Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 53
Filtrer
1.
J Vis Exp ; (189)2022 11 11.
Article de Anglais | MEDLINE | ID: mdl-36440832

RÉSUMÉ

Pulmonary arterial hypertension (PAH) is a progressive life-threatening disease, primarily affecting small pulmonary arterioles of the lung. Currently, there is no cure for PAH. It is important to discover new compounds that can be used to treat PAH. The mouse hypoxia-induced PAH model is a widely used model for PAH research. This model recapitulates human clinical manifestations of PAH Group 3 disease and is an important research tool to evaluate the effectiveness of new experimental therapies for PAH. Research using this model often requires the administration of compounds in mice. For a compound that needs to be given directly into the bloodstream, optimizing intravenous (IV) administration is a key part of the experimental procedures. Ideally, the IV injection system should permit multiple injections over a set time course. Although the mouse hypoxia-induced PAH model is very popular in many laboratories, it is technically challenging to perform multiple IV bolus dosing and invasive hemodynamic assessment in this model. In this protocol, we present step-by-step instructions on how to carry out multiple IV bolus dosing via mouse jugular vein and perform arterial and right ventricle catheterization for hemodynamic assessment in mouse hypoxia-induced PAH model.


Sujet(s)
Hypertension pulmonaire , Hypertension artérielle pulmonaire , Humains , Souris , Animaux , Hypertension pulmonaire/traitement médicamenteux , Hypertension pulmonaire/étiologie , Artère pulmonaire , Hémodynamique , Hypoxie , Modèles animaux de maladie humaine
2.
Int J Cardiol ; 326: 131-138, 2021 03 01.
Article de Anglais | MEDLINE | ID: mdl-33091520

RÉSUMÉ

AIM: Heart failure following myocardial infarction (MI) is a potentially lethal problem with a staggering incidence. The CardiAMP Heart Failure trial represents the first attempt to personalize marrow-derived cell-based therapy to individuals with cell characteristics associated with beneficial responses in prior trials. Before the initiation of the randomized pivotal trial, an open-label "roll-in cohort" was completed to ensure the feasibility of the protocol's procedures. METHODS: Patients with chronic post-MI heart failure (NYHA class II-III) receiving stable, guideline-directed medical therapy with a left ventricular ejection fraction between 20 and 40% were eligible. Two weeks prior to treatment, a ~ 5 mL bone marrow aspiration was performed to examine "cell potency". On treatment day, a 60 mL bone marrow aspiration, bone marrow mononuclear cell (BM MNC) enrichment and transendocardial injection of 200 million BM MNC's was performed in a single, point of care encounter. Patients were then followed to assess clinical outcomes. RESULTS: The cell potency small volume bone marrow aspirate, the 60 mL bone marrow aspirate, and transendocardial injections were well tolerated in 10 patients enrolled. There were no serious adverse events related to bone marrow aspiration or cell delivery. Improvement in 6-min walk distance was observed at 6 months (+47.8 m, P = 0.01) and trended to improvement at 12 months (+46.4, P = 0.06). Similarly, trends to improved NYHA heart failure functional class, quality of life, left ventricular ejection fraction and recruitment of previously akinetic left ventricular wall segments were observed. CONCLUSION: All CardiAMP HF protocol procedures were feasible and well tolerated. Favorable functional, echo and quality of life trends suggest this approach may offer promise for patients with post MI heart failure. The randomized CardiAMP Heart Failure pivotal trial is underway to confirm the efficacy of this approach. CLINICAL TRIAL REGISTRATION: https://clinicaltrials.gov/ct2/show/NCT02438306.


Sujet(s)
Défaillance cardiaque , Ischémie myocardique , Moelle osseuse , Transplantation de moelle osseuse , Thérapie cellulaire et tissulaire , Études de faisabilité , Défaillance cardiaque/diagnostic , Défaillance cardiaque/thérapie , Humains , Systèmes automatisés lit malade , Qualité de vie , Débit systolique , Résultat thérapeutique , Fonction ventriculaire gauche
4.
Am Heart J ; 201: 141-148, 2018 07.
Article de Anglais | MEDLINE | ID: mdl-29803986

RÉSUMÉ

BACKGROUND: Heart failure following myocardial infarction is a common, disabling, and deadly condition. Direct injection of autologous bone marrow mononuclear cells into the myocardium may result in improved functional recovery, relieve symptoms, and improve other cardiovascular outcomes. METHODS: CardiAMP-HF is a randomized, double-blind, sham-controlled, pivotal trial designed to investigate the safety and efficacy of autologous bone marrow mononuclear cells treatment for patients with medically refractory and symptomatic ischemic cardiomyopathy. The primary end point is change in 6-minute walk distance adjusted for major adverse cardiovascular events at 12 months following treatment. Particularly novel aspects of this trial include a cell potency assay to screen subjects who have bone marrow cell characteristics that suggest a favorable response to treatment, a point-of-care treatment method, a high target dose of 200 million cells, and an efficient transcatheter intramyocardial delivery method that is associated with high cell retention. CONCLUSIONS: This novel approach may lead to a new treatment for those with ischemic heart disease suffering from medically refractory heart failure.


