molecule was dependent of TF-inhibition. We show that TF signals through ERK1/2 to activate Ets-1 and induce CCL2 gene expression by binding to its promoter region. We conclude that endothelial cell TF signals through ERK1/2 and Ets-1 to trigger microvessel formation.


Asunto(s)
Microvasos/fisiología , Proteína Proto-Oncogénica c-ets-1/genética , Tromboplastina/fisiología , Transcripción Genética , Animales , Antígenos CD/metabolismo , Secuencia de Bases , Cadherinas/metabolismo , Línea Celular Transformada , Quimiocina CCL2/metabolismo , Cartilla de ADN , Humanos , Ratones , Ratones Desnudos , Reacción en Cadena en Tiempo Real de la Polimerasa
13.
Arterioscler Thromb Vasc Biol ; 31(11): 2560-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21836071

RESUMEN

OBJECTIVE: Growing insight into the antiplatelet properties of new nitric oxide (NO) donors has expanded their potential use in cardiovascular diseases. As such, we reported that oral administration of a new exogenous NO donor (LA419) induced significant inhibition of platelet deposition on damaged vascular wall without provoking hypotension in an in vivo experimental model. Thrombin is one of the major triggers of platelet deposition and thrombosis on injured vessels; however, the effects of NO on thrombin-induced platelet activation are not fully known. Here, our aim was to investigate the inhibitory effects of exogenous NO administration on the major changes in platelet proteins induced by thrombin. METHODS AND RESULTS: Platelets were obtained from a group of swine orally treated with LA419 (0.9 mg kg(-1)) or placebo for 8 days. Washed platelets were incubated with thrombin (0.4 NIH U/mL). Platelet proteins were then sequentially extracted based on differential solubility and studied by two-dimensional electrophoresis, mass spectrometry (matrix-assisted laser desorption ionization/time of flight), Western blot, and confocal immunofluorescence. NO treatment abrogated thrombin effects on 24 proteins involved in actin assembly, signaling, and metabolic activity. NO treatment prevented thrombin-induced translocation of gelsolin, filamin, 14-3-3ζ, phosphatidylinositol 3-kinase-γ isoform, and growth factor receptor-bound protein 2 (Grb2). CONCLUSION: Our results show that exogenous NO donor treatment renders platelets less sensitive to thrombin activation and inhibits thrombosis by interfering with the platelet shape change machinery.


Asunto(s)
Plaquetas/efectos de los fármacos , Proteínas del Citoesqueleto/metabolismo , Proteína Adaptadora GRB2/metabolismo , Donantes de Óxido Nítrico/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteómica , Transducción de Señal/efectos de los fármacos , Trombina/farmacología , Administración Oral , Animales , Plaquetas/fisiología , Proteínas Contráctiles/metabolismo , Proteínas del Citoesqueleto/efectos de los fármacos , Femenino , Filaminas , Gelsolina/metabolismo , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Dinitrato de Isosorbide/administración & dosificación , Dinitrato de Isosorbide/análogos & derivados , Dinitrato de Isosorbide/farmacología , Proteínas de Microfilamentos/metabolismo , Modelos Animales , Donantes de Óxido Nítrico/administración & dosificación , Inhibidores de las Quinasa Fosfoinosítidos-3 , Transducción de Señal/fisiología , Porcinos
14.
Arterioscler Thromb Vasc Biol ; 31(11): 2607-15, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21868706

RESUMEN

OBJECTIVE: Tissue factor (TF) triggers arterial thrombosis. TF is also able to initiate cellular signaling mechanisms leading to angiogenesis. Because high cardiovascular risk atherosclerotic plaques show significant angiogenesis, our objective was to investigate whether TF is able to trigger and stabilize atherosclerotic plaque neovessel formation. METHODS AND RESULTS: In this study, we showed, by real-time confocal microscopy in 3-dimensional basement membrane cocultures, that TF in human microvascular endothelial cells (HMEC-1) and in human vascular smooth muscle cells (HVSMCs) plays an important role in the formation of capillary-like networks. TF silencing in endothelial cells and smooth muscle cells inhibits the formation of tube-like structures with stable phenotype. Using an in vivo model, we observed that TF inhibition in either HMEC-1 or HVSMCs reduced their shared ability to form new capillaries. The phenotypic changes induced by TF silencing were linked to reduced chemokine (C-C motif) ligand 2 (CCL2) expression in endothelial cells. Wound healing and chemotactic assays demonstrated that TF-induced release of CCL2 stimulated HVSMC migration to HMEC-1. CONCLUSION: Endogenous TF regulates CCL2 production in endothelial cells. Secreted CCL2 mediates the angiogenic effect of TF by recruiting smooth muscle cells toward endothelial cells and facilitates the maturation of newly formed microvessels.


