Single epicardial cell transcriptome sequencing identifies Caveolin 1 as an essential factor in zebrafish heart regeneration.

TitleSingle epicardial cell transcriptome sequencing identifies Caveolin 1 as an essential factor in zebrafish heart regeneration.
Publication TypeJournal Article
Year of Publication2016
AuthorsCao, J, Navis, A, Cox, BD, Dickson, AL, Gemberling, M, Karra, R, Bagnat, M, Poss, KD
JournalDevelopment
Volume143
Issue2
Pagination232-43
Date Published2016 Jan 15
ISSN1477-9129
KeywordsAnimals, Caveolin 1, Heart, Myocytes, Cardiac, Pericardium, Regeneration, Zebrafish, Zebrafish Proteins
Abstract

In contrast to mammals, adult zebrafish have a high capacity to regenerate damaged or lost myocardium through proliferation of cardiomyocytes spared from damage. The epicardial sheet covering the heart is activated by injury and aids muscle regeneration through paracrine effects and as a multipotent cell source, and has received recent attention as a target in cardiac repair strategies. Although it is recognized that epicardium is required for muscle regeneration and itself has high regenerative potential, the extent of cellular heterogeneity within epicardial tissue is largely unexplored. Here, we performed transcriptome analysis on dozens of epicardial lineage cells purified from zebrafish harboring a transgenic reporter for the pan-epicardial gene tcf21. Hierarchical clustering analysis suggested the presence of at least three epicardial cell subsets defined by expression signatures. We validated many new pan-epicardial and epicardial markers by alternative expression assays. Additionally, we explored the function of the scaffolding protein and main component of caveolae, caveolin 1 (cav1), which was present in each epicardial subset. In BAC transgenic zebrafish, cav1 regulatory sequences drove strong expression in ostensibly all epicardial cells and in coronary vascular endothelial cells. Moreover, cav1 mutant zebrafish generated by genome editing showed grossly normal heart development and adult cardiac anatomy, but displayed profound defects in injury-induced cardiomyocyte proliferation and heart regeneration. Our study defines a new platform for the discovery of epicardial lineage markers, genetic tools, and mechanisms of heart regeneration.

DOI10.1242/dev.130534
Alternate JournalDevelopment
PubMed ID26657776
PubMed Central IDPMC4725347
Grant ListT32 HD040372 / HD / NICHD NIH HHS / United States
HL081674 / HL / NHLBI NIH HHS / United States
K08-HL116485 / HL / NHLBI NIH HHS / United States
R01 HL081674 / HL / NHLBI NIH HHS / United States
K08 HL116485 / HL / NHLBI NIH HHS / United States
R01 AR065439 / AR / NIAMS NIH HHS / United States
R01AR065439-02 / AR / NIAMS NIH HHS / United States