Sujet(s)
Transplantation de moelle osseuse/méthodes , Défaillance cardiaque/thérapie , Monocytes/transplantation , Infarctus du myocarde/complications , Débit systolique/physiologie , Fonction ventriculaire gauche/physiologie , Adulte , Sujet âgé , Méthode en double aveugle , Femelle , Études de suivi , Défaillance cardiaque/étiologie , Défaillance cardiaque/physiopathologie , Humains , Mâle , Adulte d'âge moyen , Monocytes/cytologie , Infarctus du myocarde/physiopathologie , Infarctus du myocarde/thérapie , Transplantation autologue , Résultat thérapeutique , Jeune adulte
5.
Cardiovasc Res ; 113(14): 1776-1788, 2017 Dec 01.
Article de Anglais | MEDLINE | ID: mdl-29016873

RÉSUMÉ

AIMS: The formation of cell-cell and cell-extra cellular matrix (ECM) contacts by endothelial cells (ECs) is crucial for the stability and integrity of a vascular network. We previously identified cingulin-like 1 (Cgnl1) in a transcriptomic screen for new angiogenic modulators. Here we aim to study the function of the cell-cell junction associated protein Cgnl1 during vessel formation. METHODS AND RESULTS: Unlike family member cingulin, Cgnl1 expression is enriched in ECs during vascular growth. Cgnl1 is important for the formation of multicellular tubule structures, as shown in vitro using loss-of function assays in a 3D matrix co-culture system that uses primary human ECs and supporting mural cells. Further studies revealed that Cgnl1 regulates vascular growth by promoting Ve-cadherin association with the actin cytoskeleton, thereby stabilizing adherens junctions. Cgnl1 also regulates focal adhesion assembly in response to ECM contact, promoting vinculin and paxillin recruitment and focal adhesion kinase signalling. In vivo, we demonstrate in a postnatal retinal vascular development model in mice that Cgnl1 function is crucial for sustaining neovascular growth and stability. CONCLUSIONS: Our data demonstrate a functional relevance for Cgnl1 as a defining factor in new vessel formation both in vitro and in vivo.


Sujet(s)
Jonctions adhérentes/métabolisme , Protéines du cytosquelette/métabolisme , Cellules endothéliales/métabolisme , dGTPases/métabolisme , Protéines membranaires/métabolisme , Néovascularisation physiologique/physiologie , Cytosquelette d'actine/métabolisme , Animaux , Adhérence cellulaire/physiologie , Protéines du cytosquelette/génétique , Endothélium vasculaire/métabolisme , Humains , Jonctions intercellulaires/métabolisme , Protéines membranaires/génétique , Souris de lignée C57BL
6.
Cardiovasc Res ; 110(1): 129-39, 2016 May 01.
Article de Anglais | MEDLINE | ID: mdl-26822228

RÉSUMÉ

AIMS: Impairment of the endothelial barrier leads to microvascular breakdown in cardiovascular disease and is involved in intraplaque haemorrhaging and the progression of advanced atherosclerotic lesions that are vulnerable to rupture. The exact mechanism that regulates vascular integrity requires further definition. Using a microarray screen for angiogenesis-associated genes during murine embryogenesis, we identified thrombospondin type I domain 1 (THSD1) as a new putative angiopotent factor with unknown biological function. We sought to characterize the role of THSD1 in endothelial cells during vascular development and cardiovascular disease. METHODS AND RESULTS: Functional knockdown of Thsd1 in zebrafish embryos and in a murine retina vascularization model induced severe haemorrhaging without affecting neovascular growth. In human carotid endarterectomy specimens, THSD1 expression by endothelial cells was detected in advanced atherosclerotic lesions with intraplaque haemorrhaging, but was absent in stable lesions, implying involvement of THSD1 in neovascular bleeding. In vitro, stimulation with pro-atherogenic factors (3% O2 and TNFα) decreased THSD1 expression in human endothelial cells, whereas stimulation with an anti-atherogenic factor (IL10) showed opposite effect. Therapeutic evaluation in a murine advanced atherosclerosis model showed that Thsd1 overexpression decreased plaque vulnerability by attenuating intraplaque vascular leakage, subsequently reducing macrophage accumulation and necrotic core size. Mechanistic studies in human endothelial cells demonstrated that THSD1 activates FAK-PI3K, leading to Rac1-mediated actin cytoskeleton regulation of adherens junctions and focal adhesion assembly. CONCLUSION: THSD1 is a new regulator of endothelial barrier function during vascular development and protects intraplaque microvessels against haemorrhaging in advanced atherosclerotic lesions.


