309 results on '"Patrick, Lacolley"'
Search Results
2. Increased atherosclerotic plaque in AOC3 knock-out in ApoE−/− mice and characterization of AOC3 in atherosclerotic human coronary arteries
- Author
-
Anna Filip, Soraya Taleb, Rümeyza Bascetin, Mohammad Jahangiri, Matthieu Bardin, Cindy Lerognon, Bruno Fève, Patrick Lacolley, Sirpa Jalkanen, and Nathalie Mercier
- Subjects
semicarbazide-sensitive amine oxidase ,vascular smooth muscle cells ,atherosclerosis ,inflammation ,vascular adhesion protein-1 ,amine oxidase copper containing 3 ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
IntroductionAmine oxidase copper containing 3 (AOC3) displays adhesion between leukocytes and endothelial cells and enzymatic functions. Given its controversial role in atherogenesis, we proposed to investigate the involvement of AOC3 in the formation of atherosclerotic plaques in ApoE−/−AOC3−/− mice and human coronary arteries.MethodsLesions, contractile markers, and AOC3 were studied in aortic tissues from 15- and 25-week-old mice and different stages of human coronary atherosclerotic arteries by immunohistochemistry (IHC) and/or western blot. Human VSMCs, treated or not with LJP1586, an AOC3 inhibitor, were used to measure differentiation markers by qPCR. AOC3 co-localization with specific cell markers was studied by using confocal microscopy in mice and human samples.ResultsAt 15 weeks old, the absence of AOC3 was associated with increased lesion size, α-SMA, and CD3 staining in the plaque independently of a cholesterol modification. At 25 weeks old, advanced plaques were larger with equivalent staining for α-SMA while CD3 increased in the media from ApoE−/−AOC3−/− mice. At both ages, the macrophage content of the lesion was not modified. Contractile markers decreased whereas MCP-1 appeared augmented only in the 15-week-old ApoE−/−AOC3. AOC3 is mainly expressed by mice and human VSMC is slightly expressed by endothelium but not by macrophages.ConclusionAOC3 knock-out increased atherosclerotic plaques at an early stage related to a VSMC dedifferentiation associated with a higher T cells recruitment in plaques explained by the MCP-1 augmentation. This suggests that AOC3 may have an important role in atherosclerosis independent of its canonical inflammatory effect. The dual role of AOC3 impacts therapeutic strategies using pharmacological regulators of SSAO activity.
- Published
- 2022
- Full Text
- View/download PDF
3. Estrogen Receptor and Vascular Aging
- Author
-
Morgane Davezac, Melissa Buscato, Rana Zahreddine, Patrick Lacolley, Daniel Henrion, Francoise Lenfant, Jean-Francois Arnal, and Coralie Fontaine
- Subjects
estradiol ,estrogen receptor ,menopause ,atherosclerosis ,endothelium ,vascular aging ,Geriatrics ,RC952-954.6 - Abstract
Cardiovascular diseases remain an age-related pathology in both men and women. These pathologies are 3-fold more frequent in men than in women before menopause, although this difference progressively decreases after menopause. The vasculoprotective role of estrogens are well established before menopause, but the consequences of their abrupt decline on the cardiovascular risk at menopause remain debated. In this review, we will attempt to summarize the main clinical and experimental studies reporting the protective effects of estrogens against cardiovascular diseases, with a particular focus on atherosclerosis, and the impact of aging and estrogen deprivation on their endothelial actions. The arterial actions of estrogens, but also part of that of androgens through their aromatization into estrogens, are mediated by the estrogen receptor (ER)α and ERβ. ERs belong to the nuclear receptor family and act by transcriptional regulation in the nucleus, but also exert non-genomic/extranuclear actions. Beside the decline of estrogens at menopause, abnormalities in the expression and/or function of ERs in the tissues, and particularly in arteries, could contribute to the failure of classic estrogens to protect arteries during aging. Finally, we will discuss how recent insights in the mechanisms of action of ERα could contribute to optimize the hormonal treatment of the menopause.
- Published
- 2021
- Full Text
- View/download PDF
4. Number and Replating Capacity of Endothelial Colony‐Forming Cells are Telomere Length Dependent: Implication for Human Atherogenesis
- Author
-
Simon Toupance, Stéphanie Simonici, Carlos Labat, Chloé Dumoulin, Tsung‐Po Lai, Cécile Lakomy, Véronique Regnault, Patrick Lacolley, Françoise Dignat George, Florence Sabatier, Abraham Aviv, and Athanase Benetos
- Subjects
aging ,endothelial progenitor cell ,telomere ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Background Short leukocyte telomere length (TL) is associated with atherosclerotic cardiovascular disease. Endothelial repair plays a key role in the development of atherosclerosis. The objective was to examine associations between TL and proliferative dynamics of endothelial colony‐forming cells (ECFCs), which behave as progenitor cells displaying endothelial repair activity. Methods and Results To isolate ECFCs, we performed a clonogenic assay on blood samples from 116 participants (aged 24–94 years) in the TELARTA (Telomere in Arterial Aging) cohort study. We detected no ECFC clones in 29 (group 1), clones with no replating capacity in other 29 (group 2), and clones with replating capacity in the additional 58 (group 3). Leukocyte TL was measured by Southern blotting and ECFCs (ECFC‐TL). Age‐ and sex‐adjusted leukocyte TL (mean±SEM) was the shortest in group 1 (6.51±0.13 kb), longer in group 2 (6.69±0.13 kb), and the longest in group 3 (6.78±0.09 kb) (P
- Published
- 2021
- Full Text
- View/download PDF
5. Covid-19 Effects on ARTErial StIffness and Vascular AgeiNg: CARTESIAN Study Rationale and Protocol
- Author
-
Rosa Maria Bruno, Bart Spronck, Bernhard Hametner, Alun Hughes, Patrick Lacolley, Christopher C. Mayer, Maria Lorenza Muiesan, Chakravarthi Rajkumar, Dimitrios Terentes-Printzios, Thomas Weber, Tine Willum Hansen, and Pierre Boutouyrie
- Subjects
COVID-19 ,coronavirus ,inflammation ,vascular ageing ,arterial stiffness ,Specialties of internal medicine ,RC581-951 ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
In December 2019, an outbreak of pneumonia caused by a novel Coronavirus (COVID-19) spread rapidly worldwide. Although the clinical manifestations of COVID-19 are dominated by respiratory symptoms, the cardiovascular system is extensively affected at multiple levels. Due to the unprecedented consequences of the COVID-19 pandemic, the ARTERY society decided to launch the Covid-19 effects on ARTErial StIffness and vascular AgeiNg (CARTESIAN) study — the first international multicentre study into the effects of COVID-19 on non-invasive biomarkers of vascular ageing. The main study objective is to evaluate the presence of Early Vascular Ageing (EVA) 6 and 12 months after COVID-19 infection. Secondary objectives are to study the effect of COVID-19 disease severity on EVA, to investigate the role of psychosocial factors in COVID-19 induced EVA, and to investigate the potential modifying effect of comorbidities and chronic treatments. In the CARTESIAN study, a broad array of cardiovascular measurements, including carotid-femoral pulse wave velocity, central blood pressure, carotid ultrasound, brachial flow-mediated dilatation, will be performed. To date, 43 centres from 21 countries have agreed to participate, with an expected study population of >2500 individuals. To our knowledge, CARTESIAN will be the first study to provide insight into the relationship between COVID-19, its severity, and early vascular ageing in a large cohort, potentially enabling future care and diagnostics to be more focused on the most vulnerable.
- Published
- 2020
- Full Text
- View/download PDF
6. P.31 Von Willebrand Factor Induces Vascular Smooth Muscle Cell Proliferation and Migration Through Low Density Lipoprotein-Related Receptor Protein 4 and αvβ3 Integrin
- Author
-
Cécile V. Denis, Patrick Lacolley, Jeremy Lagrange, Peter J. Lenting, Jean-Baptiste Michel, Alexandre Raoul, and Veronique Regnault
- Subjects
VWF ,VSMC ,αvβ3 ,LRP4 ,Specialties of internal medicine ,RC581-951 ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Background and Objectives: Von Willebrand factor (VWF) is a plasma glycoprotein involved in primary hemostasis but recent data suggest additional roles beyond hemostasis in angiogenesis and potentially in vascular smooth muscle cell (VSMC) proliferation. Our aim was to investigate how VWF can modulate VSMC proliferation and identified the underlying mechanisms and the in vivo pathophysiological relevance. Methods and Results: Cultured aortic VSMCs proliferation and migration were increased in the presence of VWF. VSMCs treatment with a siRNA targeting αv integrin or the RGT-peptide blocking αvβ3 signaling completely inhibited proliferation. VWF did not bind directly to αvβ3 on VSMCs. We identified that VWF A2 domain was able to bind VSMCs. Since the low-density lipoprotein-related receptor protein (LRP) family are known to act as co-receptors we hypothesized the involvement of a member in the signaling pathway. Using the universal LRP-inhibitor (RAP), we confirmed LRP-mediated VSMC proliferation. siRNA experiments and proximity ligation assay staining identified LRP4 as the VWF-counterreceptor on VSMCs and showed co-localization between αvβ3 and LRP4. Carotid ligations were applied to VWF +/+ and −/− mice and intimal hyperplasia (IH) was measured. Less VWF−/− mice developed IH compared to VWF +/+ mice. Finally, the proliferative effect of VWF was confirmed in human atherosclerotic lesions from different vessels (aortas, carotids) showing a proximity between VWF and a-SM actin positive cells. Conclusions: VWF mediates VSMC proliferation through its A2 domain binding to the LRP4 receptor and integrin αvβ3 signaling. The decreased IH following vascular injury suggests that targeting VWF-LRP4 interactions may contribute to limit remodeling.
- Published
- 2020
- Full Text
- View/download PDF
7. Vimentin as a target for the treatment of COVID-19
- Author
-
Zhenlin Li, Denise Paulin, Patrick Lacolley, Dario Coletti, and Onnik Agbulut
- Subjects
Medicine ,Diseases of the respiratory system ,RC705-779 - Abstract
We and others propose vimentin as a possible cellular target for the treatment of COVID-19. This innovative idea is so recent that it requires further attention and debate. The significant role played by vimentin in virus-induced infection however is well established: (1) vimentin has been reported as a co-receptor and/or attachment site for SARS-CoV; (2) vimentin is involved in viral replication in cells; (3) vimentin plays a fundamental role in both the viral infection and the consequent explosive immune-inflammatory response and (4) a lower vimentin expression is associated with the inhibition of epithelial to mesenchymal transition and fibrosis. Moreover, the absence of vimentin in mice makes them resistant to lung injury. Since vimentin has a twofold role in the disease, not only being involved in the viral infection but also in the associated life-threatening lung inflammation, the use of vimentin-targeted drugs may offer a synergistic advantage as compared with other treatments not targeting vimentin. Consequently, we speculate here that drugs which decrease the expression of vimentin can be used for the treatment of patients with COVID-19 and advise that several Food and Drug Administration-approved drugs be immediately tested in clinical trials against SARS-CoV-2, thus broadening therapeutic options for this type of viral infection.
- Published
- 2020
- Full Text
- View/download PDF
8. P12 Proliferation and Procoagulant Activity of Vascular Smooth Muscle Cells from Thoracic and Abdominal Aortic Aneurysms
- Author
-
Melusine Didelot, Jeremy Lagrange, Jean-Baptiste Michel, Patrick Lacolley, and Veronique Regnault
- Subjects
Specialties of internal medicine ,RC581-951 ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
In contrast to abdominal aortic aneurysm (AAA), aneurysms of the thoracic ascending aorta (TAA) are not associated with a mural thrombus. Mechanistic understanding of the involvement of vascular smooth muscle cells (VSMCs) in thrombus formation is lacking. We aim to determine (i) whether thrombin generation at the surface of VSMCs is increased in AAA, (ii) the contribution of the avb3 integrin as a receptor for prothrombin, (iii) the implication of protease activated receptor (PAR) activation in the thrombin generation, and (iv) the capacity of thrombin to induce VSMC proliferation. Primary cultures of VSMCs were prepared from human biopsies of TAA, AAA and healthy aorta (n = 11/group). In the presence of prothrombin deficient plasma, similar low levels of thrombin were formed on the 3 types of VSMCs. In the presence of healthy plasma, thrombin generation was higher on VSMCs from AAA and lower on VSMCs from TAA compared with controls. Incubation of VSMCs with a RGT peptide inhibiting the b3 signaling reduced the amount of thrombin in the 3 groups. Incubation of healthy VSMCs with a selective PAR-1 antagonist decreased thrombin generation. This inhibition was more pronounced for AAA (36%) than from TAA (17%). VSMCs from AAA were more sensitive to thrombin-induced proliferation than VSMCs from TAA. The prothrombotic phenotype and proliferation of VSMCs from AAA compared to TAA is mediated partly by PAR-1. This argues for a contribution of VSMC in the occurrence of thrombotic events in AAA. Thus, inhibition of PAR signaling represents a new target in this complex disease.
- Published
- 2020
- Full Text
- View/download PDF
9. P11 The Role of Smooth Muscle Integrin Alpha V and TGF-Beta Pathways in Vascular Fibrosis
- Author
-
Alexandre Raoul, Ekaterina Belozertseva, Huguette Louis, Zhenlin Li, Veronique Regnault, and Patrick Lacolley
- Subjects
Specialties of internal medicine ,RC581-951 ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
It has been demonstrated that arterial stiffness is linked to arterial fibrosis manifested by increased collagen and other extracellular matrix synthesis in smooth muscle cells (SMC). Transmembrane receptors integrins mediating cell-cell and cell-matrix signalling pathways are involved in tissue fibrosis. We study the role of one integrin subunit av in angiotensin II (AngII)-induced SMC proliferation and arterial fibrosis and stiffness using SMC specific knock-out av mouse model (avSMKO) induced in adult mice by injection of tamoxifen. There is no difference in vascular fibrosis in basal conditions between control and mutant mice. However, decreased arterial fibrosis is observed in avSMKO mutant mice after 28-day perfusion of AngII. Analysis of RNA from aorta of control and mutant mice by Affymetrix microarrays indicated an alteration of the TGF-β pathway in AngII-treated mutant mice. In order to examine the mechanism associated to the decreased fibrosis in vascular SMC from avSMKO mice, isolated VSMC from the aorta of control and avSMKO mice were treated with TGF-β1 or AngII. The results indicated that these two treatments increased the expression of collagen, fibronectin and integrin αν at the protein and RNA levels as well as the phosphorylation of ERK and smad2/3 in the control cells. Inactivation of integrin αν partly inhibited all above effects induced by TGF-β1 and AngII. Our study indicates a role of av and TGF-β1 in the arterial fibrogenesis. Therefore, av signaling could be a therapeutic target against arterial fibrosis and stiffness in pathological conditions.
