Proteasome-dependent regulation of distinct metabolic states during long-term culture of human iPSC-derived cardiomyocytes

A Ebert, AU Joshi, S Andorf, Y Dai… - Circulation …, 2019 - Am Heart Assoc
A Ebert, AU Joshi, S Andorf, Y Dai, S Sampathkumar, H Chen, Y Li, P Garg, K Toischer…
Circulation research, 2019Am Heart Assoc
Rationale: The immature presentation of human induced pluripotent stem cell–derived
cardiomyocytes (iPSC-CMs) is currently a challenge for their application in disease
modeling, drug screening, and regenerative medicine. Long-term culture is known to
achieve partial maturation of iPSC-CMs. However, little is known about the molecular
signaling circuitries that govern functional changes, metabolic output, and cellular
homeostasis during long-term culture of iPSC-CMs. Objective: We aimed to identify and …
Rationale:
The immature presentation of human induced pluripotent stem cell–derived cardiomyocytes (iPSC-CMs) is currently a challenge for their application in disease modeling, drug screening, and regenerative medicine. Long-term culture is known to achieve partial maturation of iPSC-CMs. However, little is known about the molecular signaling circuitries that govern functional changes, metabolic output, and cellular homeostasis during long-term culture of iPSC-CMs.
Objective:
We aimed to identify and characterize critical signaling events that control functional and metabolic transitions of cardiac cells during developmental progression, as recapitulated by long-term culture of iPSC-CMs.
Methods and Results:
We combined transcriptomic sequencing with pathway network mapping in iPSC-CMs that were cultured until a late time point, day 200, in comparison to a medium time point, day 90, and an early time point, day 30. Transcriptomic landscapes of long-term cultured iPSC-CMs allowed mapping of distinct metabolic stages during development of maturing iPSC-CMs. Temporally divergent control of mitochondrial metabolism was found to be regulated by cAMP/PKA (protein kinase A)- and proteasome-dependent signaling events. The PKA/proteasome-dependent signaling cascade was mediated downstream by Hsp90 (heat shock protein 90), which in turn modulated mitochondrial respiratory chain proteins and their metabolic output. During long-term culture, this circuitry was found to initiate upregulation of iPSC-CM metabolism, resulting in increased cell contractility that reached a maximum at the day 200 time point.
Conclusions:
Our results reveal a PKA/proteasome- and Hsp90-dependent signaling pathway that regulates mitochondrial respiratory chain proteins and determines cardiomyocyte energy production and functional output. These findings provide deeper insight into signaling circuitries governing metabolic homeostasis in iPSC-CMs during developmental progression.
Am Heart Assoc