Asunto(s)
Quimiocina CCL2/metabolismo , Microvasos/metabolismo , Neovascularización Fisiológica/fisiología , Tromboplastina/metabolismo , Técnicas de Cocultivo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/fisiopatología , Transducción de Señal/fisiología
15.
Cardiovasc Res ; 118(10): 2354-2366, 2022 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34406379

RESUMEN

AIMS: Despite increasing evidence that monocytes may acquire endothelial features, it remains unclear how monocytes participate in angiogenesis after ischaemic damage. We investigated whether ischaemic cells can release microvesicles (MVs) and promote neovascularization in a model of peripheral artery disease (PAD). METHODS AND RESULTS: To model PAD, we used an in vivo experimental model of hind-limb ischaemia (HLI) in mice. MVs were isolated from the ischaemic muscle and from peripheral blood at different times after unilateral femoral artery ligation. MVs were phenotypically characterized to identify cell origin. HLI in mice induced the release of MVs with a much higher content of tissue factor (TF) than non-HLI control mice both in the MVs isolated from the affected limb muscle area and from blood. MVs were mainly released from endothelial cells (ECs) and induced Mo differentiation to endothelial cell-like (ECL) cells. Differentiation to ECL cells encompassed highly strict hierarchical transcription factor activation, initiated by ETS1 activation. MVs secreted by microvascular ECs over-expressing TF (upTF-EMVs), were injected in the ischaemic hind-limb in parallel with control EMVs (from random siRNA-treated cells) or EMVs released by silenced TF ECs. In animals treated with upTF-EMVs in the ischaemic zone, there was a highly significant increase in functional new vessels formation (seen by magnetic resonance angiography), a concomitant increase in the pool of circulating Ly6Clow Mo expressing vascular EC markers, and a significantly higher number of Mo/macrophages surrounding and integrating the newly formed collaterals. CONCLUSION: Ischaemia-activated ECs release EMVs rich in TF that induce monocyte differentiation into ECL cells and the formation of new vessels in the ischaemic zone. TF by this mechanism of formation of new blood microvessels can contribute to ischaemic tissue repair.


Asunto(s)
Micropartículas Derivadas de Células , Tromboplastina , Animales , Células Endoteliales , Isquemia , Ratones , Monocitos
16.
Front Immunol ; 13: 836662, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35251029

RESUMEN

We have recently shown that in ischemic tissue, the hypoxic endothelial cells (EC) release extracellular microvesicles (EMVs) that are rich in tissue factor (TF). These TF-EMVs induce monocyte (Mo) homing to the ischemic zone, their differentiation into EC-like cells, and the formation of new blood vessels increasing tissue perfusion. In addition to membrane proteins, EMVs contain noncoding RNAs that can modulate cellular signaling pathways in the recipient cells. Here, we have investigated whether miRNA contained into secreted EMVs may be transferred into Mo where they could modulate EC-like cell differentiation and angiogenic responses. Our results indicated that EMVs released from activated ECs contain high levels of miR-126 and that the levels are directly proportional to TF expression in EMVs. Interestingly, miR-126 is transferred to Mo when they are incubated with TF-EMVs. Increased levels of miR-126 in Mo do not promote EC-like cell differentiation but regulate angiogenesis by targeting several components of the VEGF pathway, as SPRED1 and PI3KR2. Our findings reveal that activated ECs secrete EMVs carrying miR-126, which can modulate Mo reprogramming of angiogenic genes.