Sujet(s)
Athérosclérose/métabolisme , Cellules endothéliales/métabolisme , Microvaisseaux/métabolisme , Néovascularisation pathologique/métabolisme , Thrombospondines/métabolisme , Animaux , Apolipoprotéines E/déficit , Apolipoprotéines E/métabolisme , Artériopathies carotidiennes/métabolisme , Femelle , Humains , Mâle , Souris de lignée C57BL , Phosphatidylinositol 3-kinases/métabolisme , Plaque d'athérosclérose/anatomopathologie , Thrombospondine-1/métabolisme
7.
Development ; 142(9): 1695-704, 2015 May 01.
Article de Anglais | MEDLINE | ID: mdl-25834021

RÉSUMÉ

SoxF family members have been linked to arterio-venous specification events and human pathological conditions, but in contrast to Sox17 and Sox18, a detailed in vivo analysis of a Sox7 mutant model is still lacking. In this study we generated zebrafish sox7 mutants to understand the role of Sox7 during vascular development. By in vivo imaging of transgenic zebrafish lines we show that sox7 mutants display a short circulatory loop around the heart as a result of aberrant connections between the lateral dorsal aorta (LDA) and either the venous primary head sinus (PHS) or the common cardinal vein (CCV). In situ hybridization and live observations in flt4:mCitrine transgenic embryos revealed increased expression levels of flt4 in arterial endothelial cells at the exact location of the aberrant vascular connections in sox7 mutants. An identical circulatory short loop could also be observed in newly generated mutants for hey2 and efnb2. By genetically modulating levels of sox7, hey2 and efnb2 we demonstrate a genetic interaction of sox7 with hey2 and efnb2. The specific spatially confined effect of loss of Sox7 function can be rescued by overexpressing the Notch intracellular domain (NICD) in arterial cells of sox7 mutants, placing Sox7 upstream of Notch in this aspect of arterial development. Hence, sox7 levels are crucial in arterial specification in conjunction with hey2 and efnb2 function, with mutants in all three genes displaying shunt formation and an arterial block.


Sujet(s)
Animal génétiquement modifié/génétique , Artères/embryologie , Régulation de l'expression des gènes au cours du développement/physiologie , Morphogenèse/physiologie , Facteurs de transcription SOX-F/génétique , Protéines de poisson-zèbre/génétique , Danio zébré/embryologie , Angiographie , Animaux , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Amorces ADN/génétique , Régulation de l'expression des gènes au cours du développement/génétique , Hybridation in situ , Morpholinos/génétique , Mutation/génétique , Débit sanguin régional/physiologie , RT-PCR , Facteurs de transcription SOX-F/métabolisme , Récepteur-3 au facteur croissance endothéliale vasculaire/métabolisme , Danio zébré/génétique , Protéines de poisson-zèbre/métabolisme
8.
Biomed Res Int ; 2015: 209315, 2015.
Article de Anglais | MEDLINE | ID: mdl-25802838

RÉSUMÉ

BACKGROUND: Novel therapies need to be evaluated in a relevant large animal model that mimics the clinical course and treatment in a reasonable time frame. To reliably assess therapeutic efficacy, knowledge regarding the translational model and the course of disease is needed. METHODS: Landrace pigs were subjected to a transient occlusion of the proximal left circumflex artery (LCx) (n = 6) or mid-left anterior descending artery (LAD) (n = 6) for 150 min. Cardiac function was evaluated before by 2D echocardiography or 3D echocardiography and pressure-volume loop analysis. At 12 weeks of follow-up the heart was excised for histological analysis and infarct size calculations. RESULTS: Directly following AMI, LVEF was severely reduced compared to baseline in the LAD group (-17.1 ± 1.6%, P = 0.009) compared to only a moderate reduction in the LCx group (-5.9 ± 1.5%, P = 0.02) and this effect remained unchanged during 12 weeks of follow-up. CONCLUSION: Two models of chronic MI, representative for different patient groups, can reproducibly be created through clinically relevant ischemia-reperfusion of the mid-LAD and proximal LCx.


Sujet(s)
Coeur/physiopathologie , Infarctus du myocarde/physiopathologie , Animaux , Modèles animaux de maladie humaine , Échocardiographie/méthodes , Échocardiographie tridimensionnelle/méthodes , Femelle , Études de suivi , Suidae
9.
Circ Cardiovasc Interv ; 7(5): 673-83, 2014 Oct.
Article de Anglais | MEDLINE | ID: mdl-25294400

