Abstract 123: Optical Mapping of Host and Human Embryonic Stem Cell-Derived Cardiomyocyte Graft Electrical Activity in Injured Hearts

2014 ◽  
Vol 115 (suppl_1) ◽  
Author(s):  
Dominic Filice ◽  
Wei-Zhong Zhu ◽  
Benjamin Van Biber ◽  
Kip D Hauch ◽  
Michael A Laflamme

Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) show tremendous promise for cardiac repair, but more information is required as to their electrical behavior in vivo. hESC-CMs expressing the protein calcium sensor GCaMP3 provide a graft autonomous reporter of activation, which we have used to show hESC-CM grafts can couple with host myocardium in injured hearts. When we sought to assess host-graft electrical interactions by optical voltage mapping, we found the commonly used lipophilic voltage dyes RH237 and di-4-ANEPPS label host but not graft tissue. We hypothesized the water-soluble voltage dye di-2-ANEPEQ could overcome this limitation and efficiently label both host and graft. After confirming good spectral separation of GCaMP3 and di-2-ANEPEQ signals by spectrofluorimetry and confocal spectral imaging, we transplanted 1x10^8 GCaMP3+ hESC-CMs into guinea pig hearts (n=6) at 4 weeks following cardiac injury and then imaged engrafted hearts at 2 weeks post-transplantation with a dual-channel CCD-based system. We found a differential time-course of di-2-ANEPEQ labeling between host and graft tissues, with stable optical action potentials (APs) obtained in host and graft tissue after ~4 and ~13 minutes of dye perfusion, respectively. This differential labeling kinetics, which was also observed on washout, presumably reflects sluggish graft perfusion and provides another tool for distinguishing host and graft signals. No regions of 1:1 host-graft coupling were identified, and graft tissue had spontaneous rates from 0.1-2 Hz and long optical AP durations from 500-1090 ms. Activation maps based on di-2-ANEPEQ and GCaMP3 signals indicated relatively slow conduction velocities in graft tissue, as well as patterns of propagation that commonly occurred along a vector distinct from that in host tissue. These imaging studies reveal multiple potentially pro-arrhythmic properties in hESC-CM graft tissue including slow propagation, ultralong AP duration, as well as aberrant patterns of activation that can vary from beat to beat. We conclude the electrical behavior of both graft and host myocardium can be reliably assessed by the simultaneous imaging of a graft-autonomous fluorescent reporter of activation and a water-soluble voltage dye.

2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Dominic Filice ◽  
Wahiba Dhahri ◽  
Joell L. Solan ◽  
Paul D. Lampe ◽  
Erin Steele ◽  
...  

Abstract Background Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) show tremendous promise for cardiac regeneration, but the successful development of hESC-CM-based therapies requires improved tools to investigate their electrical behavior in recipient hearts. While optical voltage mapping is a powerful technique for studying myocardial electrical activity ex vivo, we have previously shown that intra-cardiac hESC-CM grafts are not labeled by conventional voltage-sensitive fluorescent dyes. We hypothesized that the water-soluble voltage-sensitive dye di-2-ANEPEQ would label engrafted hESC-CMs and thereby facilitate characterization of graft electrical function and integration. Methods We developed and validated a novel optical voltage mapping strategy based on the simultaneous imaging of the calcium-sensitive fluorescent protein GCaMP3, a graft-autonomous reporter of graft activation, and optical action potentials (oAPs) derived from di-2-ANEPEQ, which labels both graft and host myocardium. Cardiomyocytes from three different GCaMP3+ hESC lines (H7, RUES2, or ESI-17) were transplanted into guinea pig models of subacute and chronic infarction, followed by optical mapping at 2 weeks post-transplantation. Results Use of a water-soluble voltage-sensitive dye revealed pro-arrhythmic properties of GCaMP3+ hESC-CM grafts from all three lines including slow conduction velocity, incomplete host-graft coupling, and spatially heterogeneous patterns of activation that varied beat-to-beat. GCaMP3+ hESC-CMs from the RUES2 and ESI-17 lines both showed prolonged oAP durations both in vitro and in vivo. Although hESC-CMs partially remuscularize the injured hearts, histological evaluation revealed immature graft structure and impaired gap junction expression at this early timepoint. Conclusion Simultaneous imaging of GCaMP3 and di-2-ANEPEQ allowed us to acquire the first unambiguously graft-derived oAPs from hESC-CM-engrafted hearts and yielded critical insights into their arrhythmogenic potential and line-to-line variation.


Author(s):  
Wamaitha SE ◽  
Grybel KJ ◽  
Alanis-Lobato G ◽  
Gerri C ◽  
Ogushi S ◽  
...  

2020 ◽  
Vol 47 ◽  
pp. 101885
Author(s):  
Yongli Shan ◽  
Lishi Ma ◽  
Cong Zhang ◽  
Yanqi Zhang ◽  
Jingyuan Zhang ◽  
...  

2021 ◽  
Vol 52 ◽  
pp. 102234
Author(s):  
Cong Zhang ◽  
Yanxing Wei ◽  
Yanqi Zhang ◽  
Jingyuan Zhang ◽  
Qi Xing ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document