23
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Difference in mobilization of progenitor cells after myocardial infarction in smoking versus non-smoking patients: insights from the BONAMI trial

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Introduction

          Although autologous bone marrow cell (BMC) therapy has emerged as a promising treatment for acute myocardial infarction (AMI), trials reported mixed results. In the BONAMI trial, active smoking reduced cardiac function recovery after reperfused AMI. Therefore, we hypothesized that variability in the functionality of BMCs retrieved from patients with cardiovascular risk factors may partly explain these mixed results. We investigated the characteristics of progenitor cells in active smokers and non-smokers with AMI and their potential impact on BMC therapy efficacy.

          Methods

          Bone marrow and blood samples from 54 smoking and 47 non-smoking patients enrolled in the BONAMI cell therapy trial were analyzed.

          Results

          The white BMC and CD45dimCD34+ cell numbers were higher in active smokers ( P = 0.001, P = 0.03, respectively). In marked contrast, either bone marrow or blood endothelial progenitor CD45dimCD34 + KDR + cells (EPCs) were decreased in active smokers ( P = 0.005, P = 0.04, respectively). Importantly, a multivariate analysis including cardiovascular risk factors confirmed the association between active smoking and lower EPC number in bone marrow ( P = 0.04) and blood ( P = 0.04). Furthermore, baseline circulating EPC count predicted infarct size decrease at three months post-AMI in non-smokers ( P = 0.01) but not in active smokers. Interestingly, baseline circulating EPCs were no longer predictive of cardiac function improvement in the BMC therapy group.

          Conclusions

          These data suggest that circulating EPCs play an important role in cardiac repair post-AMI only in non-smokers and that active smoking-associated EPC alterations may participate in the impairment of cardiac function recovery observed in smokers after AMI, an effect that was overridden by BMC therapy.

          Related collections

          Most cited references33

          • Record: found
          • Abstract: found
          • Article: not found

          Number and migratory activity of circulating endothelial progenitor cells inversely correlate with risk factors for coronary artery disease.

          Recent studies provide increasing evidence that postnatal neovascularization involves bone marrow-derived circulating endothelial progenitor cells (EPCs). The regulation of EPCs in patients with coronary artery disease (CAD) is unclear at present. Therefore, we determined the number and functional activity of EPCs in 45 patients with CAD and 15 healthy volunteers. The numbers of isolated EPCs and circulating CD34/kinase insert domain receptor (KDR)-positive precursor cells were significantly reduced in patients with CAD by approximately 40% and 48%, respectively. To determine the influence of atherosclerotic risk factors, a risk factor score including age, sex, hypertension, diabetes, smoking, positive family history of CAD, and LDL cholesterol levels was used. The number of risk factors was significantly correlated with a reduction of EPC levels (R=-0.394, P=0.002) and CD34-/KDR-positive cells (R=-0.537, P<0.001). Analysis of the individual risk factors demonstrated that smokers had significantly reduced levels of EPCs (P<0.001) and CD34-/KDR-positive cells (P=0.003). Moreover, a positive family history of CAD was associated with reduced CD34-/KDR-positive cells (P=0.011). Most importantly, EPCs isolated from patients with CAD also revealed an impaired migratory response, which was inversely correlated with the number of risk factors (R=-0.484, P=0.002). By multivariate analysis, hypertension was identified as a major independent predictor for impaired EPC migration (P=0.043). The present study demonstrates that patients with CAD revealed reduced levels and functional impairment of EPCs, which correlated with risk factors for CAD. Given the important role of EPCs for neovascularization of ischemic tissue, the decrease of EPC numbers and activity may contribute to impaired vascularization in patients with CAD. The full text of this article is available at http://www.circresaha.org.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Mesenchymal stem cells: biology, pathophysiology, translational findings, and therapeutic implications for cardiac disease.

            Mesenchymal stem cells (MSCs) are a prototypical adult stem cell with capacity for self-renewal and differentiation with a broad tissue distribution. Initially described in bone marrow, MSCs have the capacity to differentiate into mesoderm- and nonmesoderm-derived tissues. The endogenous role for MSCs is maintenance of stem cell niches (classically the hematopoietic), and as such, MSCs participate in organ homeostasis, wound healing, and successful aging. From a therapeutic perspective, and facilitated by the ease of preparation and immunologic privilege, MSCs are emerging as an extremely promising therapeutic agent for tissue regeneration. Studies in animal models of myocardial infarction have demonstrated the ability of transplanted MSCs to engraft and differentiate into cardiomyocytes and vasculature cells, recruit endogenous cardiac stem cells, and secrete a wide array of paracrine factors. Together, these properties can be harnessed to both prevent and reverse remodeling in the ischemically injured ventricle. In proof-of-concept and phase I clinical trials, MSC therapy improved left ventricular function, induced reverse remodeling, and decreased scar size. This article reviews the current understanding of MSC biology, mechanism of action in cardiac repair, translational findings, and early clinical trial data of MSC therapy for cardiac disease.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Critical reevaluation of endothelial progenitor cell phenotypes for therapeutic and diagnostic use.

              Diverse subsets of endothelial progenitor cells (EPCs) are used for the treatment of ischemic diseases in clinical trials, and circulating EPCs levels are considered as biomarkers for coronary and peripheral artery disease. However, despite significant steps forward in defining their potential for both therapeutic and diagnostic purposes, further progress has been mired by unresolved questions around the definition and the mechanism of action of EPCs. Diverse culturing methods and detection of various combinations of different surface antigens were used to enrich and identify EPCs. These attempts were particularly challenged by the close relationship and overlapping markers of the endothelial and hematopoietic lineages. This article will critically review the most commonly used protocols to define EPCs by culture assays or by fluorescence-activated cell sorter in the context of their therapeutic or diagnostic use. We also delineate new research avenues to move forward our knowledge on EPC biology.
                Bookmark

                Author and article information

                Contributors
                Journal
                Stem Cell Res Ther
                Stem Cell Res Ther
                Stem Cell Research & Therapy
                BioMed Central
                1757-6512
                2013
                24 December 2013
                : 4
                : 6
                : 152
                Affiliations
                [1 ]INSERM UMR1087/CNRS UMR6291, l’institut du thorax, IRS-UN, 8 quai Moncousu, BP 70721 44007 Nantes, cedex 1, France
                [2 ]CNRS, UMR 6291 Nantes, France
                [3 ]Université de Nantes, Nantes, France
                [4 ]CHU Nantes, Laboratoire d’immunologie, CIC 4 Nantes, France
                [5 ]Univ Lille Nord de France, UDSL, IFR 114, EA 2693, Faculté de Médecine, Lille, France
                [6 ]CHRU, Institut d’Hématologie-Transfusion, Pôle de Pathologie cardiologie-vasculaire, Lille, France
                [7 ]INSERM UMR 1048, Inst Metab et Cardiovasc Dis I2MC, Université Toulouse III-Paul Sabatier, Service de Pharmacie CHU Rangueil, Toulouse, France
                [8 ]Inserm, CIC-BT 504, CIC-P 006 and U955 team 3, AP-HP, Henri Mondor University Hospital, Créteil, France
                [9 ]Department of Cardiology, INSERM U661, CHU de Montpellier, Université Montpellier 1, Montpellier, France
                [10 ]Unité Mixte de Thérapie Cellulaire EFS RA, UM biochimie des cancers et Biothérapies, Pôle de Biologie CHU de Grenoble, Grenoble, France
                [11 ]Cell therapy facility EFS Ile de France and CIC-BT N°504, Créteil, France
                [12 ]Department of Cardiology and CIC-Biotherapies 511, CHU de Toulouse, Toulouse, France
                Article
                scrt382
                10.1186/scrt382
                4054959
                24423369
                3b12cea2-6066-499c-8467-00a780379f28
                Copyright © 2013 Lamirault et al.; licensee BioMed Central Ltd.

                This is an open access article distributed under the terms of the Creative Commons Attribution License ( http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

                History
                : 11 September 2013
                : 14 November 2013
                : 10 December 2013
                Categories
                Research

                Molecular medicine
                Molecular medicine

                Comments

                Comment on this article