Asunto(s)
Micropartículas Derivadas de Células , MicroARNs , Micropartículas Derivadas de Células/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Monocitos/metabolismo , Tromboplastina/metabolismo
17.
J Mol Cell Cardiol ; 50(3): 522-33, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21219908

RESUMEN

The extent of cardiac remodeling determines survival after acute MI. However, the mechanisms driving cardiac remodeling remain unknown. We examined the effect of ischemia and reperfusion (R) on myocardial changes up to 6 days post-MI. Pigs underwent 1.5h or 4h mid-LAD balloon occlusion and sacrificed or 1.5h occlusion followed by R and sacrificed at 2.5h, 1 day, 3 days, and 6 days. Ischemic- (IM) and non-ischemic myocardium (NIM) was obtained for molecular analysis of: 1) apoptosis (P-Bcl2, Bax, P-p53, active-caspase-3); 2) the TLR-4-MyD88-dependent and independent pathways; 3) Akt/mTOR/P70(S6K) axis activation; and, 4) fibrosis (TGF-ß, collagen1-A1/A3). Histopathology for inflammation, collagen, and fibroblast content, TUNEL staining, and metalloproteinase activity was performed. Apoptosis is only detected upon R in IM cardiomyocytes and progresses up to 6 days post-R mainly associated with infiltrated macrophages. The Akt/mTOR/P70(s6K) pathway is also activated upon R (IM) and remains elevated up to 6 days-R (P<0.05). Ischemia activates the TLR-4-MyD88-dependent (cytokines/chemokines) and -independent (IRF-3) pathways in IM and NIM and remains high up to 6 days post-R (P<0.05). Accordingly, leukocytes and macrophages are progressively recruited to the IM (P<0.05). Ischemia up-regulates pro-fibrotic TGF-ß that gradually rises collagen1-A1/-A3 mRNA with subsequent increase in total collagen fibrils and fibroblasts from 3 days-R onwards (P<0.005). MMP-2 activity increases from ischemia to 3 days post-R (P<0.05). We report that there is a timely coordinated cellular and molecular response to myocardial ischemia and R within the first 6 days after MI. In-depth understanding of the mechanisms involved in tissue repair is warranted to timely intervene and better define novel cardioprotective strategies.


Asunto(s)
Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Remodelación Ventricular/fisiología , Animales , Apoptosis/fisiología , Colágeno/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Etiquetado Corte-Fin in Situ/métodos , Inflamación/fisiopatología , Leucocitos/patología , Macrófagos/patología , Metaloproteinasa 2 de la Matriz/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Porcinos , Serina-Treonina Quinasas TOR/metabolismo , Receptor Toll-Like 4/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
18.
Arterioscler Thromb Vasc Biol ; 30(6): 1246-52, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20360535

RESUMEN

OBJECTIVE: To investigate the effect of rosuvastatin on platelet deposition under controlled shear rate conditions and to identify new platelet proteins involved in the interaction with the activating substrate. METHODS AND RESULTS: Platelet-vessel wall interaction and thrombosis take place under dynamic conditions involving the interaction of the exposed damaged vascular wall with the circulating blood cells and proteins. Blood was perfused over type I collagen at different wall shear rates, and platelet deposition was measured by confocal microscopy. Perfused effluent blood was collected, platelets were sequentially extracted based on differential protein solubility, and proteins were separated by 2D gel electrophoresis. Blockade of 3-hydroxy-3-methylglutaryl-coenzyme A reductase significantly reduced platelet deposition and modulated the expression pattern of 18 proteins in the platelet subproteome. Among them, an increase in platelet surface 78-kDa glucose-regulated protein (GRP78), a stress-inducible multifunctional endoplasmic reticulum protein, was clearly apparent. Immunoprecipitation of platelet GRP78 revealed its interaction with tissue factor. Moreover, blockade of surface GRP78 resulted in a substantial increase in platelet deposition and tissue factor procoagulant activity and in a decrease in clotting time. CONCLUSIONS: These findings demonstrate that blockade of 3-hydroxy-3-methylglutaryl-coenzyme A reductase reduces platelet deposition and inhibits GRP78 translocation from the platelet surface after shear and collagen activation. For the first time to our knowledge, this study reports on the presence and functional role of GRP78 in platelets and indicates that GRP78 has additional functions beyond those of a molecular chaperone.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Colágeno Tipo I/metabolismo , Fluorobencenos/farmacología , Proteínas de Choque Térmico/sangre , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Adhesividad Plaquetaria/efectos de los fármacos , Pirimidinas/farmacología , Sulfonamidas/farmacología , Animales , Plaquetas/metabolismo , Relación Dosis-Respuesta a Droga , Electroforesis en Gel Bidimensional , Chaperón BiP del Retículo Endoplásmico , Humanos , Inmunoprecipitación , Microscopía Confocal , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Transporte de Proteínas , Proteómica/métodos , Flujo Sanguíneo Regional , Rosuvastatina Cálcica , Estrés Mecánico , Porcinos , Tromboplastina/metabolismo
19.
Cardiovasc Res ; 117(9): 2054-2068, 2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-32991689