- Published
- 2020
- Full Text
- View/download PDF
10. Correction: Exome-wide association study reveals novel susceptibility genes to sporadic dilated cardiomyopathy.
- Author
-
Ulrike Esslinger, Sophie Garnier, Agathe Korniat, Carole Proust, Georgios Kararigas, Martina Müller-Nurasyid, Jean-Philippe Empana, Michael P Morley, Claire Perret, Klaus Stark, Alexander G Bick, Sanjay K Prasad, Jennifer Kriebel, Jin Li, Laurence Tiret, Konstantin Strauch, Declan P O'Regan, Kenneth B Marguiles, Jonathan G Seidman, Pierre Boutouyrie, Patrick Lacolley, Xavier Jouven, Christian Hengstenberg, Michel Komajda, Hakon Hakonarson, Richard Isnard, Eloisa Arbustini, Harald Grallert, Stuart A Cook, Christine E Seidman, Vera Regitz-Zagrosek, Thomas P Cappola, Philippe Charron, François Cambien, and Eric Villard
- Subjects
Medicine ,Science - Abstract
[This corrects the article DOI: 10.1371/journal.pone.0172995.].
- Published
- 2020
- Full Text
- View/download PDF
11. Molecular determinants of arterial stiffness
- Author
-
Stéphane Laurent, Céline Fassot, Patrick Lacolley, and Pierre Boutouyrie
- Subjects
Hypertension ,Monogenic disease ,Extra-cellular matrix ,Biomechanics ,Gene profile ,Specialties of internal medicine ,RC581-951 ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Arterial stiffness has an independent predictive value for cardiovascular events. This review proposes an integrated view of the molecular determinants of arterial stiffness, based on a candidate gene approach, an analysis of the structure–function relationship in hypertension, and studies on gene expression profile in humans. In monogenic diseases of connective tissue (Marfan, Williams, and Ehlers–Danlos syndromes) and corresponding animal models, the precise characterization of arterial phenotype allows understanding the influence of abnormal, genetically determined, wall components on arterial stiffness. These studies underline the role of extra-cellular matrix signaling in the vascular wall and the fact that elastin and collagen have not only passive elastic or rigid properties, but also are implicated in the control of SMC function. In animal models of essential hypertension (SHR and SHR-SP), the structural modifications of the arterial wall include a higher number of elastin/SMC connections, and smaller fenestrations of the internal elastic lamina, which could redistribute the mechanical load towards elastic materials. Thus, the changes in arterial wall material which accompany wall hypertrophy in these animals are not associated with an increased stiffness. Taken together, these data afford strong arguments to consider that arterial stiffness is not only influenced by the amount and density of stiff wall material, but mainly by its spatial organization.
- Published
- 2019
- Full Text
- View/download PDF
12. Platelet aggregation impacts thrombin generation assessed by calibrated automated thrombography
- Author
-
Mélusine Didelot, Clémence Docq, Denis Wahl, Patrick Lacolley, Véronique Regnault, and Jérémy Lagrange
- Subjects
blood coagulation disorders ,blood coagulation tests ,blood platelet disorders ,platelet activation ,platelet aggregation ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
A calibrated automated thrombogram (CAT) is performed usually with human platelet-free plasma (PFP) but may be more relevant with platelet-rich plasma (PRP). In this case, platelets are not stimulated by subendothelial molecules like collagen. Our aim was to assess the consequence of strong (collagen) or weak (ADP) induction of platelet release and aggregation on thrombin generation. Platelet aggregation in PRP was triggered with 10 µg/mL collagen or 10 µM ADP using a lumi-aggregometer. Thrombin generation curves were monitored by CAT in different conditions: PRP, PRP with activated platelets (actPRP), aggregated PRP (agPRP), aggregated platelets resuspended in autologous PFP (resPRP), PFP and PFP obtained after aggregation (agPFP). We found a 3-fold shortening of the lag time and time to peak and a marked increase in velocity and thrombin peak without changes in endogenous thrombin potential (ETP) in agPRP with both agonists compared with PRP. The same holds true in agPFP but with a marked increase in ETP compared with PFP. Similar changes in the kinetics of thrombin generation were observed with actPRP-collagen and to a lesser extent in resPRP-collagen compared with PRP. By contrast, there were no modifications of the thrombin generation curves in actPRP-ADP. Alpha-2-macroglobin-thrombin complexes were unchanged in the different PRP conditions but were increased in PFP prepared from agPFP compared to control PFP. Platelet aggregation during activation by agonists other than thrombin did not increase thrombin generation but accelerated its kinetics mainly via platelet content release and platelet-derived extracellular vesicules formation. In diseases characterized by altered platelet granule content or release as well as altered platelet activation, a platelet aggregation step prior to CAT analysis may be clinically relevant to improve laboratory estimation of the bleeding/thrombotic balance.
- Published
- 2018
- Full Text
- View/download PDF
13. Circulating Endothelial Cells are Associated with Thromboembolic Events in Patients with Antiphospholipid Antibodies
- Author
-
Thomas Foret, Virginie Dufrost, Marie Heymonet, Jessie Risse, Gilbert C. Faure, Huguette Louis, Jeremy Lagrange, Patrick Lacolley, Katrien Devreese, Sébastien Gibot, Veronique Regnault, Stéphane Zuily, and Denis Wahl
- Subjects
Hematology - Abstract
Background Endothelial damage has been described in antiphospholipid antibody (aPL)-positive patients. However, it is uncertain whether circulating endothelial cells (CECs)—which are released when endothelial injury occurs—can be a marker of patients at high risk for thrombosis. Methods Ninety-seven patients with aPL and/or systemic lupus erythematosus (SLE) were included. CECs were determined by an automated CellSearch system. We also assayed plasma levels of tissue factor-bearing extracellular vesicles (TF+/EVs) and soluble triggering receptor expressed on myeloid cells 1 (sTREM-1) as markers of endothelial dysfunction/damage. Results Patients' mean age was 46.1 ± 13.9 years, 77 were women. Thirty-seven had SLE and 75 patients were suffering from antiphospholipid syndrome. Thirty-seven percent of patients presented a medical history of arterial thrombosis and 46% a history of venous thromboembolism (VTE). Thirteen patients had increased levels of CECs (>20/mL), with a mean CEC level of 48.3 ± 21.3 per mL. In univariate analysis, patients with obesity or medical history of myocardial infarction (MI), VTE, or nephropathy had a significant increased CEC level. In multivariate analysis, obesity (odds ratio [OR] = 6.07, 95% confidence interval [CI]: 1.42–25.94), VTE (OR = 7.59 [95% CI: 1.38–41.66]), and MI (OR = 5.5 [95% CI: 1.1–26.6)] were independently and significantly associated with elevated CECs. We also identified significant correlations between CECs and other markers of endothelial dysfunction: sTREM-1 and TF+/EVs. Conclusion This study demonstrated that endothelial injury assessed by the levels of CECs was associated with thromboembolic events in patients with aPL and/or autoimmune diseases.
- Published
- 2022
14. Shared Heritability of Blood Pressure and Pulse Wave Velocity: Insights From the STANISLAS Cohort
- Author
-
Constance Xhaard, Pierre de Villemereuil, Athanase Benetos, Erwan Bozec, Claire Dandine-Roulland, Edith Le Floch, Véronique Regnault, Patrick Lacolley, Faiez Zannad, Patrick Rossignol, and Nicolas Girerd
- Subjects
Internal Medicine - Abstract
Background: Pulse wave velocity (PWV) is a marker of arterial stiffness, which is intrinsically highly correlated with blood pressure (BP). However, the interplay of PWV and BP heritability has not been extensively studied. This study aimed to estimate the heritability of PWV and BP and determine the genetic correlation between PWV and BP. Methods: The heritability of PWV and BP was estimated in 1080 subjects from the STANISLAS (Suivi Temporaire Annuel Non-Invasif de la Santé des Lorrains Assurés Sociaux) cohort with at least one relative using a linear mixed model within one frequentist and one Bayesian framework implemented, respectively, in the Gaston and MCMCglmm R packages. Then their genetic correlations were also estimated. Results: The heritability estimations for PWV were within the same range of the heritability of systolic BP and diastolic BP (23%, 19%, and 27%, respectively). Daytime heritability of BP was higher than nighttime BP. In addition, phenotypic correlations between PWV and systolic BP/diastolic BP were, respectively, 0.34 and 0.23, whereas nonsignificant genetic correlations were 0.08 and 0.22 respectively, indicating that PWV and diastolic BP shared more polygenic codeterminants than PWV and systolic BP. Conclusions: Our results suggest that the heritability of PWV is >20% and within the same range as BP heritability. It also suggests that the link between PWV and BP goes beyond phenotypic association: PWV and BP (in particular diastolic BP) share common genetic determinants. This genetic interdependence of PWV and BP appears largely polygenic.
- Published
- 2023
15. The β1-Adrenergic Receptor Contributes to Sepsis-Induced Immunosuppression Through Modulation of Regulatory T-Cell Inhibitory Function*
- Author
-
Manon, Durand, Eugénie, Hagimont, Huguette, Louis, Pierre, Asfar, Jean-Pol, Frippiat, Mervyn, Singer, Guillaume, Gauchotte, Carlos, Labat, Patrick, Lacolley, Bruno, Levy, Benjamin, Glenn Chousterman, and Antoine, Kimmoun
- Subjects
Immunosuppression Therapy ,Mice, Inbred C57BL ,Disease Models, Animal ,Mice ,Sepsis ,Animals ,Humans ,Critical Care and Intensive Care Medicine ,Adrenergic beta-1 Receptor Antagonists ,T-Lymphocytes, Regulatory - Abstract
Although cardiovascular benefits of β 1 -adrenergic receptor blockade have been described in sepsis, little is known about its impact on the adaptive immune response, specifically CD4 T cells. Herein, we study the effects of β 1 -adrenergic receptor modulation on CD4 T-cell function in a murine model of sepsis.Experimental study.University laboratory.C57BL/6 mice.High-grade sepsis was induced by cecal ligation and puncture in wild-type mice (β 1+/+ ) with or without esmolol (a selective β 1 -adrenergic receptor blocker) or in β 1 -adrenergic receptor knockout mice (β 1-/- ). At 18 hours after surgery, echocardiography was performed with blood and spleen collected to analyze lymphocyte function.At 18 hours, β 1+/+ cecal ligation and puncture mice exhibited characteristics of high-grade sepsis and three surrogate markers of immunosuppression, namely decreased splenic CD4 T cells, reduced CD4 T-cell proliferation, and increased regulatory T lymphocyte cell proportions. Pharmacologic and genetic β 1 -adrenergic receptor blockade reversed the impact of sepsis on CD4 T and regulatory T lymphocyte proportions and maintained CD4 T-cell proliferative capacity. β 1 -adrenergic receptor blocked cecal ligation and puncture mice also exhibited a global decrease in both pro- and anti-inflammatory mediators and improved in vivo cardiovascular efficiency with maintained cardiac power index despite the expected decrease in heart rate.β 1 -adrenergic receptor activation enhances regulatory T lymphocyte inhibitory function and thus contributes to sepsis-induced immunosuppression. This can be attenuated by β 1 -adrenergic receptor blockade, suggesting a potential immunoregulatory role for this therapy in the management of sepsis.
- Published
- 2022
16. Smooth muscle αv integrins regulate vascular fibrosis via CD109 downregulation of TGF-β signalling
- Author
-
Zhenlin Li, Ekaterina Belozertseva, Ara Parlakian, Rümeyza Bascetin, Huguette Louis, Yuki Kawamura, Jocelyne Blanc, Jacqueline Gao-Li, Florence Pinet, Adam Lacy-Hulbert, Pascal Challande, Jay D Humphrey, Veronique Regnault, Patrick Lacolley, Adaptation Biologique et Vieillissement = Biological Adaptation and Ageing (B2A), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Institut de Biologie Paris Seine (IBPS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Défaillance Cardiovasculaire Aiguë et Chronique (DCAC), Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lorraine (UL), Yale University [New Haven], Facteurs de Risque et Déterminants Moléculaires des Maladies liées au Vieillissement - U 1167 (RID-AGE), Institut Pasteur de Lille, Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), University of Washington [Seattle], Institut Jean Le Rond d'Alembert (DALEMBERT), Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), ANR-15-RHUS-0004,FIGHT-HF,Combattre l'insuffisance cardiaque(2015), and ANR-13-BSV1-0026,EVASION,Intégrines alphav des cellules musculaires lisses vasculaires : liens mécanistiques entre rigidité artérielle et génération de thrombine intrapariétale(2013)
- Subjects
[SDV]Life Sciences [q-bio] ,Vascular smooth muscle cell ,Alpha v integrin ,Collagen ,CD109 - Abstract
Aimsαv integrins are implicated in fibrosis in a number of organs through their ability to activate TGF-β. However their role in vascular fibrosis and collagen accumulation is only partially understood. Here we have used αv conditional knockout mice and cell lines to determine how αv contributes to vascular smooth muscle cell (VSMC) function in vascular fibrosis and the role of TGF-β in that process.Methods and resultsAngiotensin II (Ang II) treatment causes upregulation of αv and β3 expression in the vessel wall, associated with increased collagen deposition. We found that deletion of αv integrin subunit from VSMCs (αvSMKO) protected mice against angiotensin II-induced collagen production and assembly. Transcriptomic analysis of the vessel wall in αvSMKO mice and controls identified a significant reduction in expression of fibrosis and related genes in αvSMKO mice. In contrast, αvSMKO mice showed prolonged expression of CD109, which is known to affect TGF-β signalling. Using cultured mouse and human VSMCs, we showed that overexpression of CD109 phenocopied knockdown of αv integrin, attenuating collagen expression, TGF-β activation, and Smad2/3 signalling in response to angiotensin II or TGF-β stimulation. CD109 and TGF-β receptor were internalized in early endosomes.ConclusionWe identify a role for VSMC αv integrin in vascular fibrosis and show that αv acts in concert with CD109 to regulate TGF-β signalling.