RÉSUMÉ

BACKGROUND: Engraftment and survival of stem cells in the infarcted myocardium remain problematic in cell-based therapy for cardiovascular disease. To overcome these issues, encapsulated mesenchymal stem cells (eMSCs) were developed that were transfected to produce glucagon-like peptide-1, an incretin hormone with known cardioprotective effects, alongside MSC endogenous paracrine factors. This study was designed to investigate the efficacy of different doses of intracoronary infusion of eMSC in a porcine model of acute myocardial infarction (AMI). METHODS AND RESULTS: One hundred pigs were subjected to a moderate AMI (posterolateral AMI; n=50) or a severe AMI (anterior AMI; n=50), whereupon surviving animals (n=36 moderate, n=33 severe) were randomized to receive either intracoronary infusion of 3 incremental doses of eMSC or Ringers' lactate control. Cardiac function was assessed using invasive hemodynamics, echocardiography, and histological analysis. A trend was observed in the moderate AMI model, whereas in the severe AMI model, left ventricular ejection fraction improved by +9.3% (P=0.004) in the best responding eMSC group, because of a preservation of left ventricular end-systolic volume. Arteriolar density increased 3-fold in the infarct area (8.4±0.9/mm(2) in controls versus 22.2±2.6/mm(2) in eMSC group; P<0.001). Although not statistically significant, capillary density was 30% higher in the border zone (908.1±99.7/mm(2) in control versus 1209.0±64.6/mm(2) in eMSC group; P=ns). CONCLUSIONS: eMSCs enable sustained local delivery of cardioprotective proteins to the heart, thereby enhancing angiogenesis and preserving contractile function in an animal AMI model.


Sujet(s)
Glucagon-like peptide 1/métabolisme , Transplantation de cellules souches mésenchymateuses/méthodes , Cellules souches mésenchymateuses/métabolisme , Infarctus du myocarde/thérapie , Myocarde/anatomopathologie , Animaux , Vaisseaux coronaires/chirurgie , Modèles animaux de maladie humaine , Échocardiographie , Glucagon-like peptide 1/génétique , Humains , Perfusions artérielles , Suidae , Transgènes/génétique , Fonction ventriculaire gauche
10.
Vasc Med ; 19(2): 94-102, 2014 Apr.
Article de Anglais | MEDLINE | ID: mdl-24829311

RÉSUMÉ

Previously, we created an experimental murine model for the induction of vulnerable plaque (VP). Although this murine model offers the opportunity to study the different molecular biological pathways that regulate plaque destabilization, the size of the animals severely limits the use of the model for in vivo diagnostics and percutaneous interventions. This study aimed to create a VP model in the rabbit, based on the murine model, to aid the assessment and development of novel diagnostic and interventional tools. New Zealand white rabbits were fed on a 2% cholesterol diet. After 1 week, a shear stress-altering device was implanted around the right carotid artery. Twelve weeks after cast placement, the carotid artery was isolated and processed for (immuno-)histological analysis to evaluate the presence of a VP phenotype. Atherosclerotic plaques with high lipid and macrophage content, low vascular smooth muscle cell content and intimal neovascularization were located upstream and downstream of the cast. The plaques lacked a significant necrotic core. In conclusion, we were able to create atherosclerotic plaques with a phenotype beyond that of a fatty streak, with a high percentage of lipids and macrophages, a thick cap with some vascular smooth muscle cells and neovascularization. However, as there was only a small necrotic core, the overall phenotype seems less vulnerable as compared to the thin fibrous cap atheroma in patients.

11.
Circ Cardiovasc Interv ; 7(2): 156-67, 2014 Apr.
Article de Anglais | MEDLINE | ID: mdl-24668227

RÉSUMÉ

BACKGROUND: Several cell-based therapies for adjunctive treatment of acute myocardial infarction have been investigated in multiple clinical trials, but the benefits still remain controversial. This meta-analysis aims to evaluate the efficacy of bone marrow-derived mononuclear cell (BMMNC) therapy in patients with acute myocardial infarction, but also explores the effect of newer generations of stem cells. METHODS AND RESULTS: A random-effects meta-analysis was performed on randomized controlled trials investigating the effects of stem cell therapy in patients with acute myocardial infarction that were published between January 2002 and September 2013. The defined end points were left ventricular (LV) ejection fraction, LV end-systolic and end-diastolic volumes, infarct size, and major adverse cardiac and cerebrovascular event rates. Also, several subgroup analyses were performed on BMMNC trials. Overall, combining the results of 22 randomized controlled trials (RCTs), LV ejection fraction increased by +2.10% (95% confidence interval [CI], 0.68-3.52; P=0.004) in the BMMNC group as compared with controls, evoked by a preservation of LV end-systolic volume (-4.05 mL; 95% CI, -6.91 to -1.18; P=0.006) and a reduction in infarct size (-2.69%; 95% CI, -4.83 to -0.56; P=0.01). However, there is no effect on cardiac function, volumes, or infarct size, when only RCTs (n=9) that used MRI-derived end points were analyzed. Moreover, no beneficial effect could be detected on major adverse cardiac and cerebrovascular event rates after BMMNC infusion after a median follow-up duration of 6 months. CONCLUSIONS: Intracoronary infusion of BMMNC is safe, but does not enhance cardiac function on MRI-derived parameters, nor does it improve clinical outcome. New and possibly more potent stem cells are emerging in the field, but their clinical efficacy still needs to be defined in future trials.


