Stromal vascular fraction transplantation as an alternative therapy for ischemic heart failure: anti-inflammatory role

Goditha U Premaratne, Li-Ping Ma, Masatoshi Fujita, Xue Lin, Entela Bollano, Michael Fu, Goditha U Premaratne, Li-Ping Ma, Masatoshi Fujita, Xue Lin, Entela Bollano, Michael Fu

Abstract

Background: The aims of this study were: (1) to show the feasibility of using adipose-derived stromal vascular fraction (SVF) as an alternative to bone marrow mono nuclear cell (BM-MNC) for cell transplantation into chronic ischemic myocardium; and (2) to explore underlying mechanisms with focus on anti-inflammation role of engrafted SVF and BM-MNC post chronic myocardial infarction (MI) against left ventricular (LV) remodelling and cardiac dysfunction.

Methods: Four weeks after left anterior descending coronary artery ligation, 32 Male Lewis rats with moderate MI were divided into 3 groups. SVF group (n = 12) had SVF cell transplantation (6 × 10(6) cells). BM-MNC group (n = 12) received BM-MNCs (6 × 10(6)) and the control (n = 10) had culture medium. At 4 weeks, after the final echocardiography, histological sections were stained with Styrus red and immunohistochemical staining was performed for α-smooth muscle actin, von Willebrand factor, CD3, CD8 and CD20.

Results: At 4 weeks, in SVF and BM-MNC groups, LV diastolic dimension and LV systolic dimension were smaller and fractional shortening was increased in echocardiography, compared to control group. Histology revealed highest vascular density, CD3+ and CD20+ cells in SVF transplanted group. SVF transplantation decreased myocardial mRNA expression of inflammatory cytokines TNF-α, IL-6, MMP-1, TIMP-1 and inhibited collagen deposition.

Conclusions: Transplantation of adipose derived SVF cells might be a useful therapeutic option for angiogenesis in chronic ischemic heart disease. Anti-inflammation role for SVF and BM transplantation might partly benefit for the cardioprotective effect for chronic ischemic myocardium.

Figures

Figure 1
Figure 1
Transplanted cells. PKH26 labeled donor cells (red fluorescence, x200) in SVF and BM-MNC transplanted groups. Bars represent a distance of 50 μm.
Figure 2
Figure 2
Vascular density. 2a (A-C) Immunohistochemistry for von Willebrand factor (brown, x100). Representative pictures in the peri-MI area from SVF, BM-MNC and Control groups, respectively. (D-F) Immunohistochemistry with α-smooth muscle actin antibody (brown, x100). Representative pictures in the peri-MI area from SVF, BM-MNC and Control groups, respectively. Scale bars indicate distances of 100 μm. 2b Graphs: the number of vessels (number/mm2) in the peri-MI area, micro-vessel density (density of vessels <30 μm in diameter) (A), and large-vessel density (density of vessels >30 μm in diameter) (B). Data are given as the mean ± SEM. *p < 0.05 vs. Control group, **p < 0.05 vs. BM-MNC group, p < 0.001 vs. Control group.
Figure 3
Figure 3
Fibrotic area. Representative pictures from groups SVF, BM-MNC and Control, respectively. Bars represent a distance of 100 μm. Graphs: Percentage of fibrotic area inside the infarct (A) and peri-infarct area (B). Data are given as the mean ± SEM. *p < 0.05 vs. Control group, p < 0.01 vs. Control group.
Figure 4
Figure 4
Expression of mRNA. Expression of mRNA levels of tumor necrosis factor α (A, TNFα); interleukin 6 (B, IL-6); matrix metalloproteinase 1 (C, MMP-1); tissue inhibitor of metalloproteinase 1 (D, TIMP-1), brain natriuretic peptide (E, BNP) and vascular endothelial growth factor (F, VEGF) in the left ventricular myocardium as measured by reverse transcription polymerase chain reaction in the rat left ventricular myocardium, 4 weeks after treatment. mRNA expressions were calculated via a standard curve and normalized to an endogen control. Data are given as the mean ± SEM. *p < 0.05 vs. Control group, **p < 0.01 vs. BM-MNC group, †p < 0.001 vs. Control group.
Figure 5
Figure 5
Immunohistochemistry for CD3+ (T lymphocytes), (brown, × 100). Representative pictures in the infarct area from SVF, BM-MNC and Control groups, respectively. Bars represent a distance of 100μm. Graph: the number of CD3+ (number/mm2) in the infarct area. Data are given as the mean ± SEM. *p < 0.05 vs. Control group.
Figure 6
Figure 6
Immunohistochemistry for CD20+ (B lymphocytes), (brown, × 100). Representative pictures in the infarct area from SVF, BM-MNC and Control groups, respectively. Bars represent a distance of 50μm. Graph: the number of CD20+ (number/mm2) in the infarct area. Data are given as the mean ± SEM. †p < 0.001 vs. Control group, **p < 0.01 vs. Control group.
Figure 7
Figure 7
Localization of IL-6 (brown) by immunohistochemical analysis in cell transplanted and control hearts. Magnification × 100. Representative pictures in the infarct area from SVF, BM-MNC and Control groups, respectively. Bars represent a distance of 100μm. Graph: Percentage of IL-6 positive area inside the infarct. Data are given as the mean ± SEM. p < 0.001 vs. Control group.

