Abstract

Human pluripotent stem cell (hPSC)-derived cardiomyocytes provide a promising regenerative cell therapy for cardiovascular patients and an important model system to accelerate drug discovery. However, cost-effective and time-efficient platforms must be developed to evaluate the quality of hPSC-derived cardiomyocytes during biomanufacturing. Here, we develop a non-invasive label-free live cell imaging platform to predict the efficiency of hPSC differentiation into cardiomyocytes. Autofluorescence imaging of metabolic co-enzymes is performed under varying differentiation conditions (cell density, concentration of Wnt signaling activator) across five hPSC lines. Live cell autofluorescence imaging and multivariate classification models provide high accuracy to separate low (< 50%) and high (≥ 50%) differentiation efficiency groups (quantified by cTnT expression on day 12) within 1 day after initiating differentiation (area under the receiver operating characteristic curve, 0.91). This non-invasive and label-free method could be used to avoid batch-to-batch and line-to-line variability in cell manufacturing from hPSCs.

Highlights

  • Human pluripotent stem cell-derived cardiomyocytes provide a promising regenerative cell therapy for cardiovascular patients and an important model system to accelerate drug discovery

  • We recorded the autofluorescence dynamics of NAD (P)H and flavin adenine dinucleotide (FAD) by Optical metabolic imaging (OMI) during the process of Human pluripotent stem cell (hPSC) differentiation into CMs. hPSCs were differentiated following a previous protocol[11], and cells were imaged on differentiation day 0, day 1 (24 h post-treatment with CHIR99021), day 3, and day 5 (48 h post-treatment with IWP2)

  • Differentiation of CMs from hPSCs critically relies on the timing and the state of Wnt signaling[11]

Read more

Summary

Introduction

Human pluripotent stem cell (hPSC)-derived cardiomyocytes provide a promising regenerative cell therapy for cardiovascular patients and an important model system to accelerate drug discovery. Cost-effective and time-efficient platforms must be developed to evaluate the quality of hPSC-derived cardiomyocytes during biomanufacturing. We develop a non-invasive label-free live cell imaging platform to predict the efficiency of hPSC differentiation into cardiomyocytes. Live cell autofluorescence imaging and multivariate classification models provide high accuracy to separate low (< 50%) and high (≥ 50%) differentiation efficiency groups (quantified by cTnT expression on day 12) within 1 day after initiating differentiation (area under the receiver operating characteristic curve, 0.91). Cost-effective and time-efficient platforms to generate functional CMs with high quality has emerged as an urgent need for cardiac medicine in drug screening, toxicity testing, disease modeling, and regenerative cell therapy. New technologies that can non-invasively monitor CM differentiation in real time and evaluate the differentiation outcome at early stages are needed to effectively optimize the biomanufacturing of CMs from stem cells

Objectives
Methods
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