2019
DOI: 10.3791/59906-v
|View full text |Cite
|
Sign up to set email alerts
|

Human iPSC-Derived Cardiomyocyte Networks on Multiwell Micro-electrode Arrays for Recurrent Action Potential Recordings

Abstract: Cardiac safety screening is of paramount importance for drug discovery and therapeutics. Therefore, the development of novel high-throughput electrophysiological approaches for hiPSC-derived cardiomyocyte (hiPSC-CM) preparations is much needed for efficient drug testing. Although multielectrode arrays (MEAs) are frequently employed for field potential measurements of excitable cells, a recent publication by Joshi-Mukherjee and colleagues described and validated its application for recurrent action potential (A… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
1

Citation Types

0
2
0

Year Published

2021
2021
2024
2024

Publication Types

Select...
3

Relationship

0
3

Authors

Journals

citations
Cited by 3 publications
(4 citation statements)
references
References 20 publications
(28 reference statements)
0
2
0
Order By: Relevance
“…High-purity iPSC-CM cultures were produced from human cardiac fibroblasts as described in (Zlochiver et al, 2019). hiPSCs cultures were differentiated using the feederfree monolayer differentiation protocol.…”
Section: Methodsmentioning
confidence: 99%
See 1 more Smart Citation
“…High-purity iPSC-CM cultures were produced from human cardiac fibroblasts as described in (Zlochiver et al, 2019). hiPSCs cultures were differentiated using the feederfree monolayer differentiation protocol.…”
Section: Methodsmentioning
confidence: 99%
“…Much like a medical electrocardiogram, FP recordings measure the potential change across the entire bulk of tissue and have been observed to be closely correlated to the underlying AP (Tertoolen et al, 2018). Recent work has shown the feasibility of multi-electrode arrays (MEA) (Meyer et al, 2012) in the large-scale efficient recording of both AP and FP readings (Edwards et al, 2018;Zlochiver et al, 2019;Hayes et al, 2019;Balafkan et al, 2020).…”
Section: Introductionmentioning
confidence: 99%
“…Therefore, several scalable methods for electrophysiological signal recording have been explored, including passive electrodes [31][32][33][34][35][36][37] and active micro/nanobioelectronic devices [38][39][40][41][42][43][44][45][46]. Microelectrode arrays (MEAs) [31,[47][48][49][50][51][52] are passive electrodes that are commonly produced on insulating substrates. These electrodes receive electrical signals from cells and subsequently transmit them to external amplifiers through leads that are coated with passivation layers.…”
Section: Introductionmentioning
confidence: 99%
“…Cardiomyocytes are participating in the regulation of the contraction–relaxation period of the heart. Their self-regulation of contractility and the generation of action potential involve transportation by multiple ion channels and exchangers, which are of high sensitivity to external electrical stimulation. Microelectrode array (MEA)-based extracellular recording featured noninvasive, dynamic, long-term, and large-scale electrophysiological studies, but it does not provide important information such as resting membrane potential and ion channel conductance. Electroporation is a well-established technique that induces the generation of nanopores in the cell membrane for intracellular action potential recording access. Due to the accessibility and high throughput, electroporation is also one of the most common physical techniques for biomolecule delivery (e.g., mRNA, DNA, and proteins) which holds great potential for genetic engineering, cellular imaging, and medical clinical applications. Compared to virus-mediated and chemical methods, electroporation prevents toxicity to cells . However, bulk electroporation (BEP)-based delivery of cardiomyocytes remains challenging.…”
mentioning
confidence: 99%