9 research outputs found

    Lymphatic and Immune Cell Cross-Talk Regulates Cardiac Recovery After Experimental Myocardial Infarction

    Get PDF
    Objective: Lymphatics play an essential pathophysiological role in promoting fluid and immune cell tissue clearance. Conversely, immune cells may influence lymphatic function and remodeling. Recently, cardiac lymphangiogenesis has been proposed as a therapeutic target to prevent heart failure after myocardial infarction (MI). We investigated the effects of gene therapy to modulate cardiac lymphangiogenesis post-MI in rodents. Second, we determined the impact of cardiac-infiltrating T cells on lymphatic remodeling in the heart. Approach and Results: Comparing adenoviral versus adeno-associated viral gene delivery in mice, we found that only sustained VEGF (vascular endothelial growth factor)-C(C156S)therapy, achieved by adeno-associated viral vectors, increased cardiac lymphangiogenesis, and led to reduced cardiac inflammation and dysfunction by 3 weeks post-MI. Conversely, inhibition of VEGF-C/-D signaling, through adeno-associated viral delivery of soluble VEGFR3 (vascular endothelial growth factor receptor 3), limited infarct lymphangiogenesis. Unexpectedly, this treatment improved cardiac function post-MI in both mice and rats, linked to reduced infarct thinning due to acute suppression of T-cell infiltration. Finally, using pharmacological, genetic, and antibody-mediated prevention of cardiac T-cell recruitment in mice, we discovered that both CD4(+)and CD8(+)T cells potently suppress, in part through interferon-gamma, cardiac lymphangiogenesis post-MI. Conclusions: We show that resolution of cardiac inflammation after MI may be accelerated by therapeutic lymphangiogenesis based on adeno-associated viral gene delivery of VEGF-C-C156S. Conversely, our work uncovers a major negative role of cardiac-recruited T cells on lymphatic remodeling. Our results give new insight into the interconnection between immune cells and lymphatics in orchestration of cardiac repair after injury.Peer reviewe

    Role of lymphangiogenesis in cardiac inflammation and the impact of inflammation on cardiac lymphatic network remodeling

    No full text
    Les vaisseaux lymphatiques jouent un rôle essentiel durant l’inflammation en réabsorbant lesantigènes et les cellules immunitaires. En revanche les médiateurs inflammatoires peuventimpacter les lymphatiques. Nous étudions si la lymphangiogenèse thérapeutique cardiaquepermet de restaurer la réabsorption des cellules immunitaires après un infarctus du myocarde(IDM) et d’autre part l’impact des cellules immunitaires sur le remodelage du réseaulymphatique post-IDM chez les rongeurs.La délivrance du vascular endothelial growth factor-CC152S (VEGF-CC152S) spécifique duVEGFR3 et encapsulé dans des microparticules d’albumine-alginate a accéléré lalymphangiogenèse d’une façon dose-dépendante et a limité le remodelage post-IDM des précollecteurs chez le rat. Par conséquent, l’œdème myocardique, la fibrose, l’inflammation et ladysfonction ont été atténués. D’autre part, nous avons montré que la thérapie VEGF-Créalisée par un vecteur viral adéno-associé (AAV9) a permis de stimuler également lalymphangiogenèse cardiaque, de réduire l’inflammation et d’atténuer la dysfonction cardiaqueà 3 semaines post-IDM chez la souris. Finalement, en utilisant un agoniste de la S1P,favorisant la séquestration ganglionnaire des lymphocytes T, nous avons trouvé que leslymphocytes TCD4+ contribuent à un remodelage lymphatique délétère durant les premièressemaines post-IDM.En conclusion, la stimulation de la lymphangiogenèse cardiaque atténue l’œdème ainsi quel’inflammation cardiaque chronique et améliore la fonction cardiaque post-IDM. Au contraire,les lymphocytes T jouent un rôle négatif sur le remodelage lymphatique cardiaque affectant lafonction cardiaque post-IDM.Lymphatic vessels play an essential role during inflammation by promoting both antigen andimmune cell reuptake. Conversely, inflammatory mediators may impact lymphatics. Here, weinvestigated whether therapeutic cardiac lymphangiogenesis restores cardiac immune celltrafficking after myocardial infarction (MI) in mice, and conversely the impact of immunecells on cardiac lymphatic remodeling post-MI.We investigated cardiac lymphatic structure and function in rodents post-MI. Intramyocardialtargeted delivery in rats of the vascular endothelial growth factor receptor 3–selectivedesigner protein VEGF-CC152S, using albumin-alginate microparticles, accelerated cardiaclymphangiogenesis in a dose-dependent manner and limited precollector remodeling post-MI.As a result, myocardial fluid balance was improved, and cardiac inflammation, fibrosis, anddysfunction were attenuated. Similarly, in mice we found that sustained VEGF-C genetherapy, achieved by adeno-associated viral (AAV9) gene delivery, increased cardiaclymphangiogenesis. This selective lymphatic expansion reduced cardiac inflammation andimproved cardiac function by 3 weeks post-MI. Finally, using an S1P agonist to acutely prevent Tcell mobilization post-MI in mice, we found that cardiac-infiltrating T lymphocytes, notably CD4+cells, contribute to the poor endogenous lymphangiogenic response during the first weeks post-MI.Our data show that while therapeutic lymphangiogenesis accelerates resolution of cardiacedema and inflammation, cardiac-infiltrating T cells limit endogenous lymphangiogenesispost-MI. Our findings reveal that inflammation and lymphangiogenesis are mechanisticallyinterconnected in remodeling hearts, and suggest that beneficial cardiac lymphatic repair maybe achieved by sustained lymphangiogenic protein or gene therapy or by limiting cardiac Tcell recruitment after cardiac injury

