15 research outputs found

    Relation entre la structure et la fonction des artères cérébrales dans l’athérosclérose : impact des traitements cardioprotecteurs

    Full text link
    Thèse réalisée en cotutelle avec Dre Christine Des RosiersLe processus de l’athérosclérose est associé à des changements vasculaires structuraux et mécaniques dont la rigidification carotidienne et aortique. Ce phénomème est bien connu et contraste avec l’augmentation paradoxale de la distensibilité cérébrovasculaire observée dans les artères cérébrales exposées aux facteurs de risque cardiovasculaire, tels que l’hypertension. L’impact de l’athérosclérose sur le remodelage, la compliance et la fonction des artères cérébrales est inconnu. En ciblant l’endothélium, l’athérosclérose induit une dysfonction endothéliale cérébrale sévère qui interfère avec le contrôle du débit sanguin cérébral et ultimement avec les fonctions cognitives. Dans les artères cérébrales, le remodelage de la paroi artérielle est toujours accompagné d’une perte des fonctions vasodilatatrices, ce qui suggère que ces deux évènements sont au cœur d’un cercle vicieux. Nos études visent à vérifier l’hypothèse selon laquelle le remodelage de la paroi est déterminé par la fonction endothéliale au niveau cérébrovasculaire alors qu’au niveau de la carotide, le stress mécanique du pouls sanguin régule les propriétés structurales et biomécaniques. Afin de vérifier cette hypothèse, dans une première étude, nous avons sélectionné trois interventions thérapeutiques aux mécanismes d’action différents qui modulent la fonction endothéliale indirectement en diminuant le stress mécanique exercé sur la paroi via une diminution de la fréquence cardiaque. Suite à un traitement chronique de trois mois chez la souris athérosclérotique, LDLr-/-; hApoB-100+/+, l’efficacité de l’ivabradine, du métoprolol et de l’exercice physique volontaire dans la prévention de l’augmentation de la compliance cérébrovasculaire s’est avérée proportionnelle à l’étendue des bénéfices sur la fonction endothéliale. La rigidification carotidienne n’a été prévenue que par les interventions qui réduisent vraiment la fréquence cardiaque, c’est-à-dire l’ivabradine et le métoprolol. Dans une deuxième étude, nous avons confirmé nos résultats en utilisant un traitement antioxydant dans le but de cibler plus directement l’endothélium. La catéchine ne réduit pas la fréquence cardiaque, mais elle est reconnue pour protéger l’endothélium cérébral en neutralisant le stress oxydant. Ainsi, la carotide est restée rigide alors que le remodelage cérébral a été prévenu. Une technique d’imagerie novatrice, la tomographie par cohérence optique, nous a permis de valider nos observations in vivo et de proposer que la catéchine prévient l’hypoperfusion du cerveau en protégeant la fonction endothéliale et l’intégrité de la paroi vasculaire cérébrale. Finalement, les deux études identifient la métalloprotéinase de type 9 comme un joueur potentiellement impliqué dans l’augmentation de la compliance cérébrovasculaire. Nos études démontrent que les changements structuraux et biomécaniques affectant la paroi des artères cérébrales sont indubitablement dépendants de l’endothélium alors que dans la carotide, le stress mécanique est le paramètre le plus déterminant. Somme toute, en protégeant indirectement l’endothélium cérébral on empêche les processus de remodelage, telle que l’activation de la métalloprotéinase de type 9. De nombreuses études ont suggéré l’implication des dysfonctions cérébrovasculaires dans la maladie d’Alzheimer. En effet, les affections vasculaires qui compromettent chroniquement le débit sanguin cérébral, telles la dysfonction endothéliale et la réduction de la lumière artérielle, vont entraîner un déficit métabolique des neurones à l’origine de la neurodégénérescence. Les traitements préventifs cardioprotecteurs, tels que l’ivabradine, l’exercice physique et la catéchine améliorent la fonction endothéliale, la structure et la biomécanique des artères cérébrales, et pourraient donc prévenir l’hypoperfusion chronique du cerveau et le déclin cognitif dans l’athérosclérose.Large artery stiffness and endothelial dysfunction are markers of atherosclerosis. Stiffening of the carotid arteries contrast with the paradoxical increase in distensibility of cerebral arteries that was reported in the presence of risk factors for cardiovascular diseases, such as hypertension. However, our knowledge concerning the influence of atherosclerosis on cerebrovascular compliance and structure remains incomplete. By targeting the endothelium, atherosclerosis induces a severe cerebral endothelial dysfunction affecting chronically the cerebral blood flow and potentially leading to cognitive dysfunctions. Few studies have shown that the paradoxical increase in cerebrovascular distensibility is consistently reported in animal model of risk factors for cardiovascular diseases exhibiting a cerebral endothelial dysfunction. That being said, we hypothesized that the compliance and structure of cerebral arteries is essentially controlled by the endothelium. To validate our hypothesis, in a first study, we selected three distinct therapeutic approaches that modulated the cerebral endothelial function and the mechanical stress imposed to the vascular wall by lowering heart rate in a mouse model of atherosclerosis, LDLr-/-; hApoB-100+/+ during three months. Ivabradine, metroprolol and voluntary physical training protected, with different efficiencies, the cerebral flow-mediated dilation and this was reflected by a prevention, or not, of the increase in compliance. A 13.5 % heart rate reduction with ivabradine and metoprolol limited carotid artery stiffening. Voluntary physical training did not induce an overall reduction of heart rate explaining the lack of effect on carotid mechanics and suggesting that carotids compliance is more influenced by the mechanical stress imposed to the vascular wall by the cardiac cycle. In a second study, we confirmed our previous findings using a diatery approach that targeted more directly the endothelium, the polyphenol antioxidant catechin. Catechin was previously proven, by us and others, to reverse endothelial dysfunction, reduce inflammation and neutralize reactive oxygen species in diverse vascular beds from animal models of atherosclerosis. Accordingly, we found that catechin prevents adverse cerebral wall remodeling but, again, without a significant heart rate reduction, carotids remained stiff. We also integrated a new live imaging technology allowing us to confirm our findings in vivo and to demonstrate that endothelial, structural and mechanical protection by catechin can result in an improvement of basal cerebral blood flow. Finally, both studies identified metalloproteinase -9 as a potential player in the process leading the weakening of the cerebral artery walls. Taken together, our studies highlight that structural and biomechanical alterations are genuinely triggered by endothelial dysfunction. In carotids, mechanicals stress seems to be the main factor controlling remodeling. In essence, indirect protection of the endothelium impedes in cerebral vessels the remodeling processes, such as the activation of metalloproteinase -9. Numerous studies have revealed that vascular, especially cerebral endothelial dysfunction is implicated in the pathogenesis of Alzheimer’s disease. When brain perfusion is compromised, the suboptimal energy delivery causes neuronal death. Deleterious cerebrovascular outcomes that promote the impairment of vasodilation and the encroachment of the lumen will limit cerebral blood flow in a chronic manner. Chronic treatment with ivabradine, voluntary physical training and catechin preserved the endothelial function, the structure and the mechanics of cerebral arteries, which guarantees a closer management of cerebral blow flow in atherosclerotic mice and a reduce propensity to develop cognitive deficiency

    Pulse pressure-dependent cerebrovascular eNOS regulation in mice

    No full text
    Arterial blood pressure is oscillatory; whether pulse pressure (PP) regulates cerebral artery myogenic tone (MT) and endothelial function is currently unknown. To test the impact of PP on MT and dilation to flow (FMD) or to acetylcholine (Ach), isolated pressurized mouse posterior cerebral arteries were subjected to either static pressure (SP) or a physiological PP (amplitude: 30 mm Hg; frequency: 550 bpm). Under PP, MT was significantly higher than in SP conditions (p < 0.05) and was not affected by eNOS inhibition. In contrast, under SP, eNOS inhibition increased (p < 0.05) MT to levels observed under PP, suggesting that PP may inhibit eNOS. At a shear stress of 20 dyn/cm(2), FMD was lower (p < 0.05) under SP than PP. Under SP, eNOS-dependent [Formula: see text] production contributed to FMD, while under PP, eNOS-dependent NO was responsible for FMD, indicating that PP favours eNOS coupling. Differences in FMD between pressure conditions were abolished after NOX2 inhibition. In contrast to FMD, Ach-induced dilations were higher (p < 0.05) under SP than PP. Reactive oxygen species scavenging reduced (p < 0.05) Ach-dependent dilations under SP, but increased (p < 0.05) them under PP; hence, under PP, Ach promotes ROS production and limits eNOS-derived NO activity. In conclusion, PP finely regulates eNOS, controlling cerebral artery reactivity

    Catechin Prevents Severe Dyslipidemia-Associated Changes in Wall Biomechanics of Cerebral Arteries in Ldlr-/-:Hapob(+/+) Mice and Improves Cerebral Blood Flow

