Abstract

Pluripotent stem cells are known to shift their mitochondrial metabolism upon differentiation, but the mechanisms underlying such metabolic rewiring are not fully understood. We hypothesized that during differentiation of human induced pluripotent stem cells (hiPSCs), mitochondria undergo mitophagy and are then replenished by the biogenesis of new mitochondria adapted to the metabolic needs of the differentiated cell. To evaluate mitophagy during iPSC differentiation, we performed live cell imaging of mitochondria and lysosomes in hiPSCs differentiating into vascular endothelial cells using confocal microscopy. We observed a burst of mitophagy during the initial phases of hiPSC differentiation into the endothelial lineage, followed by subsequent mitochondrial biogenesis as assessed by the mitochondrial biogenesis biosensor MitoTimer. Furthermore, hiPSCs undergoing differentiation showed greater mitochondrial oxidation of fatty acids and an increase in ATP levels as assessed by an ATP biosensor. We also found that during mitophagy, the mitochondrial phosphatase PGAM5 is cleaved in hiPSC-derived endothelial progenitor cells and in turn activates β-catenin-mediated transcription of the transcriptional coactivator PGC-1α, which upregulates mitochondrial biogenesis. These data suggest that mitophagy itself initiates the increase in mitochondrial biogenesis and oxidative metabolism through transcriptional changes during endothelial cell differentiation. In summary, these findings reveal a mitophagy-mediated mechanism for metabolic rewiring and maturation of differentiating cells via the β-catenin signaling pathway. We propose that such mitochondrial-nuclear cross talk during hiPSC differentiation could be leveraged to enhance the metabolic maturation of differentiated cells.

Highlights

  • Pluripotent stem cells such as human induced pluripotent stem cells rely mainly on glycolysis for cellular ATP [1,2,3] and use glutamine metabolism to sustain pluripotency [4, 5]

  • While mitophagy has primarily been studied in the setting of culling damaged mitochondria in the setting of injury or disease, we posited that mitophagy may serve as a physiologic mechanism for the metabolic rewiring of differentiating stem cells by removing mitochondria adapted to the pluripotent state and replacing them with new mitochondria configured for the differentiated cell state

  • We show that mitophagy and subsequent mitochondrial biogenesis are required for the differentiation of human induced pluripotent stem cells (hiPSCs) to endothelial cells through the interaction of cleaved phosphoglycerate mutase family member 5 (PGAM5) and β-catenin

Read more

Summary

Introduction

Pluripotent stem cells such as human induced pluripotent stem cells (hiPSCs) rely mainly on glycolysis for cellular ATP [1,2,3] and use glutamine metabolism to sustain pluripotency [4, 5]. Mitophagy in iPSC differentiation communication between mitochondria and the nucleus One such potential mediator is the mitochondrial protein PGAM5 (Phosphoglycerate Mutase Family Member 5), a serine/threonine phosphatase that is embedded in the mitochondrial outer membrane [16] and is cleaved and released into the cytosol during mitophagy [17, 18]. It enhances signaling via the β-catenin pathway by dephosphorylating β-catenin, stabilizing it and allowing it to translocate to the nucleus to transcribe Wnt/β-catenin pathway genes [17]. This novel feedback mechanism of mitochondrial-nuclear cross talk in differentiating pluripotent stem cells provides new insights into the intersection of metabolic and developmental signaling

Objectives
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call