- Published
- 2023
17. P13 ARHGEF1/RHOA SIGNALING PARTICIPATE IN AGEING-INDUCED ARTERIAL STIFFNESS AND HYPERCOAGULABILITY
- Author
-
Camille Rouillon, Nathalie Mercier, Patrick Lacolley, Gervaise Loirand, and Véronique Regnault
- Subjects
Specialties of internal medicine ,RC581-951 ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
The RhoA signaling pathway is a master regulator of mechanostransduction and plasticity of vascular smooth muscle cells (VSMCs) that controls arterial stiffening. The RhoA exchange factor Arhgef1 is causally involved in the development of angiotensin II-dependent hypertension. Our aim was to determine whether Arhgef1 plays a key role in age-associated arterial stiffness and the coupling with modifications of the procoagulant properties of blood and VSMCs. We used 65 week-old transgenic mice invalidated for Arhgef1 (Arhgef1−/−) and age-matched controls (Arhgef1+/+). In vivo arterial diameter pressure, distensibility/arterial pressure and elastic modulus/circumferential stress curves at the level of carotid artery were recorded using an echotracking system (VEVO 770 Visualsonics Imaging) in anesthetized animals. Systolic blood pressure, pulse pressure and heart rate were not different between mutant and control mice. Isobaric carotid distensibility was increased in Arhgef1−/− mice compared to Arhgef1+/+ mice. The elastic modulus/circumferential stress curves were shifted significantly rightwards in Arhgef1−/− mice compared to Arhgef1+/+ mice. Thrombin generation in blood and at the surface of VSMCs cultured from aorta was reduced in Arhgef1−/− mice. Anticoagulant markers secreted by the vascular wall (tissue factor pathway inhibitor and thrombomodulin) were increased in plasma of Arhgef1−/− mice. The time of formation of an occlusive thrombus induced by FeCl3 in the carotid artery was prolonged in Arhgef1−/− mice. In conclusion, the Arhgef1/RhoA contractile pathway contributes to arterial stiffening and VSMC procoagulant properties in aging. Whether this reduced procoagulant properties of the vascular wall is a cause or consequence of arterial stiffness remains to be elucidated.
- Published
- 2017
- Full Text
- View/download PDF
18. TARGETING VASCULAR SMOOTH MUSCLE CELL TO IMPROVE ARTERIAL STIFFNESS
- Author
-
Patrick Lacolley
- Subjects
Specialties of internal medicine ,RC581-951 ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Hypertension and arterial aging engage a plethora of key signaling pathways that act in concert to induce vascular smooth muscle cell (VSMC) phenotypic changes leading to vascular degeneration and extracellular matrix (ECM) changes responsible for alterations of the mechanical properties of the vascular wall. This review highlights proof-of-concept examples of components of the extracellular matrix, VSMC receptors which connect extracellular and intracellular structures and signaling pathways regulating changes in mechanotransduction and vascular homeostasis. This presentation presents new directions in the role of vascular smooth muscle cells VSMCs traditionally limited to regulation of contractile properties and synthesis of ECM proteins. VSMCs may exert negative feedback or positive feedback on ECM stiffness and mechanical load via stabilized focal adhesions, activated Rho-ROCK signaling pathways or actomyosin contraction. Understanding the mechanisms of cellular stiffness are also important to appreciate its contribution to mechanical properties at the tissue-level. Many other cell types, including macrophages, could participate to inflammation and VSMC stiffness leading to fibrosis of the arterial wall. In view of the multitude roles of VSMCs and feedback controls, only omic approaches and computational models may extrapolate the overall effects on the vascular wall in light of hemodynamics and complex interactions amongst differentially sized vessels. The use of novel animal models with multiple genetic manipulations of VSMC signaling pathways can provide further insight into the link between large vessel stiffness and small vessel dysfunction.
- Published
- 2017
- Full Text
- View/download PDF
19. A new pro-thrombotic mechanism of neutrophil extracellular traps in antiphospholipid syndrome: impact on activated protein C resistance
- Author
-
Virginie Dufrost, Thomas Foret, Véronique Regnault, Jeremy Lagrange, Patricia Costa, Denis Wahl, Patrick Lacolley, Stéphane Zuily, Cécile Lakomy, and Lucie Salomon du Mont
- Subjects
Lupus anticoagulant ,biology ,business.industry ,Thrombosis ,Neutrophil extracellular traps ,Antiphospholipid Syndrome ,medicine.disease ,Extracellular Traps ,Cross-Sectional Studies ,Rheumatology ,Antiphospholipid syndrome ,Immunology ,Antithrombotic ,medicine ,biology.protein ,Humans ,Pharmacology (medical) ,Antibody ,Activated protein C resistance ,business ,Protein C ,Activated Protein C Resistance ,medicine.drug - Abstract
Objectives In APS, precise evaluation of thrombotic risk is a major challenge. Different players, such as activated protein C (APC) resistance or neutrophil extracellular traps (NETs) contribute to the risk of thrombosis. Nevertheless, no study has investigated the interaction between these actors. The main objective of this study was to investigate the relation between NETs and APC resistance. Methods We designed a cross-sectional study including APS/antiphospholipid antibodies (aPL) patients and patients with autoimmune diseases (AID). We performed thrombin generation tests without and with APC to determine APC resistance. To evaluate circulating NETs, we measured plasma levels of MPO-DNA complexes and cell-free DNA with ELISA. Results We recruited 117 patients with definite APS/aPL or AID. We found a positive correlation between NETs and APC resistance, in APS patients and specifically in patients with high thrombotic risk, displaying LA or positivity of all three aPL tests (triple+), or anti-domain I IgG (aDI+). All these patient subgroups had increased NETs concentrations and APC resistance. As the risk profile for thrombosis increased, the relationship between NETs and APC resistance was stronger. Conclusion We have shown that NETs participate in the hypercoagulable state of APS patients by contributing to APC resistance, in particular in high-risk patients. In these most at-risk patients, a targeted action on NETs could reduce APC resistance and constitute a new therapeutic approach in the treatment of APS patients in addition to antithrombotic therapy.
- Published
- 2021
20. Smooth Muscle Cell Molecular Underpinnings of Vascular Ageing
- Author
-
Celia Schellenberg, Alexandre Raoul, Patrick Lacolley, Véronique Regnault, Défaillance Cardiovasculaire Aiguë et Chronique (DCAC), Université de Lorraine (UL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy), Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lorraine (UL), and REGNAULT, Véronique
- Subjects
Pulmonary and Respiratory Medicine ,0303 health sciences ,business.industry ,[SDV]Life Sciences [q-bio] ,Myocytes, Smooth Muscle ,Cell ,030204 cardiovascular system & hematology ,medicine.disease ,Muscle, Smooth, Vascular ,Cell biology ,Vascular ageing ,[SDV] Life Sciences [q-bio] ,03 medical and health sciences ,Vascular Stiffness ,0302 clinical medicine ,medicine.anatomical_structure ,Smooth muscle ,Ageing ,Arterial stiffness ,Medicine ,Mechanotransduction ,Cardiology and Cardiovascular Medicine ,business ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology - Abstract
International audience
- Published
- 2021
21. International consensus on the prevention of venous and arterial thrombotic events in patients with inflammatory bowel disease
- Author
-
Tim Raine, Siew C. Ng, Edouard Louis, David T. Rubin, Peter Bossuyt, Fernando Magro, Laurent Peyrin-Biroulet, Subrata Ghosh, Silvio Danese, Paulo Gustavo Kotze, Véronique Regnault, Fábio Vieira Teixeira, Patrick Lacolley, Alfredo Papa, Taku Kobayashi, Stéphane Zuily, Richard B. Gearry, Sameer Al Awadhi, Pablo Olivera, Zuily, Stephane [0000-0002-9326-6881], Kotze, Paulo G [0000-0002-9632-6691], Bossuyt, Peter [0000-0003-4027-7365], Ghosh, Subrata [0000-0002-1713-7797], Kobayashi, Taku [0000-0002-2073-4234], Ng, Siew C [0000-0002-6850-4454], Papa, Alfredo [0000-0002-4186-7298], Raine, Tim [0000-0002-5855-9873], Rubin, David T [0000-0001-5647-1723], Danese, Silvio [0000-0001-7341-1351], Peyrin-Biroulet, Laurent [0000-0003-2536-6618], Apollo - University of Cambridge Repository, Centro de Educación Médica e Investigaciones Clínicas (CEMIC), Défaillance Cardiovasculaire Aiguë et Chronique (DCAC), Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lorraine (UL), Pontifical Catholic University of Paraná (PUCPR), Pontifical Catholic University of Paraná, Rashid Hospital, Imelda General Hospital, University of Otago [Dunedin, Nouvelle-Zélande], University of Birmingham [Birmingham], Kitasato University, NIHR Biomedical Research Centre [London], Guy's and St Thomas' NHS Foundation Trust-King‘s College London, Centre Hospitalier Universitaire de Liège (CHU-Liège), Hospital de São João [Porto], The Chinese University of Hong Kong [Hong Kong], Fondazione 'Policlinico Universitario A. Gemelli' [Rome], Cambridge University Hospitals - NHS (CUH), University of Cambridge [UK] (CAM), The University of Chicago Medicine [Chicago], Istituto Clinico Humanitas [Milan] (IRCCS Milan), Humanitas University [Milan] (Hunimed), Nutrition-Génétique et Exposition aux Risques Environnementaux (NGERE), and Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lorraine (UL)
- Subjects
Evidence-Based Guidelines ,medicine.medical_specialty ,[SDV]Life Sciences [q-bio] ,International Cooperation ,Settore MED/12 - GASTROENTEROLOGIA ,education ,Anti-Inflammatory Agents ,MEDLINE ,030204 cardiovascular system & hematology ,Risk Assessment ,Inflammatory bowel disease ,03 medical and health sciences ,0302 clinical medicine ,Fibrinolytic Agents ,Risk Factors ,Epidemiology ,Humans ,Medicine ,In patient ,Risk factor ,Intensive care medicine ,Hepatology ,business.industry ,Patient Acuity ,Gastroenterology ,Thrombosis ,Guideline ,Evidence-based medicine ,Inflammatory Bowel Diseases ,medicine.disease ,digestive system diseases ,3. Good health ,Hospitalization ,Cardiovascular diseases ,030211 gastroenterology & hepatology ,business - Abstract
Patients with inflammatory bowel disease (IBD) are at increased risk of thrombotic events. Therapies for IBD have the potential to modulate this risk. The aims of this Evidence-Based Guideline were to summarize available evidence and to provide practical recommendations regarding epidemiological aspects, prevention and drug-related risks of venous and arterial thrombotic events in patients with IBD. A virtual meeting took place in May 2020 involving 14 international IBD experts and 3 thrombosis experts from 12 countries. Proposed statements were voted upon in an anonymous manner. Agreement was defined as at least 75% of participants voting as ‘fully agree’ or ‘mostly agree’ with each statement. For each statement, the level of evidence was graded according to the Scottish Intercollegiate Guidelines Network (SIGN) grading system. Consensus was reached for 19 statements. Patients with IBD harbour an increased risk of venous and arterial thrombotic events. Thromboprophylaxis is indicated during hospitalization of any cause in patients with IBD. Disease activity is a modifiable risk factor in patients with IBD, and physicians should aim to achieve deep remission to reduce the risk. Exposure to steroids should be limited. Antitumour necrosis factor agents might be associated with a reduced risk of thrombotic events., Patients with inflammatory bowel disease (IBD) are at increased risk of thrombotic events. This Evidence-Based Guideline presents an international consensus on the prevention of venous and arterial thrombotic events in patients with IBD, and includes 19 recommendations for clinical practice.
- Published
- 2021
22. Implication of Free Fatty Acids in Thrombin Generation and Fibrinolysis in Vascular Inflammation in Zucker Rats and Evolution with Aging
- Author
-
Jérémy Lagrange, Mélusine Didelot, Amel Mohamadi, Lucy A. Walton, Saartje Bloemen, Bas de Laat, Huguette Louis, Simon N. Thornton, Brian Derby, Michael J. Sherratt, Bruno Fève, Pascal Challande, Riaz Akhtar, J. Kennedy Cruickshank, Patrick Lacolley, and Véronique Regnault
- Subjects
vascular aging ,blood coagulation test ,obesity ,fatty acids ,thrombin generation ,Physiology ,QP1-981 - Abstract
Background: The metabolic syndrome (MetS) and aging are associated with modifications in blood coagulation factors, vascular inflammation, and increased risk of thrombosis.Objectives: Our aim was to determine concomitant changes in thrombin generation in the blood compartment and at the surface of vascular smooth muscle cells (VSMCs) and its interplay with adipokines, free fatty acids (FFA), and metalloproteinases (MMPs) in obese Zucker rats that share features of the human MetS.Methods: Obese and age-matched lean Zucker rats were compared at 25 and 80 weeks of age. Thrombin generation was assessed by calibrated automated thrombography (CAT).Results: Endogenous thrombin potential (ETP) was increased in obese rats independent of platelets and age. Clot half-lysis time was delayed with obesity and age. Interleukin (IL)-1β and IL-13 were increased with obesity and age respectively. Addition of exogenous fibrinogen, leptin, linoleic, or palmitic acid increased thrombin generation in plasma whereas adiponectin had an opposite effect. ETP was increased at the surface of VSMCs from obese rats and addition of exogenous palmitic acid further enhanced ETP values. Gelatinase activity was increased in aorta at both ages in obese rats and MMP-2 activity was increased in VSMCs from obese rats.Conclusions: Our study demonstrated in MetS an early prothrombotic phenotype of the blood compartment reinforced by procoagulant properties of dedifferentiated and inflammatory VSMCs. Mechanisms involved (1) increased fibrinogen and impaired fibrinolysis and (2) increased saturated fatty acids responsible for additive procoagulant effects. Whether specifically targeting this hypercoagulability using direct thrombin inhibitors would improve outcome in MetS is worth investigating.
