Neuregulin/ErbB signaling regulates cardiac subtype specification in differentiating human embryonic stem cells

WZ Zhu, Y Xie, KW Moyes, JD Gold, B Askari… - Circulation …, 2010 - Am Heart Assoc
WZ Zhu, Y Xie, KW Moyes, JD Gold, B Askari, MA Laflamme
Circulation research, 2010Am Heart Assoc
Rationale: Human embryonic stem cell–derived cardiomyocytes (hESC-CMs) exhibit either
a “working” chamber or a nodal-like phenotype. To generate optimal hESC-CM preparations
for eventual clinical application in cell-based therapies, we will need to control their
differentiation into these specialized cardiac subtypes. Objective: To demonstrate intact
neuregulin (NRG)-1β/ErbB signaling in hESC-CMs and test the hypothesis that this
signaling pathway regulates cardiac subtype abundance in hESC-CM cultures. Methods …
Rationale:
Human embryonic stem cell–derived cardiomyocytes (hESC-CMs) exhibit either a “working” chamber or a nodal-like phenotype. To generate optimal hESC-CM preparations for eventual clinical application in cell-based therapies, we will need to control their differentiation into these specialized cardiac subtypes.
Objective:
To demonstrate intact neuregulin (NRG)-1β/ErbB signaling in hESC-CMs and test the hypothesis that this signaling pathway regulates cardiac subtype abundance in hESC-CM cultures.
Methods and Results:
All experiments used hESC-CM cultures generated using our recently reported directed differentiation protocol. To support subsequent action potential phenotyping approaches and provide a higher-throughput method of determining cardiac subtype, we first developed and validated a novel genetic label that identifies nodal-type hESC-CMs. Next, control hESC-CM preparations were compared to those differentiated in the presence of exogenous NRG-1β, an anti–NRG-1β neutralizing antibody, or the ErbB antagonist AG1478. We used 3 independent approaches to determine the ratio of cardiac subtypes in the resultant populations: direct action potential phenotyping under current-clamp, activation of the aforementioned genetic label, and subtype-specific marker expression by RT-PCR. Using all 3 end points, we found that inhibition of NRG-1β/ErbB signaling greatly enhanced the proportion of cells showing the nodal phenotype.
Conclusions:
NRG-1β/ErbB signaling regulates the ratio of nodal- to working-type cells in differentiating hESC-CM cultures and presumably functions similarly during early human heart development. We speculate that, by manipulating NRG-1β/ErbB signaling, it will be possible to generate preparations of enriched working-type myocytes for infarct repair, or, conversely, nodal cells for potential use in a biological pacemaker.
Am Heart Assoc