Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Int J Mol Sci ; 22(10)2021 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-34066088

RESUMO

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.


Assuntos
Aterosclerose/patologia , Complemento C3/metabolismo , Hiperlipoproteinemia Tipo II/patologia , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Proteoma/metabolismo , Adulto , Aterosclerose/imunologia , Aterosclerose/metabolismo , Estudos de Casos e Controles , Adesão Celular , Células Cultivadas , Feminino , Humanos , Hiperlipoproteinemia Tipo II/imunologia , Hiperlipoproteinemia Tipo II/metabolismo , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/imunologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/imunologia , Miócitos de Músculo Liso/metabolismo , Proteoma/análise , Remodelação Vascular , Cicatrização , Adulto Jovem
2.
Sci Rep ; 11(1): 10545, 2021 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-34006889

RESUMO

The aims of this study were to evaluate the feasibility of the New Zealand White (NZW) rabbit for studying implanted biomaterials in pelvic reconstructive surgery; and to compare the occurrence of graft-related complications of a commercial polypropylene (PP) mesh and new developed human dermal matrix implanted at vaginal and abdominal level. 20 white female NZW rabbits were randomized into two groups, experimental group (human acellular dermal matrices-hADM-graft) and control group (commercial PP graft). In each animal, grafts were surgically implanted subcutaneously in the abdominal wall and in the vaginal submucosa layer for 180 days. The graft segments were then removed and the surgical and clinical results were analyzed. The main surgical challenges during graft implantation were: (a) an adequate vaginal exposure while maintaining the integrity of the vaginal mucosa layer; (b) to keep aseptic conditions; (c) to locate and dissect the breast vein abdominal surgery; and (d) to withdraw blood samples from the ear artery. The most abnormal findings during the explant surgery were found in the PP group (33% of vaginal mesh extrusion) in comparison with the hADM group (0% of vaginal graft extrusion), p = 0.015. Interestingly, macroscopic observation showed that the integration of the vaginal grafts was more common in the hADM group (40%) than in the PP group, in which the vaginal mesh was identified in 100% of the animals (p = 0.014). The NZW rabbit is a good model for assessing materials to be used as grafts for pelvic reconstructive surgery and vaginal surgery. Animals are easily managed during the procedures, including surgical intervention and vaginal mucosa approach. Additionally, hADM is associated with fewer clinical complications, as well as better macroscopic tissue integration, compared to PP mesh.


Assuntos
Diafragma da Pelve/fisiopatologia , Diafragma da Pelve/cirurgia , Animais , Materiais Biocompatíveis , Modelos Animais de Doenças , Feminino , Coelhos
3.
Stem Cell Res Ther ; 10(1): 361, 2019 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-31783922

RESUMO

BACKGROUND: The composition and function of the adipose tissue covering the heart are poorly known. In this study, we have investigated the epicardial adipose tissue (EAT) covering the cardiac ventricular muscle and the EAT covering the left anterior descending artery (LAD) on the human heart, to identify their resident stem cell functional activity. METHODS: EAT covering the cardiac ventricular muscle was isolated from the apex (avoiding areas irrigated by major vessels) of the heart (ventricular myocardium adipose tissue (VMAT)) and from the area covering the epicardial arterial sulcus of the LAD (PVAT) in human hearts excised during heart transplant surgery. Adipose stem cells (ASCs) from both adipose tissue depots were immediately isolated and phenotypically characterized by flow cytometry. The different behavior of these ASCs and their released secretome microvesicles (MVs) were investigated by molecular and cellular analysis. RESULTS: ASCs from both VMAT (mASCs) and the PVAT (pASCs) were characterized by the expression of CD105, CD44, CD29, CD90, and CD73. The angiogenic-related genes VEGFA, COL18A1, and TF, as well as the miRNA126-3p and miRNA145-5p, were analyzed in both ASC types. Both ASCs were functionally able to form tube-like structures in three-dimensional basement membrane substrates. Interestingly, pASCs showed a higher level of expression of VEGFA and reduced level of COL18A1 than mASCs. Furthermore, MVs released by mASCs significantly induced human microvascular endothelial cell migration. CONCLUSION: Our study indicates for the first time that the resident ASCs in human epicardial adipose tissue display a depot-specific angiogenic function. Additionally, we have demonstrated that resident stem cells are able to regulate microvascular endothelial cell function by the release of MVs.


Assuntos
Tecido Adiposo/citologia , Expressão Gênica , Células-Tronco/metabolismo , Movimento Celular , Micropartículas Derivadas de Células/metabolismo , Colágeno Tipo VIII/genética , Colágeno Tipo VIII/metabolismo , Colágeno Tipo XVIII , Vasos Coronários/citologia , Meios de Cultivo Condicionados/farmacologia , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Pericárdio/citologia , Células-Tronco/citologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
Circ Res ; 125(1): 74-89, 2019 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-31219744

RESUMO

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.


Assuntos
Adipócitos/metabolismo , Diferenciação Celular/fisiologia , Células Endoteliais/metabolismo , MicroRNAs/metabolismo , Microvasos/metabolismo , Neovascularização Fisiológica/fisiologia , Adipócitos/patologia , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Células Cultivadas , Células Endoteliais/patologia , Células HeLa , Humanos , Isquemia/metabolismo , Isquemia/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , MicroRNAs/antagonistas & inibidores , Microvasos/patologia
5.
PLoS One ; 14(6): e0218098, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31220102

RESUMO

AIMS: Myocardial infarction induces myocardial injury and tissue damage. During myocardial infarction strong cellular response is initiated to salvage the damaged tissues. This response is associated with the induction of different signaling pathways. Of these, the canonical Wnt signaling is increasingly important for its prosurvival cellular role, making it a good candidate for the search of new molecular targets to develop therapies to prevent heart failure in infarcted patients. METHODS: Herein we report that GSK3ß regulates the canonical Wnt signaling in C57Bl6 mice hearts. GSK3ß is a canonical Wnt pathway inhibitor. Using GSK3ß inhibitors and inducing myocardial injury (MI) in Lrp5-/- mice model we show that GSK3ß phosphorylation levels regulate downstream canonical Wnt pathway genes in the ischemic heart. In the setting of MI, myocardial damage assessment usually correlates with functional and clinical outcomes. Therefore, we measured myocardial injury size in Wt and Lrp5-/- mice in the presence and absence of two different GSK3 inhibitors prior to MI. Myocardial injury was independent of GSK3 inhibitor treatments and GSK3ß expression levels. RESULTS: These studies support a central role for GSK3ß in the activation of the canonical Wnt pathway in the Wt heart. Although LRP5 is protective against myocardial injury, GSK3ß expression levels do not regulate heart damage.


Assuntos
Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Miocárdio/metabolismo , Via de Sinalização Wnt , Animais , Expressão Gênica , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Isquemia Miocárdica/tratamento farmacológico , Isquemia Miocárdica/enzimologia , Miocárdio/enzimologia , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Piridinas/uso terapêutico , Pirimidinas/uso terapêutico , Via de Sinalização Wnt/genética
7.
Int J Cardiol ; 270: 28-35, 2018 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-29936043

RESUMO

AIMS: Milk thistle (Silybum marianum; SM) is an herb commonly used for hepatoprotection with antioxidant and antifibrotic properties. We investigated in pigs the cardiac effects of SM intake during the acute phase of myocardial infarction (MI) and remodeling period post-MI. METHODS: Study-1 tested the effect of SM use on the acute phase of MI. Hence, animals were distributed to a control group or to receive SM prior infarction (1.5 h ischemia). Animals were sacrificed after 2.5 h of reperfusion. Study-2 tested the effect of SM use in the cardiac remodeling phase. Accordingly, animals received for 10 d diet ±â€¯SM prior MI and followed the same regime for 3 weeks and then sacrificed. Study-3 tested the effect of SM in a non-infarcted heart; therefore, animals received for 10 d diet ±â€¯SM and then sacrificed. RESULTS: Animals taking SM before MI showed a reduction in cardiac damage (decreased oxidative damage, ROS production and xanthine oxidase levels; preserved mitochondrial function; and increased myocardial salvage; p < 0.05) versus controls. Animals that remained on chronic SM intake post-MI improved left ventricular remodeling. This was associated with the attenuation of the TGFß1/TßRs/SMAD2/3 signaling, lower myofibroblast transdifferentiation and collagen content in the border zone (p < 0.05 vs. all other groups). Cardiac contractility improved in animals taking SM (p < 0.05 vs. post-MI-control). No changes in cardiac function or fibrosis were detected in animals on SM but without MI. CONCLUSION: Intake of SM protects the heart against the deleterious effects of an MI and favors cardiac healing. These benefits may be attributed to the antioxidant and antifibrotic properties of SM.