Sujet(s)
Thérapie cellulaire et tissulaire/méthodes , Perfusions artérielles/méthodes , Infarctus du myocarde/thérapie , Transplantation de cellules souches/méthodes , Études de suivi , Humains , Hypertrophie ventriculaire gauche/anatomopathologie , Perfusions artérielles/effets indésirables , Imagerie par résonance magnétique , Infarctus du myocarde/anatomopathologie , Infarctus du myocarde/physiopathologie , Essais contrôlés randomisés comme sujet , Transplantation de cellules souches/effets indésirables , Débit systolique/physiologie , Résultat thérapeutique
12.
Arterioscler Thromb Vasc Biol ; 34(3): 594-602, 2014 Mar.
Article de Anglais | MEDLINE | ID: mdl-24407030

RÉSUMÉ

OBJECTIVE: In arteriogenesis, pre-existing anastomoses undergo enlargement to restore blood flow in ischemic tissues. Chemokine (C-X-C motif) ligand 10 (CXCL10) is secreted after Toll-like receptor activation. Toll-like receptors are involved in arteriogenesis; however, the role of CXCL10 is still unclear. In this study, we investigated the role for CXCL10 in a murine hindlimb ischemia model. APPROACH AND RESULTS: Unilateral femoral artery ligation was performed in wild-type (WT) and CXCL10(-/-) knockout (KO) mice and perfusion recovery was measured using laser-Doppler perfusion analysis. Perfusion recovery was significantly lower in KO mice compared with WT at days 4 and 7 after surgery (KO versus WT: 28±5% versus 81±13% at day 4; P=0.003 and 57±12% versus 107±8% at day 7; P=0.003). Vessel measurements of α-smooth muscle actin-positive vessels revealed increasing numbers in time after surgery, which was significantly higher in WT when compared with that in KO. Furthermore, α-smooth muscle actin-positive vessels were significantly larger in WT when compared with those in KO at day 7 (wall thickness, P<0.001; lumen area, P=0.003). Local inflammation was assessed in hindlimb muscles, but this did not differ between WT and KO. Chimerization experiments analyzing perfusion recovery and histology revealed an equal contribution for bone marrow-derived and circulating CXCL10. Migration assays showed a stimulating role for both intrinsic and extrinsic CXCL10 in vascular smooth muscle cell migration. CONCLUSIONS: CXCL10 plays a causal role in arteriogenesis. Bone marrow-derived CXCL10 and tissue-derived CXCL10 play a critical role in accelerating perfusion recovery after arterial occlusion in mice probably by promoting vascular smooth muscle cell recruitment and maturation of pre-existing anastomoses.


Sujet(s)
Chimiokine CXCL10/déficit , Circulation collatérale/physiologie , Néovascularisation physiologique/physiologie , Lésion d'ischémie-reperfusion/physiopathologie , Animaux , Aorte/cytologie , Moelle osseuse/métabolisme , Cellules cultivées , Chimiokine CXCL10/antagonistes et inhibiteurs , Chimiokine CXCL10/sang , Chimiokine CXCL10/génétique , Chimiokine CXCL10/pharmacologie , Chimiokine CXCL10/physiologie , Femelle , Artère fémorale , Membre pelvien/vascularisation , Cellules endothéliales de la veine ombilicale humaine , Humains , Inflammation , Ischémie/physiopathologie , Fluxmétrie laser Doppler , Ligature , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Muscles squelettiques/vascularisation , Muscles squelettiques/métabolisme , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/métabolisme , Interférence par ARN , Petit ARN interférent/pharmacologie , Chimère post-radique , Protéines recombinantes/pharmacologie
13.
Cell Transplant ; 23(12): 1525-35, 2014.
Article de Anglais | MEDLINE | ID: mdl-23561893

