Back to Journals » Biologics: Targets and Therapy » Volume 18

Gene Expression, Morphology, and Electrophysiology During the Dynamic Development of Human Induced Pluripotent Stem Cell-Derived Atrial- and Ventricular-Like Cardiomyocytes [Letter]

Authors Wulandari M, Prihatono A, Rusdi AJ 

Received 27 May 2024

Accepted for publication 29 May 2024

Published 5 June 2024 Volume 2024:18 Pages 145—146

DOI https://doi.org/10.2147/BTT.S480180

Checked for plagiarism Yes

Editor who approved publication: Professor Shein-Chung Chow



Mayang Wulandari,1 Amal Prihatono,1 Achmad Jaelani Rusdi2

1Diploma 3 in Acupuncture Studies, ITSK RS DR Soepraoen Malang, Kota Malang, Jawa Timur, Indonesia; 2Medical Record and Health Information Department, ITSK RS DR Soepraoen Malang, Kota Malang, Jawa Timur, Indonesia

Correspondence: Mayang Wulandari, Prodi Akupunktur ITSK RS dr Soepraoen Jalan Majapahit No. 1, Kota Malang, Jawa Timur, 65119, Indonesia, Tel +628123315617, Email [email protected]


View the original paper by Dr Zhou and colleagues

A Response to Letter has been published for this article.


Dear editor

The study titled “Gene Expression, Morphology, and Electrophysiology During the Dynamic Development of Human Induced Pluripotent Stem Cell-Derived Atrial- and Ventricular-Like Cardiomyocytes” presents a valuable investigation into the dynamic development of human induced pluripotent stem cell-derived atrial-like (iPS-AM) and ventricular-like (iPS-VM) cardiomyocytes.1 The authors employ a small molecule-based approach to modulate retinoic acid and bone morphogenetic protein signaling pathways during differentiation. Their comprehensive assessment, integrating gene expression, morphology, and electrophysiological function,2 offers valuable insights into the maturation process of these cell types. A key strength of this work lies in its multifaceted approach. By employing immunofluorescence, qRT-PCR, flow cytometry, and electron microscopy, the authors demonstrate a coordinated increase in pan-cardiomyocyte markers3 (TNNT2, ACTN2) alongside chamber-specific markers (NPPA, MYL7, KCNJ5 for iPS-AM; GJA1, MYL2, CACNA1C for iPS-VM). This detailed characterization underscores the progressive acquisition of chamber-specific phenotypes during differentiation. Furthermore, the study effectively captures the dynamic changes in cell morphology. The observed transformation of cTnT organization from a scattered pattern to a well-defined sarcomeric structure in both iPS-AMs and iPS-VMs highlights the maturation of contractile machinery.4 Additionally, the dynamic shift in mitochondrial abundance and lipid droplet content suggests a metabolic reprogramming that aligns with functional development.5 While the study effectively demonstrates dynamic changes in gene expression and morphology, a deeper exploration of the electrophysiological maturation is warranted. Although resting and action potential amplitudes remained similar, the observed prolongation of action potential duration during development requires further investigation. Future studies could explore the underlying ionic currents and channel expression profiles to provide a more comprehensive understanding of electrophysiological maturation. In conclusion, the work by Yafei Zhou sheds light on the dynamic development of human iPSC-derived cardiomyocytes. Their findings hold significant promise for advancing our understanding of heart development and potentially paving the way for future applications in disease modeling and regenerative medicine. Further research delving deeper into electrophysiological maturation and potentially incorporating functional assays could offer even richer insights into this vital process.

Disclosure

The authors report no conflicts of interest in this communication.

References

1. Zhou Y, Zhou R, Huang W, et al. Gene expression, morphology, and electrophysiology during the dynamic development of human induced pluripotent stem cell-derived atrial- and ventricular-like cardiomyocytes. Biologics. 2024;18:115–127. doi:10.2147/BTT.S448054

2. Camunas-Soler J. Integrating single-cell transcriptomics with cellular phenotypes: cell morphology, Ca2+ imaging and electrophysiology. Biophys Rev. 2024;16(1):89–107. doi:10.1007/s12551-023-01174-2

3. Cui C, Wang J, Qian D, et al. binary colloidal crystals drive spheroid formation and accelerate maturation of human-induced pluripotent stem cell-derived cardiomyocytes. ACS Appl Mater Interfaces. 2019;11(4):3679–3689. doi:10.1021/acsami.8b17090

4. Mao M, Qu X, Zhang Y, et al. Leaf-venation-directed cellular alignment for macroscale cardiac constructs with tissue-like functionalities. Nat Commun. 2023;14(1):2077. doi:10.1038/s41467-023-37716-1

5. Mehta A, Ratre YK, Soni VK, et al. Orchestral role of lipid metabolic reprogramming in T-cell malignancy. Front Oncol. 2023;2023:13.

Creative Commons License © 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.