44 results on '"Guerin CL"'
Search Results
2. Early-onset ventilator-associated pneumonia incidence in intensive care units: a surveillance-based study
- Author
-
Lepape Alain, Januel Jean-Marie, Voirin Nicolas, Bénet Thomas, Vanhems Philippe, Allaouchiche Bernard, Argaud Laurent, Chassard Dominique, and Guérin Claude
- Subjects
Infectious and parasitic diseases ,RC109-216 - Abstract
Abstract Background The incidence of ventilator-associated pneumonia (VAP) within the first 48 hours of intensive care unit (ICU) stay has been poorly investigated. The objective was to estimate early-onset VAP occurrence in ICUs within 48 hours after admission. Methods We analyzed data from prospective surveillance between 01/01/2001 and 31/12/2009 in 11 ICUs of Lyon hospitals (France). The inclusion criteria were: first ICU admission, not hospitalized before admission, invasive mechanical ventilation during first ICU day, free of antibiotics at admission, and ICU stay ≥ 48 hours. VAP was defined according to a national protocol. Its incidence was the number of events per 1,000 invasive mechanical ventilation-days. The Poisson regression model was fitted from day 2 (D2) to D8 to incident VAP to estimate the expected VAP incidence from D0 to D1 of ICU stay. Results Totally, 367 (10.8%) of 3,387 patients in 45,760 patient-days developed VAP within the first 9 days. The predicted cumulative VAP incidence at D0 and D1 was 5.3 (2.6-9.8) and 8.3 (6.1-11.1), respectively. The predicted cumulative VAP incidence was 23.0 (20.8-25.3) at D8. The proportion of missed VAP within 48 hours from admission was 11% (9%-17%). Conclusions Our study indicates underestimation of early-onset VAP incidence in ICUs, if only VAP occurring ≥ 48 hours are considered to be hospital-acquired. Clinicians should be encouraged to develop a strategy for early detection after ICU admission.
- Published
- 2011
- Full Text
- View/download PDF
3. The Onset of Intussusceptive Angiogenesis in COVID-19 Patients Might Come from the Mobilization of Stem Cell Sub-Populations Expressing the Hemangioblast Marker CD143.
- Author
-
Soret L, Guerin CL, Goudot G, Guyonnet L, Diehl JL, Philippe A, Gaussem P, and Smadja DM
- Subjects
- Humans, Male, Female, Middle Aged, Cross-Sectional Studies, Hemangioblasts metabolism, Aged, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic pathology, Biomarkers metabolism, Adult, Leukocytes, Mononuclear metabolism, Stem Cells metabolism, Angiogenesis, Basigin, COVID-19 pathology, COVID-19 metabolism, SARS-CoV-2
- Abstract
COVID-19 and infectious diseases have been included in strategic development goals (SDG) of United Nations (UN). The SARS-CoV-2 pandemic has unveiled complex pathophysiological mechanisms underpinning COVID-19, notably inducing a systemic acquired vascular hemopathy characterized by endothelial dysfunction and intussusceptive angiogenesis, a rapid vascular remodeling process identified as a hallmark in severe COVID-19 cases affecting pulmonary and cardiac tissues. Stem cell migration have been proposed as significant regulators of this neoangiogenic process. In a monocentric cross-sectional study, through spectral flow cytometry analysis of peripheral blood mononuclear cells, we identified a distinct stem cell subpopulation mobilized in critical COVID-19. Indeed, by an unsupervised analysis generating a UMAP representation we highlighted eleven different clusters in critical and non-critical COVID-19 patients. Only one cluster was significantly associated to critical COVID-19 compared to non-critical patients. This cluster expressed the markers: CD45dim, CD34+, CD117+, CD147+, and CD143+, and were negative for CD133. Higher level of expression of hemangioblast markers CD143 were found in critical COVID-19 patients. This population, indicative of hemangioblast-like cells, suggests a key role in COVID-19-related neoangiogenesis, potentially driving the severe vascular complications observed. Our findings underscore the need for further investigation into the contributions of adult stem cells in COVID-19 pathology, offering new insights into therapeutic targets and interventions., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
4. Platelet Activation and Severe Bleeding During Extracorporeal Carbon Dioxide Removal in Chronic Obstructive Pulmonary Disease Patients.
- Author
-
Smadja DM, Chocron R, Rivet N, Ortuno S, Guerin CL, and Diehl JL
- Abstract
Competing Interests: Disclosure: J.-L.D. reports grants and nonfinancial research support from ALung, nonfinancial research support from General Electric Healthcare, and personal fees and nonfinancial research support from Xenios/Novalung (Fresenius Medical Care), all outside the submitted work. D.M.S. reports personal fees from Carmat and Léo Pharma all outside the submitted work. The other authors have no conflicts of interest to report.
- Published
- 2024
- Full Text
- View/download PDF
5. Repeated peripheral infusions of anti-EGFRvIII CAR T cells in combination with pembrolizumab show no efficacy in glioblastoma: a phase 1 trial.
- Author
-
Bagley SJ, Binder ZA, Lamrani L, Marinari E, Desai AS, Nasrallah MP, Maloney E, Brem S, Lustig RA, Kurtz G, Alonso-Basanta M, Bonté PE, Goudot C, Richer W, Piaggio E, Kothari S, Guyonnet L, Guerin CL, Waterfall JJ, Mohan S, Hwang WT, Tang OY, Logun M, Bhattacharyya M, Markowitz K, Delman D, Marshall A, Wherry EJ, Amigorena S, Beatty GL, Brogdon JL, Hexner E, Migliorini D, Alanio C, and O'Rourke DM
- Subjects
- Humans, ErbB Receptors, Neoplasm Recurrence, Local metabolism, T-Lymphocytes, Tumor Microenvironment, Glioblastoma therapy, Antibodies, Monoclonal, Humanized
- Abstract
We previously showed that chimeric antigen receptor (CAR) T-cell therapy targeting epidermal growth factor receptor variant III (EGFRvIII) produces upregulation of programmed death-ligand 1 (PD-L1) in the tumor microenvironment (TME). Here we conducted a phase 1 trial (NCT03726515) of CAR T-EGFRvIII cells administered concomitantly with the anti-PD1 (aPD1) monoclonal antibody pembrolizumab in patients with newly diagnosed, EGFRvIII
+ glioblastoma (GBM) (n = 7). The primary outcome was safety, and no dose-limiting toxicity was observed. Secondary outcomes included median progression-free survival (5.2 months; 90% confidence interval (CI), 2.9-6.0 months) and median overall survival (11.8 months; 90% CI, 9.2-14.2 months). In exploratory analyses, comparison of the TME in tumors harvested before versus after CAR + aPD1 administration demonstrated substantial evolution of the infiltrating myeloid and T cells, with more exhausted, regulatory, and interferon (IFN)-stimulated T cells at relapse. Our study suggests that the combination of CAR T cells and PD-1 inhibition in GBM is safe and biologically active but, given the lack of efficacy, also indicates a need to consider alternative strategies., (© 2024. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2024
- Full Text
- View/download PDF
6. Results of an international survey about methods used to isolate human endothelial colony-forming cells: guidance from the SSC on Vascular Biology of the ISTH.
- Author
-
Blandinières A, Randi AM, Paschalaki KE, Guerin CL, Melero-Martin JM, and Smadja DM
- Subjects
- Adult, Humans, Biology, Australia, Cells, Cultured, Neovascularization, Physiologic, Endothelial Cells, Cell Culture Techniques
- Abstract
Background: Assessment of endothelial colony-forming cell (ECFC) number and vasculogenic properties is crucial for exploring vascular diseases and regeneration strategies. A previous survey of the Scientific and Standardization Committee on Vascular Biology of the International Society on Thrombosis and Haemostasis clarified key methodological points but highlighted a lack of standardization associated with ECFC culture., Objectives: The aim of this study was to provide expert consensus guidance on ECFC isolation and culture., Methods: We surveyed 21 experts from 10 different countries using a questionnaire proposed during the 2019 International Society on Thrombosis and Haemostasis Congress in Melbourne (Australia) to attain a consensus on ECFC isolation and culture., Results: We report here the consolidated results of the questionnaire. There was agreement on several general statements, mainly the technical aspects of ECFC isolation and cell culture. In contrast, on the points concerning the definition of a colony of ECFCs, the quantification of ECFCs, and the estimation of their age (in days or number of passages), the expert opinions were widely dispersed., Conclusion: Our survey clearly indicates an unmet need for rigorous standardization, multicenter comparison of results, and validation of ECFC isolation and culture procedures for clinical laboratory practice and robustness of results. To this end, we propose a standardized protocol for the isolation and expansion of ECFCs from umbilical cord and adult peripheral blood., Competing Interests: Declaration of competing interests There are no competing interests to disclose., (Copyright © 2023 International Society on Thrombosis and Haemostasis. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
7. Platelet activation and coronavirus disease 2019 mortality: Insights from coagulopathy, antiplatelet therapy and inflammation.
- Author
-
Philippe A, Chocron R, Bonnet G, Yatim N, Sutter W, Hadjadj J, Weizman O, Guerin CL, Mirault T, Fauvel C, Hauw-Berlemont C, Samama CM, Terrier B, Planquette B, Waldmann V, Fontenay M, Sanchez O, Diehl JL, Gaussem P, Cohen A, Gendron N, and Smadja DM
- Subjects
- Humans, Platelet Activation, Inflammation diagnosis, Inflammation drug therapy, Biomarkers, Platelet Aggregation Inhibitors adverse effects, COVID-19
- Abstract
Background: Coronavirus disease 2019 (COVID-19) is associated with an inflammatory cytokine burst and a prothrombotic coagulopathy. Platelets may contribute to microthrombosis, and constitute a therapeutic target in COVID-19 therapy., Aim: To assess if platelet activation influences mortality in COVID-19., Methods: We explored two cohorts of patients with COVID-19. Cohort A included 208 ambulatory and hospitalized patients with varying clinical severities and non-COVID patients as controls, in whom plasma concentrations of the soluble platelet activation biomarkers CD40 ligand (sCD40L) and P-selectin (sP-sel) were quantified within the first 48hours following hospitalization. Cohort B was a multicentre cohort of 2878 patients initially admitted to a medical ward. In both cohorts, the primary outcome was in-hospital mortality., Results: In cohort A, median circulating concentrations of sCD40L and sP-sel were only increased in the 89 critical patients compared with non-COVID controls: sP-sel 40,059 (interquartile range 26,876-54,678)pg/mL; sCD40L 1914 (interquartile range 1410-2367)pg/mL (P<0.001 for both). A strong association existed between sP-sel concentration and in-hospital mortality (Kaplan-Meier log-rank P=0.004). However, in a Cox model considering biomarkers of immunothrombosis, sP-sel was no longer associated with mortality, in contrast to coagulopathy evaluated with D-dimer concentration (hazard ratio 4.86, 95% confidence interval 1.64-12.50). Moreover, in cohort B, a Cox model adjusted for co-morbidities suggested that prehospitalization antiplatelet agents had no significant impact on in-hospital mortality (hazard ratio 1.05, 95% CI 0.80-1.37; P=0.73)., Conclusions: Although we observed an association between excessive biomarkers of platelet activation and in-hospital mortality, our findings rather suggest that coagulopathy is more central in driving disease progression, which may explain why prehospitalization antiplatelet drugs were not a protective factor against mortality in our multicentre cohort., (Copyright © 2023 Elsevier Masson SAS. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
8. Combined Transplantation of Human MSCs and ECFCs Improves Cardiac Function and Decrease Cardiomyocyte Apoptosis After Acute Myocardial Infarction.
