HOMEPRODUCTSCOMPANYCONTACTFAQResearchDictionaryPharmaSign Up FREE or Login

Microfluidic single-cell analysis shows that porcine induced pluripotent stem cell-derived endothelial cells improve myocardial function by paracrine activation.

AbstractRATIONALE:
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.
AuthorsMingxia Gu, Patricia K Nguyen, Andrew S Lee, Dan Xu, Shijun Hu, Jordan R Plews, Leng Han, Bruno C Huber, Won Hee Lee, Yongquan Gong, Patricia E de Almeida, Jennifer Lyons, Fumi Ikeno, Cholawat Pacharinsak, Andrew J Connolly, Sanjiv S Gambhir, Robert C Robbins, Michael T Longaker, Joseph C Wu
JournalCirculation research (Circ Res) Vol. 111 Issue 7 Pg. 882-93 (Sep 14 2012) ISSN: 1524-4571 [Electronic] United States
PMID22821929 (Publication Type: Journal Article, Research Support, N.I.H., Extramural, Research Support, Non-U.S. Gov't)
Topics
  • Animals
  • Cell Differentiation (physiology)
  • Cell Survival (physiology)
  • Cell Transplantation
  • Cells, Cultured
  • Echocardiography
  • Endothelium, Vascular (cytology, physiology, transplantation)
  • Female
  • Heart (physiopathology)
  • Magnetic Resonance Imaging
  • Mice
  • Mice, SCID
  • Microfluidic Analytical Techniques
  • Models, Animal
  • Myocardial Infarction (pathology, physiopathology, therapy)
  • Myocytes, Cardiac (pathology)
  • Neovascularization, Physiologic
  • Paracrine Communication (physiology)
  • Pluripotent Stem Cells (cytology, physiology)
  • Swine
  • Swine, Miniature

Join CureHunter, for free Research Interface BASIC access!

Take advantage of free CureHunter research engine access to explore the best drug and treatment options for any disease. Find out why thousands of doctors, pharma researchers and patient activists around the world use CureHunter every day.
Realize the full power of the drug-disease research graph!


Choose Username:
Email:
Password:
Verify Password:
Enter Code Shown: