Summary

Establishing a Swine Model of Post-myocardial Infarction Heart Failure for Stem Cell Treatment

Published: May 25, 2020
doi:

Summary

We sought to establish a swine model of heart failure induced by left circumflex artery blockage and rapid pacing to test the effect and safety of intramyocardial administration of stem cells for cell-based therapies.

Abstract

Although advances have been achieved in the treatment of heart failure (HF) following myocardial infarction (MI), HF following MI remains one of the major causes of mortality and morbidity around the world. Cell-based therapies for cardiac repair and improvement of left ventricular function after MI have attracted considerable attention. Accordingly, the safety and efficacy of these cell transplantations should be tested in a preclinical large animal model of HF prior to clinical use. Pigs are widely used for cardiovascular disease research due to their similarity to humans in terms of heart size and coronary anatomy. Therefore, we sought to present an effective protocol for the establishment of a porcine chronic HF model using closed-chest coronary balloon occlusion of the left circumflex artery (LCX), followed by rapid ventricular pacing induced with pacemaker implantation. Eight weeks later, the stem cells were administered by intramyocardial injection in the peri-infarct area. Then the infarct size, cell survival, and left ventricular function (including echocardiography, hemodynamic parameters, and electrophysiology) were evaluated. This study helps establish a stable preclinical large animal HF model for stem cell treatment.

Introduction

Cardiovascular diseases, coronary artery disease (CAD) in particular, remain the major cause of morbidity and mortality in Hong Kong and worldwide1. In Hong Kong, a 26% increase from 2012 to 2017 of the number of CAD patients treated under the Hospital Authority was projected2. Among all CADs, acute myocardial infarction (MI) is a leading cause of death and subsequent complications, such as heart failure (HF). These contribute to significant medical, social, and financial burdens. In patients with MI, thrombolytic therapy or primary percutaneous coronary intervention (PCI) is an effective therapy in preserving life, but these therapies can only reduce cardiomyocyte (CM) loss during MI. The treatments available are unable to replenish the permanent loss of CMs, which leads to cardiac fibrosis, myocardial remodeling, cardiac arrhythmia, and eventually heart failure. The mortality rate at 1-year post-MI is around 7% with more than 20% patients developing HF3. In end-stage HF patients, heart transplantation is the only available effective therapy, but it is limited by a shortage of available organs. Novel therapies are necessary to reverse the development of post-MI HF. As a result, cell-based therapy is considered an attractive approach to repair the impaired CMs and ameliorate left ventricular (LV) function in HF following MI. Our previous studies found stem cell transplantation to be beneficial for heart function improvement after direct intramyocardial transplantation in small animal models of MI4,5. Standardized preclinical large animal HF protocols are thus needed to further test the efficacy and safety of stem cell transplantation before clinical use.

Recent decades have witnessed the widespread use of pigs in cardiovascular research for stem cell therapy. HF pigs are a promising model of translational research due to their similarity to humans in terms of cardiac size, weight, rhythm, function, and coronary artery anatomy. Moreover, porcine HF models can mimic post-MI HF patients in terms of CM metabolism, electrophysiological properties, and neuroendocrine changes under ischemic conditions6. The protocol presented here uses such a standardized pig HF model, employing a closed-chest coronary balloon occlusion of the left circumflex artery (LCX) followed by rapid pacing induced by pacemaker implantation. The study also optimizes the route of intramyocardial administration of stem cells for the treatment of post-MI HF. The purpose is to produce a porcine animal model of chronic myocardial infarction that can be used to develop treatments that are clinically relevant for patients with severe CAD.

Protocol

All animal experiments were performed in accordance with the Guide for the Care and Use of Laboratory Animals published by the US National Institutes of Health and regulations of the University of Hong Kong, and the protocol was approved by the Committee on the Use of Live Animals in Teaching and Research (CULTAR) at the University of Hong Kong. NOTE: Female farm pigs weighing 35-40 kg (9-12 months old) were used for this study. The flowchart of this experiment is shown in <strong class="xfig"…

Representative Results

Mortality A total of 24 pigs were used in this study. Three of them died during MI induction because of sustained VT. One animal died in the open-heart surgery for cell injection because of wound bleeding. Two animals died because of severe infection. Two animals were excluded because of slight EF reduction (LVEF reduction > 40% of baseline). As a result, 16 animals completed the whole study protocol. …

Discussion

Standard animal models are of paramount importance to understand the pathophysiology and mechanisms of diseases and test novel therapeutics. Our protocol establishes a porcine model of HF induced by left circumflex artery blockage and rapid pacing. Eight weeks after the induction of MI, the animals developed significant impairment of LVEF, LVEDD, LVESD, +dP/dt, and ESPVR. This protocol also tests the administration method of stem cell therapy for heart regeneration by intramyocardial injection. The infarct size, and card…

Divulgaciones

The authors have nothing to disclose.

Acknowledgements

The authors acknowledge Alfreda and Kung Tak Chung for their excellent technical support during the animal experiments.

Materials

Amiodarone Mylan
Anaesthetic machines and respirator Drager Fabius plus XL
Angiocath Becton Dickinson 381147
Anti-human nuclear antigen abcam ab19118
Axio Plus image capturing system Zeiss Axioskop 2 PLUS Axioskop 2 plus
AxioVision Rel. 4.5 software Zeiss
Baytril Bayer enrofloxacin
Betadine Mundipharma
CardioLab Electrophysiology Recording Systems GE Healthcare G220f
Culture media MesenCult 05420
Cyclosporine Novartis
Defibrillator GE Healthcare CardioServ
Dorminal TEVA
Echocardiographic system GE Vingmed Vivid i
EchoPac software GE Vingmed
Electrophysiological catheter Cordis Corp
Embozene Microsphere Boston Scientific 17020-S1 700 μm
Endotracheal tube Vet Care VCPET70PCW Size 7
Ethanol VWR chemicals 20821.33
Formalin Sigma HT501320 10%
IVC balloon Dilatation Catheter Boston Scientific 3917112041 Mustang
JR4 guiding catheter Cordis Corp 67208200 6F
Lidocaine Quala
Mersilk Ethicon W584 2-0
Metoprolol succinate Wockhardt
Microtome Leica RM2125RT
Mobile C arm fluoroscopy equipment GE Healthcare OEC 9900 Elite
Pacemaker St Jude Medical PM1272 Assurity MRI pacemaker
Pacemaker generator St Jude Medical Merlln model 3330
Pressure-volume catheter CD Leycom CA-71103-PL 7F
Pressure–volume signal processor CD Leycom SIGMA-M
Programmable Stimulator Medtronic Inc 5328
PTCA Dilatation balloon Catheter Boston Scientific H7493919120250 MAVERICK over the wire
Ramipril TEVA
Sheath introducer Cordis Corp 504608X 8F, 9F, 12F
Steroid Versus Arthritis
Temgesic Nindivior buprenorphine
Venous indwelling needle TERUMO SR+OX2225C 22G
Vicryl Ethicon VCP320H 2-0
Xylazine Alfasan International B.V.
Zoletil Virbac New Zealand Limited tiletamine+zolezepam

Referencias

  1. Mozaffarian, D., et al. Heart disease and stroke statistics-2015 update: a report from the American Heart Association. Circulation. 131, e29 (2015).
  2. Hospital Authority. . Hospital Authority Statistical Report 2013. , (2013).
  3. Cung, T. T., et al. Cyclosporine before PCI in Patients with Acute Myocardial Infarction. The New England Journal of Medicine. 373 (11), 1021-1031 (2015).
  4. Liao, S. Y., et al. Proarrhythmic risk of embryonic stem cell-derived cardiomyocyte transplantation in infarcted myocardium. Heart Rhythm. 7, 1852-1859 (2010).
  5. Liao, S. Y., et al. Overexpression of Kir2.1 channel in embryonic stem cell-derived cardiomyocytes attenuates posttransplantation proarrhythmic risk in myocardial infarction. Heart Rhythm. 10, 273-282 (2013).
  6. Liu, Y., et al. Thoracic spinal cord stimulation improves cardiac contractile function and myocardial oxygen consumption in a porcine model of ischemic heart failure. Journal of Cardiovascular Electrophysiology. 23, 534-540 (2012).
  7. Liao, S. Y., et al. Improvement of Myocardial Function Following Catheter-Based Renal Denervation in Heart Failure. JACC: Basic to Translational Science. 2 (3), 270-281 (2017).
  8. Liao, S. Y., et al. Remodelling of cardiac sympathetic re-innervation with thoracic spinal cord stimulation improves left ventricular function in a porcine model of heart failure. Europace. 17 (12), 1875-1883 (2015).
  9. Daehnert, I., Rotzsch, C., Wiener, M., Schneider, P. Rapid right ventricular pacing is an alternative to adenosine in catheter interventional procedures for congenital heart disease. Heart. 90 (9), 1047-1050 (2004).
  10. Hála, P., et al. Tachycardia-Induced Cardiomyopathy as a Chronic Heart Failure Model in Swine. Journal of Visualized Experiments. (132), e57030 (2018).
  11. Santoso, T., et al. Endomyocardial implantation of autologous bone marrow mononuclear cells in advanced ischemic heart failure: a randomized placebo-controlled trial (END-HF). Journal of Cardiovascular Translational Research. 7, 545-552 (2014).
  12. Traverse, J. H., et al. Cardiovascular Cell Therapy Research Network. Effect of intracoronary delivery of autologous bone marrow mononuclear cells 2 to 3 weeks following acute myocardial infarction on left ventricular function: the LateTIME randomized trial. Journal of the American Medical Association. 306, 2110-2119 (2011).
  13. Traverse, J. H., et al. Cardiovascular Cell Therapy Research Network (CCTRN). Effect of the use and timing of bone marrow mononuclear cell delivery on left ventricular function after acute myocardial infarction: the TIME randomized trial. Journal of the American Medical Association. 308, 2380-2389 (2012).
  14. de Jong, R., Houtgraaf, J. H., Samiei, S., Boersma, E., Duckers, H. J. Intracoronary stem cell infusion after myocardial infarction. A meta-analysis and update on clinical trials. Circulation: Cardiovascular Interventions. 7, 156-167 (2014).
  15. Nowbar, A. N., et al. DAMASCENE writing group. Discrepancies in autologous bone marrow stem cell trials and enhancement of ejection fraction (DAMASCENE): weighted regression and meta-analysis. British Medical Journal. 348, g2688 (2014).
  16. Kanelidis, A. J., Premer, C., Lopez, J., Balkan, W., Hare, J. M. Route of Delivery Modulates the Efficacy of Mesenchymal Stem Cell Therapy for Myocardial Infarction: A Meta-Analysis of Preclinical Studies and Clinical Trials. Circulation Research. 120 (7), 1139-1150 (2017).
  17. Hou, D., et al. Radiolabeled cell distribution after intramyocardial, intracoronary, and interstitial retrograde coronary venous delivery: implications for current clinical trials. Circulation. 112 (9 Suppl), I150-I156 (2005).
  18. Hu, X., et al. A Large-Scale Investigation of Hypoxia-Preconditioned Allogeneic Mesenchymal Stem Cells for Myocardial Repair in Nonhuman Primates: Paracrine Activity Without Remuscularization. Circulation Research. 118, 970-983 (2016).
  19. Chong, J. J., et al. Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts. Nature. 510, 273-277 (2014).
  20. Martens, A., et al. Substantial early loss of induced pluripotent stem cells following transplantation in myocardial infarction. Artificial Organs. 38, 978-984 (2014).
  21. Shiba, Y., et al. Allogeneic transplantation of iPS cell-derived cardiomyocytes regenerates primate hearts. Nature. 538, 388-391 (2016).
check_url/es/60392?article_type=t

Play Video

Citar este artículo
Sun, S., Jiang, Y., Zhen, Z., Lai, W., Liao, S., Tse, H. Establishing a Swine Model of Post-myocardial Infarction Heart Failure for Stem Cell Treatment. J. Vis. Exp. (159), e60392, doi:10.3791/60392 (2020).

View Video