- Author
-
Tripathi H, Domingues A, Donahue R, Cras A, Guerin CL, Gao E, Levitan B, Ratajczak MZ, Smadja DM, Abdel-Latif A, and Tarhuni WM
- Subjects
- Mice, Animals, Humans, Myocytes, Cardiac metabolism, Mice, Inbred NOD, Mice, SCID, Apoptosis, Ischemia metabolism, Mesenchymal Stem Cell Transplantation methods, Myocardial Infarction, Mesenchymal Stem Cells metabolism
- Abstract
Background: Ischemic heart disease, often caused by an acute myocardial infarction (AMI) is one of the leading causes of morbidity and mortality worldwide. Despite significant advances in medical and procedural therapies, millions of AMI patients progress to develop heart failure every year., Methods: Here, we examine the combination therapy of human mesenchymal stromal cells (MSCs) and endothelial colony-forming cells (ECFCs) to reduce the early ischemic damage (MSCs) and enhance angiogenesis (ECFCs) in a pre-clinical model of acute myocardial infarction. NOD/SCID mice were subjected to AMI followed by transplantation of MSCs and ECFCs either alone or in combination. Cardiomyocyte apoptosis and cardiac functional recovery were assessed in short- and long-term follow-up studies., Results: At 1 day after AMI, MSC- and ECFC-treated animals demonstrated significantly lower cardiomyocyte apoptosis compared to vehicle-treated animals. This phenomenon was associated with a significant reduction in infarct size, cardiac fibrosis, and improvement in functional cardiac recovery 4 weeks after AMI., Conclusions: The use of ECFCs, MSCs, and the combination of both cell types reduce cardiomyocyte apoptosis, scar size, and adverse cardiac remodeling, compared to vehicle, in a pre-clinical model of AMI. These results support the use of this combined cell therapy approach in future human studies during the acute phase of ischemic cardiac injury., (© 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
9. Increased Circulating CD62E + Endothelial Extracellular Vesicles Predict Severity and in- Hospital Mortality of COVID-19 Patients.
- Author
-
Mezine F, Guerin CL, Philippe A, Gendron N, Soret L, Sanchez O, Mirault T, Diehl JL, Chocron R, Boulanger CM, and Smadja DM
- Subjects
- Humans, Hospital Mortality, E-Selectin, COVID-19, Extracellular Vesicles
- Abstract
COVID-19 and infectious diseases have been included in strategic development goals (SDG) of United Nations (UN). Severe form of COVID-19 has been described as an endothelial disease. In order to better evaluate Covid-19 endotheliopathy, we characterized several subsets of circulating endothelial extracellular vesicles (EVs) at hospital admission among a cohort of 60 patients whose severity of COVID-19 was classified at the time of inclusion. Degree of COVID-19 severity was determined upon inclusion and categorized as moderate to severe in 40 patients and critical in 20 patients. We measured citrated plasma EVs expressing endothelial membrane markers. Endothelial EVs were defined as harboring VE-cadherin (CD144+), PECAM-1 (CD31 + CD41-) or E-selectin (CD62E+). An increase in CD62E + EV levels on admission to the hospital was significantly associated with critical disease. Moreover, Kaplan-Meier survival curves for CD62E + EV level showed that level ≥ 88,053 EVs/μL at admission was a significant predictor of in hospital mortality (p = 0.004). Moreover, CD62E + EV level ≥ 88,053 EV/μL was significantly associated with higher in-hospital mortality (OR 6.98, 95% CI 2.1-26.4, p = 0.002) in a univariate logistic regression model, while after adjustment to BMI CD62E + EV level ≥ 88,053 EV/μL was always significantly associated with higher in-hospital mortality (OR 5.1, 95% CI 1.4-20.0, p = 0.01). The present findings highlight the potential interest of detecting EVs expressing E-selectin (CD62) to discriminate Covid-19 patients at the time of hospital admission and identify individuals with higher risk of fatal outcome., (© 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
10. Phenotyping polarization dynamics of immune cells using a lipid droplet-cell pairing microfluidic platform.
- Author
-
Pinon L, Ruyssen N, Pineau J, Mesdjian O, Cuvelier D, Chipont A, Allena R, Guerin CL, Asnacios S, Asnacios A, Pierobon P, and Fattaccioli J
- Subjects
- Immunological Synapses, Signal Transduction, B-Lymphocytes, Antigens metabolism, Microfluidics, Lipid Droplets metabolism
- Abstract
The immune synapse is the tight contact zone between a lymphocyte and a cell presenting its cognate antigen. This structure serves as a signaling platform and entails a polarization of intracellular components necessary to the immunological function of the cell. While the surface properties of the presenting cell are known to control the formation of the synapse, their impact on polarization has not yet been studied. Using functional lipid droplets as tunable artificial presenting cells combined with a microfluidic pairing device, we simultaneously observe synchronized synapses and dynamically quantify polarization patterns of individual B cells. By assessing how ligand concentration, surface fluidity, and substrate rigidity impact lysosome polarization, we show that its onset and kinetics depend on the local antigen concentration at the synapse and on substrate rigidity. Our experimental system enables a fine phenotyping of monoclonal cell populations based on their synaptic readout., Competing Interests: The authors declare no competing interests., (© 2022 The Authors.)
- Published
- 2022
- Full Text
- View/download PDF
11. Bioprosthetic Total Artificial Heart Implantation Does Not Induce Chronic Inflammation.
- Author
-
Peronino C, Guerin CL, Ivak P, Guyonnet L, Chocron R, Detriché G, Latremouille C, Gruest M, Philippe A, Capel A, Pya Y, Martin AC, Jansen P, Gendron N, Netuka I, and Smadja DM
- Subjects
- Humans, Leukocytes, Mononuclear, Inflammation etiology, Cytokines, Heart Transplantation adverse effects, Heart, Artificial adverse effects, Heart Failure surgery
- Abstract
The Aeson total artificial heart (A-TAH) has been developed for patients at risk of death from biventricular failure. We aimed to assess the inflammatory status in nine subjects implanted with the A-TAH in kinetics over one year. Laboratory assessment of leukocyte counts, inflammatory cytokines assay, and peripheral blood mononuclear cell collection before and after A-TAH implantation. Leukocyte counts were not significantly modulated according to time after A-TAH implantation (coefficient of the linear mixed effect model with 95% CI, -0.05 (-0.71 to -0.61); p = 0.44). We explored inflammatory cytokine after A-TAH and did not observe, at any time, a modified profile compared to pre-implantation values (all p -values > 0.05). Finally, we compared the distribution of circulating immune cell subpopulations identified based on sequential expression patterns for multiple clusters of differentiation. None of the population explored had significant modulation during the 12-month follow-up (all p -values > 0.05). In conclusion, using a cytokine multiplex assay combined with a flow cytometry approach, we demonstrated the absence of inflammatory signals in peripheral blood over a period of 12 months following A-TAH implantation., Competing Interests: D.M.S., P.I., Y.P. and I.N. received consulting fees from CARMAT. C.P., C.L., A.C. and P.J. are employed by CARMAT-SAS. Other authors have no conflicts of interest to report., (Copyright © ASAIO 2022.)
- Published
- 2022
- Full Text
- View/download PDF
12. Extracellular vesicles from triple negative breast cancer promote pro-inflammatory macrophages associated with better clinical outcome.
- Author
-
Tkach M, Thalmensi J, Timperi E, Gueguen P, Névo N, Grisard E, Sirven P, Cocozza F, Gouronnec A, Martin-Jaular L, Jouve M, Delisle F, Manel N, Rookhuizen DC, Guerin CL, Soumelis V, Romano E, Segura E, and Théry C
- Subjects
- Humans, Macrophages, Extracellular Vesicles physiology, Triple Negative Breast Neoplasms pathology
- Abstract
Tumor associated macrophages (TAMs), which differentiate from circulating monocytes, are pervasive across human cancers and comprise heterogeneous populations. The contribution of tumor-derived signals to TAM heterogeneity is not well understood. In particular, tumors release both soluble factors and extracellular vesicles (EVs), whose respective impact on TAM precursors may be different. Here, we show that triple negative breast cancer cells (TNBCs) release EVs and soluble molecules promoting monocyte differentiation toward distinct macrophage fates. EVs specifically promoted proinflammatory macrophages bearing an interferon response signature. The combination in TNBC EVs of surface CSF-1 promoting survival and cargoes promoting cGAS/STING or other activation pathways led to differentiation of this particular macrophage subset. Notably, macrophages expressing the EV-induced signature were found among patients’ TAMs. Furthermore, higher expression of this signature was associated with T cell infiltration and extended patient survival. Together, this data indicates that TNBC-released CSF-1-bearing EVs promote a tumor immune microenvironment associated with a better prognosis in TNBC patients.
- Published
- 2022
- Full Text
- View/download PDF
13. Ret kinase-mediated mechanical induction of colon stem cells by tumor growth pressure stimulates cancer progression in vivo.
- Author
-
Nguyen Ho-Bouldoires TH, Sollier K, Zamfirov L, Broders-Bondon F, Mitrossilis D, Bermeo S, Guerin CL, Chipont A, Champenois G, Leclère R, André N, Ranno L, Michel A, Ménager C, Meseure D, Demené C, Tanter M, Fernández-Sánchez ME, and Farge E
- Subjects
- Animals, Biomarkers, Tumor, Cell Line, Tumor, Cell Transformation, Neoplastic, Female, Gene Expression Regulation, Enzymologic, Gene Expression Regulation, Neoplastic, Humans, Male, Mice, Mice, Inbred Strains, Neoplastic Stem Cells, Proto-Oncogene Proteins c-ret genetics, Colonic Neoplasms metabolism, Proto-Oncogene Proteins c-ret metabolism
- Abstract
How mechanical stress actively impacts the physiology and pathophysiology of cells and tissues is little investigated in vivo. The colon is constantly submitted to multi-frequency spontaneous pulsatile mechanical waves, which highest frequency functions, of 2 s period, remain poorly understood. Here we find in vivo that high frequency pulsatile mechanical stresses maintain the physiological level of mice colon stem cells (SC) through the mechanosensitive Ret kinase. When permanently stimulated by a magnetic mimicking-tumor growth analogue pressure, we find that SC levels pathologically increase and undergo mechanically induced hyperproliferation and tumorigenic transformation. To mimic the high frequency pulsatile mechanical waves, we used a generator of pulsed magnetic force stimulation in colonic tissues pre-magnetized with ultra-magnetic liposomes. We observed the pulsatile stresses using last generation ultra-wave dynamical high-resolution imaging. Finally, we find that the specific pharmacological inhibition of Ret mechanical activation induces the regression of spontaneous formation of SC, of CSC markers, and of spontaneous sporadic tumorigenesis in Apc mutated mice colons. Consistently, in human colon cancer tissues, Ret activation in epithelial cells increases with tumor grade, and partially decreases in leaking invasive carcinoma. High frequency pulsatile physiological mechanical stresses thus constitute a new niche that Ret-dependently fuels mice colon physiological SC level. This process is pathologically over-activated in the presence of permanent pressure due to the growth of tumors initiated by pre-existing genetic alteration, leading to mechanotransductive self-enhanced tumor progression in vivo, and repressed by pharmacological inhibition of Ret., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
14. Gonadotropins as novel active partners in vascular diseases: Insight from angiogenic properties and thrombotic potential of endothelial colony-forming cells.