    No full text
    Endothelial dysfunction and oxidative stress contribute to the atherosclerotic process that includes stiffening of large peripheral arteries. In contrast, our laboratory previously reported a paradoxical increase in cerebro-vascular compliance in LDLr(−/−):hApoB(+/+) atherosclerotic (ATX) mice (7). We hypothesized that prevention of cerebral artery endothelial dysfunction with a chronic dietary antioxidant intake would normalize the changes in cerebral artery wall structure and biomechanics and prevent the decline in basal cerebral blood flow associated with atherosclerosis. Three-month-old ATX mice were treated, or not, for 3 mo with the polyphenol (+)-catechin (CAT; 30 mg·kg(−1)·day(−1)) and compared with wild-type controls. In isolated, pressurized cerebral arteries from ATX mice, CAT prevented endothelial dysfunction (deterioration of endothelium-dependent, flow-mediated dilations; P < 0.05), the inward hypertrophic structural remodeling (increase in the wall-to-lumen ratio; P < 0.05), and the rise in cerebrovascular compliance (rightward shift of the stress-strain curve measured in passive conditions, reflecting mechanical properties of the arterial wall; P < 0.05). Doppler optical coherence tomography imaging in vivo confirmed these findings, showing that cerebral compliance was higher in ATX mice and normalized by CAT (P < 0.05). CAT also prevented basal cerebral hypoperfusion in ATX mice (P < 0.05). Active remodeling of the cerebrovascular wall in ATX mice was further suggested by the increase (P < 0.05) in pro-metalloproteinase-9 activity, which was normalized by CAT. We conclude that, by preserving the endothelial function, a chronic treatment with CAT prevents the deleterious effect of severe dyslipidemia on cerebral artery wall structure and biomechanical properties, contributing to preserving resting cerebral blood flow

    Ivabradine and metoprolol differentially affect cardiac glucose metabolism despite similar heart rate reduction in a mouse model of dyslipidemia

    No full text
    International audienceWhile heart rate reduction (HRR) is a target for the management of patients with heart disease, contradictory results were reported using ivabradine, which selectively inhibits the pacemaker If current, vs. β-blockers like metoprolol. This study aimed at testing whether similar HRR with ivabradine vs. metoprolol differentially modulates cardiac energy substrate metabolism, a factor determinant for cardiac function, in a mouse model of dyslipidemia (hApoB(+/+);LDLR(-/-)). Following a longitudinal study design, we used 3- and 6-mo-old mice, untreated or treated for 3 mo with ivabradine or metoprolol. Cardiac function was evaluated in vivo and ex vivo in working hearts perfused with (13)C-labeled substrates to assess substrate fluxes through energy metabolic pathways. Compared with 3-mo-old, 6-mo-old dyslipidemic mice had similar cardiac hemodynamics in vivo but impaired (P \textless 0.001) contractile function (aortic flow: -45%; cardiac output: -34%; stroke volume: -35%) and glycolysis (-24%) ex vivo. Despite inducing a similar 10% HRR, ivabradine-treated hearts displayed significantly higher stroke volume values and glycolysis vs. their metoprolol-treated counterparts ex vivo, values for the ivabradine group being often not significantly different from 3-mo-old mice. Further analyses highlighted additional significant cardiac alterations with disease progression, namely in the total tissue level of proteins modified by O-linked N-acetylglucosamine (O-GlcNAc), whose formation is governed by glucose metabolism via the hexosamine biosynthetic pathway, which showed a similar pattern with ivabradine vs. metoprolol treatment. Collectively, our results emphasize the implication of alterations in cardiac glucose metabolism and signaling linked to disease progression in our mouse model. Despite similar HRR, ivabradine, but not metoprolol, preserved cardiac function and glucose metabolism during disease progression

    MINI-FOCUS: INFLAMMATION IN CARDIAC INJURY The IL-1b Antibody Gevokizumab Limits Cardiac Remodeling and Coronary Dysfunction in Rats With Heart Failure

    No full text
    International audienceHIGHLIGHTS Immediate IL-1b antibody gevokizumab administration reduces ischemia/ reperfusion related infarct size. Immediate and late IL-1b antibody gevokizumab administration improves heart failure related left ventricular remodeling. IL-1b antibody gevokizumab improves heart failure related coronary dysfunction. Fro

    The IL-1β Antibody Gevokizumab Limits Cardiac Remodeling and Coronary Dysfunction in Rats With Heart Failure

    No full text
    This study reports preclinical data showing that the interleukin (IL)-1β modulation is a new promising target in the pathophysiological context of heart failure. Indeed, in nondiabetic Wistar and diabetic Goto-Kakizaki rats with chronic heart failure induced by myocardial infarction, administration of the IL-1β antibody gevokizumab improves ‘surrogate’ markers of survival (i.e., left ventricular remodeling, hemodynamics, and function as well as coronary function). However, whether IL-1β modulation per se or in combination with standard treatments of heart failure improves long-term outcome in human heart failure remains to be determined
    corecore