References

    1. Shabbir A, Zisa D, Suzuki G, Lee T. Heart failure therapy mediated by the trophic activities by the bone marrow mesenchymal stem cells: a noninvasive therapeutic regimen. Am J Physiol Heart Circ Physiol. 2009;296(6):1888–1997. doi: 10.1152/ajpheart.00186.2009.
    1. Tse H-F, Siu C-W, Zhu S-G, Songyan L, Zhang Q-Y, Lai W-H, Kwong Y-L, Nicholls J, Lau C-P. Paracrine effects of direct intramyocardial implantation of bone marrow derived cells to enhance neovascularization in chronic ischaemic myocardium. Eur J Heart Fail. 2007;9(8):747–753. doi: 10.1016/j.ejheart.2007.03.008.
    1. Makkar RR, Price MJ, Lill M, Frantzen M, Takizawa K, Kleisli T, Zheng J, Kar S, McClelan Miyamota T, Bick-Forrester J, Fishbein MC, Shah PK, Forrester JS, Sharifi B, Cheng P-S, Qayyum M. Intramyocardial injection of allogenic bone marrow-derived mesenchymal stem cells without immunosuppression preserves cardiac function in a porcine model of myocardial infarction. J Cardiovasc Pharmacol Ther. 2005;10(4):225–233. doi: 10.1177/107424840501000403.
    1. Baba S, Heike T, Yoshimoto M, Umeda K, Doi H, Iwasa T, Lin X, Matsuoka S, Komeda M, Nakahata T. Flk1+ cardiac stem/progenitor cells derived from embryonic stem cells improve cardiac function in a dilated cardiomyopathy mouse model. Cardiovasc Res. 2007;76(1):119–131. doi: 10.1016/j.cardiores.2007.05.013.
    1. Tse H-F, Thamber S, Kwong Y-L, Rowlings P, Bellamy G, McCrohon J, Thomas P, Bastian B, Chan JKF, Lo G, Ho C-L, Chan W-S, Kwong RY, Parker A, Hauser Chan J, Fong DYT, Lau C-P. Prospective randomized trial of direct endomyocardial implantation of bone marrow cells for treatment of severe coronary artery diseases (PROTECT-CAD trial) Eur Heart J. 2007;28:2998–3005. doi: 10.1093/eurheartj/ehm485.
    1. Dill T, Schächinger V, Rolf A, Möllmann S, Thiele H, Tillmanns H, Assmus B, Dimmeler S, Zeiher AM, Hamm C. Intracoronary administration of bone marrow-derived progenitor cells improves left ventricular function in patients at risk for adverse remodeling after acute ST-segment elevation myocardial infarction: Results of the Reinfusion of Enriched Progenitor cells And Infarct Remodeling in Acute Myocardial Infarction study (REPAIR-AMI) cardiac magnetic resonance imaging substudy. Am Heart J. 2009;157(3):541–547. doi: 10.1016/j.ahj.2008.11.011.
    1. Yamamoto N, Akamatsu H, Hasegawa S, Yamada T, Nakata S, Ohkuma M, Miyachi EI, Marunouchi T, Matsunaga K. Isolation of multipotent stem cells from mouse adipose tissue. J Dermatol Sci. 2007;48(1):43–52. doi: 10.1016/j.jdermsci.2007.05.015.
    1. Astori G, Vignati F, Bardelli S, Tubio M, Gola M, Albertini V, Bambi F, Scali G, Castelli D, Rasini V, Soldati G, Moccetti. "In vitro" and multicolor phenotypic characterization of cell subpopulations identified in fresh human adipose tissue stromal vascular fraction and in the derived mesenchymal stem cells. J Transl Med. 2007;5(5):844–851.
    1. Palpant NJ, Yasuda S, MacDougald O, Metzger JM. Non-canonical Wnt signaling enhances differentiation of Sca+/c-kit+ adipose-derived murine stromal vascular cells into spontaneously beating cardiac myocytes. J Mol Cell Cardiol. 2007;43:362–370. doi: 10.1016/j.yjmcc.2007.06.012.
    1. Planat-Bénard V, Menard C, André M, Puceat M, Perez A, Garcia-Verdugo JM, Pérnicaud L, Casteilla L. Spontaneous cardiomyocyte differentiation from adipose tissue stroma cells. Circ Res. 2004;94:1–10.
    1. Planat-Bénard V, Silvestre JS, Cousin B, Andre M, Nibbelink M, Tamarat R, Clergue M, Manneville C, Saillan-Barreau C, Duriez M, Tedgui A, Levy B, Pérnicaud L, Casteilla. Plasticity of human adipose lineage cells toward endothelial cells: physiological and therapeutic perspectives. Circulation. 2004;109:656–663.
    1. Nagaya N, Kangawa K, Itoh T, Iwase T, Murakami S, Miyahara Y, Fujii T, Uematsu M, Ohgushi H, Yamagishi M, Tokudome T, Mori H, Miyatake K, Kitamura S. Transplantation of mesenchymal stem cells improves cardiac function in a rat dilated cardiomyopathy. Circulation. 2005;112:1128–1135. doi: 10.1161/CIRCULATIONAHA.104.500447.
    1. Mazo M, Planat-Bénard V, Abizanda G, Pelacho B, Léobon B, Gavira JJ, Penuelas I, Cemborain A, Pérnicaud L, Laharrague P, Joffre C, Boisson M, Ecay M, Collantes M, Barba J, Casteilla L, Prosper F. Transplantation of adipose derived stromal cells is associated with functional improvement in a rat model of chronic myocardial infarction. Eur J Heart Fail. 2008;10:454–462. doi: 10.1016/j.ejheart.2008.03.017.
    1. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, Benhaim P, Lorenz HP, Hedrick MH. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7:211–228. doi: 10.1089/107632701300062859.
    1. Tambara K, Premaratne GU, Sakaguchi G, Kanemitsu N, Lin X, Nakajima H, Sakakibara Y, Kimura Y, Yamamoto M, Tabata Y, Ikeda T, Komeda M. Administration of control-released hepatocyte growth factor enhances the efficacy of skeletal myoblast transplantation in rat infarcted hearts by greatly increasing both quantity and quality of the graft. Circulation. 2005;112(Suppl I):129–134.
    1. Nakajima H, Sakakibara Y, Tambara K, Marui A, Yoshimoto M, Premaratne GU, Lin X, Kanemitsu N, Sakaguchi G, Ikeda T, Nishimura K, Nakahata T, Komeda M. Delivery route in bone marrow cell transplantation should be optimized according to the etiology of heart disease. Circ J. 2008;72:1528–1535. doi: 10.1253/circj.CJ-06-0430.
    1. Premaratne GU, Tambara K, Fujita M, Lin X, Kanemitsu N, Tomita S, Sakaguchi G, Nakajima H, Ikeda T, Komeda M. Repeated implantation is a more effective cell delivery method in skeletal myoblast transplantation for rat myocardial infarction. Circ J. 2006;70:1184–1189. doi: 10.1253/circj.70.1184.
    1. Fazel S, Cimini M, Chen L, Li S, Angoulvant D, Fedak P, Verma S, Weisel RD, Keating A, Li RK. Cardioprotective c-kit+ cells are from the bone marrow and regulate the myocardial balance of angiogenic cytokines. J Clin Invest. 2006;116(7):1865–1877. doi: 10.1172/JCI27019.
    1. Wang JS, Shum-Tim D, Galipeau J, Chedrawy E, Eliopoulos N, Chiu RC. Marrow stromal cells for cellular cardiomyoplasty: feasibility and potential clinical advantages. J Thorac Cardiovasc Surg. 2000;120:999–1005. doi: 10.1067/mtc.2000.110250.
