Combined lineage tracing and scRNA-seq reveals unexpected first heart field predominance of human iPSC differentiation

Author:

Galdos Francisco X12ORCID,Lee Carissa1,Lee Soah3,Paige Sharon14,Goodyer William14,Xu Sidra1,Samad Tahmina1,Escobar Gabriela V1,Darsha Adrija5,Beck Aimee1,Bak Rasmus O6ORCID,Porteus Matthew H17ORCID,Wu Sean M128ORCID

Affiliation:

1. Stanford Cardiovascular Institute, Stanford University

2. Institute for Stem Cell Biology and Regenerative Medicine, Stanford University

3. Department of Pharmacy, Sungkyunkwan University

4. Division of Pediatric Cardiology, Department of Pediatrics, Stanford University

5. School of Medicine, University of California, San Diego

6. Department of Biomedicine, Aarhus University

7. Department of Pediatrics, Stanford University

8. Division of Cardiovascular of Medicine, Department of Medicine, Stanford University

Abstract

During mammalian development, the left and right ventricles arise from early populations of cardiac progenitors known as the first and second heart fields, respectively. While these populations have been extensively studied in non-human model systems, their identification and study in vivo human tissues have been limited due to the ethical and technical limitations of accessing gastrulation-stage human embryos. Human-induced pluripotent stem cells (hiPSCs) present an exciting alternative for modeling early human embryogenesis due to their well-established ability to differentiate into all embryonic germ layers. Here, we describe the development of a TBX5/MYL2 lineage tracing reporter system that allows for the identification of FHF- progenitors and their descendants including left ventricular cardiomyocytes. Furthermore, using single-cell RNA sequencing (scRNA-seq) with oligonucleotide-based sample multiplexing, we extensively profiled differentiating hiPSCs across 12 timepoints in two independent iPSC lines. Surprisingly, our reporter system and scRNA-seq analysis revealed a predominance of FHF differentiation using the small molecule Wnt-based 2D differentiation protocol. We compared this data with existing murine and 3D cardiac organoid scRNA-seq data and confirmed the dominance of left ventricular cardiomyocytes (>90%) in our hiPSC-derived progeny. Together, our work provides the scientific community with a powerful new genetic lineage tracing approach as well as a single-cell transcriptomic atlas of hiPSCs undergoing cardiac differentiation.

Funder

NHLBI Division of Intramural Research

NIH Office of the Director

NIH K08 Mentored Clinical Scientist Research Career Development Award

NIH/NHLBI

The Dorothy Dee and Marjorie Boring Trust Award

Stanford Medical Scientist Training Program

Publisher

eLife Sciences Publications, Ltd

Subject

General Immunology and Microbiology,General Biochemistry, Genetics and Molecular Biology,General Medicine,General Neuroscience

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3