Assuntos
Cardiotônicos/uso terapêutico , Infarto do Miocárdio/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/uso terapêutico , Silybum marianum , Remodelação Ventricular/efeitos dos fármacos , Animais , Cardiotônicos/isolamento & purificação , Cardiotônicos/farmacologia , Células Cultivadas , Fibrose , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Estresse Oxidativo/fisiologia , Extratos Vegetais/isolamento & purificação , Extratos Vegetais/farmacologia , Suínos , Remodelação Ventricular/fisiologia
8.
Thromb Haemost ; 117(9): 1816-1827, 2017 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-28640320

RESUMO

The clinical impact of in-stent thrombosis is high because it is associated with high mortality and 20 % of the patients suffer a recurrent event within the two following years. The aim of this study was to characterise the morphologic and proteomic profile of in-stent thrombi (IST) in comparison to thrombi developed on native coronary arteries (CT) to identify a differential molecular signature. The study included 45 patients with ST-elevation-myocardial-infarction (STEMI) treated by primary-percutaneous-intervention and thrombus aspiration: 21 had IST and 24 had CT. Thrombi were characterised by morphologic immunohistochemical analysis and differential proteomic profiling (2-DE+MALDI-TOF/TOF). Bioinformatic analysis revealed differences in proteins related to oxidative-stress and cell death/survival. IST showed a higher content of structural proteins (gelsolin, actin-cytoplasmic-1, tropomyosin, and myosin) together with an imbalance in redox-homeostasis related proteins (increased superoxide-dismutase and decreased peroxiredoxin-2 thrombus content), and a coordinated increase of chaperones (HSP60 and HSC70) and cellular quality control-related proteins (26S-protease-regulatory-subunit-7). These changes were reflected into a significant decrease in HSC70 systemic levels and a significant increase in advanced-oxidation-protein-products (AOPP) indicative of increased oxidative stress-mediated protein damage in IST. Our results reveal an imbalance in redox-related proteins indicative of an exacerbated oxidative-stress that leads to an accumulation of AOPP serum levels in IST. Moreover, the coordinated increase in chaperones and regulatory proteins reflects the activation of intracellular protection mechanisms to maintain protein integrity in IST. The failure to counterbalance the stress situation could trigger cellular apoptosis leading to the destabilization of the thrombus and to a worse prognosis of IST-STEMI-patients.


Assuntos
Trombose Coronária/metabolismo , Imunidade Inata , Estresse Oxidativo , Intervenção Coronária Percutânea/instrumentação , Infarto do Miocárdio com Supradesnível do Segmento ST/terapia , Stents , Biomarcadores/metabolismo , Biologia Computacional , Trombose Coronária/diagnóstico por imagem , Trombose Coronária/etiologia , Trombose Coronária/terapia , Feminino , Fibrinogênio/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Chaperonas Moleculares/metabolismo , Intervenção Coronária Percutânea/efeitos adversos , Proteômica/métodos , Infarto do Miocárdio com Supradesnível do Segmento ST/diagnóstico por imagem , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Trombectomia , Resultado do Tratamento
9.
Int J Cardiol ; 215: 406-16, 2016 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-27128573

RESUMO

AIMS: Molecular chaperones constitute protectors of intracellular protein integrity and seem to confer short-term defence against various cell insults. Myocardial damage is associated to a loss of protective chaperones. Ischemic post-conditioning (IPost-Co) is a procedure that seems to protect against reperfusion injury. However, little is known on alpha-crystallin-B-chain (cryab/HspB5) evolution in IPost-Co. Here we have investigated cryab in myocardial ischemia and IPost-Co. METHODS AND RESULTS: Pigs underwent closed-chest 1.5h mid-left anterior descending (LAD) balloon occlusion and were either sacrificed without reperfusion (I;N=10), subjected to 2.5h of reperfusion and sacrificed (I/R; N=5); or subjected to IPost-Co before reperfusion and sacrificed 2.5h afterwards (IPost-Co; N=5). A sham-operated group was included (N=6). Proteomic analysis (2-D-electrophoresis/MALDI-TOF/TOF) revealed cryab as a single spot (20kDa; pI7.6). Myocardial cryab-20-protein and cryab-gene expression levels were decreased after ischemia and I/R(P<0.05). After IPost-Co, cryab-20-protein and cryab-gene expression levels were similar to those found in the heart of sham-operated animals (P<0.05). There was a direct correlation between LVEF-improvement after IPost-Co and myocardial cryab-20-protein levels. In a mice proof-of-principle study, cryab-20-peptide was synthesized and administered 1h before LAD-ligation and ECG-proven MI. A 59% reduction in infarct size was achieved in cryab-20-treated animals (P<0.05). CONCLUSIONS: Ischemia and reperfusion induce a decrease in myocardial cryab-20-protein levels together with a clinical impairment of cardiac function. IPost-Co induces a clinical improvement of cardiac function and a preservation of cryab-20 levels. Intervention studies on a mice-MI model showed that cryab-20-peptide administration reduces infarct size. All together our results show a significant cardioprotective effect of cryab.


