Combined transplantation of skeletal myoblasts and angiopoietic progenitor cells reduces infarct size and apoptosis and improves cardiac function in chronic.

Slides:



Advertisements
Similar presentations
Paracrine Action Enhances the Effects of Autologous Mesenchymal Stem Cell Transplantation on Vascular Regeneration in Rat Model of Myocardial Infarction 
Advertisements

Manuel J. Antunes, MD, PhD, DSc 
Transplantation of hypoxia-preconditioned mesenchymal stem cells improves infarcted heart function via enhanced survival of implanted cells and angiogenesis 
Therapeutic Effect of Midkine on Cardiac Remodeling in Infarcted Rat Hearts  Shinya Fukui, MD, Satoru Kitagawa-Sakakida, MD, PhD, Sin Kawamata, MD, PhD,
Epicardial infarct repair with basic fibroblast growth factor–enhanced CorMatrix-ECM biomaterial attenuates postischemic cardiac remodeling  Holly E.M.
Association of CD14+ monocyte-derived progenitor cells with cardiac allograft vasculopathy  Mohamed Salama, MD, PhD, Olena Andrukhova, PhD, Susanne Roedler,
Direct epicardial shock wave therapy improves ventricular function and induces angiogenesis in ischemic heart failure  Daniel Zimpfer, MD, Seyedhossein.
Electron transport chain dysfunction in neonatal pressure-overload hypertrophy precedes cardiomyocyte apoptosis independent of oxidative stress  Eric.
Comparative effects of mesenchymal progenitor cells, endothelial progenitor cells, or their combination on myocardial infarct regeneration and cardiac.
Cell-based gene therapy modifies matrix remodeling after a myocardial infarction in tissue inhibitor of matrix metalloproteinase-3–deficient mice  Denis.
Downregulation of the CXC chemokine receptor 4/stromal cell–derived factor 1 pathway enhances myocardial neovascularization, cardiomyocyte survival, and.
Paracrine Action Enhances the Effects of Autologous Mesenchymal Stem Cell Transplantation on Vascular Regeneration in Rat Model of Myocardial Infarction 
An ovine model of postinfarction dilated cardiomyopathy
Regulatory T cells enhance mesenchymal stem cell survival and proliferation following autologous cotransplantation in ischemic myocardium  Yifu Zhou,
Volume 3, Issue 6, Pages (December 2014)
The reduction of hemodynamic loading assists self-regeneration of the injured heart by increasing cell proliferation, inhibiting cell apoptosis, and inducing.
Edaravone promotes activation of resident cardiac stem cells by transplanted mesenchymal stem cells in a rat myocardial infarction model  Guang-Wei Zhang,
Mechanical preconditioning enables electrophysiologic coupling of skeletal myoblast cells to myocardium  Klaus Neef, PhD, Yeong-Hoon Choi, MD, Sureshkumar.
Cell transplantation preserves cardiac function after infarction by infarct stabilization: Augmentation by stem cell factor  Shafie Fazel, MD, MSc, Liwen.
Bone marrow–derived mononuclear cell transplantation improves myocardial recovery by enhancing cellular recruitment and differentiation at the infarction.
Changfa Guo, MD, Husnain Kh. Haider, PhD, Winston S. N
Changfa Guo, MD, Husnain Kh
Persistence of marrow stromal cells implanted into acutely infarcted myocardium: Observations in a xenotransplant model  Derek J. MacDonald, MD, Jun Luo,
Building a bioartificial heart: A 3-song saga
A novel vascularized patch enhances cell survival and modifies ventricular remodeling in a rat myocardial infarction model  Qi Zhou, MD, PhD, Jian-Ye.
Myocardial regeneration for chronic heart failure: Not as easy as it sounds  Richard A. Hopkins, MD  The Journal of Thoracic and Cardiovascular Surgery 
Aging impairs the angiogenic response to ischemic injury and the activity of implanted cells: Combined consequences for cell therapy in older recipients 
Preventing cardiac remodeling: The combination of cell-based therapy and cardiac support therapy preserves left ventricular function in rodent model of.
Targeted overexpression of leukemia inhibitory factor to preserve myocardium in a rat model of postinfarction heart failure  Mark F. Berry, MD, Timothy.
Reversible pulmonary trunk banding III: Assessment of myocardial adaptive mechanisms—contribution of cell proliferation  Maria C.D. Abduch, DVM, PhD,