    1. Meyer GP, Wollert KC, Lotz J, Steffens J, Lippolt P, Fichtner S, Hecker H, Schaefer A, Arseniev L, Hertenstein B, Ganser A, Drexler H. Intracoronary bone marrow cell transfer after myocardial infarction: Eighteen months' follow-up data from the randomized, controlled BOOST (Bone marrow transfer to enhance ST-elevation infarct regeneration) Trial. Circulation. 2006;113:1287–1294. doi: 10.1161/CIRCULATIONAHA.105.575118. 4.
    1. Miyahara Y, Nagaya N, Kataoka M, Yanagawa B, Tanaka K, Hao H, Ishino K, Ishida H, Shimizu T, Kangawa K, Sano S, Okano T, Kitamura S, Mori H. Monolayered mesenchymal stem cells repair scarred myocardium after myocardial infarction. Nature Med. 2006;12:459–465. doi: 10.1038/nm1391.
    1. DiNicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P, Grisanti S, Gianni AM. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood. 2002;99:3838–3843. doi: 10.1182/blood.V99.10.3838.
    1. Tse WT, Pendleton JD, Beyer WM, Egalka MC, Guinan EC. Suppression of allogeneic T-cell proliferation by human marrow stromal cells: implications in transplantation. Transplantation. 2003;75:389–397. doi: 10.1097/01.TP.0000045055.63901.A9.
    1. Prabhu SD. Cytokine-induced modulation of cardiac function. Circ Res. 2004;95:1140–1153. doi: 10.1161/01.RES.0000150734.79804.92.
    1. Torre-Amione G, Kapadiya S, Lee J, Durans JB, Bies RD, Young JB, Mann DL. Tumor necrosis factor-α and tumor necrosis factor receptors in the failing human heart. Circulation. 1996;93:704–711.
    1. Ono K, Matsumori A, Shioi T, Furukawa Y, Sasayama S. Cytokine gene expression after myocardial infarction in rat hearts. Possible implication in left ventricular remodeling. Circulation. 1998;98:149–156.
    1. Testa M, Yeh M, Lee P, Fanelli R, Loperfido F, Berman JW, LeJemtel TH. Circulating levels of cytokines and their endogenous modulators in patients with mild to severe congestive heart failure due to coronary artery disease or hypertension. J Am Coll Cardiol. 1996;28:964–971. doi: 10.1016/S0735-1097(96)00268-9.
    1. Torre-Amione G, Kapadiya SR, Benedict C, Oral H, Young JB, Mann DL. Proinflammatory cytokine levels in patients with depressed left ventricular ejection fraction: a report from the Studies of Left Ventricular Dysfunction (SOLVD) J Am Coll Cardiol. 1996;27:1201–1206. doi: 10.1016/0735-1097(95)00589-7.
    1. Haugen E, Chen J, Wikström J, Grönros J, Gan LM, Fu M. Parallel gene expressions of IL-6 and BNP during cardiac hypertrophy complicated with diastolic dysfunction in spontaneously hypertensive rats. Int J Cardiol. 2007;115(1):24–28. doi: 10.1016/j.ijcard.2006.01.031.
    1. Guo J, Lin G, Bao C, Hu Z, Hu M. Anti-inflammation role for mesenchymal stem cells transplantation in myocardial infarction. Inflammation. 2007;30:97–104. doi: 10.1007/s10753-007-9025-3.
    1. Kerckhoven V, Kalkman REA, Sexena PR, Schoemaker RG. Altered cardiac collagen and associated changes in diastolic function of infarcted rat hearts. Cardiovasc Res. 2000;46:316–323. doi: 10.1016/S0008-6363(99)00427-7.
    1. Assmus B, Walter DH, Lehmann R, Honold J, Martin H, Dimmeler S, Zeiher AM, Schächinger V. Intracoronary infusion of progenitor cells is not associated with aggravated restenosis development or atherosclerotic disease progression in patients with acute myocardial infarction. Eur Heart J. 2006;27(24):2989–2995. doi: 10.1093/eurheartj/ehl235.

Source: PubMed

3
Abonneren