Assuntos
Pós-Condicionamento Isquêmico/métodos , Isquemia Miocárdica/terapia , alfa-Cristalinas/farmacologia , Animais , Cardiotônicos/farmacologia , Hipóxia Celular/fisiologia , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos C3H , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Proteômica/métodos , Distribuição Aleatória , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Suínos
10.
Eur Heart J ; 36(16): 965-75, 2015 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-25217443

RESUMO

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.


Assuntos
Trombose Coronária/patologia , Infarto do Miocárdio/patologia , Profilinas/metabolismo , Análise de Variância , Plaquetas/patologia , Clopidogrel , Trombose Coronária/metabolismo , Trombose Coronária/terapia , Feminino , Fibrinogênio/metabolismo , Humanos , Leucócitos/patologia , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/terapia , Intervenção Coronária Percutânea , Agregação Plaquetária/fisiologia , Inibidores da Agregação Plaquetária/uso terapêutico , Trombectomia/métodos , Trombina/fisiologia , Ticlopidina/análogos & derivados , Ticlopidina/uso terapêutico , Tempo para o Tratamento
11.
Toxicol Pathol ; 43(4): 569-80, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25311372

RESUMO

Drug-induced vascular injury (DIVI) is commonly associated with phosphodiesterase (PDE) inhibitors. Despite histological characterization, qualified biomarkers for DIVI detection are lacking. We investigated whether a single administration of roflumilast (PDE-IV inhibitor) induces vascular damage and identified novel surrogate biomarkers of acute vascular injury. Pigs received postoperative 250, 375, or 500 µg of roflumilast or placebo/control. After 1.5 hr, coronary reactivity was determined by catheter-based administration of acetylcholine and sodium nitroprusside (SNP) in the coronary sinus. Immunohistochemical analysis of vessel integrity (von Willebrand factor [vWF]) and fibrin(ogen) deposition was performed in the coronary artery and aorta. Peripheral blood was collected for differential proteomics and microparticles analysis. Circulating interleukin (IL)-6 was analyzed. Roflumilast-treated animals displayed higher vasodilation to acetylcholine and SNP versus controls (p < .05). Roflumilast-treated animals showed a dose-dependent (p < .05) decrease in vessel integrity and dose-dependent increase in fibrin deposition forming a continuous layer at roflumilast-500 µg. Peripheral blood of roflumilast-500-µg-treated animals showed increased levels of total and endothelial-derived microparticles and exhibited a coordinated change in proteins kininogen-1, endothelin-1, gelsolin, apolipoprotein A-I, and apolipoprotein-J associated with vascular injury (p < .05 vs. controls). IL-6 remained unaltered. Roflumilast-induced vascular injury can be detected by novel markers in peripheral blood. Validation of these surrogate markers in human samples seems required.


Assuntos
Aminopiridinas/toxicidade , Benzamidas/toxicidade , Micropartículas Derivadas de Células/efeitos dos fármacos , Proteoma/efeitos dos fármacos , Lesões do Sistema Vascular/sangue , Lesões do Sistema Vascular/induzido quimicamente , Animais , Biomarcadores/sangue , Proteínas Sanguíneas/análise , Proteínas Sanguíneas/metabolismo , Micropartículas Derivadas de Células/metabolismo , Ciclopropanos/toxicidade , Feminino , Interleucina-6/sangue , Inibidores da Fosfodiesterase 4/toxicidade , Proteoma/análise , Proteoma/metabolismo , Proteômica/métodos , Suínos
12.
Int J Cardiol ; 175(3): 528-38, 2014 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-25023790

