[HTML][HTML] A universal system for highly efficient cardiac differentiation of human induced pluripotent stem cells that eliminates interline variability

PW Burridge, S Thompson, MA Millrod, S Weinberg… - PloS one, 2011 - journals.plos.org
PW Burridge, S Thompson, MA Millrod, S Weinberg, X Yuan, A Peters, V Mahairaki…
PloS one, 2011journals.plos.org
Background The production of cardiomyocytes from human induced pluripotent stem cells
(hiPSC) holds great promise for patient-specific cardiotoxicity drug testing, disease
modeling, and cardiac regeneration. However, existing protocols for the differentiation of
hiPSC to the cardiac lineage are inefficient and highly variable. We describe a highly
efficient system for differentiation of human embryonic stem cells (hESC) and hiPSC to the
cardiac lineage. This system eliminated the variability in cardiac differentiation capacity of a …
Background
The production of cardiomyocytes from human induced pluripotent stem cells (hiPSC) holds great promise for patient-specific cardiotoxicity drug testing, disease modeling, and cardiac regeneration. However, existing protocols for the differentiation of hiPSC to the cardiac lineage are inefficient and highly variable. We describe a highly efficient system for differentiation of human embryonic stem cells (hESC) and hiPSC to the cardiac lineage. This system eliminated the variability in cardiac differentiation capacity of a variety of human pluripotent stem cells (hPSC), including hiPSC generated from CD34+ cord blood using non-viral, non-integrating methods.
Methodology/Principal Findings
We systematically and rigorously optimized >45 experimental variables to develop a universal cardiac differentiation system that produced contracting human embryoid bodies (hEB) with an improved efficiency of 94.7±2.4% in an accelerated nine days from four hESC and seven hiPSC lines tested, including hiPSC derived from neonatal CD34+ cord blood and adult fibroblasts using non-integrating episomal plasmids. This cost-effective differentiation method employed forced aggregation hEB formation in a chemically defined medium, along with staged exposure to physiological (5%) oxygen, and optimized concentrations of mesodermal morphogens BMP4 and FGF2, polyvinyl alcohol, serum, and insulin. The contracting hEB derived using these methods were composed of high percentages (64–89%) of cardiac troponin I+ cells that displayed ultrastructural properties of functional cardiomyocytes and uniform electrophysiological profiles responsive to cardioactive drugs.
Conclusion/Significance
This efficient and cost-effective universal system for cardiac differentiation of hiPSC allows a potentially unlimited production of functional cardiomyocytes suitable for application to hPSC-based drug development, cardiac disease modeling, and the future generation of clinically-safe nonviral human cardiac cells for regenerative medicine.
PLOS