    Role de la lymphangiogenèse dans l'inflammation cardiaque et effet de l'inflammation sur le réseau lymphatique cardiaque

    No full text
    Lymphatic vessels play an essential role during inflammation by promoting both antigen andimmune cell reuptake. Conversely, inflammatory mediators may impact lymphatics. Here, weinvestigated whether therapeutic cardiac lymphangiogenesis restores cardiac immune celltrafficking after myocardial infarction (MI) in mice, and conversely the impact of immunecells on cardiac lymphatic remodeling post-MI.We investigated cardiac lymphatic structure and function in rodents post-MI. Intramyocardialtargeted delivery in rats of the vascular endothelial growth factor receptor 3–selectivedesigner protein VEGF-CC152S, using albumin-alginate microparticles, accelerated cardiaclymphangiogenesis in a dose-dependent manner and limited precollector remodeling post-MI.As a result, myocardial fluid balance was improved, and cardiac inflammation, fibrosis, anddysfunction were attenuated. Similarly, in mice we found that sustained VEGF-C genetherapy, achieved by adeno-associated viral (AAV9) gene delivery, increased cardiaclymphangiogenesis. This selective lymphatic expansion reduced cardiac inflammation andimproved cardiac function by 3 weeks post-MI. Finally, using an S1P agonist to acutely prevent Tcell mobilization post-MI in mice, we found that cardiac-infiltrating T lymphocytes, notably CD4+cells, contribute to the poor endogenous lymphangiogenic response during the first weeks post-MI.Our data show that while therapeutic lymphangiogenesis accelerates resolution of cardiacedema and inflammation, cardiac-infiltrating T cells limit endogenous lymphangiogenesispost-MI. Our findings reveal that inflammation and lymphangiogenesis are mechanisticallyinterconnected in remodeling hearts, and suggest that beneficial cardiac lymphatic repair maybe achieved by sustained lymphangiogenic protein or gene therapy or by limiting cardiac Tcell recruitment after cardiac injury.Les vaisseaux lymphatiques jouent un rôle essentiel durant l’inflammation en réabsorbant lesantigènes et les cellules immunitaires. En revanche les médiateurs inflammatoires peuventimpacter les lymphatiques. Nous étudions si la lymphangiogenèse thérapeutique cardiaquepermet de restaurer la réabsorption des cellules immunitaires après un infarctus du myocarde(IDM) et d’autre part l’impact des cellules immunitaires sur le remodelage du réseaulymphatique post-IDM chez les rongeurs.La délivrance du vascular endothelial growth factor-CC152S (VEGF-CC152S) spécifique duVEGFR3 et encapsulé dans des microparticules d’albumine-alginate a accéléré lalymphangiogenèse d’une façon dose-dépendante et a limité le remodelage post-IDM des précollecteurs chez le rat. Par conséquent, l’œdème myocardique, la fibrose, l’inflammation et ladysfonction ont été atténués. D’autre part, nous avons montré que la thérapie VEGF-Créalisée par un vecteur viral adéno-associé (AAV9) a permis de stimuler également lalymphangiogenèse cardiaque, de réduire l’inflammation et d’atténuer la dysfonction cardiaqueà 3 semaines post-IDM chez la souris. Finalement, en utilisant un agoniste de la S1P,favorisant la séquestration ganglionnaire des lymphocytes T, nous avons trouvé que leslymphocytes TCD4+ contribuent à un remodelage lymphatique délétère durant les premièressemaines post-IDM.En conclusion, la stimulation de la lymphangiogenèse cardiaque atténue l’œdème ainsi quel’inflammation cardiaque chronique et améliore la fonction cardiaque post-IDM. Au contraire,les lymphocytes T jouent un rôle négatif sur le remodelage lymphatique cardiaque affectant lafonction cardiaque post-IDM