RESUMO

BACKGROUND: Studies in patients support a beneficial effect of statin treatment early after acute coronary syndrome and/or prior percutaneous coronary intervention. However, statin effect during total occlusion remains unknown. OBJECTIVES: To investigate whether infusion of activated simvastatin during ischemia and prior reperfusion and oral administration thereafter confers cardioprotection and improves cardiac healing in a preclinical model of myocardial infarction. METHODS: Pigs (n=24) fed a 10 day Western-type diet underwent a 90 min coronary-balloon occlusion (MI) being randomized to a single intravenous infusion of active ß-hydroxy acid derivative of simvastatin (ß-OH-S; 0.3 mg/kg) 15 min prior to reperfusion or vehicle. Animals were either sacrificed 2.5 h post-reperfusion or kept under the same regime ± simvastatin (p.o., 20 mg/day) for 3 weeks. Jeopardized and remote myocardium was obtained for molecular/histological studies. Echocardiography was assessed. RESULTS: ß-OH-S infusion prior to reperfusion reduced coronary and cardiac oxidative DNA-damage, diminished neutrophil infiltration at the site of ischemia, preserved mitochondrial membrane potential and reduced apoptosis in the ischemic myocardium (lower mRNA levels of Fas, casp8, p53, and casp3 and mitochondrial-p-Bcl2; and reduced TUNEL and active caspase-3; p<0.05 vs. vehicle/control). This treatment regime attenuated reperfusion-related arrhythmias and stunning leading to a 40% increased myocardial salvage (p<0.05 vs. vehicle/control). 3 weeks post-MI simvastatin-treated animals showed P-PKCε increase, lower intramyocardial lipotoxicity, TßRII/Smad2/3 signaling restoration and subsequent myofibroblast differentiation and collagen-fibril formation in the evolving scar (p<0.05 vs. control). Simvastatin suppressed cardiac RhoA mobilization and triggered Akt/eNOS signaling. CONCLUSIONS: Acute HMG-CoA-reductase inhibition during total ischemia and prior reperfusion limits reperfusion injury and prolonged oral simvastatin treatment thereafter improves cardiac healing post-MI.


Assuntos
Modelos Animais de Doenças , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Infarto do Miocárdio/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Animais , Colesterol na Dieta/efeitos adversos , Feminino , Fibrose/tratamento farmacológico , Fibrose/patologia , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/etiologia , Traumatismo por Reperfusão Miocárdica/patologia , Suínos
13.
Atherosclerosis ; 224(2): 368-76, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22882904

RESUMO

OBJECTIVES: Lipids have been detected in the ischemic myocardium of patients' post-myocardial infarction (MI). However, their effect on the cardiac healing process remains unknown. We investigated whether intramyocardial lipids affect the signaling pathways involved in the fibrotic reparative response impairing cardiac healing post-MI. METHODS: Pigs, fed either a high-cholesterol diet (HC) or a regular-chow (NC), were subjected to experimentally-induced acute MI (90 min mid-LAD balloon occlusion) and then, upon reperfusion (R), maintained for 21 days with the same diet regime (HC/R(+) and NC/R(+), respectively). A group of hypercholesterolemic animals were sacrificed after ischemia without reperfusion (HC/R(-)). Cardiac tissue was obtained for molecular/cellular/histological analysis. Infarct size and echocardiography were assessed. RESULTS: At the time of acute MI, hypercholesterolemic animals showed a higher incidence of ventricular dysrhythmias. At sacrifice, intramyocardial lipids were absent in HC/R(-). HC/R(+) showed higher lipid content (ApoB, cholesteryl-ester and triglycerides) and lower expression/activity of the TGFß/TßRII/Smad2/3 pathway (involved in scar reparative fibrosis) than NC/R(+) in the forming scar. Collagen synthesis was accordingly reduced in the scar of HC/R(+). Infarct size was 44% larger in HC/R(+) which had higher apoptosis and lower Akt/eNOS activity in the jeopardized myocardium. Systolic function was similarly deteriorated post-MI in all animals whereas no changes were detected in diastolic-related parameters. No changes were detected in systolic parameters 21 days post-MI in NC/R(+) animals. In contrast, both systolic- and diastolic-related parameters were further deteriorated in HC/R(+) animals. CONCLUSION: Intramyocardial lipid accumulation impairs TGFß/TßRII/Smad2/3 signaling altering the fibrotic reparative process of the scar resulting in larger infarcts and cardiac dysfunction.