RÉSUMÉ

The objective of this study was to assess the effect of high-dose atorvastatin treatment on endothelial progenitor cell (EPC) recruitment and angiographic and clinical outcome in coronary artery disease (CAD) patients treated with the Genous EPC-capturing stent. The HEALING IIB study was a multicenter, open-label, prospective trial that enrolled 100 patients. Patients were started on 80 mg atorvastatin qd, at least 2 weeks before the index procedure and continued for at least 4 weeks after the index procedure. Eighty-seven patients were included in this analysis. EPC levels significantly increased as early as 2 weeks after the start of atorvastatin. Remarkably, among this group, 31 patients proved to be nonresponders to atorvastatin treatment (i.e., no increase in EPC levels), while 56 patients were responders (i.e., rise in EPC count between week -2 and 0). Compared to responders, nonresponders had a significantly higher baseline EPC count (76 ± 10 vs. 41 ± 5, p < 0.01) with a lower late luminal loss (LLL) at 6- and 18-month follow-up (FU) (0.61 ± 0.07 vs. 0.88 ± 0.08, p < 0.05, and 0.50 ± 0.08 vs. 0.82 ± 0.08, p < 0.01 respectively). Furthermore, baseline EPC count was inversely correlated with LLL at 6-month FU (R = -0.42, p < 0.001). Patients with a higher EPC count at baseline showed no increase in EPC recruitment in response to statin treatment but had favorable LLL at 6- and 18-month FU, whereas patients with lower EPC count were responsive to statin therapy, but EPCs might be less functional as they had higher LLL at 6- and 18-month FU. These data imply that although statin treatment can enhance EPC titer in patients with low baseline levels, there is no indication for a possible beneficial clinical effect with EPC capture stents.


Sujet(s)
Coronarographie , Maladie des artères coronaires/imagerie diagnostique , Maladie des artères coronaires/anatomopathologie , Progéniteurs endothéliaux/effets des médicaments et des substances chimiques , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/pharmacologie , Endoprothèses , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Atorvastatine , Maladie des artères coronaires/sang , Démographie , Femelle , Acides heptanoïques/pharmacologie , Humains , Lipides/sang , Mâle , Adulte d'âge moyen , Analyse multifactorielle , Pyrroles/pharmacologie , Analyse de régression , Résultat thérapeutique
14.
Methods Mol Biol ; 1036: 207-12, 2013.
Article de Anglais | MEDLINE | ID: mdl-23807797

RÉSUMÉ

Adipose tissue represents an abundant, accessible source of regenerative cells that can be easily obtained in sufficient amount for therapy. Adipose-derived regenerative cells (ADRC) are comprised of leukocytes, smooth muscles, endothelial cells, and mesenchymal stem cells. In contrast to bone-marrow-derived MSC, the abundance of adipose tissue in patients and the higher frequency per unit mass of regenerative cells allow for the isolation of cells in therapeutic meaningful amounts in less than 2h after donor tissue acquisition.Harvest of adipose tissue can thus follow primary PCI, allowing efficient treatment within 24h. This obviates the need for extensive cell culturing in GMP clean room facilities and makes ADSCs a promising and practical autologous cell source. In the following chapter, we will describe the liposuction procedure for stem cell harvest, two cell delivery techniques, and pressure/volume loop analysis for the follow-up of our patients enrolled in the clinical studies.


Sujet(s)
Tissu adipeux/transplantation , Séparation cellulaire/méthodes , Défaillance cardiaque/thérapie , Lipectomie/méthodes , Transplantation de cellules souches mésenchymateuses/méthodes , Cellules souches mésenchymateuses/cytologie , Infarctus du myocarde/thérapie , Maladie aigüe , Différenciation cellulaire , Cellules cultivées , Essais cliniques comme sujet , Humains
15.
Circ Res ; 113(2): 153-66, 2013 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-23658436

RÉSUMÉ

RATIONALE: Mesenchymal precursor cells (MPCs) are a specific Stro-3+ subpopulation of mesenchymal stem cells isolated from bone marrow. MPCs exert extensive cardioprotective effects, and are considered to be immune privileged. OBJECTIVE: This study assessed the safety, feasibility, and efficacy of intracoronary delivery of allogeneic MPCs directly after acute myocardial infarction in sheep. METHODS AND RESULTS: Initially, intracoronary delivery conditions were optimized in 20 sheep. These conditions were applied in a randomized study of 68 sheep with an anterior acute myocardial infarction. Coronary flow was monitored during MPC infusion, and cardiac function was assessed using invasive hemodynamics and echocardiography at baseline and during 8 weeks follow-up. Coronary flow remained within thrombolysis in myocardial infarction III definitions in all sheep during MPC infusion. Global left ventricular ejection fraction as measured by pressure-volume loop analysis deteriorated in controls to 40.7±2.6% after 8 weeks. In contrast, MPC treatment improved cardiac function to 52.8±0.7%. Echocardiography revealed significant improvement of both global and regional cardiac functions. Infarct size decreased by 40% in treated sheep, whereas infarct and border zone thickness were enhanced. Left ventricular adverse remodeling was abrogated by MPC therapy, resulting in a marked reduction of left ventricular volumes. Blood vessel density increased by >50% in the infarct and border areas. Compensatory cardiomyocyte hypertrophy was reduced in border and remote segments, accompanied by reduced collagen deposition and apoptosis. No microinfarctions in remote myocardial segments or histological abnormalities in unrelated organs were found. CONCLUSIONS: Intracoronary infusion of allogeneic MPCs is safe, feasible, and markedly effective in a large animal model of acute myocardial infarction.