    Role de la lymphangiogenèse dans l'inflammation cardiaque et effet de l'inflammation sur le réseau lymphatique cardiaque

    No full text
    Lymphatic vessels play an essential role during inflammation by promoting both antigen andimmune cell reuptake. Conversely, inflammatory mediators may impact lymphatics. Here, weinvestigated whether therapeutic cardiac lymphangiogenesis restores cardiac immune celltrafficking after myocardial infarction (MI) in mice, and conversely the impact of immunecells on cardiac lymphatic remodeling post-MI.We investigated cardiac lymphatic structure and function in rodents post-MI. Intramyocardialtargeted delivery in rats of the vascular endothelial growth factor receptor 3–selectivedesigner protein VEGF-CC152S, using albumin-alginate microparticles, accelerated cardiaclymphangiogenesis in a dose-dependent manner and limited precollector remodeling post-MI.As a result, myocardial fluid balance was improved, and cardiac inflammation, fibrosis, anddysfunction were attenuated. Similarly, in mice we found that sustained VEGF-C genetherapy, achieved by adeno-associated viral (AAV9) gene delivery, increased cardiaclymphangiogenesis. This selective lymphatic expansion reduced cardiac inflammation andimproved cardiac function by 3 weeks post-MI. Finally, using an S1P agonist to acutely prevent Tcell mobilization post-MI in mice, we found that cardiac-infiltrating T lymphocytes, notably CD4+cells, contribute to the poor endogenous lymphangiogenic response during the first weeks post-MI.Our data show that while therapeutic lymphangiogenesis accelerates resolution of cardiacedema and inflammation, cardiac-infiltrating T cells limit endogenous lymphangiogenesispost-MI. Our findings reveal that inflammation and lymphangiogenesis are mechanisticallyinterconnected in remodeling hearts, and suggest that beneficial cardiac lymphatic repair maybe achieved by sustained lymphangiogenic protein or gene therapy or by limiting cardiac Tcell recruitment after cardiac injury.Les vaisseaux lymphatiques jouent un rôle essentiel durant l’inflammation en réabsorbant lesantigènes et les cellules immunitaires. En revanche les médiateurs inflammatoires peuventimpacter les lymphatiques. Nous étudions si la lymphangiogenèse thérapeutique cardiaquepermet de restaurer la réabsorption des cellules immunitaires après un infarctus du myocarde(IDM) et d’autre part l’impact des cellules immunitaires sur le remodelage du réseaulymphatique post-IDM chez les rongeurs.La délivrance du vascular endothelial growth factor-CC152S (VEGF-CC152S) spécifique duVEGFR3 et encapsulé dans des microparticules d’albumine-alginate a accéléré lalymphangiogenèse d’une façon dose-dépendante et a limité le remodelage post-IDM des précollecteurs chez le rat. Par conséquent, l’œdème myocardique, la fibrose, l’inflammation et ladysfonction ont été atténués. D’autre part, nous avons montré que la thérapie VEGF-Créalisée par un vecteur viral adéno-associé (AAV9) a permis de stimuler également lalymphangiogenèse cardiaque, de réduire l’inflammation et d’atténuer la dysfonction cardiaqueà 3 semaines post-IDM chez la souris. Finalement, en utilisant un agoniste de la S1P,favorisant la séquestration ganglionnaire des lymphocytes T, nous avons trouvé que leslymphocytes TCD4+ contribuent à un remodelage lymphatique délétère durant les premièressemaines post-IDM.En conclusion, la stimulation de la lymphangiogenèse cardiaque atténue l’œdème ainsi quel’inflammation cardiaque chronique et améliore la fonction cardiaque post-IDM. Au contraire,les lymphocytes T jouent un rôle négatif sur le remodelage lymphatique cardiaque affectant lafonction cardiaque post-IDM