Assuntos
Cicatriz/metabolismo , Metabolismo dos Lipídeos , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Miofibroblastos/metabolismo , Cicatrização , Animais , Apolipoproteínas B/metabolismo , Apoptose , Ésteres do Colesterol/metabolismo , Cicatriz/etiologia , Cicatriz/patologia , Cicatriz/fisiopatologia , Colágeno/biossíntese , Modelos Animais de Doenças , Hipercolesterolemia/complicações , Hipercolesterolemia/metabolismo , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Miofibroblastos/patologia , Óxido Nítrico Sintase Tipo III/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Suínos , Fatores de Tempo , Fator de Crescimento Transformador beta/metabolismo , Triglicerídeos/metabolismo , Fibrilação Ventricular/etiologia , Fibrilação Ventricular/metabolismo , Função Ventricular Esquerda
14.
J Transl Med ; 10: 160, 2012 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-22873206

RESUMO

Our hypothesis was that overexpression of certain lipoprotein receptors might be related to lipid accumulation in the human ischemic myocardium. Intramyocardial lipid overload contributes to contractile dysfunction and arrhythmias in cardiomyopathy. Thus, the purpose of this study was to assess the effect of hypercholesterolemic LDL and hypertrigliceridemic VLDL dose on LRP1 expression in cardiomyocytes, as well as the potential correlation between LRP1 expression and neutral lipid accumulation in the left ventricle tissue from ischemic cardiomyopathy patients. Cell culture experiments include control and LRP1-deficient cardiomyocytes exposed to lipoproteins under normoxic and hypoxic conditions. Explanted hearts from 18 ICM patients and eight non-diseased hearts (CNT) were included. Low density lipoprotein receptor-related protein 1 (LRP1), very low density lipoprotein receptor (VLDLR) and low density lipoprotein receptor (LDLR) expression was analyzed by real time PCR and Western blotting. Cholesteryl ester (CE), triglyceride (TG) and free cholesterol (FC) content was assess by thin layer chromatography following lipid extraction. Western blotting experiments showed that protein levels of LRP1, VLDLR and HIF-1α were significantly upregulated in ischemic hearts. Immunohistochemistry and confocal microscopy analysis showed that LRP1 and HIF-1α were upregulated in cardiomyocytes of ICM patients. In vitro studies showed that VLDL, LDL and hypoxia exerted an upregulatory effect on LRP1 expression and that LRP1 played a major role in cholesteryl ester accumulation from lipoproteins in cardiomyocytes. Myocardial CE accumulation strongly correlated with LRP1 levels in ischemic hearts. Taken together, our results suggest that LRP1 upregulation is key for myocardial cholesterol ester accumulation in ischemic human hearts and that LRP1 may be a target to prevent the deleterious effects of myocardial cholesterol accumulation in ischemic cardiomyopathy.


Assuntos
Ésteres do Colesterol/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Isquemia Miocárdica/metabolismo , Animais , Western Blotting , Linhagem Celular , Feminino , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase em Tempo Real
15.
Arterioscler Thromb Vasc Biol ; 31(11): 2607-15, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21868706

RESUMO

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.


Assuntos
Quimiocina CCL2/metabolismo , Microvasos/metabolismo , Neovascularização Fisiológica/fisiologia , Tromboplastina/metabolismo , Técnicas de Cocultura , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Humanos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/fisiopatologia , Transdução de Sinais/fisiologia
16.
Arterioscler Thromb Vasc Biol ; 31(11): 2733-41, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21852561

RESUMO

OBJECTIVE: Our aim was to analyze the regulation of CC Chemokine ligand 20 (CCL20) by LDL in human vascular smooth muscle cells (VSMC). METHODS AND RESULTS: In asymptomatic subjects, circulating CCL20 levels were higher in patients with hypercholesterolemia (18.5±3.2 versus 9.1±1.3 pg/mL; P<0.01). LDL induced the expression of CCL20 in VSMC in a dose- and time-dependent manner. Increased levels of CCL20 secreted by LDL-treated VSMC significantly induced human lymphocyte migration, an effect reduced by CCL20 silencing. The upregulation of CCL20 by LDL was dependent on the activation of kinase signaling pathways and NF-κB. By site-directed mutagenesis, electrophoretic mobility shift assay, and chromatin immunoprecipitation, we identified a NF-κB site (-80/-71) in CCL20 promoter critical for LDL responsiveness. Lysophosphatidic acid mimicked the upregulation of CCL20 induced by LDL, and minimal oxidation of LDL increased the ability of LDL to induce CCL20 through a mechanism that involves lysophosphatidic acid receptors. CCL20 was overexpressed in atherosclerotic lesions from coronary artery patients, colocalizing with VSMC. CCL20 was detected in conditioned media from healthy human aorta and its levels were significantly higher in secretomes from carotid endarterectomy specimens. CONCLUSION: This study identifies CCL20 in atherosclerotic lesions and recognizes this chemokine as a mediator highly sensitive to the inflammatory response elicited by LDL.


Assuntos
Quimiocina CCL20/metabolismo , Hipercolesterolemia/metabolismo , Lipoproteínas LDL/farmacologia , Músculo Liso Vascular/metabolismo , NF-kappa B/metabolismo , Regulação para Cima/efeitos dos fármacos , Adulto , Idoso , Aorta/metabolismo , Aorta/patologia , Células Cultivadas , Doença da Artéria Coronariana/metabolismo , Doença da Artéria Coronariana/patologia , Doença da Artéria Coronariana/cirurgia , Relação Dose-Resposta a Droga , Endarterectomia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Transdução de Sinais , Fatores de Tempo , Regulação para Cima/fisiologia
17.
Arterioscler Thromb Vasc Biol ; 31(6): 1411-20, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21454812

RESUMO

OBJECTIVE: Hypoxia is considered a key factor in the progression of atherosclerotic lesions. Low-density lipoprotein receptor-related protein (LRP1) plays a pivotal role in the vasculature. The aim of this study was to investigate the effect of hypoxia on LRP1 expression and function in vascular smooth muscle cells (VSMC) and the role of hypoxia-inducible factor-α (HIF-1α). METHODS AND RESULTS: Real-time polymerase chain reaction and Western blot analysis demonstrated that hypoxia (1% O(2)) time-dependently induced LRP1 mRNA (maximum levels at 1 to 2 hours) and protein expression (maximum levels at 12 to 24 hours). The delayed hypoxic upregulation of LRP1 protein versus mRNA may be explained by the long half-life of LRP1 protein. Luciferase assays demonstrated that hypoxia and HIF-1α overaccumulation induced LRP1 promoter activity and that 2 consensus hypoxia response element sites located at -1072/-1069 and -695/-692 participate in the induction. Chromatin immunoprecipitation showed the in vivo binding of HIF-1α to LRP1 promoter in hypoxic VSMC. Hypoxia effects on LRP1 protein expression were functionally translated into an increased cholesteryl ester (CE) accumulation from aggregated low-density lipoprotein (agLDL) uptake. The blockade of HIF-1α expression inhibited the upregulatory effect of hypoxia on LRP1 expression and agLDL-derived intracellular CE overaccumulation, suggesting that both LRP1 overexpression and CE overaccumulation in hypoxic vascular cells are dependent on HIF-1α. Immunohistochemical analysis showed the colocalization of LRP1 and HIF-1α in vascular cells of human advanced atherosclerotic plaques. CONCLUSION: Hypoxia upregulates LRP1 expression and agLDL-derived intracellular CE accumulation in human VSMC through HIF-1α induction.


Assuntos
Antígenos CD/genética , Hipóxia Celular , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Aterosclerose/metabolismo , Células Cultivadas , Ésteres do Colesterol/metabolismo , Regulação da Expressão Gênica , Humanos , Lipoproteínas LDL/metabolismo , Músculo Liso Vascular/citologia , Regiões Promotoras Genéticas , RNA Mensageiro/análise , Proteína de Ligação a Elemento Regulador de Esterol 1/análise , Proteína de Ligação a Elemento Regulador de Esterol 2/análise
18.
Eur Heart J ; 32(22): 2841-50, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21398644

