Title Microfluidic Single-Cell Analysis Shows That Porcine Induced Pluripotent Stem Cell-Derived Endothelial Cells Improve Myocardial Function by Paracrine Activation
Authors Gu, Mingxia
Nguyen, Patricia K.
Lee, Andrew S.
Xu, Dan
Hu, Shijun
Plews, Jordan R.
Han, Leng
Huber, Bruno C.
Lee, Won Hee
Gong, Yongquan
de Almeida, Patricia E.
Lyons, Jennifer
Ikeno, Fumi
Pacharinsak, Cholawat
Connolly, Andrew J.
Gambhir, Sanjiv S.
Robbins, Robert C.
Longaker, Michael T.
Wu, Joseph C.
Affiliation Stanford Univ, Dept Med, Div Cardiol, Sch Med, Stanford, CA 94305 USA.
Stanford Univ, Mol Imaging Program, Dept Radiol, Stanford, CA 94305 USA.
Peking Univ, Dept Physiol & Pathophysiol, Key Lab Mol Cardiovasc Sci, Educ Minist,Hlth Sci Ctr, Beijing 100871, Peoples R China.
Stanford Univ, Dept Cardiothorac Surg, Stanford, CA 94305 USA.
Stanford Univ, Dept Comparat Med, Stanford, CA 94305 USA.
Stanford Univ, Dept Pathol, Stanford, CA 94305 USA.
Stanford Univ, Dept Plast Surg, Stanford, CA 94305 USA.
Stanford Univ, Inst Stem Cell Biol & Regenerat Med, Stanford, CA 94305 USA.
265 Campus Dr,Room 1120B, Stanford, CA 94305 USA.
Keywords induced pluripotent stem cells
large-animal models
paracrine activation
myocardial infarction
molecular imaging
ischemic heart disease
vascular biology
CARDIAC-FUNCTION
GENE-EXPRESSION
SOMATIC-CELLS
CLINICAL TRANSLATION
INFARCTION MODEL
ISCHEMIC-HEART
GENERATION
THERAPY
DIFFERENTIATION
DERIVATION
Issue Date 2012
Publisher circulation research
Citation CIRCULATION RESEARCH.2012,111,(7),882-893.
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. (Circ Res. 2012;111:882-893.)
URI http://hdl.handle.net/20.500.11897/392808
ISSN 0009-7330
DOI 10.1161/CIRCRESAHA.112.269001
Indexed SCI(E)
Appears in Collections: 分子心血管学教育部重点实验室

Files in This Work
There are no files associated with this item.

Web of Science®


75

Checked on Last Week

Scopus®



Checked on Current Time

百度学术™


0

Checked on Current Time

Google Scholar™





License: See PKU IR operational policies.