RESUMEN

AIMS: Atherosclerosis, the leading cause of cardiovascular diseases, is driven by high blood cholesterol levels and chronic inflammation. Low-density lipoprotein receptors (LDLR) play a critical role in regulating blood cholesterol levels by binding to and clearing LDLs from the circulation. The disruption of the interaction between proprotein convertase subtilisin/kexin 9 (PCSK9) and LDLR reduces blood cholesterol levels. It is not well known whether other members of the LDLR superfamily may be targets of PCSK9. The aim of this work was to determine if LDLR-related protein 5 (LRP5) is a PCSK9 target and to study the role of PCSK9 and LRP5 in foam cell formation and lipid accumulation. METHODS AND RESULTS: Primary cultures of human inflammatory cells (monocytes and macrophages) were silenced for LRP5 or PCSK9 and challenged with LDLs. We first show that LRP5 is needed for macrophage lipid uptake since LRP5-silenced macrophages show less intracellular CE accumulation. In macrophages, internalization of LRP5-bound LDL is already highly evident after 5 h of LDL incubation and lasts up to 24 h; however, in the absence of both LRP5 and PCSK9, there is a strong reduction of CE accumulation indicating a role for both proteins in lipid uptake. Immunoprecipitation experiments show that LRP5 forms a complex with PCSK9 in lipid-loaded macrophages. Finally, PCSK9 participates in TLR4/NFkB signalling; a decreased TLR4 protein expression levels and a decreased nuclear translocation of NFκB were detected in PCSK9 silenced cells after lipid loading, indicating a downregulation of the TLR4/NFκB pathway. CONCLUSION: Our results show that both LRP5 and PCSK9 participate in lipid uptake in macrophages. In the absence of LRP5, there is a reduced release of PCSK9 indicating that LRP5 also participates in the mechanism of release of soluble PCSK9. Furthermore, PCSK9 up-regulates TLR4/NFκB favouring inflammation.


Asunto(s)
Aterosclerosis/enzimología , Inflamación/enzimología , Metabolismo de los Lípidos , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Macrófagos/enzimología , Monocitos/enzimología , Proproteína Convertasa 9/metabolismo , Aterosclerosis/genética , Aterosclerosis/inmunología , Transporte Biológico , Células Cultivadas , Colesterol/metabolismo , Células Espumosas/enzimología , Células Espumosas/inmunología , Humanos , Inmunidad Innata , Inflamación/genética , Inflamación/inmunología , Lipoproteínas LDL/metabolismo , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Macrófagos/inmunología , Monocitos/inmunología , FN-kappa B/metabolismo , Proproteína Convertasa 9/genética , Transducción de Señal , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Receptor Toll-Like 4/metabolismo , Proteína Wnt3A
20.
Curr Pharm Des ; 27(29): 3186-3197, 2021 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-33213315

RESUMEN

Cardiovascular diseases (CVD) remain the world's leading cause of death and disability in both men and women, but with different prognostics and outcomes between sexes. Although the burden of CVD is generally related to the conventional risk factors, the relevance of non-traditional risk factors is increasingly being recognized to explain the so-called "residual risk". Men and women share many similarities regarding classical cardiovascular risk factors but have different disease pathophysiology, clinical presentations, prevalence, and outcomes of CVDs. How sex-specificities regarding the effects of non-traditional risk factors may contribute to the evolution of atherosclerosis and its clinical manifestations in males and females remain largely underanalyzed. The present review summarizes the current knowledge for sex differences in atherosclerotic plaque composition and clinical evolution in association with risk factors, such as inflammation, lipoprotein(a), hemostasis, intraplaque calcification, and depression. We further discuss the potential sex-differential impact of chronic infectious diseases, gut microbiome and, epigenetic gene expression regulation for atherosclerosis and the effect of female-specific disorders in CVD.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Aterosclerosis/epidemiología , Femenino , Humanos , Masculino , Factores de Riesgo , Caracteres Sexuales , Factores Sexuales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA