Intravenously-Delivered Mesenchymal Stem Cells: Systemic Anti-Inflammatory Effects Improve Left Ventricular Dysfunction in Acute Myocardial Infarction and Ischemic Cardiomyopathy.

MedStar author(s):
Citation: Circulation Research. 120(10):1598-1613, 2017 May 12PMID: 28232595Institution: MedStar Health Research Institute | MedStar Heart & Vascular InstituteForm of publication: Journal ArticleMedline article type(s): Journal ArticleSubject headings: *Cardiomyopathies/th [Therapy] | *Mesenchymal Stem Cell Transplantation/mt [Methods] | *Myocardial Infarction/th [Therapy] | *Myocardial Ischemia/th [Therapy] | *Ventricular Dysfunction, Left/th [Therapy] | Administration, Intravenous | Animals | Cardiomyopathies/im [Immunology] | Cardiomyopathies/pp [Physiopathology] | Cells, Cultured | Humans | Male | Mesenchymal Stromal Cells/im [Immunology] | Mice | Myocardial Infarction/im [Immunology] | Myocardial Infarction/pp [Physiopathology] | Myocardial Ischemia/im [Immunology] | Myocardial Ischemia/pp [Physiopathology] | Treatment Outcome | Ventricular Dysfunction, Left/im [Immunology] | Ventricular Dysfunction, Left/pp [Physiopathology]Year: 2017Local holdings: Available online from MWHC library: 1953 - presentISSN:
  • 0009-7330
Abstract: Rationale:Virtually all mesenchymal stem cell (MSC) studies assume therapeutic effects accrue from local myocardial effects of engrafted MSCs. Since few intravenously-administered MSCs engraft in the myocardium, studies have mainly utilized direct myocardial delivery. We adopted a different paradigm. <b><u>Objective:</u></b> To test whether intravenously-administered MSCs reduce LV dysfunction both post-AMI and in ischemic cardiomyopathy, and that these effects are caused, at least partly, by systemic anti-inflammatory activities. <b><u>Methods and Results:</u></b> Mice underwent 45min of left anterior descending artery occlusion. Human MSCs, grown chronically at 5% O<sub>2</sub>, were administered intravenously. LV function was assessed by serial echocardiography, TTC staining determined infarct size, and FACS assessed cell composition. Fluorescent and radiolabeled MSCs (1x10<sup>6</sup>) were injected 24h post-MI and homed to regions of myocardial injury; however, the myocardium contained only a small proportion of total MSCs. Mice received 2x10<sup>6</sup> MSCs or saline intravenously 24h post-MI (n=16/group). At day 21, we harvested blood and spleens for FACS and hearts for TTC staining. Adverse LV remodeling and deteriorating LVEF occurred in control mice with large infarcts (>25% LV). Intravenous MSCs eliminated the progressive deterioration in LVEF, LVEDV and LVESV. MSCs significantly decreased natural killer (NK) cells in the heart and spleen, and neutrophils in the heart. Specific NK cell depletion 24h pre-AMI significantly improved infarct size, LVEF, and adverse LV remodeling, changes associated with decreased neutrophils in the heart. In an ischemic cardiomyopathy model, mice 4 weeks post-MI were randomized to tail-vein injection of 2x10<sup>6</sup> MSCs, with injection repeated at week 3 (n=16) vs PBS control (n=16). MSCs significantly increased LVEF and decreased LVESV. <b><u>Conclusions:</u></b> Intravenously-administered MSCs for AMI attenuate the progressive deterioration in LV function and adverse remodeling in mice with large infarcts, and in ischemic cardiomyopathy they improve LV function, effects apparently modulated in part by systemic anti-inflammatory activities.All authors: Albelda MT, Dimastromatteo J, Epstein SE, Frias JC, Glover DK, Kharazi A, Lipinski MJ, Luger D, Sikora S, Vertelov G, Waksman R, Westman PCFiscal year: FY2017Digital Object Identifier: Date added to catalog: 2017-05-06
Holdings
Item type Current library Collection Call number Status Date due Barcode
Journal Article MedStar Authors Catalog Article 28232595 Available 28232595

Available online from MWHC library: 1953 - present

Rationale:Virtually all mesenchymal stem cell (MSC) studies assume therapeutic effects accrue from local myocardial effects of engrafted MSCs. Since few intravenously-administered MSCs engraft in the myocardium, studies have mainly utilized direct myocardial delivery. We adopted a different paradigm. <b><u>Objective:</u></b> To test whether intravenously-administered MSCs reduce LV dysfunction both post-AMI and in ischemic cardiomyopathy, and that these effects are caused, at least partly, by systemic anti-inflammatory activities. <b><u>Methods and Results:</u></b> Mice underwent 45min of left anterior descending artery occlusion. Human MSCs, grown chronically at 5% O<sub>2</sub>, were administered intravenously. LV function was assessed by serial echocardiography, TTC staining determined infarct size, and FACS assessed cell composition. Fluorescent and radiolabeled MSCs (1x10<sup>6</sup>) were injected 24h post-MI and homed to regions of myocardial injury; however, the myocardium contained only a small proportion of total MSCs. Mice received 2x10<sup>6</sup> MSCs or saline intravenously 24h post-MI (n=16/group). At day 21, we harvested blood and spleens for FACS and hearts for TTC staining. Adverse LV remodeling and deteriorating LVEF occurred in control mice with large infarcts (>25% LV). Intravenous MSCs eliminated the progressive deterioration in LVEF, LVEDV and LVESV. MSCs significantly decreased natural killer (NK) cells in the heart and spleen, and neutrophils in the heart. Specific NK cell depletion 24h pre-AMI significantly improved infarct size, LVEF, and adverse LV remodeling, changes associated with decreased neutrophils in the heart. In an ischemic cardiomyopathy model, mice 4 weeks post-MI were randomized to tail-vein injection of 2x10<sup>6</sup> MSCs, with injection repeated at week 3 (n=16) vs PBS control (n=16). MSCs significantly increased LVEF and decreased LVESV. <b><u>Conclusions:</u></b> Intravenously-administered MSCs for AMI attenuate the progressive deterioration in LV function and adverse remodeling in mice with large infarcts, and in ischemic cardiomyopathy they improve LV function, effects apparently modulated in part by systemic anti-inflammatory activities.

English

Powered by Koha