Sujet(s)
Vaisseaux coronaires/physiologie , Transplantation de cellules souches mésenchymateuses/méthodes , Infarctus du myocarde/anatomopathologie , Infarctus du myocarde/chirurgie , Fonction ventriculaire gauche/physiologie , Remodelage ventriculaire/physiologie , Animaux , Études de suivi , Perfusions artérielles , Infarctus du myocarde/physiopathologie , Répartition aléatoire , Ovis , Transplantation homologue
16.
J Am Coll Cardiol ; 61(14): 1471-81, 2013 Apr 09.
Article de Anglais | MEDLINE | ID: mdl-23500310

RÉSUMÉ

Oxidative stress greatly influences the pathogenesis of various cardiovascular disorders. Coronary interventions, including balloon angioplasty and coronary stent implantation, are associated with increased vascular levels of reactive oxygen species in conjunction with altered endothelial cell and smooth muscle cell function. These alterations potentially lead to restenosis, thrombosis, or endothelial dysfunction in the treated artery. Therefore, the understanding of the pathophysiological role of reactive oxygen species (ROS) generated during or after coronary interventions, or both, is essential to improve the success rate of these procedures. Superoxide O2(·-) anions, whether derived from uncoupled endothelial nitric oxide synthase, nicotinamide adenine dinucleotide phosphate oxidase, xanthine oxidase, or mitochondria, are among the most harmful ROS. O2(·-) can scavenge nitric oxide, modify proteins and nucleotides, and induce proinflammatory signaling, which may lead to greater ROS production. Current innovations in stent technologies, including biodegradable stents, nitric oxide donor-coated stents, and a new generation of drug-eluting stents, therefore address persistent oxidative stress and reduced nitric oxide bioavailability after percutaneous coronary interventions. This review discusses the molecular mechanisms of ROS generation after coronary interventions, the related pathological events-including restenosis, endothelial dysfunction, and stent thrombosis-and possible therapeutic ways forward.


Sujet(s)
Resténose coronaire/anatomopathologie , Stress oxydatif/physiologie , Intervention coronarienne percutanée/effets indésirables , Intervention coronarienne percutanée/méthodes , Espèces réactives de l'oxygène/métabolisme , Endoprothèses , Adulte , Sujet âgé , Angioplastie coronaire par ballonnet/effets indésirables , Angioplastie coronaire par ballonnet/méthodes , Resténose coronaire/thérapie , Sténose coronarienne/imagerie diagnostique , Sténose coronarienne/mortalité , Sténose coronarienne/thérapie , Endoprothèses à élution de substances , Endothélium vasculaire/enzymologie , Endothélium vasculaire/physiologie , Femelle , Humains , Mâle , Adulte d'âge moyen , Monitorage physiologique , Nitric oxide synthase type III/métabolisme , Soins postopératoires/méthodes , Complications postopératoires/traitement médicamenteux , Complications postopératoires/métabolisme , Complications postopératoires/anatomopathologie , Pronostic , Radiographie , Essais contrôlés randomisés comme sujet , Appréciation des risques , Analyse de survie , Résultat thérapeutique , Vasodilatateurs/usage thérapeutique
18.
Trends Cardiovasc Med ; 23(5): 153-64, 2013 Jul.
Article de Anglais | MEDLINE | ID: mdl-23375379

RÉSUMÉ

Advanced atherosclerotic lesions prone to rupture are characterized by a distinct histomorphology and pathobiology that became in recent years, increasingly related to the process of intraplaque neovascularization. Molecular mechanisms that regulate angiogenesis and that are active in the plaque region may destabilize advanced lesions by promoting microvessel growth and thus providing an entry route for inflammatory cells secondary to the luminal endothelium. In addition, angiogenic factors can also define intraplaque microvessel integrity and endothelial barrier function, determining the prevalence of intraplaque hemorrhaging. Here, we aim to compose a hypothetical model for angiogenic regulation of vulnerable plaque development, based on the evidence of clinical correlation and experimental functional studies that are provided for five of the most well-described angiogenic pathways in the current literature.


Sujet(s)
Athérosclérose/anatomopathologie , Microvaisseaux/anatomopathologie , Néovascularisation pathologique , Protéines angiogéniques/métabolisme , Animaux , Athérosclérose/métabolisme , Modèles animaux de maladie humaine , Évolution de la maladie , Humains , Microvaisseaux/métabolisme , Modèles biologiques , Plaque d'athérosclérose , Transduction du signal
19.
Cell Transplant ; 22(3): 535-43, 2013.
Article de Anglais | MEDLINE | ID: mdl-22507673