- Published
- 2017
- Full Text
- View/download PDF
23. Exome-wide association study reveals novel susceptibility genes to sporadic dilated cardiomyopathy.
- Author
-
Ulrike Esslinger, Sophie Garnier, Agathe Korniat, Carole Proust, Georgios Kararigas, Martina Müller-Nurasyid, Jean-Philippe Empana, Michael P Morley, Claire Perret, Klaus Stark, Alexander G Bick, Sanjay K Prasad, Jennifer Kriebel, Jin Li, Laurence Tiret, Konstantin Strauch, Declan P O'Regan, Kenneth B Marguiles, Jonathan G Seidman, Pierre Boutouyrie, Patrick Lacolley, Xavier Jouven, Christian Hengstenberg, Michel Komajda, Hakon Hakonarson, Richard Isnard, Eloisa Arbustini, Harald Grallert, Stuart A Cook, Christine E Seidman, Vera Regitz-Zagrosek, Thomas P Cappola, Philippe Charron, François Cambien, and Eric Villard
- Subjects
Medicine ,Science - Abstract
AimsDilated cardiomyopathy (DCM) is an important cause of heart failure with a strong familial component. We performed an exome-wide array-based association study (EWAS) to assess the contribution of missense variants to sporadic DCM.Methods and results116,855 single nucleotide variants (SNVs) were analyzed in 2796 DCM patients and 6877 control subjects from 6 populations of European ancestry. We confirmed two previously identified associations with SNVs in BAG3 and ZBTB17 and discovered six novel DCM-associated loci (Q-valueConclusionWe identified eight loci independently associated with sporadic DCM. The functions of the best candidate genes at these loci suggest that proteostasis regulation might play a role in DCM pathophysiology.
- Published
- 2017
- Full Text
- View/download PDF
24. Conductance Artery Wall Layers and Their Respective Roles in the Clearance Functions
- Author
-
Jean-Baptiste Michel, Jeremy Lagrange, Veronique Regnault, and Patrick Lacolley
- Subjects
Adventitia ,Adipose Tissue ,Myocytes, Smooth Muscle ,Humans ,Arteries ,Vascular Diseases ,Cardiology and Cardiovascular Medicine - Abstract
Evolutionary organization of the arterial wall into layers occurred concomitantly with the emergence of a highly muscularized, pressurized arterial system that facilitates outward hydraulic conductance and mass transport of soluble substances across the arterial wall. Although colliding circulating cells disperse potential energy within the arterial wall, the different layers counteract this effect: (1) the endothelium ensures a partial barrier function; (2) the media comprises smooth muscle cells capable of endocytosis/phagocytosis; (3) the outer adventitia and perivascular adipocytic tissue are the final receptacles of convected substances. While the endothelium forms a physical and a biochemical barrier, the medial layer is avascular, relying on the specific permeability properties of the endothelium for metabolic support. Different components of the media interact with convected molecules: medial smooth muscle cells take up numerous molecules via scavenger receptors and are capable of phagocytosis of macro/micro particles. The outer layers—the highly microvascularized innervated adventitia and perivascular adipose tissue—are also involved in the clearance functions of the media: the adventitia is the seat of immune response development, inward angiogenesis, macromolecular lymphatic drainage, and neuronal stimulation. Consequently, the clearance functions of the arterial wall are physiologically essential, but also may favor the development of arterial wall pathologies. This review describes how the walls of large conductance arteries have acquired physiological clearance functions, how this is determined by the attributes of the endothelial barrier, governed by endocytic and phagocytic capacities of smooth muscle cells, impacting adventitial functions, and the role of these clearance functions in arterial wall diseases.
- Published
- 2022
25. Evidences for Inotropic Effects Induced by Simultaneous Antegrade and Retrograde Myocardial Perfusion in a Swine Model: Potential Applications for ECMO
- Author
-
Daniel Grandmougin, Antoine Chalon, Aude Falanga, Vanessa Marie, Fréderique Groubatch-Joineau, Brice Mourer, Pierre-Yves Marie, Patrick Lacolley, and Nguyen Tran
- Published
- 2022
26. The VWF/LRP4/αVβ3-axis represents a novel pathway regulating proliferation of human vascular smooth muscle cells
- Author
-
Paulette Legendre, Melusine Didelot, Jeremy Lagrange, Olivier D. Christophe, Vincent Muczynski, Morel E Worou, Cécile V. Denis, Guillaume Gauchotte, Patrick Lacolley, François Plénat, Alexandre Raoul, Peter J. Lenting, Véronique Regnault, Jean-Baptiste Michel, Marc-Damien Lourenco-Rodrigues, Défaillance Cardiovasculaire Aiguë et Chronique (DCAC), Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lorraine (UL), Laboratoire de Recherche Vasculaire Translationnelle (LVTS (UMR_S_1148 / U1148)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP)-Université Sorbonne Paris Nord, Hémostase, Inflammation, Thrombose (HITH - U1176 Inserm - CHU Bicêtre), Institut National de la Santé et de la Recherche Médicale (INSERM)-AP-HP Hôpital Bicêtre (Le Kremlin-Bicêtre)-Université Paris-Saclay, Université de Lorraine (UL), Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy), Université de Lorraine (UL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy), Institut Carnot IFPEN Transports Energie, IFP Energies nouvelles (IFPEN), IFP Energies nouvelles (IFPEN)-IFP Energies nouvelles (IFPEN), Laboratoire des Interactions Ecotoxicologie, Biodiversité, Ecosystèmes (LIEBE), Université Paul Verlaine - Metz (UPVM)-Centre National de la Recherche Scientifique (CNRS), Service de Pathologie [CHRU Nancy], Denis, Cécile, and Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité)-Université Sorbonne Paris Nord
- Subjects
[SDV.MHEP.HEM] Life Sciences [q-bio]/Human health and pathology/Hematology ,Male ,Vascular smooth muscle ,Physiology ,Angiogenesis ,[SDV]Life Sciences [q-bio] ,Proliferation ,Cell ,030204 cardiovascular system & hematology ,Vascular biology ,Muscle, Smooth, Vascular ,0302 clinical medicine ,Cell Movement ,hemic and lymphatic diseases ,Receptor ,Cells, Cultured ,Mice, Knockout ,0303 health sciences ,biology ,Chemistry ,[SDV.MHEP.HEM]Life Sciences [q-bio]/Human health and pathology/Hematology ,Hyperplasia ,Plaque, Atherosclerotic ,Cell biology ,medicine.anatomical_structure ,Smooth muscle cells ,cardiovascular system ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,Haemostasis ,Signal Transduction ,Myocytes, Smooth Muscle ,Integrin ,Inflammation ,03 medical and health sciences ,Von Willebrand factor ,Neointima ,Physiology (medical) ,von Willebrand Factor ,medicine ,Animals ,LDL-Receptor Related Proteins ,Cell Proliferation ,030304 developmental biology ,Vascular System Injuries ,Atherosclerosis ,Integrin alphaVbeta3 ,medicine.disease ,Mice, Inbred C57BL ,biology.protein ,Carotid Artery Injuries - Abstract
Aims Von Willebrand factor (VWF) is a plasma glycoprotein involved in primary hemostasis, while also having additional roles beyond hemostasis namely in cancer, inflammation, angiogenesis and potentially in vascular smooth muscle cell (VSMC) proliferation. Here, we addressed how VWF modulates VSMC proliferation and investigated the underlying molecular pathways and the in vivo pathophysiological relevance. Methods and results VWF induced proliferation of human aortic VSMCs and also promoted VSMC migration. Treatment of cells with a siRNA against αv integrin or the RGT-peptide blocking αvβ3 signaling abolished proliferation. However, VWF did not bind to αvβ3 on VSMCs through its RGD-motif. Rather, we identified the VWF A2 domain as the region mediating binding to the cells. We hypothesized the involvement of a member of the LDL-related receptor protein (LRP) family due to their known ability to act as co-receptors. Using the universal LRP-inhibitor receptor-associated protein, we confirmed LRP-mediated VSMC proliferation. siRNA experiments and confocal fluorescence microscopy identified LRP4 as the VWF-counterreceptor on VSMCs. Also co-localization between αvβ3 and LRP4 was observed via proximity ligation analysis and immuno-precipitation experiments. The pathophysiological relevance of our data was supported by VWF-deficient mice having significant reduced, if any, hyperplasia in carotid artery ligation and artery femoral denudation models. In wild-type mice, infiltration of VWF in intimal regions enriched in proliferating VSMCs was found. Interestingly, also analysis of human atherosclerotic lesions showed abundant VWF accumulation in VSMC-proliferating rich intimal areas. Conclusions VWF mediates VSMC proliferation through a mechanism involving A2 domain binding to the LRP4 receptor and integrin αvβ3 signaling. Our findings provide new insights into the mechanisms that drive physiological repair and pathological hyperplasia of the arterial vessel wall. In addition, the VWF/LRP4-axis may represent a novel therapeutic target to modulate VSMC proliferation. Translational perspective The mechanisms that drive physiological repair and pathological hyperplasia of the arterial vessel wall are complex and only partially understood. Specifically, the role of subendothelial-matrix proteins remains unclear. Here, we show that the hemostatic protein von Willebrand factor (VWF) accumulates in the vascular wall of atherosclerotic lesions and localizes to areas of vascular smooth muscle cell (VSMC) proliferation. VWF was found to use its A2-domain for binding to the VSMC-receptor LRP4, which in turn triggered outside-in signaling via integrin αVβ3, thereby inducing VSMC proliferation. Interfering with A2-domain/LRP4 interactions might offer innovative and additional therapeutical approaches to limit pathological hyperplasia.
- Published
- 2021
27. The resolvin D2 - GPR18 axis is expressed in human coronary atherosclerosis and transduces atheroprotection in apolipoprotein E deficient mice
- Author
-
Matthieu Bardin, Sven-Christian Pawelzik, Jeremy Lagrange, Ali Mahdi, Hildur Arnardottir, Véronique Regnault, Bruno Fève, Patrick Lacolley, Jean-Baptiste Michel, Nathalie Mercier, and Magnus Bäck
- Subjects
Pharmacology ,Inflammation ,Mice ,Apolipoproteins E ,Docosahexaenoic Acids ,Animals ,Humans ,Coronary Artery Disease ,Atherosclerosis ,Biochemistry ,Receptors, G-Protein-Coupled ,Signal Transduction - Abstract
Chronic inflammation in atherosclerosis reflects a failure in the resolution of inflammation. Pro-resolving lipid mediators derived from omega-3 fatty acids reduce the development of atherosclerosis in murine models. The aim of the present study was to decipher the role of the specialized proresolving mediator (SPM) resolvin D2 (RvD2) in atherosclerosis and its signaling through the G-protein coupled receptor (GPR) 18. The ligand and receptor were detected in human coronary arteries in relation to the presence of atherosclerotic lesions and its cellular components. Importantly, RvD2 levels were significantly higher in atherosclerotic compared with healthy human coronary arteries. Furthermore, apolipoprotein E (ApoE) deficient hyperlipidemic mice were treated with either RvD2 or vehicle in the absence and presence of the GPR18 antagonist O-1918. RvD2 significantly reduced atherosclerosis, necrotic core area, and pro-inflammatory macrophage marker expression. RvD2 in addition enhanced macrophage phagocytosis. The beneficial effects of RvD2 were not observed in the presence of O-1918. Taken together, these results provide evidence of atheroprotective pro-resolving signalling through the RvD2-GPR18 axis.
- Published
- 2022
28. 4.4 ARTERIAL PHENOTYPE MODULATION AND REGULATION OF VASCULAR FIBROSIS IN MICE BY CONDITIONAL INACTIVATION OF INTEGRIN AV SUBUNIT IN VASCULAR SMOOTH MUSCLE CELLS
- Author
-
Ekaterina Belozertseva, Huguette Louis, Zhenlin Li, Mustapha Bourhim, Dominique Dumas, Veronique Regnault, and Patrick Lacolley
- Subjects
Specialties of internal medicine ,RC581-951 ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Integrin αv functions as a receptor for adhesion proteins and is expressed at high density in vascular smooth muscle cells (VSMC)1,2,3,4,5 whose phenotypic modulation plays a crucial role in arterial ageing and atherosclerosis.6,7. Our aim was to define the arterial phenotype in mice conditionally inactivated for the integrin αv subunit in VSMC8,9,10(αv SMKO) and its role in angiotensin II (AngII)-induced arterial fibrosis. Transgenic mice αv SMKO and their control littermates (WT) were treated with two doses of AngII, low (0.3 mg/kg/day) and high (1.5 mg/kg/day), for 4 weeks. At baseline, blood pressure was lower in αvSMKO compared to WT mice. Carotid distensibility was increased in αv SMKO mice (13.3±0.7 vs 10.3±0.6 mmHg−1.10−3). With low dose AngII isobaric distensibility remained higher in αvSMKOmice (12.4±1.2 vs 10.7±1.0 mmHg−1.10−3).With high dose AngII the increase in collagen content in carotid media was lower in αvSMKOthan in WT (19 vs 35%) for a similar increase in blood pressure (30 mmHg) and arterial wall hypertrophy. Collagen immunostaining and fluorescence measurements (multiphoton microscopy second harmonic generation) confirmed that high dose AngII induced lower increases in collagen content in αvSMKO mice versus WT (8.9±1.7 vs 14.2±1.4 greyscale mean/pixel). The combination of similar arterial wall hypertrophy with less fibrosis in mutant mice explains an increased distensibility in response to AngII. The αv subunit regulates AngII-induced arterial fibrosis as determined by collagen staining, immunostaining and fluorescence. Pharmacological targeting of vascular αv integrin may have clinical applications in the treatment of patients with fibrosis associated with hypertension and atherosclerosis.
- Published
- 2016
- Full Text
- View/download PDF
29. 9.2 DELETION OF CHROMOSOME 9P21 NONCODING CARDIOVASCULAR RISK INTERVAL IN MICE INDUCES A PROTHROMBOTIC PHENOTYPE
- Author
-
Amel Mohamadi, Mustapha Bourhim, Gemma Basatemur, Huguette Louis, Athanase Benetos, Patrick Lacolley, Ziad Mallat, and Veronique Regnault
- Subjects
Specialties of internal medicine ,RC581-951 ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Background: SNPs on chromosome 9p21.3 risk locus have been associated with cardiovascular diseases. We have established a direct mechanistic link between 9p21 noncoding risk interval and susceptibility to aneurysm in a mouse model with a targeted deletion of the 9p21 noncoding cardiovascular disease risk interval. The deficiency of transcripts encoded by this locus predisposes to a pro-thrombotic phenotype and arterial stiffening in this mouse model and in humans with 9p21 DNA variants. Methods: Carotid blood flow following FeCl3 application was monitored via Doppler profiles. Results: The deletion of the orthologous 70-kb noncoding interval on mouse chromosome 4 (chr4Δ70kb/Δ70kb), synthetic to human chromosome 9p21, predisposes to arterial thrombosis. The time to occlusion in a FeCl3-induced carotid thrombosis model was significantly decreased by 30% in the absence of the locus and confirmed by a new model of physiological thrombosis. There was no difference between groups in blood pressure, carotid stiffness parameters (diameter and distensibility for a given level of arterial pressure) or in vascular structure. We explored the potential impact of the deletion locus on thrombin generation as well as on platelet aggregation and reactivity all were increased compared to controls. In 100 healthy carriers of the 9p21 risk T allele display an increased aortic arterial stiffness compared with carriers of the C allele. Conclusion: These results establish a direct link between variants or deletion in the 9p21 non-coding risk interval and increased platelet reactivity and thrombin generation predisposing to thrombosis in mouse and increased arterial stiffness in aged population.