    Role of M2-like macrophage recruitment during angiogenic growth factor therapy

    No full text
    International audienceTherapeutic angiogenesis has yet to fulfill its promise for the clinical treatment of ischemic diseases. Given the impact of macrophages during pathophysiological angiogenesis, we asked whether macrophages may similarly modulate vascular responses to targeted angiogenic therapies. Mouse matrigel plug assay and rat myocardial infarction (MI) model were used to assess angiogenic therapy with either VEGF-A or FGF-2 with HGF (F+H) delivered locally via albumin-alginate microcapsules. The infiltration of classical M1-type and alternative M2-like macrophages was assessed. Clodronate was used to prevent macrophage recruitment, and the VEGFR2 blocking antibody, DC101, to prevent VEGF-A signaling. At 3 weeks after matrigel implantation, the combination therapy (F+H) led to increased total, and specifically M2-like, macrophage infiltration versus control and VEGF-A plugs, correlating with the angiogenic response. In contrast, VEGF-A preferential recruited M1-type macrophages. In agreement with a direct role of M2-like macrophages in F+H-induced vessel growth, clodronate radically decreased angiogenesis. Further, DC101 reduced F+H-induced angiogenesis, without altering macrophage infiltration, revealing macrophage-derived VEGF-A as a crucial determinant of tissue responsiveness. Similarly, increased cardiac M2-like macrophage infiltration was found following F+H therapy post-MI, with strong correlation between macrophage levels and angiogenic and arteriogenic responses. In conclusion, M2-like macrophages play a decisive role, linked to VEGF-A production, in regulation of tissue responsiveness to angiogenic therapies including the combination of F+H. Our data suggest that future attempts at therapeutic revascularization in ischemic patients might benefit from coupling targeted growth factor delivery with either direct or indirect approaches to recruit pro-angiogenic macrophages in order to maximize therapeutic angiogenic/arteriogenic responses

    Regulation and impact of cardiac lymphangiogenesis in pressure-overload-induced heart failure

    No full text
    International audienceAims: Lymphatics are essential for cardiac health, and insufficient lymphatic expansion (lymphangiogenesis) contributes to development of heart failure (HF) after myocardial infarction. However, the regulation and impact of lymphangiogenesis in non-ischaemic cardiomyopathy following pressure-overload remains to be determined. Here, we investigated cardiac lymphangiogenesis following transversal aortic constriction (TAC) in C57Bl/6 and Balb/c mice, and in end-stage HF patients.Methods and results: Cardiac function was evaluated by echocardiography, and cardiac hypertrophy, lymphatics, inflammation, oedema, and fibrosis by immunohistochemistry, flow cytometry, microgravimetry, and gene expression analysis. Treatment with neutralizing anti-VEGFR3 antibodies was applied to inhibit cardiac lymphangiogenesis in mice. We found that VEGFR3-signalling was essential to prevent cardiac lymphatic rarefaction after TAC in C57Bl/6 mice. While anti-VEGFR3-induced lymphatic rarefaction did not significantly aggravate myocardial oedema post-TAC, cardiac immune cell levels were increased, notably myeloid cells at 3 weeks and T lymphocytes at 8 weeks. Moreover, whereas inhibition of lymphangiogenesis did not aggravate interstitial fibrosis, it increased perivascular fibrosis and accelerated development of left ventricular (LV) dilation and dysfunction. In clinical HF samples, cardiac lymphatic density tended to increase, although lymphatic sizes decreased, notably in patients with dilated cardiomyopathy. Similarly, comparing C57Bl/6 and Balb/c mice, lymphatic remodelling post-TAC was linked to LV dilation rather than to hypertrophy. The striking lymphangiogenesis in Balb/c was associated with reduced cardiac levels of macrophages, B cells, and perivascular fibrosis at 8 weeks post-TAC, as compared with C57Bl/6 mice that displayed weak lymphangiogenesis. Surprisingly, however, it did not suffice to resolve myocardial oedema, nor prevent HF development.Conclusions: We demonstrate for the first time that endogenous lymphangiogenesis limits TAC-induced cardiac inflammation and perivascular fibrosis, delaying HF development in C57Bl/6 but not in Balb/c mice. While the functional impact of lymphatic remodelling remains to be determined in HF patients, our findings suggest that under settings of pressure-overload poor cardiac lymphangiogenesis may accelerate HF development
    corecore