Microfluidic Single-Cell Analysis Shows That Porcine Induced Pluripotent Stem Cell–Derived Endothelial Cells Improve Myocardial Function by Paracrine Activation

Author:

Gu Mingxia1,Nguyen Patricia K.1,Lee Andrew S.1,Xu Dan1,Hu Shijun1,Plews Jordan R.1,Han Leng1,Huber Bruno C.1,Lee Won Hee1,Gong Yongquan1,de Almeida Patricia E.1,Lyons Jennifer1,Ikeno Fumi1,Pacharinsak Cholawat1,Connolly Andrew J.1,Gambhir Sanjiv S.1,Robbins Robert C.1,Longaker Michael T.1,Wu Joseph C.1

Affiliation:

1. From the Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA (M.G., P.K.N., A.S.L., D.X., S.H., J.R.P., L.H., B.C.H., W.H.L., Y.G., P.E.d.A., J.L., F.I., J.C.W.); the Department of Radiology, Molecular Imaging Program, Stanford University, Stanford, CA (A.S.L., D.X., S.H., J.R.P., L.H., B.C.H., W.H.L., Y.G., P.E.d.A., S.S.G., J.C.W.); the Department of Physiology and Pathophysiology, Key Laboratory of Molecular Cardiovascular Sciences of Education...

Abstract

Rationale: Induced pluripotent stem cells (iPSCs) hold great promise for the development of patient-specific therapies for cardiovascular disease. However, clinical translation will require preclinical optimization and validation of large-animal iPSC models. Objective: To successfully derive endothelial cells from porcine iPSCs and demonstrate their potential utility for the treatment of myocardial ischemia. Methods and Results: Porcine adipose stromal cells were reprogrammed to generate porcine iPSCs (piPSCs). Immunohistochemistry, quantitative PCR, microarray hybridization, and angiogenic assays confirmed that piPSC-derived endothelial cells (piPSC-ECs) shared similar morphological and functional properties as endothelial cells isolated from the autologous pig aorta. To demonstrate their therapeutic potential, piPSC-ECs were transplanted into mice with myocardial infarction. Compared with control, animals transplanted with piPSC-ECs showed significant functional improvement measured by echocardiography (fractional shortening at week 4: 27.2±1.3% versus 22.3±1.1%; P <0.001) and MRI (ejection fraction at week 4: 45.8±1.3% versus 42.3±0.9%; P <0.05). Quantitative protein assays and microfluidic single-cell PCR profiling showed that piPSC-ECs released proangiogenic and antiapoptotic factors in the ischemic microenvironment, which promoted neovascularization and cardiomyocyte survival, respectively. Release of paracrine factors varied significantly among subpopulations of transplanted cells, suggesting that transplantation of specific cell populations may result in greater functional recovery. Conclusions: In summary, this is the first study to successfully differentiate piPSCs-ECs from piPSCs and demonstrate that transplantation of piPSC-ECs improved cardiac function after myocardial infarction via paracrine activation. Further development of these large animal iPSC models will yield significant insights into their therapeutic potential and accelerate the clinical translation of autologous iPSC-based therapy.

Publisher

Ovid Technologies (Wolters Kluwer Health)

Subject

Cardiology and Cardiovascular Medicine,Physiology

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3