- Published
- 2016
- Full Text
- View/download PDF
30. 9.5 COAGULATION CONTROL BY THE RHOA PATHWAY AND THE EXCHANGE FACTOR ARHGEF1
- Author
-
Camille Rouillon, Amel Mohammadi, Gervaise Loirand, Veronique Regnault, and Patrick Lacolley
- Subjects
Specialties of internal medicine ,RC581-951 ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Platelet activation by thrombin is an auto-amplification loop of thrombin generation, a major factor in the formation of atherosclerotic plaques. The small G protein RhoA, under the direct control of the exchange factor Arhgef1, modulates several cellular functions in inflammation. The objective was to study the RhoA pathway and its control by Arhgef1 in platelet aggregation and thrombin generation due to PAR receptor activation by thrombin. We used a knockout mouse model for the exchange factor Arhgef1 (Arhgef1 −/−). In response to an agonist (collagen, ADP and thrombin), the expression of surface glycoproteins and the aggregation of washed platelets were not altered in the Arhgef1 −/− mice compared to Argef1 +/+ mice. In contrast, platelet activation studied by the secretion of granules a, exposure to phosphatidylserine and release of microparticles were decreased in the Arhgef1 −/− mice. Thrombin generation in whole platelet-rich blood was also reduced by 25%. These changes result in a lengthening of the time of occurrence of an occlusive thrombus in the carotid induced by FeCl3. In conclusion, the results confirm the involvement of the RhoA pathway in platelet activation and demonstrate an Arhgef1-dependent mechanism. The results in mice show a new auto-amplification mechanism of thrombin generation by platelets through PAR and membrane phospholipids. Redistribution of phospholipid linked rearrangements of the membrane complex induced by inflammation suggests that the RhoA pathway potentiates the deleterious effects of thrombin in atherothrombosis.
- Published
- 2016
- Full Text
- View/download PDF
31. 5.5 AGE-DEPENDENT TELOMERE ATTRITION, SHORT TELOMERES AND ATHEROSCLEROSIS
- Author
-
Simon Toupance, Anna Kearney-Schwartz, Mohamed Temmar, Cécile Lakomy, Carlos Labat, Patrick Rossignol, Faiez Zannad, Patrick Lacolley, Abraham Aviv, and Athanase Benetos
- Subjects
Specialties of internal medicine ,RC581-951 ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Background: Short leukocyte telomere length (LTL) is associated with atherosclerosis. The prevailing view is this association exists since LTL is a biomarker of cumulative inflammation and oxidative stress during adult life. However recent studies show that LTL in adults is defined mainly by LTL at birth and attrition during childhood. Therefore we can suggest that short LTL might precede clinical expression of atherosclerosis. Objectives: To examine the directionality in the relation between carotid atheroma and LTL dynamics. Methods: LTL was measured by TRF in samples donated 9 years apart on average by 257 men and women aged 41 to 80 at the inclusion. Results: LTL attrition during follow-up (FU) period was 25±17 bp/year. No relation was observed between LTL attrition and presence of carotid atherosclerotic plaques (PCAP). Baseline (BL)-LTL was highly correlated (r=0.96, p
- Published
- 2016
- Full Text
- View/download PDF
32. 9.4 EVOLUTION OF CARDIAC FUNCTION AND METABOLISM DURING AGING IN A MURINE ANIMAL MODEL OF OBESITY
- Author
-
Delphine Lambert, Fatiha Maskali, Sylvain Poussier, Alexandra Clement, Jean-Loup Machu, Pierre-Yves Marie, Patrick Lacolley, and Athanase Benetos
- Subjects
Specialties of internal medicine ,RC581-951 ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Purpose/Background/Objectives: Obesity is a well-known risk factor of cardiovascular diseases and a potentially modifiable determinant of arterial ageing. The objectives of this experimental study were to assess the effects of a long-term high fat diet (HFD) on metabolism, adipose tissues and phenotypes of cardiovascular aging. Methods: Murine model chosen was C57BL/6J mice receiving during one year HFD or control diet (CD). Longitudinal follow-up of weight, systolic bloodpressure, heart rate and metabolic parameters was performed. An echocardiographic system was used to study cardiac function. Metabolism at the level of the adipose tissues was studied with FDG positron emission tomography (PET). Results: After 12 months of diet the whole mice showed a positive correlation between plasma leptin level and left ventricular thickness and mass (both p
- Published
- 2016
- Full Text
- View/download PDF
33. Cell senescence: basic mechanisms and the need for computational networks in vascular ageing
- Author
-
Véronique Regnault, Florence Pinet, Pascal Challande, Zhenlin Li, Patrick Lacolley, Défaillance Cardiovasculaire Aiguë et Chronique (DCAC), Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lorraine (UL), Sorbonne Université (SU), Facteurs de Risque et Déterminants Moléculaires des Maladies liées au Vieillissement - U 1167 (RID-AGE), Institut Pasteur de Lille, Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Adaptation Biologique et Vieillissement = Biological Adaptation and Ageing (B2A), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Institut de Biologie Paris Seine (IBPS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut Jean Le Rond d'Alembert (DALEMBERT), Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), and LACOLLEY, Patrick
- Subjects
Proteomics ,Senescence ,Physiology ,[SDV]Life Sciences [q-bio] ,Cell ,Cell Cycle Proteins ,030204 cardiovascular system & hematology ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Physiology (medical) ,medicine ,Animals ,Humans ,Gene Regulatory Networks ,Protein Interaction Maps ,Vascular Diseases ,Epigenetics ,Senolytic ,Cellular Senescence ,Cell Proliferation ,030304 developmental biology ,0303 health sciences ,Systems Biology ,Computational Biology ,Cell Cycle Checkpoints ,[SDV] Life Sciences [q-bio] ,medicine.anatomical_structure ,Signalling ,Gene Expression Regulation ,Blood Vessels ,Cardiology and Cardiovascular Medicine ,Cell aging ,Neuroscience ,Algorithms ,Function (biology) ,Biological network ,Signal Transduction - Abstract
This review seeks to provide an update of the mechanisms of vascular cell senescence, from newly identified molecules to arterial ageing phenotypes, and finally to present a computational approach to connect these selected proteins in biological networks. We will discuss current key signalling and gene expression pathways by which these focus proteins and networks drive normal and accelerated vascular ageing. We also review the possibility that senolytic drugs, designed to restore normal cell differentiation and function, could effectively treat multiple age-related vascular diseases. Finally, we discuss how cell senescence is both a cause and a consequence of vascular ageing because of the possible feedback controls between identified networks.
- Published
- 2020
34. Anti-Domain I beta(2)-Glycoprotein I Antibodies and Activated Protein C Resistance Predict Thrombosis in Antiphospholipid Syndrome
- Author
-
Patrick Lacolley, Véronique Regnault, Marc Lambert, Philip G. de Groot, Hilde Kelchtermans, Emmanuel de Maistre, Zakera Shums, Nadine Magy-Bertrand, Vincent Poindron, Francis Guillemin, Virginie Dufrost, Gary L. Norman, Thomas Lecompte, Jessie Risse, Bas de Laat, Denis Wahl, Hélène Desmurs-Clavel, Stéphane Zuily, RS: Carim - B01 Blood proteins & engineering, Biochemie, Défaillance Cardiovasculaire Aiguë et Chronique (DCAC), Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lorraine (UL), Synapse Research Institute, Maladies chroniques, santé perçue, et processus d'adaptation (APEMAC), Université de Lorraine (UL), Centre Hospitalier Régional Universitaire de Besançon (CHRU Besançon), Hospices Civils de Lyon (HCL), Lille Inflammation Research International Center - U 995 (LIRIC), Institut Pasteur de Lille, Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Département de Médecine Interne et Immunologie Clinique (DMIIC - STRASBOURG), CHU Strasbourg, Franche-Comté Électronique Mécanique, Thermique et Optique - Sciences et Technologies (UMR 6174) (FEMTO-ST), Université de Franche-Comté (UFC), Université Bourgogne Franche-Comté [COMUE] (UBFC)-Université Bourgogne Franche-Comté [COMUE] (UBFC)-Ecole Nationale Supérieure de Mécanique et des Microtechniques (ENSMM)-Centre National de la Recherche Scientifique (CNRS)-Université de Technologie de Belfort-Montbeliard (UTBM), Centre régional de compétence des Maladies systémiques et auto-immunes rares de l'adulte et Maladies vasculaires rares, Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy), INOVA Diagnostics, San Diego, and Université de Genève (UNIGE)
- Subjects
medicine.medical_specialty ,[SDV]Life Sciences [q-bio] ,1ST EPISODE ,030204 cardiovascular system & hematology ,Gastroenterology ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Antiphospholipid syndrome ,Internal medicine ,medicine ,Humans ,CLASSIFICATION CRITERIA ,Prospective Studies ,Activated Protein C Resistance ,LUPUS ANTICOAGULANTS ,030203 arthritis & rheumatology ,RISK ,Lupus anticoagulant ,REVISED CRITERIA ,biology ,business.industry ,RECOGNIZE ,Thrombosis ,General Medicine ,ASSOCIATION ,STANDARDIZATION ,medicine.disease ,Antiphospholipid Syndrome ,Cross-Sectional Studies ,beta 2-Glycoprotein I ,biology.protein ,Female ,AUTOANTIBODIES ,Activated protein C resistance ,Antibody ,business ,Protein C ,β2 glycoprotein i ,Cohort study ,medicine.drug ,GENERATION - Abstract
Background Antibodies binding to domain I of β2-glycoprotein I (aDI) and activated protein C (APC) resistance are associated with an increased risk of thrombosis in cross-sectional studies. The objective of this study was to assess their predictive value for future thromboembolic events in patients with antiphospholipid antibodies (aPL) or antiphospholipid syndrome. Methods This prospective multicenter cohort study included consecutive patients with aPL or systemic lupus erythematosus. We followed 137 patients (43.5 ± 15.4 year old; 107 women) for a mean duration of 43.1 ± 20.7 months. Results We detected aDI IgG antibodies by ELISA in 21 patients. An APC sensitivity ratio (APCsr) was determined using a thrombin generation–based test. The APCsr was higher in patients with anti–domain I antibodies demonstrating APC resistance (0.75 ± 0.13 vs 0.48 ± 0.20, P 95th percentile; HR, 6.07 [95% CI, 1.69–21.87]; P = 0.006). A sensitivity analysis showed an increased risk of higher aDI IgG levels up to HR 5.61 (95% CI, 1.93–16.31; P = 0.01). In multivariate analysis, aDI IgG (HR, 3.90 [95% CI, 1.33–11.46]; P = 0.01) and APC resistance (HR, 4.98 [95% CI, 1.36–18.28]; P = 0.02) remained significant predictors of thrombosis over time. Conclusions Our study shows that novel tests for antibodies recognizing domain I of β2-glycoprotein I and functional tests identifying APC resistance are significant predictors of thrombosis over time and may be useful for risk stratification.
- Published
- 2020
35. Alpha-2-macroglobulin in hemostasis and thrombosis: An underestimated old double-edged sword
- Author
-
Jeremy Lagrange, Thomas Lecompte, Tanja Knopp, Patrick Lacolley, and Véronique Regnault
- Subjects
Hemostasis ,Pregnancy ,Thrombin ,Cytokines ,Humans ,Female ,Thrombosis ,alpha-Macroglobulins ,Hematology ,Pregnancy-Associated alpha 2-Macroglobulins ,Transcription Factors - Abstract
Antiproteinases such as alpha-2-macroglobulin (A2M) play a role in hemostasis. A2M is highly conserved throughout evolution and is a high molecular weight homo-tetrameric glycoprotein. A2M proteinase inhibitor activity is possible via a unique cage structure leading to proteinase entrapment without direct enzymatic activity inhibition. Following this entrapment, proteinase clearance is possible through A2M binding to the low-density lipoprotein receptor-related protein 1. A2M synthesis is regulated by pro-inflammatory cytokines and increases during several chronic or acute inflammatory diseases and varies with age. For instance, A2M plasma levels are known to be increased in patients with diabetes mellitus, nephrotic syndrome, or sepsis. Concerning hemostasis, A2M can trap many proteinases involved in coagulation and fibrinolysis. Because of its pleiotropic effects A2M can be seen as both anti- and pro-hemostatic. A2M can inhibit thrombin, factor Xa, activated protein C, plasmin, tissue-plasminogen activator, and urokinase. Through its many different functions A2M is generally put apart in the balanced regulation of hemostasis. In addition, the fact that A2M plasma levels are differently regulated during inflammatory-related diseases and that A2M can neutralize cytokines that also modify hemostasis could explain why it is difficult to link common proteins and parameters of hemostasis with the mechanisms of thrombosis in such diseases. Thus, we propose in the present review to summarize known functions of A2M, give a brief overview about diseases, and then to focus on the roles of this antiproteinase in hemostasis and thrombosis.