CCR3- and CXCR4-mediated interactions regulate migration of CD34+ human bone marrow progenitors to ischemic myocardium and subsequent tissue repair  N.
Vascular endothelial growth factor transgene expression in cell-transplanted hearts  Terrence M. Yau, MD, MSc, Guangming Li, MD, Richard D Weisel, MD,
Tissue-engineered, hydrogel-based endothelial progenitor cell therapy robustly revascularizes ischemic myocardium and preserves ventricular function 
J. Bonatti, MD, T. Schachner, MD, N. Bonaros, MD, A. Öhlinger, MD, M
Left ventricular unloading before reperfusion reduces endothelin-1 release and calcium overload in porcine myocardial infarction  Sophie Tamareille, PhD,
Right ventricular failure secondary to chronic overload in congenital heart diseases: Benefits of cell therapy using human embryonic stem cell–derived.
Therapeutic Potential of Human Umbilical Cord Derived Stem Cells in a Rat Myocardial Infarction Model  Kai Hong Wu, MD, PhD, Bin Zhou, PhD, Cun Tao Yu,
Renoprotective immunosuppression by pioglitazone with low-dose cyclosporine in rat heart transplantation  Yosuke Tanaka, MD, Tomomi Hasegawa, MD, PhD,
Improvement of cardiac function in the failing rat heart after transfer of skeletal myoblasts engineered to overexpress placental growth factor  Matthias.
The lord of the rings  Antonio Miceli, MD, PhD 
Novel regenerative therapy using cell-sheet covered with omentum flap delivers a huge number of cells in a porcine myocardial infarction model  Yasuhiro.
Myocyte apoptosis occurs early during the development of pressure-overload hypertrophy in infant myocardium  Yeong-Hoon Choi, MD, Douglas B. Cowan, PhD,
Anish Krishnan, BSc, Douglas Drak, BSc, Shisan Bao, PhD, David S
Volume 12, Issue 6, Pages (December 2005)
Impairment of human cell–based vasculogenesis in rats by hypercholesterolemia- induced endothelial dysfunction and rescue with l-arginine supplementation 
A first start for lung transplantation?
Robotic minimally invasive cell transplantation for heart failure
Cellular therapy reverses myocardial dysfunction
Xianyao Xu, MS, Jennifer L. Philip, MD, Md
Layered implantation of myoblast sheets attenuates adverse cardiac remodeling of the infarcted heart  Naosumi Sekiya, MD, Goro Matsumiya, MD, PhD, Shigeru.
Restoration of left ventricular geometry and improvement of left ventricular function in a rodent model of chronic ischemic cardiomyopathy  Jiashing Yu,
Association of electrostimulation with cell transplantation in ischemic heart disease  Abdel Shafy, MD, Thomas Lavergne, MD, Christian Latremouille, MD,
Niv Ad, MD, Lawrence M. Wei, MD 
Prevascularization with gelatin microspheres containing basic fibroblast growth factor enhances the benefits of cardiomyocyte transplantation  Yutaka.
Grafted skeletal myoblast sheets attenuate myocardial remodeling in pacing-induced canine heart failure model  Hiroki Hata, MD, Goro Matsumiya, MD, PhD,
Volume 21, Issue 3, Pages (March 2013)
Volume 19, Issue 4, Pages (April 2011)
Passing the torch The Journal of Thoracic and Cardiovascular Surgery
Zhi Cui, MSc, Ren-Ke Li, MD, PhD 
Myoblast-seeded biodegradable scaffolds to prevent post–myocardial infarction evolution toward heart failure  Matthias Siepe, MD, Marie-Noëlle Giraud,
The Journal of Thoracic and Cardiovascular Surgery
Discussion The Journal of Thoracic and Cardiovascular Surgery
The future of cardiac surgery training: A survival guide
Optimized ventricular restraint therapy: Adjustable restraint is superior to standard restraint in an ovine model of ischemic cardiomyopathy  Lawrence.
Implantation of fetal rat lung fragments into bleomycin-induced pulmonary fibrosis  Hiroaki Toba, MD, Shoji Sakiyama, MD, PhD, Koichiro Kenzaki, MD, PhD,
Volume 20, Issue 1, Pages (January 2012)
Optimizing cardiac cell therapy: From processing to delivery
Stromal cell-derived factor and granulocyte-monocyte colony-stimulating factor form a combined neovasculogenic therapy for ischemic cardiomyopathy  Y.
Impact of preoperative left ventricular function and time from infarction on the long-term benefits after intramyocardial CD133+ bone marrow stem cell.
Prolonged mechanical unloading preserves myocardial contractility but impairs relaxation in rat heart of dilated cardiomyopathy accompanied by myocardial.
Presentation transcript:

Combined transplantation of skeletal myoblasts and angiopoietic progenitor cells reduces infarct size and apoptosis and improves cardiac function in chronic ischemic heart failure  Nikolaos Bonaros, MD, Rauend Rauf, MD, Dominik Wolf, MD, Eva Margreiter, PhD, Alexandar Tzankov, MD, Bernhard Schlechta, MD, Alfred Kocher, MD, Harald Ott, MD, Thomas Schachner, MD, Steffen Hering, PhD, Johannes Bonatti, MD, Guenther Laufer, MD  The Journal of Thoracic and Cardiovascular Surgery  Volume 132, Issue 6, Pages 1321-1328.e2 (December 2006) DOI: 10.1016/j.jtcvs.2006.07.023 Copyright © 2006 The American Association for Thoracic Surgery Terms and Conditions

Figure 1 Transplanted skeletal myoblasts and AC-133+ angiopoietic progenitors survive in chronic ischemic myocardium. A, Fluorescent staining of YFP-labeled skeletal myoblasts (green) in the infarct area detected 4 weeks after cell injection (40× magnification). B, Immunohistochemical staining of skeletal myoblasts by My-32 antibody confirms fluorescence-based cell detection in the infarct scar (40× magnification). C, Immunofluorescent staining of My-32 (red) showing stable myotube formation in the scar (cell nuclei stained blue) (100× magnification). D, Fluorescent staining of DiI-labeled AC-133+ cells (red) in the border zone 4 weeks after cell transplantation (40× magnification). E, Fluorescent staining of skeletal myoblasts (green) and AC-133+ cells (red) in the infarct area 4 weeks after combined cell transplantation of skeletal myoblasts and AC-133+ cells in the scar and border zone, respectively (100× magnification). The Journal of Thoracic and Cardiovascular Surgery 2006 132, 1321-1328.e2DOI: (10.1016/j.jtcvs.2006.07.023) Copyright © 2006 The American Association for Thoracic Surgery Terms and Conditions

Figure 2 Combined transplantation of skeletal myoblasts and AC-133+ progenitors reverses left ventricular dilatation (A) and improves left ventricular function (B), as assessed by transthoracic echocardiography 4 weeks after cell transplantation (results expressed as mean ± standard deviation). M-Mode echocardiography, i., 2 days after myocardial infarction; ii., 4 weeks after double-cell therapy. Animals received Roswell Park Memorial Institute injections in the scar and peri-infarct rim (control), homologous skeletal myoblasts in the scar (SM), human-derived AC-133+ cells in the peri-infarct rim (SC), homologous skeletal myoblasts and human-derived AC-133+ cells (Comb) in the scar and peri-infarct rim, respectively. Note anterior wall hypokinesia and left ventricular dilatation after myocardial infarction (arrow). Both findings are partially reversed 4 weeks after combined cell therapy (arrow). The Journal of Thoracic and Cardiovascular Surgery 2006 132, 1321-1328.e2DOI: (10.1016/j.jtcvs.2006.07.023) Copyright © 2006 The American Association for Thoracic Surgery Terms and Conditions

Figure 3 Combined transplantation of skeletal myoblasts and angiopoietic progenitors prevents myocardial fibrosis and results in reduced myocardial scar, as assessed by Masson’s Trichrome staining 8 weeks after myocardial infarction. A, Scar area of a control animal 4 weeks after injection of culture medium (40× magnification). B, Scar area of an animal after myocardial infarction and 4 weeks after skeletal myoblasts transplantation (40× magnification). C, Quantification of myocardial fibrosis 4 weeks after injection of culture medium (control), homologous skeletal myoblasts (SM), human-derived AC-133+ cells (SC), or combination of homologous skeletal myoblasts and human-derived AC-133+ cells. The Journal of Thoracic and Cardiovascular Surgery 2006 132, 1321-1328.e2DOI: (10.1016/j.jtcvs.2006.07.023) Copyright © 2006 The American Association for Thoracic Surgery Terms and Conditions

Figure 4 Double-cell therapy with skeletal myoblasts and AC-133+ cells attenuates apoptosis in the border zone and distal area, as assessed by TUNEL staining. TUNEL staining of the border zone 4 weeks after injections of (A) culture medium and (B) combination of homologous skeletal myoblasts and human-derived AC-133+ cells (brown and blue nuclei indicating apoptotic and non-apoptotic cells, respectively). Quantification of apoptotic index, calculated as the ratio of apoptotic cardiomyocytes in the total myocardial cell population in the border zone (C) and distal area (D). Results are expressed as mean ± standard deviation. The Journal of Thoracic and Cardiovascular Surgery 2006 132, 1321-1328.e2DOI: (10.1016/j.jtcvs.2006.07.023) Copyright © 2006 The American Association for Thoracic Surgery Terms and Conditions

Figure 5 Degree of neovascularization in the peri-infarct area of chronic infarcted myocardium after injection of (A) culture medium and (B) combination of skeletal myoblasts and AC-133+ cells. Quantification of capillary density in the border zone (C) and scar (D) 4 weeks after cell transplantation. The Journal of Thoracic and Cardiovascular Surgery 2006 132, 1321-1328.e2DOI: (10.1016/j.jtcvs.2006.07.023) Copyright © 2006 The American Association for Thoracic Surgery Terms and Conditions

Figures E1 and E2 M-Mode echocardiography before myocardial infarction (E1) and 4 weeks after cell monotherapy (E2). Note anterior wall movement and normal dimensions of the left ventricle before myocardial infarction (arrow, Figure E1) and 4 weeks after cell monotherapy (arrow, Figure E2). The Journal of Thoracic and Cardiovascular Surgery 2006 132, 1321-1328.e2DOI: (10.1016/j.jtcvs.2006.07.023) Copyright © 2006 The American Association for Thoracic Surgery Terms and Conditions

Figures E3 and E4 Double-cell therapy with skeletal myoblasts and AC-133+ cells attenuates apoptosis in the border zone and distal area, as assessed by TUNEL staining. TUNEL staining of the border zone 4 weeks after injections of homologous skeletal myoblasts (E3) and human-derived AC-133+ cells (E4) (brown and blue nuclei indicating apoptotic and non-apoptotic cells, respectively). The Journal of Thoracic and Cardiovascular Surgery 2006 132, 1321-1328.e2DOI: (10.1016/j.jtcvs.2006.07.023) Copyright © 2006 The American Association for Thoracic Surgery Terms and Conditions

Figures E5 and E6 Degree of neovascularization in the peri-infarct area of chronic infarcted myocardium after injection of skeletal myoblasts (E5) and AC-133+ cells (E6) as assessed by means of CD34 staining of endothelial cells. The Journal of Thoracic and Cardiovascular Surgery 2006 132, 1321-1328.e2DOI: (10.1016/j.jtcvs.2006.07.023) Copyright © 2006 The American Association for Thoracic Surgery Terms and Conditions