- Author
-
Détriché G, Gendron N, Philippe A, Gruest M, Billoir P, Rossi E, Guerin CL, Lokajczyk A, Brabant S, Prié D, Mirault T, and Smadja DM
- Subjects
- Fetal Blood, Gonadotropins, Humans, Testosterone, Endothelial Cells, Vascular Diseases
- Abstract
Background: The impact of estrogen and testosterone on atherosclerotic cardiovascular disease is well known, but the role of the gonadotropins follicle-stimulating hormone (FSH), luteinizing hormone (LH), and prolactin (PRL) to some extent remain less studied., Objectives: To explore the angiogenic potential of gonadotropins on endothelial colony-forming cells (ECFCs)., Methods: We examined the effects of various doses of gonadotropins on ECFCs obtained from cord blood by assessing colony number, proliferation, migration, and sprouting ability. Moreover, we studied thrombin generation in ECFCs exposed to gonadotropins by performing a thrombin generation assay. Finally, we determined the levels of circulating gonadotropins in 30 men, to exclude the effect of estrogen, with lower extremity arterial disease (LEAD), in comparison with age- and sex-matched controls., Results: Exposure to FSH, LH, or PRL resulted in an increase in ECFC migration but showed no effect on proliferation or ECFC commitment from cord blood mononuclear cells. Using a three-dimensional fibrin gel assay, we showed that ECFC sprouting was significantly enhanced by gonadotropins. Exposure to FSH also increased the thrombin generation of ECFCs exposed to FSH. Finally, FSH and LH levels in men with LEAD were higher than those in controls., Conclusion: Gonadotropins increase ECFC-related angiogenesis and may be involved in thrombin generation in cardiovascular disease. Gonadotropins may act as biomarkers; moreover, we hypothesize that gonadotropin-blocking strategies may be a novel interesting therapeutic approach in atherosclerotic cardiovascular disease., (© 2021 International Society on Thrombosis and Haemostasis.)
- Published
- 2022
- Full Text
- View/download PDF
15. Type I interferon response and vascular alteration in chilblain-like lesions during the COVID-19 outbreak.
- Author
-
Frumholtz L, Bouaziz JD, Battistella M, Hadjadj J, Chocron R, Bengoufa D, Le Buanec H, Barnabei L, Meynier S, Schwartz O, Grzelak L, Smith N, Charbit B, Duffy D, Yatim N, Calugareanu A, Philippe A, Guerin CL, Joly B, Siguret V, Jaume L, Bachelez H, Bagot M, Rieux-Laucat F, Maylin S, Legoff J, Delaugerre C, Gendron N, Smadja DM, and Cassius C
- Subjects
- France, Humans, Pandemics, COVID-19 immunology, Chilblains virology, Interferon Type I immunology
- Abstract
Background: The outbreak of chilblain-like lesions (CLL) during the COVID-19 pandemic has been reported extensively, potentially related to SARS-CoV-2 infection, yet its underlying pathophysiology is unclear., Objectives: To study skin and blood endothelial and immune system activation in CLL in comparison with healthy controls and seasonal chilblains (SC), defined as cold-induced sporadic chilblains occurring during 2015 and 2019 with exclusion of chilblain lupus., Methods: This observational study was conducted during 9-16 April 2020 at Saint-Louis Hospital, Paris, France. All patients referred with CLL seen during this period of the COVID-19 pandemic were included in this study. We excluded patients with a history of chilblains or chilblain lupus. Fifty patients were included., Results: Histological patterns were similar and transcriptomic signatures overlapped in both the CLL and SC groups, with type I interferon polarization and a cytotoxic-natural killer gene signature. CLL were characterized by higher IgA tissue deposition and more significant transcriptomic activation of complement and angiogenesis factors compared with SC. We observed in CLL a systemic immune response associated with IgA antineutrophil cytoplasmic antibodies in 73% of patients, and elevated type I interferon blood signature in comparison with healthy controls. Finally, using blood biomarkers related to endothelial dysfunction and activation, and to angiogenesis or endothelial progenitor cell mobilization, we confirmed endothelial dysfunction in CLL., Conclusions: Our findings support an activation loop in the skin in CLL associated with endothelial alteration and immune infiltration of cytotoxic and type I IFN-polarized cells leading to clinical manifestations., (© 2021 British Association of Dermatologists.)
- Published
- 2021
- Full Text
- View/download PDF
16. Elevated Circulating Stem Cells Level is Observed One Month After Implantation of Carmat Bioprosthetic Total Artificial Heart.
- Author
-
Guyonnet L, Detriché G, Gendron N, Philippe A, Latremouille C, Soret L, Capel A, Peronino C, Jansen P, Ivak P, Carpentier A, Mirault T, Netuka I, Guerin CL, and Smadja DM
- Subjects
- Adult, Antigens, CD34, Humans, Leukocytes, Mononuclear, Male, Stem Cells, Endothelial Cells, Heart, Artificial
- Abstract
The Aeson® total artificial heart (A-TAH) has been developed as a total heart replacement for patients at risk of death from biventricular failure. We previously described endothelialization of the hybrid membrane inside A-TAH probably at the origin of acquired hemocompatibility. We aimed to quantify vasculogenic stem cells in peripheral blood of patients with long-term A-TAH implantation. Four male adult patients were included in this study. Peripheral blood mononuclear cells were collected before A-TAH implantation (T0) and after implantation at one month (T1), between two and five months (T2), and then between six and twelve months (T3). Supervised analysis of flow cytometry data confirmed the presence of the previously identified Lin
- CD133+ CD45- and Lin- CD34+ with different CD45 level intensities. Lin- CD133+ CD45- , Lin- CD34+ CD45- and Lin- CD34+ CD45+ were not modulated after A-TAH implantation. However, we demonstrated a significant mobilization of Lin- CD34+ CD45dim (p = 0.01) one month after A-TAH implantation regardless of the expression of CD133 or c-Kit. We then visualized data for the resulting clusters on a uniform manifold approximation and projection (UMAP) plot showing all single cells of the live Lin- and CD34+ events selected from down sampled files concatenated at T0 and T1. The three clusters upregulated at T1 are CD45dim clusters, confirming our results. In conclusion, using a flow cytometry approach, we demonstrated in A-TAH-transplanted patients a significant mobilization of Lin- CD34+ CD45dim in peripheral blood one month after A-TAH implantation. Using a flow cytometry approach, we demonstrated in A-TAH transplanted patients a significant mobilization of Lin- CD34+ CD45dim in peripheral blood one month after A-TAH implantation. This cell population could be at the origin of newly formed endothelial cells on top of hybrid membrane in Carmat bioprosthetic total artificial heart., (© 2021. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)- Published
- 2021
- Full Text
- View/download PDF
17. Current concepts on endothelial stem cells definition, location, and markers.
- Author
-
Chambers SEJ, Pathak V, Pedrini E, Soret L, Gendron N, Guerin CL, Stitt AW, Smadja DM, and Medina RJ
- Subjects
- Biomarkers metabolism, Humans, Ischemia therapy, Neovascularization, Physiologic, Stem Cells, Endothelial Progenitor Cells metabolism
- Abstract
Ischemic vascular disease is a major cause of mortality and morbidity worldwide, and regeneration of blood vessels in perfusion-deficient tissues is a worthwhile therapeutic goal. The idea of delivering endothelial stem/progenitor cells to repair damaged vasculature, reperfuse hypoxic tissue, prevent cell death, and consequently diminish tissue inflammation and fibrosis has a strong scientific basis and clinical value. Various labs have proposed endothelial stem/progenitor cell candidates. This has created confusion, as there are profound differences between these cell definitions based on isolation methodology, characterization, and reparative biology. Here, a stricter definition based on stem cell biology principles is proposed. Although preclinical studies have often been promising, results from clinical trials have been highly contradictory and served to highlight multiple challenges associated with disappointing therapeutic benefit. This article reviews recent accomplishments in the field and discusses current difficulties when developing endothelial stem cell therapies. Emerging evidence that disputes the classic view of the bone marrow as the source for these cells and supports the vascular wall as the niche for these tissue-resident endothelial stem cells is considered. In addition, novel markers to identify endothelial stem cells, including CD157, EPCR, and CD31
low VEGFR2low IL33+ Sox9+ , are described., (© 2021 The Authors. STEM CELLS TRANSLATIONAL MEDICINE published by Wiley Periodicals LLC on behalf of AlphaMed Press.)- Published
- 2021
- Full Text
- View/download PDF
18. Do Endothelial Colony-forming Cells Come From Bone Marrow or Vessels/VSELs?
- Author
-
Detriche G, Guerin CL, Gendron N, Mirault T, and Smadja DM
- Subjects
- Embryonic Stem Cells, Blood Vessels cytology, Bone Marrow, Endothelial Cells, Pluripotent Stem Cells
- Published
- 2021
- Full Text
- View/download PDF
19. Circulating Von Willebrand factor and high molecular weight multimers as markers of endothelial injury predict COVID-19 in-hospital mortality.
- Author
-
Philippe A, Chocron R, Gendron N, Bory O, Beauvais A, Peron N, Khider L, Guerin CL, Goudot G, Levasseur F, Peronino C, Duchemin J, Brichet J, Sourdeau E, Desvard F, Bertil S, Pene F, Cheurfa C, Szwebel TA, Planquette B, Rivet N, Jourdi G, Hauw-Berlemont C, Hermann B, Gaussem P, Mirault T, Terrier B, Sanchez O, Diehl JL, Fontenay M, and Smadja DM
- Subjects
- Adult, Aged, Biomarkers blood, Biomarkers chemistry, COVID-19 physiopathology, Cross-Sectional Studies, Endothelium, Vascular physiopathology, Female, Hospital Mortality, Humans, Kaplan-Meier Estimate, Male, Middle Aged, Molecular Weight, Paris epidemiology, Proportional Hazards Models, Protein Multimerization, Severity of Illness Index, Thrombosis blood, Thrombosis etiology, von Willebrand Factor chemistry, COVID-19 blood, COVID-19 mortality, Pandemics, SARS-CoV-2, von Willebrand Factor metabolism
- Abstract
Background: Coronavirus disease 2019 (COVID-19) is a respiratory disease associated with endotheliitis and microthrombosis., Objectives: To correlate endothelial dysfunction to in-hospital mortality in a bi-centric cohort of COVID-19 adult patients., Methods: Consecutive ambulatory and hospitalized patients with laboratory-confirmed COVID-19 were enrolled. A panel of endothelial biomarkers and von Willebrand factor (VWF) multimers were measured in each patient ≤ 48 h following admission., Results: Study enrolled 208 COVID-19 patients of whom 23 were mild outpatients and 189 patients hospitalized after admission. Most of endothelial biomarkers tested were found increased in the 89 critical patients transferred to intensive care unit. However, only von Willebrand factor antigen (VWF:Ag) scaled according to clinical severity, with levels significantly higher in critical patients (median 507%, IQR 428-596) compared to non-critical patients (288%, 230-350, p < 0.0001) or COVID-19 outpatients (144%, 133-198, p = 0.007). Moreover, VWF high molecular weight multimers (HMWM) were significantly higher in critical patients (median ratio 1.18, IQR 0.86-1.09) compared to non-critical patients (0.96, 1.04-1.39, p < 0.001). Among all endothelial biomarkers measured, ROC curve analysis identified a VWF:Ag cut-off of 423% as the best predictor for in-hospital mortality. The accuracy of VWF:Ag was further confirmed in a Kaplan-Meier estimator analysis and a Cox proportional Hazard model adjusted on age, BMI, C-reactive protein and D-dimer levels., Conclusion: VWF:Ag is a relevant predictive factor for in-hospital mortality in COVID-19 patients. More than a biomarker, we hypothesize that VWF, including excess of HMWM forms, drives microthrombosis in COVID-19., (© 2021. The Author(s), under exclusive licence to Springer Nature B.V. part of Springer Nature.)
- Published
- 2021
- Full Text
- View/download PDF
20. Platelet activation in critically ill COVID-19 patients.
- Author
-
Yatim N, Boussier J, Chocron R, Hadjadj J, Philippe A, Gendron N, Barnabei L, Charbit B, Szwebel TA, Carlier N, Pène F, Azoulay C, Khider L, Mirault T, Diehl JL, Guerin CL, Rieux-Laucat F, Duffy D, Kernéis S, Smadja DM, and Terrier B
- Abstract
Background: Microvascular, arterial and venous thrombotic events have been largely described during severe coronavirus disease 19 (COVID-19). However, mechanisms underlying hemostasis dysregulation remain unclear., Methods: We explored two independent cross-sectional cohorts to identify soluble markers and gene-expression signatures that discriminated COVID-19 severity and outcomes., Results: We found that elevated soluble (s)P-selectin at admission was associated with disease severity. Elevated sP-selectin was predictive of intubation and death (ROC AUC = 0.67, p = 0.028 and AUC = 0.74, p = 0.0047, respectively). An optimal cutoff value was predictive of intubation with 66% negative predictive value (NPV) and 61% positive predictive value (PPV), and of death with 90% NPV and 55% PPV. An unbiased gene set enrichment analysis revealed that critically ill patients had increased expression of genes related to platelet activation. Hierarchical clustering identified ITG2AB, GP1BB, PPBP and SELPLG to be upregulated in a grade-dependent manner. ROC curve analysis for the prediction of intubation was significant for SELPLG and PPBP (AUC = 0.8, p = 0.046 for both). An optimal cutoff value for PBPP was predictive of intubation with 100% NPV and 45% PPV, and for SELPLG with 100% NPV and 50% PPV., Conclusion: We provide evidence that platelets contribute to COVID-19 severity. Plasma sP-selectin level was associated with severity and in-hospital mortality. Transcriptional analysis identified PPBP/CXCL7 and SELPLG as biomarkers for intubation. These findings provide additional evidence for platelet activation in driving critical COVID-19. Specific studies evaluating the performance of these biomarkers are required., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
21. Placental growth factor level in plasma predicts COVID-19 severity and in-hospital mortality.
- Author
-
Smadja DM, Philippe A, Bory O, Gendron N, Beauvais A, Gruest M, Peron N, Khider L, Guerin CL, Goudot G, Levavasseur F, Duchemin J, Pene F, Cheurfa C, Szwebel TA, Sourdeau E, Planquette B, Hauw-Berlemont C, Hermann B, Gaussem P, Samama CM, Mirault T, Terrier B, Sanchez O, Rance B, Fontenay M, Diehl JL, and Chocron R
- Subjects
- Adult, Biomarkers, Female, Hospital Mortality, Humans, Placenta Growth Factor, SARS-CoV-2, COVID-19, Vascular Endothelial Growth Factor A
- Abstract
Background: Coronavirus disease 2019 (COVID-19) is a respiratory disease associated with vascular inflammation and endothelial injury., Objectives: To correlate circulating angiogenic markers vascular endothelial growth factor A (VEGF-A), placental growth factor (PlGF), and fibroblast growth factor 2 (FGF-2) to in-hospital mortality in COVID-19 adult patients., Methods: Consecutive ambulatory and hospitalized patients with COVID-19 infection were enrolled. VEGF-A, PlGF, and FGF-2 were measured in each patient ≤48 h following admission., Results: The study enrolled 237 patients with suspected COVID-19: 208 patients had a positive diagnostic for COVID-19, of whom 23 were mild outpatients and 185 patients hospitalized after admission. Levels of VEGF-A, PlGF, and FGF-2 significantly increase with the severity of the disease (P < .001). Using a logistic regression model, we found a significant association between the increase of FGF-2 or PlGF and mortality (odds ratio [OR] 1.11, 95% confidence interval [CI; 1.07-1.16], P < .001 for FGF-2 and OR 1.07 95% CI [1.04-1.10], P < .001 for PlGF) while no association were found for VEGF-A levels. Receiver operating characteristic curve analysis was performed and we identified PlGF above 30 pg/ml as the best predictor of in-hospital mortality in COVID-19 patients. Survival analysis for PlGF confirmed its interest for in-hospital mortality prediction, by using a Kaplan-Meier survival curve (P = .001) and a Cox proportional hazard model adjusted to age, body mass index, D-dimer, and C-reactive protein (3.23 95% CI [1.29-8.11], P = .001)., Conclusion: Angiogenic factor PlGF is a relevant predictive factor for in-hospital mortality in COVID-19 patients. More than a biomarker, we hypothesize that PlGF blocking strategies could be a new interesting therapeutic approach in COVID-19., (© 2021 International Society on Thrombosis and Haemostasis.)
- Published
- 2021
- Full Text
- View/download PDF
22. Multidimensional Proteomic Approach of Endothelial Progenitors Demonstrate Expression of KDR Restricted to CD19 Cells.
- Author
-
Guerin CL, Guyonnet L, Goudot G, Revets D, Konstantinou M, Chipont A, Chocron R, Blandinieres A, Khider L, Rancic J, Peronino C, Debuc B, Cras A, Knosp C, Latremouille C, Capel A, Ollert M, Diehl JL, Jansen P, Planquette B, Sanchez O, Gaussem P, Mirault T, Carpentier A, Gendron N, and Smadja DM
- Subjects
- Endothelial Progenitor Cells pathology, Female, Humans, Male, Middle Aged, Proteomics, Antigens, CD19 metabolism, COVID-19 metabolism, Endothelial Progenitor Cells metabolism, Gene Expression Regulation, Heart, Artificial, SARS-CoV-2 metabolism, Vascular Endothelial Growth Factor Receptor-2 metabolism
- Abstract
Endothelial progenitor cells (EPCs) are involved in vasculogenesis and cardiovascular diseases. However, the phenotype of circulating EPCs remains elusive but they are more often described as CD34
+ KDR+ . The aim of the study was to extensively characterize circulating potential vasculogenic stem cell candidates in two populations of patients with cardiovascular disease by powerful multidimensional single cell complementary cytometric approaches (mass, imaging and flow). We identified cellular candidates in one patient before and after bioprosthetic total artificial heart implantation and results were confirmed in healthy peripheral and cord blood by mass cytometry. We also quantified cellular candidates in 10 patients with different COVID-19 severity. Both C-TAH implantation and COVID-19 at critical stage induce a redistribution of circulating CD34+ and CD19+ sub-populations in peripheral blood. After C-TAH implantation, circulating CD34+ progenitor cells expressed c-Kit stem marker while specific subsets CD34+ CD133-/+ CD45-/dim c-Kit+ KDR- were mobilized. KDR was only expressed by CD19+ B-lymphocytes and CD14+ monocytes subpopulations in circulation. We confirmed by mass cytometry this KDR expression on CD19+ in healthy peripheral and cord blood, also with a VE-cadherin expression, confirming absence of endothelial lineage marker on CD34+ subtypes. In COVID-19, a significant mobilization of CD34+ c-Kit+ KDR- cells was observed between moderate and critical COVID-19 patients regardless CD133 or CD45 expression. In order to better evaluate EPC phenotype, we performed imaging flow cytometry measurements of immature CD34+ KDR+ cells in cord blood and showed that, after elimination of non-circular events, those cells were all CD19+ . During COVID-19, a significant mobilization of CD19+ KDR+ per million of CD45+ cells was observed between moderate and critical COVID-19 patients regardless of CD34 expression. CD34+ c-Kit+ cells are mobilized in both cardiovascular disease described here. KDR cells in peripheral blood are CD19 positive cells and are not classic vasculogenic stem and/or progenitor cells. A better evaluation of c-Kit and KDR expressing cells will lead to the redefinition of circulating endothelial progenitors.Graphical abstract Central illustration figure. Multidimensional proteomic approach of endothelial progenitors demonstrate expression of KDR restricted to CD19 cells. Endothelial progenitor cells (EPCs) are involved in cardiovascular diseases, however their phenotype remains elusive. We elucidated here EPCs phenotype by a deep characterization by multidimensional single cell complementary cytometric approaches after Bioprosthetic total artificial heart implantation and during COVID-19. We showed a redistribution of circulating CD34+ and CD19+ sub-populations in both situations. None of the immature cell population expresses KDR. Mobilized CD34+ expressed c-Kit. Imaging flow cytometry demonstrated that CD34+ KDR+ cells, after elimination of non-circular events, are all CD19+ . Our results suggest a new definition of circulating EPCs and emphasize involvement of CD19 cells in cardiovascular disease.- Published
- 2021
- Full Text
- View/download PDF
23. Endothelial Colony-Forming Cells from Idiopathic Pulmonary Fibrosis Patients Have a High Procoagulant Potential.
- Author
-
Billoir P, Blandinières A, Gendron N, Chocron R, Gunther S, Philippe A, Guerin CL, Israël-Biet D, and Smadja DM
- Subjects
- Humans, Thrombin, Thrombomodulin, Endothelial Cells cytology, Idiopathic Pulmonary Fibrosis, Stem Cells cytology
- Abstract
Idiopathic pulmonary fibrosis (IPF) is a severe, progressive and irreversible lung disease constantly associated with a major vascular remodeling process. Endothelial colony-forming cells (ECFCs) are human vasculogenic cells proposed as a cell therapy product or liquid biopsy in vascular disorders. Since the link between IPF and thrombosis has been largely proposed, the aim of our study was to explore hypercoagulability states in ECFCs from patients with IPF. We performed Thrombin generation assay (TGA) in cord blood (CB)-ECFCs, peripheral blood (PB)-ECFCs and IPF-ECFCs. Endogenous thrombin potential and peak were higher in IPF-ECFCs compared to CB-ECFCs and PB-ECFCs. As thrombin generation in ECFCs was increased, we evaluated anticoagulant proteins expressed on ECFCs membrane and identified thrombomodulin and EPCR. We found a significant decrease of both anticoagulant proteins at membrane using flow cytometry. This study is the first to examine ECFC thrombin generation in IPF. This new finding strongly argues for a role of ECFC in IPF pathophysiology and thrombotic related disorders in IPF. Graphical Abstract.
- Published
- 2021
- Full Text
- View/download PDF
24. Interleukin-8 Receptors CXCR1 and CXCR2 Are Not Expressed by Endothelial Colony-forming Cells.
- Author
-
Blandinières A, Hong X, Philippe A, Bièche I, Vacher S, Rossi E, Detriche G, Gendron N, Gaussem P, Guerin CL, Melero-Martin JM, and Smadja DM
- Subjects
- Endothelial Cells cytology, Fetal Blood cytology, Humans, Endothelial Cells metabolism, Receptors, Interleukin-8A metabolism, Receptors, Interleukin-8B metabolism
- Abstract
Endothelial colony-forming cells (ECFCs) are human vasculogenic cells described as potential cell therapy product and good candidates for being a vascular liquid biopsy. Since interleukin-8 (IL-8) is a main actor in senescence, its ability to interact with ECFCs has been explored. However, expression of CXCR1 and CXCR2, the two cellular receptors for IL-8, by ECFCs remain controversial as several teams published contradictory reports. Using complementary technical approaches, we have investigated the presence of these receptors on ECFCs isolated from cord blood. First, CXCR1 and CXCR2 were not detected on several clones of cord blood- endothelial colony-forming cell using different antibodies available, in contrast to well-known positive cells. We then compared the RT-PCR primers used in different papers to search for the presence of CXCR1 and CXCR2 mRNA and found that several primer pairs used could lead to non-specific DNA amplification. Last, we confirmed those results by RNA sequencing. CXCR1 and CXCR2 were not detected in ECFCs in contrary to human-induced pluripotent stem cell-derived endothelial cells (h-iECs). In conclusion, using three different approaches, we confirmed that CXCR1 and CXCR2 were not expressed at mRNA or protein level by ECFCs. Thus, IL-8 secretion by ECFCs, its effects in angiogenesis and their involvement in senescent process need to be reanalyzed according to this absence of CXCR-1 and - 2 in ECFCs.Graphical Abstract.