RESUMO

AIMS: Atherosclerosis plaque development includes infiltration of inflammatory cells, accumulation of lipids and fibrous cap formation. Low-density lipoprotein receptor-related protein 1 (LRP1) is expressed on atherosclerotic lesions associated with macrophages and vascular smooth muscle cells. The aim of this work is to analyse the role in atherosclerosis lesion progression of another member of the LDL receptor protein family, low-density lipoprotein receptor-related protein 5 (LRP5), a co-receptor with Frizzled known to activate the Wnt signalling pathway in several cell types. METHODS AND RESULTS: LRP5 is expressed in human vascular and innate inflammatory cells. LRP5 is transcriptionally regulated by aggregated LDL (agLDL), participating in the lipid uptake and transformation of macrophages into foam cells, a critical step in atherosclerosis progression. AgLDL-treated macrophages show up-regulated expression of ß-catenin, LEF1, c-jun, cyclinD1, bone morphogenetic protein 2 (BMP2), and osteopontin (OPN), proteins and targets of the Wnt signalling pathway, whereas LRP5-silenced macrophages show a significant down-regulation of OPN and BMP2 expression. Furthermore, LRP5-deficient macrophages exhibit an impaired migration both in wound-repair and modified Boyden chambers models. CONCLUSION: These results demonstrate the involvement of LRP5 in the innate inflammatory reaction to lipid infiltration in atherosclerosis.


Assuntos
Aterosclerose/etiologia , Movimento Celular/fisiologia , Metabolismo dos Lipídeos/fisiologia , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Macrófagos/metabolismo , Via de Sinalização Wnt/fisiologia , Apoptose/fisiologia , Aterosclerose/metabolismo , Proteína Morfogenética Óssea 2/metabolismo , Diferenciação Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Células Espumosas/fisiologia , Humanos , Lipoproteínas LDL/farmacologia , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/antagonistas & inibidores , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Monócitos/metabolismo , Osteopontina/metabolismo , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia
19.
Eur J Clin Invest ; 41(10): 1087-97, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21434892

RESUMO

BACKGROUND: Low density lipoprotein receptor-related protein (LRP1) plays a key role on vascular functionality and is upregulated by hypercholesterolemia and hypertension. To investigate the effect of cholesterol-lowering interventions on vascular LRP1 over expression and whether simvastatin influences LRP1 expression. MATERIAL AND METHODS: Male New Zealand rabbits were recruited into various groups, one group was fed a normal chow diet for 28 days (control group, n = 6), other group (n = 24) was fed a hypercholesterolemic diet (HC), six rabbits were euthanized at day 28 to test the capacity of HC diet to induce early atherosclerosis and the rest at day 60 (n = 18) after receiving either HC diet (HC group, n = 6), HC diet with simvastatin (2·5 mg/kg.day) (HC+simv group, n = 6), or a normal chow diet (NC group, n = 6) for the last 32 days. RESULTS: High-cholesterol diet raised vascular LRP1 concomitantly with increased lipid, VSMC and macrophage content in the arterial intima. Simvastatin and return to normocholesterolemic diet significantly reduced systemic cholesterol levels and vascular lipid content. Interestingly, these interventions also downregulate LRP1 overexpression in the vascular wall although to a different extent (HC+simv: 75 ± 3·6%vs NC: 50 ± 3·5% versus, P = 0·002). Immunohistochemistry studies showed that LRP1 diminushion was associated to a reduction in the number of intimal VSMC in HC+simv.group. Simvastatin per se did not exert any significant effect on LRP1 expression in rabbit aortic smooth muscle cells (rSMC). CONCLUSIONS: Our results demonstrate that cholesterol-lowering interventions exerted down regulatory effects on vascular LRP1 over expression induced by hypercholesterolemia and that simvastatin did not influence LRP1 expression beyond its cholesterol-lowering effects.


Assuntos
Aterosclerose/tratamento farmacológico , Colesterol/sangue , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Hipercolesterolemia/sangue , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/sangue , Sinvastatina/uso terapêutico , Animais , Endotélio Vascular/efeitos dos fármacos , Masculino , Coelhos , Reação em Cadeia da Polimerase em Tempo Real
20.
J Mol Cell Cardiol ; 50(3): 522-33, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21219908

RESUMO

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.


Assuntos
Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Remodelação Ventricular/fisiologia , Animais , Apoptose/fisiologia , Colágeno/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose , Marcação In Situ das Extremidades Cortadas/métodos , Inflamação/fisiopatologia , Leucócitos/patologia , Macrófagos/patologia , Metaloproteinase 2 da Matriz/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Suínos , Serina-Treonina Quinases TOR/metabolismo , Receptor 4 Toll-Like/metabolismo , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA