1. Academic Validation
  2. Inhibition of stearoyl-coA desaturase selectively eliminates tumorigenic Nanog-positive cells: improving the safety of iPS cell transplantation to myocardium

Inhibition of stearoyl-coA desaturase selectively eliminates tumorigenic Nanog-positive cells: improving the safety of iPS cell transplantation to myocardium

  • Cell Cycle. 2014;13(5):762-71. doi: 10.4161/cc.27677.
Lan Zhang 1 Yaohua Pan 2 Gangjian Qin 3 Lijuan Chen 4 Tapan K Chatterjee 5 Neal L Weintraub 5 Yaoliang Tang 5
Affiliations

Affiliations

  • 1 Department of Vascular Surgery; Renji Hospital; School of Medicine; Shanghai Jiaotong University; Shanghai, China; Vascular Biology Center; Department of Medicine; Medical College of Georgia/Georgia Regents University; Augusta, GA USA.
  • 2 Department of Neurosurgery; Renji Hospital; School of Medicine; Shanghai Jiaotong University; Shanghai, China.
  • 3 Feinberg Cardiovascular Research Institute; Department of Medicine-Cardiology; Northwestern University Feinberg School of Medicine; Chicago, IL USA.
  • 4 Department of Cardiology; Zhongda Hospital; Medical School of Southeast University; Nanjing, China.
  • 5 Vascular Biology Center; Department of Medicine; Medical College of Georgia/Georgia Regents University; Augusta, GA USA.
Abstract

Induced pluripotent stem cells (iPS) can differentiate into cardiomyocytes (CM) and represent a promising form of cellular therapy for heart regeneration. However, residual undifferentiated iPS derivates (iPSD), which are not fully eliminated by cell differentiation or purification protocols, may form tumors after transplantation, thus compromising therapeutic application. Inhibition of stearoyl-coA desaturase (SCD) has recently been reported to eliminate undifferentiated human embryonic stem cells, which share many features with iPSD. Here, we tested the effects of PluriSin#1, a small-molecule inhibitor of SCD, on iPS-derived CM. We found that plurisin#1 treatment significantly decreased the mRNA and protein level of Nanog, a marker for both cell pluripotency and tumor progression; importantly, we provide evidence that PluriSin#1 treatment at 20 µM for 1 day significantly induces the Apoptosis of Nanog-positive iPSD. In addition, PluriSin#1 treatment at 20 µM for 4 days diminished Nanog-positive stem cells in cultured iPSD while not increasing Apoptosis of iPS-derived CM. To investigate whether PluriSin#1 treatment prevents tumorigenicity of iPSD after cell transplantation, we intramyocardially injected PluriSin#1- or DMSO-treated iPSD in a mouse model of myocardial infarction (MI). DMSO-treated iPSD readily formed Nanog-expressing tumors 2 weeks after injection, which was prevented by treatment with PluriSin#1. Moreover, treatment with PluriSin#1 did not change the expression of cTnI, α-MHC, or MLC-2v, markers of cardiac differentiation (P>0.05, n = 4). Importantly, pluriSin#1-treated iPS-derived CM exhibited the ability to engraft and survive in the infarcted myocardium. We conclude that inhibition of SCD holds the potential to enhance the safety of therapeutic application of iPS cells for heart regeneration.

Keywords

Nanog; PluriSin#1; iPS; myocardial infarction; tumorigenicity.

Figures
Products