- Published
- 2021
- Full Text
- View/download PDF
25. Angiopoietin-2 as a marker of endothelial activation is a good predictor factor for intensive care unit admission of COVID-19 patients.
- Author
-
Smadja DM, Guerin CL, Chocron R, Yatim N, Boussier J, Gendron N, Khider L, Hadjadj J, Goudot G, Debuc B, Juvin P, Hauw-Berlemont C, Augy JL, Peron N, Messas E, Planquette B, Sanchez O, Charbit B, Gaussem P, Duffy D, Terrier B, Mirault T, and Diehl JL
- Subjects
- Aged, Betacoronavirus, Biomarkers blood, COVID-19, Critical Care methods, E-Selectin blood, Female, Gene Expression Profiling, Hospitalization, Humans, Male, Middle Aged, Pandemics, Patient Admission, Prospective Studies, Respiration, Artificial, SARS-CoV-2, Angiopoietin-2 blood, Coronavirus Infections blood, Coronavirus Infections therapy, Endothelium, Vascular metabolism, Intensive Care Units, Pneumonia, Viral blood, Pneumonia, Viral therapy
- Abstract
Background: Coronavirus disease-2019 (COVID-19), a respiratory disease has been associated with ischemic complications, coagulation disorders, and an endotheliitis., Objectives: To explore endothelial damage and activation-related biomarkers in COVID-19 patients with criteria of hospitalization for referral to intensive care unit (ICU) and/or respiratory worsening., Methods: Analysis of endothelial and angiogenic soluble markers in plasma from patients at admission., Results: Study enrolled 40 consecutive COVID-19 patients admitted to emergency department that fulfilled criteria for hospitalization. Half of them were admitted in conventional wards without any ICU transfer during hospitalization; whereas the 20 others were directly transferred to ICU. Patients transferred in ICU were more likely to have lymphopenia, decreased SpO2 and increased D-dimer, CRP and creatinine levels. In those patients, soluble E-selectin and angiopoietin-2 were significantly increased (p value at 0.009 and 0.003, respectively). Increase in SELE gene expression (gene coding for E-selectin protein) was confirmed in an independent cohort of 32 patients using a whole blood gene expression profile analysis. In plasma, we found a strong association between angiopoetin-2 and CRP, creatinine and D-dimers (with p value at 0.001, 0.001 and 0.003, respectively). ROC curve analysis identified an Angiopoietin-2 cut-off of 5000 pg/mL as the best predictor for ICU outcome (Se = 80.1%, Sp = 70%, PPV = 72.7%, NPV = 77%), further confirmed in multivariate analysis after adjustment for creatinine, CRP or D-dimers., Conclusion: Angiopoietin-2 is a relevant predictive factor for ICU direct admission in COVID-19 patients. This result showing an endothelial activation reinforces the hypothesis of a COVID-19-associated microvascular dysfunction.
- Published
- 2020
- Full Text
- View/download PDF
26. Valproic Acid Decreases Endothelial Colony Forming Cells Differentiation and Induces Endothelial-to-Mesenchymal Transition-like Process.
- Author
-
Nevo N, Lecourt S, Bièche I, Kucia M, Cras A, Blandinieres A, Vacher S, Gendron N, Guerin CL, Ratajczak MZ, and Smadja DM
- Subjects
- Animals, Cell Movement drug effects, Cell Proliferation drug effects, Endothelial Progenitor Cells drug effects, Humans, Male, Mice, Nude, Neovascularization, Physiologic drug effects, Phenotype, Cell Differentiation drug effects, Endothelial Progenitor Cells cytology, Mesoderm cytology, Valproic Acid pharmacology
- Abstract
Valproic acid (VPA), a histone deacetylase (HDAC) inhibitor is a widely used anticonvulsant drug. VPA is also under clinical evaluation to be employed in anticancer therapy, as an antithrombotic agent or a molecule to be used in the stem cells expansion protocols. Since endothelial colony forming cells (ECFC) has been identified as the human postnatal vasculogenic cells involved in thrombotic disorders and serve as a promising source of immature cell for vascular repair, objectives of the present study were to determine how VPA contributes to ECFC commitment and their angiogenic properties. We examined the effect of VPA on ECFC obtained from cord blood by evaluating colony number, proliferation, migration and their sprouting ability in vitro, as well as their in vivo vasculogenic properties. VPA inhibited endothelial differentiation potential from of cord blood derived stem cells associated with decreased proliferation and sprouting activity of cultured ECFC. VPA treatment significantly decreased the vessel-forming ability of ECFC transplanted together with mesenchymal stem cells (MSC) in Matrigel implants in nude mice model. Surprisingly, a microscopic evaluation revealed that VPA induces marked morphological changes from a cobblestone-like EC morphology to enlarged spindle shaped morphology of ECFC. RT-qPCR and a CD31/CD90 flow cytometry analysis confirmed a phenotypic switch of VPA-treated ECFC to mesenchymal-like phenotype. In conclusion, the pan-HDAC inhibitor VPA described for expansion of hematopoietic stem cells and very small embryonic like stem cells cannot be successfully employed for differentiation of endothelial lineage committed ECFC into functional endothelial cells. Our data also suggest that VPA based therapeutics may induce endothelial dysfunction associated with fibrosis that might induce thrombosis recurrence or venous insufficiency.
- Published
- 2020
- Full Text
- View/download PDF
27. Hemocompatibility and safety of the Carmat Total Artifical Heart hybrid membrane.
- Author
-
Richez U, De Castilla H, Guerin CL, Gendron N, Luraghi G, Grimme M, Wu W, Taverna M, Jansen P, Latremouille C, Migliavacca F, Dubini G, Capel A, Carpentier A, and Smadja DM
- Abstract
The Carmat bioprosthetic total artificial heart (C-TAH) is a biventricular pump developed to minimize drawbacks of current mechanical assist devices and improve quality of life during support. This study aims to evaluate the safety of the hybrid membrane, which plays a pivotal role in this artificial heart. We investigated in particular its blood-contacting surface layer of bovine pericardial tissue, in terms of mechanical aging, risks of calcification, and impact of the hemodynamics shear stress inside the ventricles on blood components. Hybrid membranes were aged in a custom-designed endurance bench. Mechanical, physical and chemical properties were not significantly modified from 9 months up to 4 years of aging using a simulating process. Exploration of erosion areas did not show no risk of oil diffusion through the membrane. Blood contacting materials in the ventricular cavities were subcutaneously implanted in Wistar rats for 30 days as a model for calcification and demonstrated that the in-house anti-calcification pretreatment with Formaldehyde-Ethanol-Tween 80 was able to significantly reduce the calcium concentration from 132 μg/mg to 4.42 μg/mg (p < 0.001). Hemodynamic simulations with a computational model were used to reproduce shear stress in left and right ventricles and no significant stress was able to trigger hemolysis, platelet activation nor degradation of the von Willebrand factor multimers. Moreover, explanted hybrid membranes from patients included in the feasibility clinical study were analyzed confirming preclinical results with the absence of significant membrane calcification. At last, blood plasma bank analysis from the four patients implanted with C-TAH during the feasibility study showed no residual glutaraldehyde increase in plasma and confirmed hemodynamic simulation-based results with the absence of hemolysis and platelet activation associated with normal levels of plasma free hemoglobin and platelet microparticles after C-TAH implantation. These results on mechanical aging, calcification model and hemodynamic simulations predicted the safety of the hybrid membrane used in the C-TAH, and were confirmed in the feasibility study., (© 2019 Published by Elsevier Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
28. Human Endothelial Colony Forming Cells Express Intracellular CD133 that Modulates their Vasculogenic Properties.
- Author
-
Rossi E, Poirault-Chassac S, Bieche I, Chocron R, Schnitzler A, Lokajczyk A, Bourdoncle P, Dizier B, Bacha NC, Gendron N, Blandinieres A, Guerin CL, Gaussem P, and Smadja DM
- Subjects
- Animals, Endothelial Progenitor Cells cytology, Heterografts, Hindlimb blood supply, Human Umbilical Vein Endothelial Cells cytology, Human Umbilical Vein Endothelial Cells metabolism, Humans, Ischemia metabolism, Ischemia pathology, Ischemia therapy, Male, Mice, Mice, Nude, Stem Cell Transplantation, AC133 Antigen biosynthesis, Antigens, Differentiation biosynthesis, Endothelial Progenitor Cells metabolism, Gene Expression Regulation, Neovascularization, Physiologic
- Abstract
Stem cells at the origin of endothelial progenitor cells and in particular endothelial colony forming cells (ECFCs) subtype have been largely supposed to be positive for the CD133 antigen, even though no clear correlation has been established between its expression and function in ECFCs. We postulated that CD133 in ECFCs might be expressed intracellularly, and could participate to vasculogenic properties. ECFCs extracted from cord blood were used either fresh (n = 4) or frozen (n = 4), at culture days <30, to investigate the intracellular presence of CD133 by flow cytometry and confocal analysis. Comparison with HUVEC and HAEC mature endothelial cells was carried out. Then, CD133 was silenced in ECFCs using specific siRNA (siCD133-ECFCs) or scramble siRNA (siCtrl-ECFCs). siCD133-ECFCs (n = 12), siCtrl-ECFCs (n = 12) or PBS (n = 12) were injected in a hind-limb ischemia nude mouse model and vascularization was quantified at day 14 with H&E staining and immunohistochemistry for CD31. Results of flow cytometry and confocal microscopy evidenced the positivity of CD133 in ECFCs after permeabilization compared with not permeabilized ECFCs (p < 0.001) and mature endothelial cells (p < 0.03). In the model of mouse hind-limb ischemia, silencing of CD133 in ECFCs significantly abolished post-ischemic revascularization induced by siCtrl-ECFCs; indeed, a significant reduction in cutaneous blood flows (p = 0.03), capillary density (CD31) (p = 0.01) and myofiber regeneration (p = 0.04) was observed. Also, a significant necrosis (p = 0.02) was observed in mice receiving siCD133-ECFCs compared to those treated with siCtrl-ECFCs. In conclusion, our work describes for the first time the intracellular expression of the stemness marker CD133 in ECFCs. This feature could resume the discrepancies found in the literature concerning CD133 positivity and ontogeny in endothelial progenitors.
- Published
- 2019
- Full Text
- View/download PDF
29. Treprostinil treatment decreases circulating platelet microvesicles and their procoagulant activity in pediatric pulmonary hypertension.
- Author
-
Bacha NC, Levy M, Guerin CL, Le Bonniec B, Harroche A, Szezepanski I, Renard JM, Gaussem P, Israel-Biet D, Boulanger CM, and Smadja DM
- Subjects
- Adolescent, Adult, Cell-Derived Microparticles drug effects, Child, Child, Preschool, Coagulants chemistry, Epoprostenol administration & dosage, Female, Heart Defects, Congenital complications, Heart Defects, Congenital drug therapy, Humans, Infant, Lung physiopathology, Male, Pulmonary Arterial Hypertension complications, Pulmonary Arterial Hypertension drug therapy, Pulmonary Circulation drug effects, Vasodilator Agents administration & dosage, Young Adult, Antihypertensive Agents administration & dosage, Epoprostenol analogs & derivatives, Familial Primary Pulmonary Hypertension drug therapy, Hypertension, Pulmonary drug therapy
- Abstract
Background: Pulmonary arterial hypertension (PAH) results from pulmonary vascular disease and may eventually lead to right heart failure and death. Vasodilator therapy has greatly improved PAH prognosis. Circulating microvesicles are considered as surrogate markers of endothelial and hematopoietic cell activation., Aim: Thus, our purpose was to determine if MVs are upregulated in pediatric PAH such as reported in adult patients, and to analyze the impact of vasodilator therapies on MV count and function., Patients: Population study consisted of 26 patients of median age 6.09 years, with Congenital Heart Disease (CHD) and elevated pulmonary vascular resistance (CHD-PAH) or idiopathic PAH (iPAH)., Results: Compared to healthy controls, all circulating MV subpopulations were found higher in untreated PAH patients. No significant differences of annexin-V+ total MV, endothelial, or leukocyte derived-MV counts were found between untreated patients and those receiving oral vasodilator therapies. Conversely, platelet MVs were significantly lower in the group treated with SC-treprostinil compared with both untreated PAH and oral therapy groups (P = 0.01), and exhibited a significant decrease of phospholipid procoagulant activity. Control samples treated in vitro with treprostinil at therapeutic concentrations showed as expected a significant decrease of platelet aggregation but also a reduced spontaneous MV generation., Conclusion: Our results suggest that treprostinil, besides vasodilation, might exert its beneficial effect through an inhibition of platelet activation, resulting in a decreased number and procoagulant activity of circulating MVs., (© 2018 Wiley Periodicals, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
30. Intra-Cardiac Release of Extracellular Vesicles Shapes Inflammation Following Myocardial Infarction.
- Author
-
Loyer X, Zlatanova I, Devue C, Yin M, Howangyin KY, Klaihmon P, Guerin CL, Kheloufi M, Vilar J, Zannis K, Fleischmann BK, Hwang DW, Park J, Lee H, Menasché P, Silvestre JS, and Boulanger CM
- Subjects
- Animals, Biomarkers metabolism, Chemokine CCL2 metabolism, Chemokine CCL7 metabolism, Coronary Vessels, Endothelial Cells metabolism, Exosomes, Extracellular Vesicles metabolism, Interleukin-6 metabolism, Ligation, Male, Mice, Mice, Inbred C57BL, Myocardial Infarction complications, Myocardial Infarction metabolism, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Extracellular Vesicles pathology, Myocardial Infarction pathology, Myocarditis etiology
- Abstract
Rationale: A rapid and massive influx of inflammatory cells occurs into ischemic area after myocardial infarction (MI), resulting in local release of cytokines and growth factors. Yet, the mechanisms regulating their production are not fully explored. The release of extracellular vesicles (EVs) in the interstitial space curbs important biological functions, including inflammation, and influences the development of cardiovascular diseases. To date, there is no evidence for in situ release of cardiac EVs after MI., Objective: The present study tested the hypothesis that local EV generation in the infarcted heart coordinates cardiac inflammation after MI., Methods and Results: Coronary artery ligation in mice transiently increases EV levels in the left ventricle when compared with sham animals. EVs from infarcted hearts were characterized as large vesicles (252±18 nm) expressing cardiomyocyte and endothelial markers and small EVs (118±4 nm) harboring exosomal markers, such as CD (cluster of differentiation) 63 and CD9. Cardiac large EVs generated after MI, but not small EVs or sham EVs, increased the release of IL (interleukin)-6, CCL (chemokine ligand) 2, and CCL7 from fluorescence-activated cell-sorted Ly6C
+ cardiac monocytes. EVs of similar diameter were also isolated from fragments of interventricular septum obtained from patients undergoing aortic valve replacement, thus supporting the clinical relevance of our findings in mice., Conclusions: The present study demonstrates that acute MI transiently increases the generation of cardiac EVs characterized as both exosomes and microvesicles, originating mainly from cardiomyocytes and endothelial cells. EVs accumulating in the ischemic myocardium are rapidly taken up by infiltrating monocytes and regulate local inflammatory responses., (© 2018 The Authors.)- Published
- 2018
- Full Text
- View/download PDF
31. Endothelial Microparticles are Associated to Pathogenesis of Idiopathic Pulmonary Fibrosis.
- Author
-
Bacha NC, Blandinieres A, Rossi E, Gendron N, Nevo N, Lecourt S, Guerin CL, Renard JM, Gaussem P, Angles-Cano E, Boulanger CM, Israel-Biet D, and Smadja DM
- Subjects
- Aged, Aged, 80 and over, Cell Differentiation physiology, Cells, Cultured, Collagen metabolism, Female, Fibroblasts cytology, Fibroblasts metabolism, Flow Cytometry, Healthy Volunteers, Humans, Lung cytology, Male, Middle Aged, Myofibroblasts metabolism, Myofibroblasts pathology, Urokinase-Type Plasminogen Activator metabolism, Cell-Derived Microparticles metabolism, Cell-Derived Microparticles pathology, Endothelial Progenitor Cells metabolism, Endothelial Progenitor Cells pathology, Idiopathic Pulmonary Fibrosis metabolism, Idiopathic Pulmonary Fibrosis pathology
- Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating disease characterized by obliteration of alveolar architecture, resulting in declining lung function and ultimately death. Pathogenic mechanisms remain unclear but involve a concomitant accumulation of scar tissue together with myofibroblasts activation. Microparticles (MPs) have been investigated in several human lung diseases as possible pathogenic elements, prognosis markers and therapeutic targets. We postulated that levels and cellular origins of circulating MPs might serve as biomarkers in IPF patients and/or as active players of fibrogenesis. Flow cytometry analysis showed a higher level of Annexin-V positive endothelial and platelet MPs in 41 IPF patients compared to 22 healthy volunteers. Moreover, in IPF patients with a low diffusing capacity of the lung for carbon monoxide (DL
CO <40%), endothelial MPs (EMPs) were found significantly higher compared to those with DLCO >40% (p = 0.02). We then used EMPs isolated from endothelial progenitor cells (ECFCs) extracted from IPF patients or controls to modulate normal human lung fibroblast (NHLF) properties. We showed that EMPs did not modify proliferation, collagen deposition and myofibroblast transdifferentiation. However, EMPs from IPF patients stimulated migration capacity of NHLF. We hypothesized that this effect could result from EMPs fibrinolytic properties and found indeed higher plasminogen activation potential in total circulating MPs and ECFCs derived MPs issued from IPF patients compared to those isolated from healthy controls MPs. Our study showed that IPF is associated with an increased level of EMPs in the most severe patients, highlighting an active process of endothelial activation in the latter. Endothelial microparticles might contribute to the lung fibroblast invasion mediated, at least in part, by a fibrinolytic activity.- Published
- 2018
- Full Text
- View/download PDF
32. Egfl7 Represses the Vasculogenic Potential of Human Endothelial Progenitor Cells.
- Author
-
d'Audigier C, Susen S, Blandinieres A, Mattot V, Saubamea B, Rossi E, Nevo N, Lecourt S, Guerin CL, Dizier B, Gendron N, Caetano B, Gaussem P, Soncin F, and Smadja DM
- Subjects
- Cell Differentiation physiology, Cell Movement genetics, Cell Movement physiology, Cells, Cultured, Endothelial Growth Factors genetics, Endothelial Progenitor Cells metabolism, Humans, MicroRNAs genetics, MicroRNAs metabolism, Neovascularization, Physiologic physiology, RNA Interference, Endothelial Growth Factors metabolism, Endothelial Progenitor Cells cytology
- Abstract
Egfl7 (VE-statin) is a secreted protein mostly specific to the endothelial lineage during development and in the adult and which expression is enhanced during angiogenesis. Egfl7 involvement in human postnatal vasculogenesis remains unresolved yet. Our aim was to assess Egfl7 expression in several angiogenic cell types originating from human bone marrow, peripheral blood, or cord blood. We found that only endothelial colony forming cells (ECFC), which are currently considered as the genuine endothelial precursor cells, expressed large amounts of Egfl7. In order to assess its potential roles in ECFC, Egfl7 was repressed in ECFC by RNA interference and ECFC angiogenic capacities were tested in vitro and in vivo. Cell proliferation, differentiation, and migration were significantly improved when Egfl7 was repressed in ECFC in vitro, whereas miR-126-3p levels remained unchanged. In vivo, repression of Egfl7 in ECFC significantly improved post-ischemic revascularization in a model of mouse hind-limb ischemia. In conclusion, ECFC are the sole postnatal angiogenic cells which express large amounts of Egfl7 and whose angiogenic properties are repressed by this factor. Thus, Egfl7 inhibition may be considered as a therapeutic option to improve ECFC-mediated postnatal vasculogenesis and to optimize in vitro ECFC expansion in order to develop an optimized cell therapy approach.
- Published
- 2018
- Full Text
- View/download PDF
33. Bone marrow-derived mesenchymal stem cell-loaded fibrin patches act as a reservoir of paracrine factors in chronic myocardial infarction.
- Author
-
Blondiaux E, Pidial L, Autret G, Rahmi G, Balvay D, Audureau E, Wilhelm C, Guerin CL, Bruneval P, Silvestre JS, Menasché P, and Clément O
- Subjects
- Animals, Cytokines metabolism, Female, Flow Cytometry, Heart Function Tests, Humans, Immunohistochemistry, Magnetic Resonance Imaging, Mesenchymal Stem Cells drug effects, Mesenchymal Stem Cells metabolism, Myocardial Infarction pathology, Myocardial Infarction physiopathology, Pericardium metabolism, Rats, Nude, Bone Marrow Cells cytology, Fibrin pharmacology, Mesenchymal Stem Cell Transplantation, Mesenchymal Stem Cells cytology, Myocardial Infarction therapy, Paracrine Communication
- Abstract
The combination of mesenchymal stem cells and tissue-engineered fibrin patches improves the therapeutic efficacy of stem cells. In vivo cardiac magnetic resonance (4.7 Tesla) and ex vivo high-spatial resolution CMR were used to track the fate of human bone marrow-derived mesenchymal stem cell (BMSC) delivered on an epicardial scaffold and more specifically assess their potential intramyocardial migration. Fifty-seven nude rats underwent permanent coronary artery ligation. Two months later, those with a left ventricular ejection fraction ≤55% were randomly allocated to receive a patch loaded with human BMSC (BMSC-P, n = 10), a patch loaded with BMSCs labelled with iron oxide nanoparticles (BMSC*-P, n = 12), an acellular patch (A-P, n = 8) or to serve as sham-operated animals (SHAM, n = 7). BMSC secretion of cytokines and growth factors was evaluated with flow-cytometry. Cardiac functional parameters of cell-treated groups (BMSC*-P and BMSC-P) yielded significantly better outcomes than the SHAM group (p = 0.044 and p = 0.026, respectively, for ejection fraction). Angiogenesis was higher in the cell-patch than in control groups (e.g. BMSC*P vs. SHAM: p = 0.007). No BMSCs were identified into the myocardium on cardiac magnetic resonance or histological sections, although persisting BMSCs were identified on the epicardial surface 21 days post-transplantation in 10% of rats hearts (Lamin A/C and CD90 positive). Cytokine and growth factor profiling demonstrated an increase in their release by cells seeded in patches. The absence of stem cell migration into the myocardium and the persistence of stem cells on the epicardial surface suggest that fibrin patches are likely to act predominantly as reservoirs of paracrine factors. Copyright © 2017 John Wiley & Sons, Ltd., (Copyright © 2017 John Wiley & Sons, Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
34. Co-injection of mesenchymal stem cells with endothelial progenitor cells accelerates muscle recovery in hind limb ischemia through an endoglin-dependent mechanism.
- Author
-
Rossi E, Smadja D, Goyard C, Cras A, Dizier B, Bacha N, Lokajczyk A, Guerin CL, Gendron N, Planquette B, Mignon V, Bernabéu C, Sanchez O, and Smadja DM
- Subjects
- Adipogenesis, Animals, Cell Adhesion, Cells, Cultured, Chondrogenesis, Disease Models, Animal, Endoglin genetics, Endothelial Progenitor Cells metabolism, Hindlimb, Ischemia metabolism, Ischemia pathology, Ischemia physiopathology, Male, Mice, Nude, Muscle, Skeletal pathology, Necrosis, Phenotype, RNA Interference, Recovery of Function, Regional Blood Flow, Signal Transduction, Time Factors, Transfection, Endoglin metabolism, Endothelial Progenitor Cells transplantation, Ischemia surgery, Mesenchymal Stem Cell Transplantation, Mesenchymal Stem Cells metabolism, Muscle, Skeletal blood supply, Neovascularization, Physiologic
- Abstract
Endothelial colony-forming cells (ECFCs) are progenitor cells committed to endothelial lineages and have robust vasculogenic properties. Mesenchymal stem cells (MSCs) have been described to support ECFC-mediated angiogenic processes in various matrices. However, MSC-ECFC interactions in hind limb ischemia (HLI) are largely unknown. Here we examined whether co-administration of ECFCs and MSCs bolsters vasculogenic activity in nude mice with HLI. In addition, as we have previously shown that endoglin is a key adhesion molecule, we evaluated its involvement in ECFC/MSC interaction. Foot perfusion increased on day 7 after ECFC injection and was even better at 14 days. Co-administration of MSCs significantly increased vessel density and foot perfusion on day 7 but the differences were no longer significant at day 14. Analysis of mouse and human CD31, and in situ hybridization of the human ALU sequence, showed enhanced capillary density in ECFC+MSC mice. When ECFCs were silenced for endoglin, coinjection with MSCs led to lower vessel density and foot perfusion at both 7 and 14 days (p<0.001). Endoglin silencing in ECFCs did not affect MSC differentiation into perivascular cells or other mesenchymal lineages. Endoglin silencing markedly inhibited ECFC adhesion to MSCs. Thus, MSCs, when combined with ECFCs, accelerate muscle recovery in a mouse model of hind limb ischemia, through an endoglin-dependent mechanism.
- Published
- 2017
- Full Text
- View/download PDF
35. Human very Small Embryonic-like Cells Support Vascular Maturation and Therapeutic Revascularization Induced by Endothelial Progenitor Cells.
- Author
-
Guerin CL, Rossi E, Saubamea B, Cras A, Mignon V, Silvestre JS, and Smadja DM
- Subjects
- Animals, Female, Heterografts, Humans, Male, Mice, Mice, Nude, Endothelial Progenitor Cells metabolism, Endothelial Progenitor Cells transplantation, Hindlimb blood supply, Hindlimb metabolism, Hindlimb pathology, Ischemia metabolism, Ischemia pathology, Ischemia therapy, Neovascularization, Physiologic
- Abstract
Very small embryonic-like stem cells (VSELs) are major pluripotent stem cells defined as cells of small size being Lineage- negative, CD133-positive, and CD45-negative. We previously described that human bone marrow VSELs were able to differentiate into endothelial cells and promoted post-ischemic revascularization in mice with surgically induced critical limb ischemia. In the present work, we isolated bone marrow VSELs from patients with critical limb ischemia and studied their ability to support endothelial progenitor cells therapeutic capacity and revascularization potential. Sorted bone marrow VSELs cultured in angiogenic media were co-injected with endothelial progenitor cells and have been show to trigger post-ischemic revascularization in immunodeficient mice, and support vessel formation in vivo in Matrigel implants better than human bone marrow mesenchymal stem cells. In conclusion, VSELs are a potential new source of therapeutic cells that may give rise to cells of the endothelial and perivascular lineage in humans. VSELs are the first real vasculogenic stem cells able to differentiate in endothelial and perivascular lineage in human adult described from now. Thus, because VSELs presence have been proposed in adult tissues, we think that VSELs are CD45 negative stem cells able to give rise to vascular regeneration in human tissues and vessels.
- Published
- 2017
- Full Text
- View/download PDF
36. Very Small Embryonic-like Stem Cells Are Mobilized in Human Peripheral Blood during Hypoxemic COPD Exacerbations and Pulmonary Hypertension.
- Author
-
Guerin CL, Blandinières A, Planquette B, Silvestre JS, Israel-Biet D, Sanchez O, and Smadja DM
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Humans, Hypertension, Pulmonary pathology, Hypertension, Pulmonary physiopathology, Hypoxia pathology, Hypoxia physiopathology, Male, Middle Aged, Pluripotent Stem Cells pathology, Pulmonary Disease, Chronic Obstructive pathology, Pulmonary Disease, Chronic Obstructive physiopathology, Hematopoietic Stem Cell Mobilization, Hypertension, Pulmonary blood, Hypoxia blood, Pluripotent Stem Cells metabolism, Pulmonary Disease, Chronic Obstructive blood
- Abstract
Very small embryonic-like stem cells (VSELs) are major pluripotent stem cells involved in vascular and tissue regeneration and constitute a recruitable pool of stem/progenitor cells with putative instrumental role in organ repair. Here, we hypothesized that VSELs might be mobilized from the bone marrow (BM) to peripheral blood (PB) in patients with hypoxic lung disease or pulmonary hypertension (PH). The objective of the present study was then to investigate the changes in VSELs number in peripheral blood of patients with hypoxic lung disease and PH. We enrolled 26 patients with Chronic Obstructive Pulmonary Disease (COPD) with or without hypoxemia, 13 patients with PH and 20 controls without any respiratory or cardiovascular diseases. In PH patients, VSELs levels have been determined during right heart catheterization in pulmonary blood and PB. For this purpose, mononuclear cells were separated by density gradient and VSELs have been quantified by using a multiparametric flow cytometry approach. The number of PB-VSELs in hypoxic COPD patients was significantly increased compared with non-hypoxic COPD patients or controls (p = 0.0055). In patients with PH, we did not find any difference in VSELs numbers between arterial pulmonary blood and venous PB (p = 0.93). However, we found an increase in VSELs in the peripheral blood of patients with PH (p = 0.03). In conclusion, we unraveled that circulating VSELs were increased in peripheral blood of patients with hypoxic COPD or with PH. Thus, VSELs may serve as a reservoir of pluripotent stem cells that can be recruited into PB and may play an important role in promoting lung repair.
- Published
- 2017
- Full Text
- View/download PDF
37. Liver microRNA-21 is overexpressed in non-alcoholic steatohepatitis and contributes to the disease in experimental models by inhibiting PPARα expression.
- Author
-
Loyer X, Paradis V, Hénique C, Vion AC, Colnot N, Guerin CL, Devue C, On S, Scetbun J, Romain M, Paul JL, Rothenberg ME, Marcellin P, Durand F, Bedossa P, Prip-Buus C, Baugé E, Staels B, Boulanger CM, Tedgui A, and Rautou PE
- Subjects
- Animals, Diet, High-Fat, Gene Expression Profiling methods, Hepatocytes metabolism, Hepatocytes pathology, Humans, Lipid Metabolism, Lipoproteins, LDL metabolism, Mice, MicroRNAs antagonists & inhibitors, PPAR alpha antagonists & inhibitors, MicroRNAs metabolism, Non-alcoholic Fatty Liver Disease metabolism, Non-alcoholic Fatty Liver Disease prevention & control, Oligonucleotides metabolism, Oligonucleotides pharmacology, PPAR alpha metabolism
- Abstract
Objective: Previous studies suggested that microRNA-21 may be upregulated in the liver in non-alcoholic steatohepatitis (NASH), but its role in the development of this disease remains unknown. This study aimed to determine the role of microRNA-21 in NASH., Design: We inhibited or suppressed microRNA-21 in different mouse models of NASH: (a) low-density lipoprotein receptor-deficient ( Ldlr
-/- ) mice fed a high-fat diet and treated with antagomir-21 or antagomir control; (b) microRNA-21-deficient and wild-type mice fed a methionine-choline-deficient (MCD) diet; (c) peroxisome proliferation-activator receptor α (PPARα)-deficient mice fed an MCD diet and treated with antagomir-21 or antagomir control. We assessed features of NASH and determined liver microRNA-21 levels and cell localisation. MicroRNA-21 levels were also quantified in the liver of patients with NASH, bland steatosis or normal liver and localisation was determined., Results: Inhibiting or suppressing liver microRNA-21 expression reduced liver cell injury, inflammation and fibrogenesis without affecting liver lipid accumulation in Ldlr-/- fed a high-fat diet and in wild-type mice fed an MCD diet. Liver microRNA-21 was overexpressed, primarily in biliary and inflammatory cells, in mouse models as well as in patients with NASH, but not in patients with bland steatosis. PPARα, a known microRNA-21 target, implicated in NASH, was decreased in the liver of mice with NASH and restored following microRNA-21 inhibition or suppression. The effect of antagomir-21 was lost in PPARα-deficient mice., Conclusions: MicroRNA-21 inhibition or suppression decreases liver injury, inflammation and fibrosis, by restoring PPARα expression. Antagomir-21 might be a future therapeutic strategy for NASH., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.)- Published
- 2016
- Full Text
- View/download PDF
38. Bone-marrow-derived very small embryonic-like stem cells in patients with critical leg ischaemia: evidence of vasculogenic potential.
- Author
-
Guerin CL, Loyer X, Vilar J, Cras A, Mirault T, Gaussem P, Silvestre JS, and Smadja DM
- Subjects
- Aged, Animals, Bone Marrow Cells cytology, Cell Differentiation, Cell Lineage, Flow Cytometry, Gangrene therapy, Gene Expression Regulation, Hindlimb pathology, Humans, Inflammation, Mice, Mice, Nude, Middle Aged, Neovascularization, Pathologic, Neovascularization, Physiologic, Phenotype, Platelet Endothelial Cell Adhesion Molecule-1 metabolism, Vascular Endothelial Growth Factor A metabolism, Vascular Endothelial Growth Factor B metabolism, Embryonic Stem Cells cytology, Ischemia therapy, Leg Injuries therapy, Ulcer therapy
- Abstract
Very small embryonic-like stem cells (VSELs) are multipotent stem cells localised in adult bone marrow (BM) that may be mobilised into peripheral blood (PB) in response to tissue injury. We aimed to quantify VSELs in BM and PB of patients with critical limb ischaemia (CLI) and to test their angiogenic potential in vitro as well as their therapeutic capacity in mouse model of CLI. We isolated BM VSELs from patients with CLI and studied their potential to differentiate into vascular lineages. Flow and imaging cytometry showed that VSEL counts were lower in BM (p< 0.001) and higher (p< 0.001) in PB from CLI patients compared to healthy controls, suggesting that ischaemia may trigger VSELs mobilisation in this patient population. Sorted BM-VSELs cultured in angiogenic media acquired a mesenchymal phenotype (CD90+, Thy-1 gene positive expression). VSEL-derived cells had a pattern of secretion similar to that of endothelial progenitor cells, as they released low levels of VEGF-A and inflammatory cytokines. Noteworthy, VSELs triggered post-ischaemic revascularisation in immunodeficient mice (p< 0.05 vs PBS treatment), and acquired an endothelial phenotype either in vitro when cultured in the presence of VEGF-B (Cdh-5 gene positive expression), or in vivo in Matrigel implants (human CD31+ staining in neo-vessels from plug sections). In conclusion, VSELs are a potential new source of therapeutic cells that may give rise to cells of the endothelial lineage in humans.
- Published
- 2015
- Full Text
- View/download PDF
39. Cooperation between human fibrocytes and endothelial colony-forming cells increases angiogenesis via the CXCR4 pathway.
- Author
-
Smadja DM, Dorfmüller P, Guerin CL, Bieche I, Badoual C, Boscolo E, Kambouchner M, Cazes A, Mercier O, Humbert M, Gaussem P, Bischoff J, and Israël-Biet D
- Subjects
- Angiogenic Proteins biosynthesis, Angiogenic Proteins genetics, Animals, Cell Differentiation, Cell Division, Cells, Cultured, Chemokine CXCL12 biosynthesis, Chemokine CXCL12 genetics, Collagen, Colony-Forming Units Assay, Culture Media, Conditioned pharmacology, Drug Combinations, Fetal Blood cytology, Fibrosis, Gene Expression Profiling, Humans, Idiopathic Pulmonary Fibrosis pathology, Laminin, Lung metabolism, Lung pathology, Male, Mice, Mice, Nude, Proteoglycans, Receptors, CXCR4 biosynthesis, Receptors, CXCR4 genetics, Endothelial Cells pathology, Fibroblasts pathology, Hematopoietic Stem Cells pathology, Idiopathic Pulmonary Fibrosis physiopathology, Neovascularization, Physiologic, Receptors, CXCR4 physiology
- Abstract
Fibrotic diseases of the lung are associated with a vascular remodelling process. Fibrocytes (Fy) are a distinct population of blood-borne cells that co-express haematopoietic cell antigens and fibroblast markers, and have been shown to contribute to organ fibrosis. The purpose of this study was to determine whether fibrocytes cooperate with endothelial colony-forming cells (ECFC) to induce angiogenesis. We isolated fibrocytes from blood of patient with idiopathic pulmonary fibrosis (IPF) and characterised them by flow cytometry, quantitative reverse transcriptase PCR (RTQ-PCR), and confocal microscopy. We then investigated the angiogenic interaction between fibrocytes and cord-blood-derived ECFC, both in vitro and in an in vivo Matrigel implant model. Compared to fibroblast culture medium, fibrocyte culture medium increased ECFC proliferation and differentiation via the SDF-1/CXCR4 pathway. IPF-Fy co-implanted with human ECFC in Matrigel plugs in immunodeficient mice formed functional microvascular beds, whereas fibroblasts did not. Evaluation of implants after two weeks revealed an extensive network of erythrocyte-containing blood vessels. CXCR4 blockade significantly inhibited this blood vessel formation. The clinical relevance of these data was confirmed by strong CXCR4 expression in vessels close to fibrotic areas in biopsy specimens from patients with IPF, by comparison with control lungs. In conclusion, circulating fibrocytes might contribute to the intense remodelling of the pulmonary vasculature in patients with idiopathic pulmonary fibrosis.
- Published
- 2014
- Full Text
- View/download PDF
40. α6-Integrin is required for the adhesion and vasculogenic potential of hemangioma stem cells.
- Author
-
Smadja DM, Guerin CL, Boscolo E, Bieche I, Mulliken JB, and Bischoff J
- Subjects
- Animals, Cell Adhesion drug effects, Cell Differentiation drug effects, Cell Proliferation, Child, Preschool, Female, Gene Knockdown Techniques, Humans, Infant, Laminin pharmacology, Liver drug effects, Liver metabolism, Liver pathology, Male, Mice, Hemangioma metabolism, Hemangioma pathology, Integrin alpha6 metabolism, Neovascularization, Physiologic drug effects, Stem Cells metabolism, Stem Cells pathology
- Abstract
Infantile hemangioma (IH) is the most common tumor of infancy. Hemangioma stem cells (HemSC) are a mesenchymal subpopulation isolated from IH CD133+ cells. HemSC can differentiate into endothelial and pericyte/smooth muscle cells and form vascular networks when injected in immune-deficient mice. α6-Integrin subunit has been implicated in the tumorgenicity of glioblastoma stem cells and the homing properties of hematopoietic, endothelial, and mesenchymal progenitor cells. Therefore, we investigated the possible function(s) of α6-integrin in HemSC. We documented α6-integrin expression in IH tumor specimens and HemSC by RT-qPCR and flow cytometry. We examined the effect of blocking or silencing α6-integrin on the adhesive and proliferative properties of HemSC in vitro and the vasculogenic and homing properties of HemSC in vivo. Targeting α6-integrin in cultured HemSC inhibited adhesion to laminin but had no effect on proliferation. Vessel-forming ability in Matrigel implants and hepatic homing after i.v. delivery were significantly decreased in α6-integrin siRNA-transfected HemSC. In conclusion, α6-integrin is required for HemSC adherence to laminin, vessel formation in vivo, and for homing to the liver. Thus, we uncovered an important role for α6 integrin in the vasculogenic properties of HemSC. Our results suggest that α6-integrin expression on HemSC could be a new target for antihemangioma therapy., (© AlphaMed Press.)
- Published
- 2014
- Full Text
- View/download PDF
41. PD-1-expressing tumor-infiltrating T cells are a favorable prognostic biomarker in HPV-associated head and neck cancer.
- Author
-
Badoual C, Hans S, Merillon N, Van Ryswick C, Ravel P, Benhamouda N, Levionnois E, Nizard M, Si-Mohamed A, Besnier N, Gey A, Rotem-Yehudar R, Pere H, Tran T, Guerin CL, Chauvat A, Dransart E, Alanio C, Albert S, Barry B, Sandoval F, Quintin-Colonna F, Bruneval P, Fridman WH, Lemoine FM, Oudard S, Johannes L, Olive D, Brasnu D, and Tartour E
- Subjects
- Animals, Carcinoma, Squamous Cell metabolism, Carcinoma, Squamous Cell virology, Female, Flow Cytometry, Fluorescent Antibody Technique, Head and Neck Neoplasms metabolism, Humans, Lymphocyte Activation immunology, Lymphocytes, Tumor-Infiltrating metabolism, Mice, Mice, Inbred C57BL, Papillomavirus Infections complications, Papillomavirus Infections immunology, Papillomavirus Infections metabolism, Prognosis, Programmed Cell Death 1 Receptor immunology, T-Lymphocyte Subsets immunology, Tumor Microenvironment immunology, Biomarkers, Tumor metabolism, Carcinoma, Squamous Cell immunology, Head and Neck Neoplasms immunology, Head and Neck Neoplasms virology, Lymphocytes, Tumor-Infiltrating immunology, Programmed Cell Death 1 Receptor biosynthesis, T-Lymphocytes immunology
- Abstract
Head and neck cancers positive for human papillomavirus (HPV) have a more favorable clinical outcome than HPV-negative cancers, but it is unknown why this is the case. We hypothesized that prognosis was affected by intrinsic features of HPV-infected tumor cells or differences in host immune response. In this study, we focused on a comparison of regulatory Foxp3(+) T cells and programmed death-1 (PD-1)(+) T cells in the microenvironment of tumors that were positive or negative for HPV, in two groups that were matched for various clinical and biologic parameters. HPV-positive head and neck cancers were more heavily infiltrated by regulatory T cells and PD-1(+) T cells and the levels of PD-1(+) cells were positively correlated with a favorable clinical outcome. In explaining this paradoxical result, we showed that these PD-1(+) T cells expressed activation markers and were functional after blockade of the PD-1-PD-L1 axis in vitro. Approximately 50% of PD-1(+) tumor-infiltrating T cells lacked Tim-3 expression and may indeed represent activated T cells. In mice, administration of a cancer vaccine increased PD-1 on T cells with concomitant tumor regression. In this setting, PD-1 blockade synergized with vaccine in eliciting antitumor efficacy. Our findings prompt a need to revisit the significance of PD-1-infiltrating T cells in cancer, where we suggest that PD-1 detection may reflect a previous immune response against tumors that might be reactivated by PD-1/PD-L1 blockade.
- Published
- 2013
- Full Text
- View/download PDF
42. Angiogenic potential of BM MSCs derived from patients with critical leg ischemia.
- Author
-
Smadja DM, d'Audigier C, Guerin CL, Mauge L, Dizier B, Silvestre JS, Dal Cortivo L, Gaussem P, and Emmerich J
- Subjects
- Aged, Animals, Humans, Mesenchymal Stem Cells cytology, Mice, Bone Marrow Cells cytology, Ischemia immunology, Leg blood supply, Leg pathology, Leukocytes, Mononuclear cytology, Mesenchymal Stem Cells metabolism, Peripheral Arterial Disease pathology
- Published
- 2012
- Full Text
- View/download PDF
43. The profibrotic cytokine transforming growth factor-β1 increases endothelial progenitor cell angiogenic properties.
- Author
-
Evrard SM, d'Audigier C, Mauge L, Israël-Biet D, Guerin CL, Bieche I, Kovacic JC, Fischer AM, Gaussem P, and Smadja DM
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Case-Control Studies, Cell Movement, Cell Survival, Cells, Cultured, Female, Fetal Blood cytology, France, Hemoglobins metabolism, Humans, Idiopathic Pulmonary Fibrosis blood, Idiopathic Pulmonary Fibrosis physiopathology, Male, Mice, Mice, Inbred C57BL, Middle Aged, Prospective Studies, RNA Interference, Receptors, Transforming Growth Factor beta genetics, Receptors, Transforming Growth Factor beta metabolism, Transfection, Transforming Growth Factor beta1 blood, Up-Regulation, Endothelial Cells metabolism, Idiopathic Pulmonary Fibrosis metabolism, Neovascularization, Physiologic, Stem Cells metabolism, Transforming Growth Factor beta1 metabolism
- Abstract
Background: Transforming growth factor-β1 (TGF-β1) is a profibrotic cytokine that plays a major role in vascular biology, and is known to regulate the phenotype and activity of various vascular cell populations. Because most fibrotic diseases, such as idiopathic pulmonary fibrosis (IPF), are associated with vascular remodeling, and as endothelial progenitor cells (EPCs) may be involved in this process, we investigated the impact of TGF-β1 modulation of EPC angiogenic properties., Methods: TGF-β1 plasma levels were determined in 64 patients with IPF and compared with those in controls. The effect of TGF-β1 on angiogenesis was studied in vivo in a Matrigel plug model and in vitro on endothelial colony-forming cells (ECFCs). We studied the effects of inhibiting the expression of the three main receptors of TGF-β1 in ECFCs by using short interfering RNA., Results: Total TGF-β1 plasma levels were significantly increased in patients with IPF as compared with controls (P < 0.0001). TGF-β1 had proangiogenic effects in vivo by increasing hemoglobin content and blood vessel formation in Matrigel plugs implanted in C57/Bl6 mice, and in vitro by enhancing ECFC viability and migration. The effects were abolished by silencing the three main TGF-β1 receptors., Conclusions: TGF-β1 is proangiogenic in vivo and induces ECFC angiogenic properties in vitro, suggesting that TGF-β1 may play a role during vascular remodeling in fibrotic disease states via EPCs., (© 2012 International Society on Thrombosis and Haemostasis.)
- Published
- 2012
- Full Text
- View/download PDF
44. Flow cytometry: retrospective, fundamentals and recent instrumentation.
- Author
-
Picot J, Guerin CL, Le Van Kim C, and Boulanger CM
- Abstract
Flow cytometry is a complete technology given to biologists to study cellular populations with high precision. This technology elegantly combines sample dimension, data acquisition speed, precision and measurement multiplicity. Beyond the statistical aspect, flow cytometry offers the possibility to physically separate sub-populations. These performances come from the common endeavor of physicists, biophysicists, biologists and computer engineers, who succeeded, by providing new concepts, to bring flow cytometry to current maturity. The aim of this paper is to present a complete retrospective of the technique and remind flow cytometry fundamentals before focusing on recent commercial instrumentation.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.