- Published
- 2022
36. Vascular smooth muscle cell dysfunction: role in arterial stiffening and cardiovascular disease
- Author
-
Patrick Lacolley, Jean-Baptiste Michel, Delphine Gomez, Magnus Bäck, and Véronique Regnault
- Published
- 2022
37. Contributors
- Author
-
Elena Aikawa, S.G. Anderson, Livia Silva Araújo Passos, Samsul Arefin, Per M. Arvidsson, Alberto Avolio, Martin Bachler, Magnus Bäck, Michael J. Bashline, Dakota Becker-Greene, Jamie Bellinge, Amar Bennasroune, Sébastien Blaise, Barry A. Borlaug, Pierre Boutouyrie, Y. Breet, Jerome W. Breslin, Matthew J. Budoff, Mark Butlin, Marina Cecelja, Chen-Huan Chen, Hao-Min Cheng, Yi-Bang Cheng, Julio A. Chirinos, Phil Chowienczyk, Shao-Yuan Chuang, Marie-Annick Clavel, Jordana B. Cohen, Alexis M. Corcoran, William K. Cornwell, Vicente F. Corrales–Medina, Nancy Côté, Thais Coutinho, James Cox, J.K. Cruickshank, Lu Dai, Stella S. Daskalopoulou, Kevin P. Davy, Marc L. De Buyzere, Paul B. Dieffenbach, Laurent Duca, Girish Dwivedi, David G. Edwards, William B. Farquhar, Bo Fernhall, John S. Floras, Laura E. Fredenburgh, Masafumi Fukumitsu, L. Gafane-Matemane, Nestor Gahungu, Ahmed K. Ghanem, Thierry C. Gillebert, Philippe Gillery, Delphine Gomez, Ezequiel Guzzetti, Bernhard Hametner, Junichiro Hashimoto, Kevin S. Heffernan, Brooks A. Hibner, Sam Hobson, Nien-Wen Hu, T.M. Hughes, Jay D. Humphrey, Stéphane Jaisson, Nadjia Kachenoura, Kazuomi Kario, Prasad V.G. Katakam, Goro Katsuumi, Avinash Kondiboyina, Sándor J. Kovács, R. Kruger, Karolina Kublickiene, Patrick Lacolley, Muriel Laffargue, Arinola O. Lampejo, Agne Laucyte-Cibulskiene, Stéphane Laurent, Hae-Young Lee, Wesley K. Lefferts, Elizabeth C. Lefferts, Adelino F. Leite-Moreira, Chee H. Liew, Joao A.C. Lima, André P. Lourenço, Kaisa Maki-Petaja, Marcy Maracle, Laurent Martiny, Pascal Maurice, Christopher C. Mayer, Barry J. McDonnell, John W. McEvoy, M.L. Meyer, Jean-Baptiste Michel, Philip J. Millar, Tohru Minamino, Gary F. Mitchell, Walter L. Murfee, Jonathan P. Mynard, Massimo Nardone, Peter M. Nilsson, Kevin O'Gallagher, Yoshiaki Ohyama, Kazunori Omote, Jeong Bae Park, Shayn M. Peirce, Philippe Pibarot, Gary L. Pierce, Stuart B. Prenner, Athanase Protogerou, Reed E. Pyeritz, Michael A. Quail, Yogesh N.V. Reddy, Alban Redheuil, Véronique Regnault, Rakhshinda Rehman, Ernst R. Rietzschel, Béatrice Romier-Crouzet, Jasjit Rooprai, Lucia Salvi, Paolo Salvi, Hervé Sartelet, Christian E.H. Schmelzer, A.E. Schutte, Angelina Schwarz, Patrick Segers, James E. Sharman, Ippei Shimizu, Marc A. Simon, Piera Sosa, Bart Spronck, Peter Stenvinkel, Eric J. Stöhr, M. Strauss-Kruger, Ariana Suarez-Martinez, Masayoshi Suda, Shih-Hsien Sung, Isabella Tan, Dimitrios Terentes-Printzios, Raymond R. Townsend, Andrew H. Tran, Elaine M. Urbina, Bharath Ambale Venkatesh, Charalambos Vlachopoulos, Anton Vonk Noordegraaf, Amandine Wahart, Ji-Guang Wang, Siegfried Wassertheurer, Andrew James Webb, Thomas Weber, Berend E. Westerhof, Ian B. Wilkinson, and Yohko Yoshida
- Published
- 2022
38. P1.15 CONDITIONAL INACTIVATION OF INTEGRIN AV SUBUNIT IN VASCULAR SMOOTH MUSCLE CELLS REGULATES FIBROSIS IN VESSELS
- Author
-
Ekaterina Belozertseva, Melusine Didelot, Amel Mohamadi, Zhenlin Li, Huguette Louis, Jean-Baptiste Michel, Véronique Regnault, and Patrick Lacolley
- Subjects
Specialties of internal medicine ,RC581-951 ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Integrin avb3 is expressed at high density in vascular smooth muscle cells (VSMCs). It functions as a receptor for adhesion proteins in VSMCs which phenotypic modulation plays a pivotal role in arteriosclerosis and atherosclerosis. The aim was to study the role of integrin avb3 in angiotensin II (Ang II)-induced arterial fibrosis in mice and in human samples of atherosclerotic arteries in situ. Transgenic mice conditionally inactivated for integrin av subunit in VSMCs (avSMKO) were treated with Ang II (1,5 mg/kg/day) for 4 weeks. Immunostained slices of atherosclerotic plaques at different stages of development and primary cultures of human aortic VSMCs were used. At baseline, blood pressure was lower in avSMKO compared to control (WT) mice. Isobaric carotid distensibility was increased and remained higher in avSMKO in response to Ang II. The increase in collagen content in response to Ang II was lower in avSMKO than in WT (15 vs 36%) for similar increase in blood pressure (20 mmHg) and arterial wall hypertrophy. The immunohistochemistry of aortic slices showed stronger staining for integrin avb3 in atherosclerotic plaques compared to healthy aortas. In VSMC cultures, the mRNA of av was decreased. In conclusion, these results show that avb3 is strongly expressed in neointimal proliferation and in fibrous plaques. The av integrin subunit seems to regulate arterial fibrosis in response to hypertension and plaque growth. Low RNA quantities of av subunit of VSMCs contrasted with strong protein staining in plaques suggesting the participation of inflammatory cells in the synthesis of this integrin.
- Published
- 2015
- Full Text
- View/download PDF
39. Shedding Light on Hemostasis in Patients With Inflammatory Bowel Diseases
- Author
-
Laurent Peyrin-Biroulet, Patrick Lacolley, Jeremy Lagrange, Véronique Regnault, Denis Wahl, Défaillance Cardiovasculaire Aiguë et Chronique (DCAC), Université de Lorraine (UL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy), University Medical Center [Mainz], Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy), Nutrition-Génétique et Exposition aux Risques Environnementaux (NGERE), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lorraine (UL), Faculté de Médecine [Nancy], Université de Lorraine (UL), and Service d'Hépato-gastro-entérologie [CHRU Nancy]
- Subjects
[SDV]Life Sciences [q-bio] ,Fibrinogen ,Inflammatory bowel disease ,digestive system ,Fibrin ,03 medical and health sciences ,0302 clinical medicine ,Von Willebrand factor ,medicine ,Humans ,Platelet ,Blood Coagulation ,Hemostasis ,Crohn's disease ,Hepatology ,biology ,business.industry ,Fibrinolysis ,Inflammatory Bowel Disease ,Gastroenterology ,Endothelial Cells ,Thrombosis ,Inflammatory Bowel Diseases ,medicine.disease ,digestive system diseases ,3. Good health ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,030211 gastroenterology & hepatology ,business ,medicine.drug - Abstract
International audience; Patients with inflammatory bowel diseases (IBD) have an increased risk of thrombosis, possibly due to changes in blood cells and molecules involved in hemostasis. They have increased platelet counts and reactivity as well as increased platelet-derived large extracellular vesicles. Coagulation is continuously activated in patients with IBD, based on measured markers of thrombin generation, and the anticoagulant functions of endothelial cells are damaged. Furthermore, fibrinogen is increased and fibrin clots are denser. However, pathogenesis of thrombosis in patients with IBD appears to differ from that of patients without IBD. Patients with IBD also take drugs that might contribute to risk of thrombosis, complicating the picture. We review the features of homeostasis that are altered in patients with IBD and possible mechanisms of this relationship.
- Published
- 2021
40. Evidences for Inotropism Triggered by Simultaneous Antegrade And Retrograde Myocardial Perfusion in A Swine Model
- Author
-
Juan-Pablo Maureira, Patrick Lacolley, Vanessa Marie, Pierre-Yves Marie, Fréderique Groubatch-Joineau, Nguyen Tran, Brice Mourer, Aude Falanga, Daniel Grandmougin, and Antoine Chalon
- Subjects
Cardiac output ,medicine.medical_specialty ,Ejection fraction ,business.industry ,Stroke volume ,Great cardiac vein ,Coronary circulation ,medicine.anatomical_structure ,Internal medicine ,cardiovascular system ,Retrograde perfusion ,Cardiology ,Medicine ,business ,Perfusion ,Coronary sinus - Abstract
Objectives: Retrograde perfusion into the coronary sinus is currently used to deliver cardioplegia. We developed an in-vivo porcine beating-heart model of self-myocardial retroperfusion (SMR) using the venous route to increase myocardial oxygenation of the left ventricle. Then, we sought to assess whether the association of a simultaneous antegrade and retrograde myocardial perfusion with oxygenated blood might trigger hemodynamic and cardiac responses compared with a single antegrade myocardial supply. Methods: A group of 8 pigs was dedicated to undergo SMR with a simultaneous antegrade physiological LAD perfusion. SMR was achieved with a bypass-line between the ascending aorta and the coronary sinus to perform a selective retrograde perfusion of the great cardiac vein with oxygenated blood after ligation of the left azygos vein. Cardiac output (CO), maximal pressure in the LV (Pmax in-LV), stroke volume (SV), left ventricular ejection fraction (LVEF), diastolic durations, heart rate (HR), and arterial systemic pressure were monitored with conductance catheters. These data were collected during the antegrade myocardial perfusion (baseline period) and compared with data recorded during a simultaneous antegrade and retrograde perfusion. At the end of the procedures, the hearts were harvested for histology. Results: SMR with antegrade LAD perfusion showed inotropic properties with significant improvements in CO, SV, Pmax in-LV and LVEF (p
- Published
- 2019
41. Endothelial-driven increase in plasma thrombin generation characterising a new hypercoagulable phenotype in acute heart failure
- Author
-
Batric Popovic, Patrick Lacolley, Faiez Zannad, Sébastien Gibot, Cécile V. Denis, Cécile Lakomy, Isabelle Clerc-Urmès, Huguette Louis, Véronique Regnault, Défaillance Cardiovasculaire Aiguë et Chronique (DCAC), Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lorraine (UL), Service de Cardiologie [CHRU Nancy], Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy), Ingénierie, Biologie et Santé en Lorraine (IBSLor), Université de Lorraine (UL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Nutrition-Génétique et Exposition aux Risques Environnementaux (NGERE), Université de Lorraine (UL)-Institut National de la Santé et de la Recherche Médicale (INSERM), Hémostase et biologie vasculaire, Université Paris-Sud - Paris 11 (UP11)-IFR93-Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Lorraine (UL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy), and Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lorraine (UL)
- Subjects
Male ,030204 cardiovascular system & hematology ,Ventricular Function, Left ,0302 clinical medicine ,Heart Rate ,Platelet ,Sinus rhythm ,Prospective Studies ,030212 general & internal medicine ,Endothelial dysfunction ,ComputingMilieux_MISCELLANEOUS ,Thrombin ,Venous Thromboembolism ,Flow Cytometry ,Prognosis ,Thrombosis ,Blood Coagulation Factors ,Nucleosomes ,3. Good health ,medicine.anatomical_structure ,Acute Disease ,Cardiology ,Female ,Blood Coagulation Tests ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,medicine.drug ,Blood Platelets ,medicine.medical_specialty ,Endothelium ,Enzyme-Linked Immunosorbent Assay ,Receptors, Cell Surface ,Inflammation ,03 medical and health sciences ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Internal medicine ,medicine ,Humans ,Aged ,Heart Failure ,business.industry ,Anticoagulants ,[SDV.BBM.BM]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,medicine.disease ,Heart failure ,Endothelium, Vascular ,business ,Biomarkers ,Protein C ,Follow-Up Studies - Abstract
International audience; Background: Subjects with heart failure (HF) are at higher risk of developing thrombosis. We investigated whether endothelium activation and inflammation induce a prothrombotic biological profile in patients with acute decompensated HF (ADHF) and sinus rhythm.Methods: Our prospective study included 34 ADHF patients, 30 patients with stable chronic HF (CHF) and 30 control inpatients without HF. In vitro thrombin generation and its downregulation by activated protein C (APC) was monitored by calibrated automated thrombography at hospital admission, at the day of discharge and after discharge, following at least six weeks of clinical stability. Circulating endothelium-derived extracellular vesicles (eEVs) were quantified by flow cytometry and nucleosomes by ELISA.Results: Thrombin generation is increased and APC sensitivity is decreased independently of platelets in ADHF at admission compared to controls (p < 0.01). Thrombin generation was also increased in CHF but only in the presence of platelets. Plasma markers of endothelium activation (von Willebrand factor, factor VIII, procoagulant eEVs and circulating nucleosomes) and the ability of plasmas to induce neutrophil extracellular trap formation in control neutrophils are elevated in ADHF at admission compared to controls (p < 0.001). In-hospital prothrombotic changes in ADHF improved significantly at the post-discharge time-point. Circulating nucleosomes were positively correlated with APC sensitivity (p = 0.013) and annexin-V-positive eEVs (p = 0.004).Conclusions: This proof-of-concept study identified an endothelial-driven hypercoagulable phenotype at the acute phase of decompensated HF contrasting with the platelet-dependent prothrombotic state in CHF. These results highlighted a cross-talk between circulating eEVs and nucleosomes, procoagulant factors and impairment of the APC anticoagulant activity in ADHF.
- Published
- 2019
42. The role of Bcl11b in vascular smooth muscle cell death
- Author
-
Amata El Mouhawess, Jocelyne Blanc, Jacqueline Gao-Li, Onnik Agbulut, Patrick Lacolley, Ara Parlakian, and Zhenlin Li
- Subjects
Cardiology and Cardiovascular Medicine - Published
- 2022
43. Simultaneous characterization of metabolic, cardiac, vascular and renal phenotypes of lean and obese SHHF rats.
- Author
-
Gina Youcef, Arnaud Olivier, Clément P J L'Huillier, Carlos Labat, Renaud Fay, Lina Tabcheh, Simon Toupance, Rosa-Maria Rodriguez-Guéant, Damien Bergerot, Frédéric Jaisser, Patrick Lacolley, Faiez Zannad, Laurent Vallar, and Anne Pizard
- Subjects
Medicine ,Science - Abstract
Individuals with metabolic syndrome (MetS) are prone to develop heart failure (HF). However, the deleterious effects of MetS on the continuum of events leading to cardiac remodeling and subsequently to HF are not fully understood. This study characterized simultaneously MetS and cardiac, vascular and renal phenotypes in aging Spontaneously Hypertensive Heart Failure lean (SHHF(+/?) regrouping (+/+) and (+/cp) rats) and obese (SHHF(cp/cp), "cp" defective mutant allele of the leptin receptor gene) rats. We aimed to refine the milestones and their onset during the progression from MetS to HF in this experimental model. We found that SHHF(cp/cp )but not SHHF(+/?) rats developed dyslipidemia, as early as 1.5 months of age. This early alteration in the lipidic profile was detectable concomitantly to impaired renal function (polyuria, proteinuria but no glycosuria) and reduced carotid distensibility as compared to SHHF(+/?) rats. By 3 months of age SHHFcp/cp animals developed severe obesity associated with dislipidemia and hypertension defining the onset of MetS. From 6 months of age, SHHF(+/?) rats developed concentric left ventricular hypertrophy (LVH) while SHHF(cp/cp) rats developed eccentric LVH apparent from progressive dilation of the LV dimensions. By 14 months of age only SHHF(cp/cp) rats showed significantly higher central systolic blood pressure and a reduced ejection fraction resulting in systolic dysfunction as compared to SHHF(+/?). In summary, the metabolic and hemodynamic mechanisms participating in the faster decline of cardiac functions in SHHF(cp/cp) rats are established long before their physiological consequences are detectable. Our results suggest that the molecular mechanisms triggered within the first three months after birth of SHHF(cp/cp) rats should be targeted preferentially by therapeutic interventions in order to mitigate the later HF development.
- Published
- 2014
- Full Text
- View/download PDF
44. Systematic Review of Antiphospholipid Antibodies in COVID-19 Patients: Culprits or Bystanders?
- Author
-
Patrick Lacolley, Thomas Foret, Virginie Dufrost, Denis Wahl, Véronique Regnault, Stéphane Zuily, Patricia Costa, Lucie Salomon du Mont, Benjamin Lefevre, Défaillance Cardiovasculaire Aiguë et Chronique (DCAC), Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lorraine (UL), Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy), Centre Hospitalier Régional Universitaire de Besançon (CHRU Besançon), Marqueurs pronostiques et facteurs de régulations des pathologies cardiaques et vasculaires - UFC ( UR 3920) (PCVP / CARDIO), Centre Hospitalier Régional Universitaire de Besançon (CHRU Besançon)-Université de Franche-Comté (UFC), Université Bourgogne Franche-Comté [COMUE] (UBFC)-Université Bourgogne Franche-Comté [COMUE] (UBFC), Service des Maladies Infectieuses et Tropicales [CHRU Nancy], Adaptation, mesure et évaluation en santé. Approches interdisciplinaires (APEMAC), Université de Lorraine (UL), REGNAULT, Véronique, and Marqueurs pronostiques et facteurs de régulations des pathologies cardiaques et vasculaires - UFC ( EA 3920) (PCVP / CARDIO)
- Subjects
medicine.medical_specialty ,[SDV]Life Sciences [q-bio] ,Disease ,030204 cardiovascular system & hematology ,Severity of Illness Index ,Fibrin Fibrinogen Degradation Products ,03 medical and health sciences ,0302 clinical medicine ,Rheumatology ,immune system diseases ,Internal medicine ,Severity of illness ,medicine ,Humans ,neoplasms ,ComputingMilieux_MISCELLANEOUS ,030203 arthritis & rheumatology ,Lupus anticoagulant ,biology ,business.industry ,SARS-CoV-2 ,Antiphospholipid Syndrome (S Zuily, Section Editor) ,Antiphospholipid antibodies ,COVID-19 ,Fibrinogen ,Thrombosis ,medicine.disease ,Isotype ,3. Good health ,Immunoglobulin A ,[SDV] Life Sciences [q-bio] ,C-Reactive Protein ,Immunoglobulin M ,beta 2-Glycoprotein I ,Antibodies, Anticardiolipin ,Immunoglobulin G ,Lupus Coagulation Inhibitor ,biology.protein ,Antibodies, Antiphospholipid ,Antibody ,business ,Cohort study - Abstract
International audience; Purpose of review: COVID-19 patients have a procoagulant state with a high prevalence of thrombotic events. The hypothesis of an involvement of antiphospholipid antibodies (aPL) has been suggested by several reports. Here, we reviewed 48 studies investigating aPL in COVID-19 patients.Recent findings: Prevalence of Lupus Anticoagulant (LA) ranged from 35% to 92% in ICU patients. Anti-cardiolipin (aCL) IgG and IgM were found in up to 52% and up to 40% of patients respectively. Anti-β2-glycoprotein I (aβ2-GPI) IgG and IgM were found in up to 39% and up to 34% of patients respectively. Between 1% and 12% of patients had a triple positive aPL profile. There was a high prevalence of aβ2-GPI and aCL IgA isotype. Two cohort studies found few persistent LA but more persistent solid phase assay aPL over time. aPL determination and their potential role is a real challenge for the treatment of this disease.
- Published
- 2021
45. Genome-wide association analysis in dilated cardiomyopathy reveals two new players in systolic heart failure on chromosomes 3p25.1 and 22q11.23
- Author
-
Magdalena Harakalova, Benjamin Meder, Philippe Charron, Manuel Gómez-Bueno, Jorg J. A. Calis, François Cambien, David-Alexandre Trégouët, Maurizia Grasso, Steven McGinn, Uwe Völker, Thomas Meitinger, Stefan Weiss, L. Duboscq-Bidot, Richard Dorent, Vera Regitz-Zagrosek, Folkert W. Asselbergs, Hélène Blanché, Olivier Dubourg, Patrick Lacolley, Pierre Boutouyrie, Delphine Bacq-Daian, Vincent Fontaine, Volker Ruppert, Marine Germain, K Lehnert, Jean-Noël Trochu, Stuart A. Cook, Angélique Curjol, Brendan J. Keating, Ibticem Raji, Anne Boland, J. Erdmann, Michael Morley, Jean-François Aupetit, Paloma Remior, Luigi Tavazzi, Gérard Roizès, Michal Mokry, Konstantin Strauch, Richard Isnard, Jean-Philippe Empana, Robert Olaso, Kenneth B. Marguiles, Zofia T. Bilińska, Stephan B. Felix, Marcus Dörr, Thomas P. Cappola, Stefan Blankenberg, Jan Haas, Céline Besse, Jean-François Deleuze, Christine E. Seidman, Christian Hengstenberg, Jessica van Setten, Hakon Hakonarson, Sanjay K Prasad, Daiane Hemerich, Pascal de Groote, Thomas Wichter, Alain van Mil, Michel Komajda, Renee Maas, Carole Proust, Declan P. O'Regan, Xavier Jouven, Ganapathi Varma Saripella, Georgios Kararigas, Eloisa Arbustini, Jin Li, Klaus Stark, Laurent Fauchier, Flavie Ader, Melanie Waldenberger, Martina Müller-Nurasyid, Eric Villard, Sophie Garnier, Cardiology, Imperial College Healthcare NHS Trust- BRC Funding, British Heart Foundation, Unité de Recherche sur les Maladies Cardiovasculaires, du Métabolisme et de la Nutrition = Institute of cardiometabolism and nutrition (ICAN), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Sorbonne Université (SU), Service de Cardiologie [CHU Pitié-Salpêtrière], CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), CHU Tenon [AP-HP], Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Unité Fonctionnelle de Cardiogénétique et Myogénétique Moléculaire et Cellulaire, Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-CHU Pitié-Salpêtrière [AP-HP], Centre hospitalier Saint-Joseph [Paris], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Bordeaux population health (BPH), Université de Bordeaux (UB)-Institut de Santé Publique, d'Épidémiologie et de Développement (ISPED)-Institut National de la Santé et de la Recherche Médicale (INSERM), Service de Génétique Cytogénétique et Embryologie [CHU Pitié-Salpêtrière], Assistance publique-Hôpitaux de Paris, Fondation Leducq, Société Française de Cardiologie, Deutsche Forschungsgemeinschaft, Helmholtz Zentrum München, Université de Bordeaux, Medical Research Council, ANR-10-LABX-0013,GENMED,Medical Genomics(2010), Unité de Recherche sur les Maladies Cardiovasculaires, du Métabolisme et de la Nutrition = Research Unit on Cardiovascular and Metabolic Diseases (ICAN), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Institut de Cardiométabolisme et Nutrition = Institute of Cardiometabolism and Nutrition [CHU Pitié Salpêtrière] (IHU ICAN), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), University Medical Center [Utrecht], Universität Greifswald - University of Greifswald, German Center for Cardiovascular Research (DZHK), Berlin Institute of Health (BIH), Charité - UniversitätsMedizin = Charité - University Hospital [Berlin], University of Pennsylvania, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), National Heart Centre Singapore (NHCS), Children’s Hospital of Philadelphia (CHOP ), University of Regensburg, Royal Brompton Hospital, Imperial College London, Istituti Clinici Scientifici Maugeri [Pavia] (IRCCS Pavia - ICS Maugeri), Helmholtz Zentrum München = German Research Center for Environmental Health, Ludwig-Maximilians-Universität München (LMU), Technische Universität Munchen - Université Technique de Munich [Munich, Allemagne] (TUM), University Medical Center of the Johannes Gutenberg-University Mainz, Perelman School of Medicine, Harvard Medical School [Boston] (HMS), University of Iceland [Reykjavik], Heidelberg University, Stanford University Medical School, Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO), Défaillance Cardiovasculaire Aiguë et Chronique (DCAC), Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lorraine (UL), Universität zu Lübeck = University of Lübeck [Lübeck], Universitätklinikum Gießen und Marburg GmbH, Maria Cecilia Hospital [Cotignola], Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Institut de génétique humaine (IGH), Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS), Hôpital cardiologique, Université de Lille, Droit et Santé-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), CHU Trousseau [Tours], Centre Hospitalier Régional Universitaire de Tours (CHRU Tours), Unité de recherche de l'institut du thorax (ITX-lab), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN), Centre hospitalier Saint Joseph - Saint Luc [Lyon], National Institute of Cardiology [Varsovie, Pologne], University of Medicine Greifswald, Centre de Référence Maladies Cardiaques Héréditaires, Centre National de Recherche en Génomique Humaine (CNRGH), Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Laboratory of Excellence GENMED [Paris] (Medical Genomics), Hospital Universitario Puerta de Hierro-Majadahonda [Madrid, Spain], Swedish University of Agricultural Sciences (SLU), Centre d'Etude du Polymorphisme Humain (CEPH), Institut Universitaire d'Hématologie (IUH), Université Paris Diderot - Paris 7 (UPD7)-Université Paris Diderot - Paris 7 (UPD7)-Fondation Jean Dausset-Université Paris Cité (UPCité), Faculté de Pharmacie de Paris - Université Paris Descartes (UPD5 Pharmacie), Université Paris Descartes - Paris 5 (UPD5), Groupe Hospitalier Paris Saint Joseph, Fondation Jean Dausset - Centre d’Etudes du Polymorphisme Humain [Paris] (CEPH), and University College of London [London] (UCL)
- Subjects
Cardiac & Cardiovascular Systems ,Cardiomyopathy, Dilated/genetics ,[SDV]Life Sciences [q-bio] ,Signal Transducing/genetics ,Dilated cardiomyopathy ,Genome-wide association study ,Adaptor Proteins, Signal Transducing/genetics ,030204 cardiovascular system & hematology ,TAURINE ,0302 clinical medicine ,GWAS ,Medicine ,POSITION STATEMENT ,1102 Cardiorespiratory Medicine and Haematology ,Genetics ,0303 health sciences ,education.field_of_study ,Genetic Predisposition to Disease/genetics ,Adaptor Proteins ,4C-sequencing ,Polymorphism, Single Nucleotide/genetics ,Genetic risk score ,Cardiology and Cardiovascular Medicine ,Life Sciences & Biomedicine ,Single Nucleotide/genetics ,Cardiomyopathy, Dilated ,Cardiomyopathy ,Population ,Locus (genetics) ,Single-nucleotide polymorphism ,Polymorphism, Single Nucleotide ,Chromosomes ,03 medical and health sciences ,Systolic/genetics ,Heart Failure, Systolic/genetics ,SNP ,Animals ,Humans ,Genetic Predisposition to Disease ,Allele ,Polymorphism ,education ,Imputation ,Adaptor Proteins, Signal Transducing ,030304 developmental biology ,Heart Failure ,Science & Technology ,business.industry ,WORKING GROUP ,1103 Clinical Sciences ,medicine.disease ,Genetic architecture ,Cardiovascular System & Hematology ,Dilated/genetics ,Cardiovascular System & Cardiology ,[SDV.SPEE]Life Sciences [q-bio]/Santé publique et épidémiologie ,business ,Apoptosis Regulatory Proteins ,Heart Failure, Systolic ,Genome-Wide Association Study - Abstract
Aims Our objective was to better understand the genetic bases of dilated cardiomyopathy (DCM), a leading cause of systolic heart failure. Methods and results We conducted the largest genome-wide association study performed so far in DCM, with 2719 cases and 4440 controls in the discovery population. We identified and replicated two new DCM-associated loci on chromosome 3p25.1 [lead single-nucleotide polymorphism (SNP) rs62232870, P = 8.7 × 10−11 and 7.7 × 10−4 in the discovery and replication steps, respectively] and chromosome 22q11.23 (lead SNP rs7284877, P = 3.3 × 10−8 and 1.4 × 10−3 in the discovery and replication steps, respectively), while confirming two previously identified DCM loci on chromosomes 10 and 1, BAG3 and HSPB7. A genetic risk score constructed from the number of risk alleles at these four DCM loci revealed a 3-fold increased risk of DCM for individuals with 8 risk alleles compared to individuals with 5 risk alleles (median of the referral population). In silico annotation and functional 4C-sequencing analyses on iPSC-derived cardiomyocytes identify SLC6A6 as the most likely DCM gene at the 3p25.1 locus. This gene encodes a taurine transporter whose involvement in myocardial dysfunction and DCM is supported by numerous observations in humans and animals. At the 22q11.23 locus, in silico and data mining annotations, and to a lesser extent functional analysis, strongly suggest SMARCB1 as the candidate culprit gene. Conclusion This study provides a better understanding of the genetic architecture of DCM and sheds light on novel biological pathways underlying heart failure.
- Published
- 2021
46. A case-control study indicates that coagulation imbalance is associated with arteriosclerosis and markers of endothelial dysfunction in kidney failure
- Author
-
Véronique Regnault, Gérard M. London, Athanase Benetos, Lucie Tran, Patrick Lacolley, Yvonnick Bézie, Tomas Serrato, Bruno Pannier, Défaillance Cardiovasculaire Aiguë et Chronique (DCAC), Université de Lorraine (UL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy), Centre Hospitalier Manhès [Fleury-Mérogis], Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy), Cardiovascular and Renal Clinical Trialists [Vandoeuvre-les-Nancy] (INI-CRCT), Institut Lorrain du Coeur et des Vaisseaux Louis Mathieu [Nancy], Centre hospitalier Saint-Joseph [Paris], and Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Arteriosclerosis ,[SDV]Life Sciences [q-bio] ,030232 urology & nephrology ,Pulse Wave Analysis ,03 medical and health sciences ,0302 clinical medicine ,Thrombin ,Von Willebrand factor ,Internal medicine ,medicine ,Humans ,Platelet ,Renal Insufficiency ,Endothelial dysfunction ,Pulse wave velocity ,Blood Coagulation ,ComputingMilieux_MISCELLANEOUS ,biology ,business.industry ,medicine.disease ,030104 developmental biology ,Endocrinology ,Coagulation ,Nephrology ,Case-Control Studies ,biology.protein ,Arterial stiffness ,business ,medicine.drug ,Kidney disease - Abstract
Endothelial dysfunction, one of many causes of arterial changes in end-stage kidney disease (kidney failure), is a likely link between early vascular aging and the risk of thrombosis or bleeding in this condition. To evaluate this, we compared links between arterial stiffness and endothelial/coagulation factors in 55 patients receiving hemodialysis therapy and 57 age-/sex-matched control individuals. Arterial stiffness was assessed from carotid-femoral pulse wave velocity, and coagulation status from the endogenous thrombin generating potential. Markers of endothelial dysfunction (von Willebrand factor, tissue factor pathway inhibitor), neutrophil extracellular traps and tissue factor-positive extracellular vesicles were higher in patients with kidney failure. Prothrombin fragments 1 and 2, and D-dimer markers of in vivo coagulation activation were also higher. However, in vitro in the presence of platelets, endogenous thrombin generating potential was lower and its downregulation by activated protein C impaired. Antiplatelet drugs did not affect these parameters. In multiple regression analysis, prothrombin fragments 1 and 2, D-dimer, factor VIII and monocyte-derived tissue factor-positive extracellular vesicles correlated with higher carotid-femoral pulse wave velocity. In patients with kidney failure, in vivo hypercoagulability occurred with reduced thrombin generation in platelet-rich plasma, likely explaining the opposing thrombotic and bleeding tendencies in patients with kidney failure. Importantly, arteriosclerosis is more closely related to a prothrombotic state. Thus, coagulation changes plus arterial stiffness highlight a major therapeutic challenge for anticoagulant and antiplatelet drug use.
- Published
- 2021
47. Elastase and exacerbation of neutrophil innate immunity are involved in multi‐visceral manifestations of COVID‐19
- Author
-
Mickael Gette, Jonas Callet, François Blanchecotte, Jérémie Raso, Véronique Regnault, Jean-Louis Guéant, Rosa-Maria Guéant-Rodriguez, Stanislas Gleye, Huguette Louis, Céline Chéry, Abderrahim Oussalah, Julien Fromonot, Régis Guieu, Patrick Lacolley, Nutrition-Génétique et Exposition aux Risques Environnementaux (NGERE), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lorraine (UL), Centre recherche en CardioVasculaire et Nutrition = Center for CardioVascular and Nutrition research (C2VN), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Défaillance Cardiovasculaire Aiguë et Chronique (DCAC), Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lorraine (UL), Aix-Marseille Université - École de médecine (AMU SMPM MED), Aix-Marseille Université - Faculté des sciences médicales et paramédicales (AMU SMPM), Aix Marseille Université (AMU)-Aix Marseille Université (AMU), Laboratoire d'Analyses Médicales L'Abo+, Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy), The study was funded by the research project FHU ARRIMAGE and the French PIA project 'Lorraine Université d’Excellence,' reference ANR-15-IDEX04-LUE., IMPACT GEENAGE, ANR-15-IDEX-0004,LUE,Isite LUE(2015), Lucas, Nelly, ISITE - Isite LUE - - LUE2015 - ANR-15-IDEX-0004 - IDEX - VALID, and Université de Lorraine (UL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy)
- Subjects
0301 basic medicine ,Exacerbation ,[SDV.IMM] Life Sciences [q-bio]/Immunology ,Neutrophils ,[SDV]Life Sciences [q-bio] ,Immunology ,Extracellular Traps ,Histones ,03 medical and health sciences ,DNase ,0302 clinical medicine ,COVID‐19 ,medicine ,Humans ,Immunology and Allergy ,CXC chemokine receptors ,innate immunity ,ComputingMilieux_MISCELLANEOUS ,Innate immune system ,Lung ,biology ,business.industry ,SARS-CoV-2 ,Elastase ,COVID-19 ,neutrophil extracellular traps (NETs) ,Neutrophil extracellular traps ,Immunity, Innate ,3. Good health ,myeloperoxidase ,030104 developmental biology ,medicine.anatomical_structure ,030228 respiratory system ,Neutrophil elastase ,Myeloperoxidase ,biology.protein ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Original Article ,business - Abstract
Background Many arguments suggest that neutrophils could play a prominent role in COVID‐19. However, the role of key components of neutrophil innate immunity in severe forms of COVID‐19 has deserved insufficient attention. We aimed to evaluate the involvement of neutrophil elastase, histone‐DNA, and DNases in systemic and multi‐organ manifestations of COVID‐19. Methods We performed a multicenter study of markers of neutrophil innate immunity in 155 cases consecutively recruited in a screening center, local hospitals, and two regional university hospitals. The cases were evaluated according to clinical and biological markers of severity and multi‐organ manifestations and compared to 35 healthy controls. Results Blood neutrophil elastase, histone‐DNA, myeloperoxidase‐DNA, and free dsDNA were dramatically increased, and DNase activity was decreased by 10‐fold, compared with controls. Neutrophil elastase and histone‐DNA were associated with intensive care admission, body temperature, lung damage, and markers of cardiovascular outcomes, renal failure, and increased interleukin‐6 (IL‐6), IL‐8, and CXCR2. Neutrophil elastase was an independent predictor of the computed tomography score of COVID‐19 lung damage and the number of affected organs, in multivariate analyses. The increased blood concentrations of NE and neutrophil extracellular traps were related to exacerbation of neutrophil stimulation through IL‐8 and CXCR2 increased concentrations and increased serum DAMPs, and to impaired degradation of NETs as a consequence of the dramatic decrease in blood DNase activity. Conclusion Our results point out the key role of neutrophil innate immunity exacerbation in COVID‐19. Neutrophil elastase and DNase could be potential biomarkers and therapeutic targets of severe systemic manifestations of COVID‐19., Blood levels of neutrophil elastase and histone‐DNA are associated with severe and systemic and multi‐organ manifestations of COVID‐19. Increased blood concentrations of neutrophil elastase and neutrophil extracellular traps are related to exacerbation of neutrophil stimulation through activated IL‐8/CXCR2 pathway. Neutrophil elastase and DNase could be potential biomarkers and therapeutic targets of severe systemic manifestations of COVID‐19.
- Published
- 2021
- Full Text
- View/download PDF
48. P.31 Von Willebrand Factor Induces Vascular Smooth Muscle Cell Proliferation and Migration Through Low Density Lipoprotein-Related Receptor Protein 4 and αvβ3 Integrin
- Author
-
Patrick Lacolley, Cécile V. Denis, Jean-Baptiste Michel, Peter J. Lenting, Alexandre Raoul, Véronique Regnault, and Jeremy Lagrange
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Vascular smooth muscle ,LRP4 ,Specialties of internal medicine ,chemistry.chemical_compound ,Von Willebrand factor ,hemic and lymphatic diseases ,Medicine ,VWF ,Diseases of the circulatory (Cardiovascular) system ,Receptor ,αvβ3 integrin ,biology ,Cell growth ,business.industry ,VSMC ,General Medicine ,αvβ3 ,Cell biology ,chemistry ,RC581-951 ,Low-density lipoprotein ,RC666-701 ,biology.protein ,cardiovascular system ,business ,circulatory and respiratory physiology - Abstract
Background and Objectives: Von Willebrand factor (VWF) is a plasma glycoprotein involved in primary hemostasis but recent data suggest additional roles beyond hemostasis in angiogenesis and potentially in vascular smooth muscle cell (VSMC) proliferation. Our aim was to investigate how VWF can modulate VSMC proliferation and identified the underlying mechanisms and the in vivo pathophysiological relevance. Methods and Results: Cultured aortic VSMCs proliferation and migration were increased in the presence of VWF. VSMCs treatment with a siRNA targeting αv integrin or the RGT-peptide blocking αvβ3 signaling completely inhibited proliferation. VWF did not bind directly to αvβ3 on VSMCs. We identified that VWF A2 domain was able to bind VSMCs. Since the low-density lipoprotein-related receptor protein (LRP) family are known to act as co-receptors we hypothesized the involvement of a member in the signaling pathway. Using the universal LRP-inhibitor (RAP), we confirmed LRP-mediated VSMC proliferation. siRNA experiments and proximity ligation assay staining identified LRP4 as the VWF-counterreceptor on VSMCs and showed co-localization between αvβ3 and LRP4. Carotid ligations were applied to VWF +/+ and −/− mice and intimal hyperplasia (IH) was measured. Less VWF−/− mice developed IH compared to VWF +/+ mice. Finally, the proliferative effect of VWF was confirmed in human atherosclerotic lesions from different vessels (aortas, carotids) showing a proximity between VWF and a-SM actin positive cells. Conclusions: VWF mediates VSMC proliferation through its A2 domain binding to the LRP4 receptor and integrin αvβ3 signaling. The decreased IH following vascular injury suggests that targeting VWF-LRP4 interactions may contribute to limit remodeling.
- Published
- 2020
49. Sex Differences in Arterial Stiffening and Central Pulse Pressure
- Author
-
Patrick Lacolley, Michel E. Safar, Véronique Regnault, Hôpital Hôtel-Dieu [Paris], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Défaillance Cardiovasculaire Aiguë et Chronique (DCAC), and Université de Lorraine (UL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy)
- Subjects
business.industry ,[SDV]Life Sciences [q-bio] ,Biophysics ,Medicine ,Vascular aging ,Cardiology and Cardiovascular Medicine ,business ,Pulse wave velocity ,ComputingMilieux_MISCELLANEOUS ,Pulse pressure ,Stiffening - Abstract
International audience
- Published
- 2020
50. Genome wide association analysis in dilated cardiomyopathy reveals two new key players in systolic heart failure on chromosome 3p25.1 and 22q11.23
- Author
-
Céline Besse, François Cambien, Folkert W. Asselbergs, Robert Olaso, Jeanette Erdman, Benjamin Meder, Stephan B. Felix, Stefan Weiss, Laurent Fauchier, Konstantin Strauch, Luigi Tavazzi, Anne Boland, Gérard Roizès, Pascal DeGroote, Renee Maas, Melanie Waldenberger, Ganapathi Varma Saripella, Pablo García-Pavía, Brendan J. Keating, Vera Regitz-Zagrosek, Marine Germain, Stefan Blankenberg, Jessica van Setten, Eloisa Arbustini, Pierre Boutouyrie, Carole Proust, Delphine Bacq-Daian, Hemerich Daiane, Sophie Garnier, Michal Mokry, Richard Dorent, Martina Müller-Nurasyid, Philippe Charron, Maurizia Grasso, Steven Mc Ginn, Vincent Fontaine, Uwe Völker, Patrick Lacolley, Thomas Meitinger, Christine E. Seidman, Ibticem Raji, David-Alexandre Trégouët, Jean-Noël Trochu, Thomas Wichter, Jörg Callis, Alain van Mil, Jean-François Deleuze, Declan P. O'Regan, Xavier Jouven, Jin Li, Klaus Stark, Eric Villard, Stuart A. Cook, Hakon Hakonarson, Michael Morley, Kenneth B. Marguiles, Sanjay K Prasad, Volker Ruppert, Jean-François Aupetit, Jean-Philippe Empana, Marcus Dörr, Thomas P. Cappola, Michel Komajda, Magdalena Harakalova, Christian Hengstenberg, Hélène Blanché, Angélique Curjol, L. Duboscq-Bidot, Richard Isnard, Olivier Dubourg, and K Lehnert
- Subjects
Genetics ,0303 health sciences ,education.field_of_study ,Population ,Genome-wide association study ,Dilated cardiomyopathy ,Locus (genetics) ,030204 cardiovascular system & hematology ,Biology ,medicine.disease ,Genome ,Genetic architecture ,03 medical and health sciences ,0302 clinical medicine ,medicine ,SNP ,education ,Gene ,030304 developmental biology - Abstract
SummaryWe present the results of the largest genome wide association study (GWAS) performed so far in dilated cardiomyopathy (DCM), a leading cause of systolic heart failure and cardiovascular death, with 2,719 cases and 4,440 controls in the discovery population. We identified and replicated two new DCM-associated loci, one on chromosome 3p25.1 (lead SNP rs62232870, p = 8.7 × 10−11 and 7.7 × 10−4 in the discovery and replication step, respectively) and the second on chromosome 22q11.23 (lead SNP rs7284877, p = 3.3 × 10−8 and 1.4 × 10−3 in the discovery and replication step, respectively) while confirming two previously identified DCM loci on chromosome 10 and 1, BAG3 and HSPB7. The genetic risk score constructed from the number of lead risk-alleles at these four DCM loci revealed that individuals with 8 risk-alleles were at a 27% increased risk of DCM compared to individuals with 5 risk alleles (median of the referral population). We estimated the genome wide heritability at 31% ± 8%.In silico annotation and functional 4C-sequencing analysis on iPSC-derived cardiomyocytes strongly suggest SLC6A6 as the most likely DCM gene at the 3p25.1 locus. This gene encodes a taurine and beta-alanine transporter whose involvement in myocardial dysfunction and DCM is supported by recent observations in humans and mice. Although less easy to discriminate the better candidate at the 22q11.23 locus, SMARCB1 appears as the strongest one.This study provides both a better understanding of the genetic architecture of DCM and new knowledge on novel biological pathways underlying heart failure, with the potential for a therapeutic perspective.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.