RÉSUMÉ

Cell therapy is a field of growing interest in the prevention of post acute myocardial infarction (AMI) heart failure. Stem cell retention upon local delivery to the heart, however, is still unsatisfactory. CellBeads were recently developed as a potential solution to this problem. CellBeads are 170-µm alginate microspheres that contain mesenchymal stem cells (MSCs) genetically modified to express glucagon-like peptide-1 (GLP-1) supplementary to inherent paracrine factors. GLP-1 is an incretin hormone that has both antiapoptotic and cardioprotective effects. Transplanting CellBeads in the post-AMI heart might induce cardiomyocyte salvage and ultimately abrogate adverse cardiac remodeling. We aimed to investigate the feasibility of intracoronary infusion of CellBeads in a large animal model of AMI. Four pigs were used in a pilot study to assess the maximal safe dose of CellBeads. In the remaining 21 animals, an AMI was induced by balloon occlusion of the left circumflex coronary artery for 90 min. During reperfusion, 60,000 CellBeads (n = 11), control beads (n = 4), or lactated Ringers' (n = 6) were infused. Animals were sacrificed after 2 or 7 days, and the hearts were excised for histological analyses. Intracoronary infusion did not permanently affect coronary flow in any of the groups. Histological analysis revealed CellBeads containing viable MSCs up to 7 days. Viability and activity of the MSCs was confirmed by qPCR analysis that showed expression of recombinant GLP-1 and human genes after 2 and 7 days. CellBeads reduced inflammatory infiltration by 29% (p = 0.001). In addition, they decreased the extent of apoptosis by 25% (p = 0.001) after 2 days. We show that intracoronary infusion of 5 million encapsulated MSCs is safe and feasible. Also, several parameters indicate that the cells have paracrine effects, suggesting a potential therapeutic benefit of this new approach.


Sujet(s)
Glucagon-like peptide 1/métabolisme , Transplantation de cellules souches mésenchymateuses/méthodes , Infarctus du myocarde/thérapie , Maladie aigüe , Alginates/composition chimique , Animaux , Apoptose , Occlusion par ballonnet , Thérapie cellulaire et tissulaire , Femelle , Glucagon-like peptide 1/génétique , Acide glucuronique/composition chimique , Acides hexuroniques/composition chimique , Humains , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/métabolisme , Infarctus du myocarde/anatomopathologie , Myocarde/métabolisme , Myocarde/anatomopathologie , Myocytes cardiaques/cytologie , Projets pilotes , Suidae
20.
Pharmacol Res ; 67(1): 10-7, 2013 Jan.
Article de Anglais | MEDLINE | ID: mdl-23063485

RÉSUMÉ

Uridine adenosine tetraphosphate (Up4A) has been identified as an endothelium-derived contracting factor, which acts through purinergic P2X and P2Y receptors. Since the coronary vascular actions of Up4A are unknown, we investigated the vasoactive profile of Up4A in coronary microvessels, and studied the involvement of purinergic receptor subtypes. Studies were performed in isolated porcine coronary small arteries (diameter∼250 µm), with and without endothelial denudation, mounted on a Mulvany wire myograph. Purinergic receptor expression was assessed by real-time PCR. Up4A (10(-9)-10(-5) M) failed to induce contraction at basal tone, but produced concentration-dependent vasorelaxation in precontracted microvessels. Up4A was slightly less potent than adenosine, ATP, and ADP in producing vasorelaxation, but significantly more potent than UTP and UDP. mRNA expression of P2X(4), P2Y(1), P2Y(2), P2Y(4), P2Y(6) and A(2A), but not P2X(1), receptors was observed. Up4A-induced vasodilation was unaffected by non-selective P2 receptor antagonist PPADS, P2X(1) antagonist MRS2159, P2Y(1) antagonist MRS2179 and P2Y(6) antagonist MRS2578, but was markedly attenuated by non-selective P1 receptor antagonist 8PT and A(2A) antagonist SCH58261. Up4A-induced vasodilation was not affected by ectonucleotidase inhibitor ARL67156, suggesting that A(2A) stimulation was not the result of Up4A breakdown to adenosine. Up4A-induced vasodilation was blunted in denuded vessels; additional A(2A) receptor blockade further attenuated Up4A-induced vasodilation, suggesting that A(2A) receptor-mediated vasodilation is only partly endothelium-dependent. In conclusion, Up4A exerts a vasodilator rather than a vasoconstrictor influence in coronary microvessels, which is mediated via A(2A) receptors and is partly endothelium-dependent.


Sujet(s)
Vaisseaux coronaires/effets des médicaments et des substances chimiques , Dinucléoside phosphates/pharmacologie , Récepteurs purinergiques P1/physiologie , Vasodilatateurs/pharmacologie , Animaux , Vaisseaux coronaires/physiologie , Endothélium vasculaire/effets des médicaments et des substances chimiques , Endothélium vasculaire/physiologie , Techniques in vitro , Microcirculation/effets des médicaments et des substances chimiques , Récepteurs purinergiques P2/physiologie , Suidae
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE