Back to Search Start Over

Single-cell RNA-seq analysis reveals a crucial role for Collagen Triple Helix Repeat Containing 1 (CTHRC1) cardiac fibroblasts after myocardial infarction

Authors :
Ruiz-Villalba, Adrian
Romero, Juan P.
Hernandez, Silvia C.
Vilas-Zornoza, Amaia
Fortelny, Nikolaus
Castro-Labrador, Laura
San Martin-Uriz, Patxi
Lorenzo-Vivas, Erika
García-Olloqui, Paula
Palacio, Marcel
Gavira, Juan José
Bastarrika, Gorka
Janssens, Stefan
Wu, Ming
Iglesias, Elena
Abizanda, Gloria
de Morentin, Xabier Martinez
Lasaga, Miren
Planell, Nuria
Bock, Christoph
Alignani, Diego
Medal, Gema
Pelacho, Beatriz
Prudovsky, Igor
Jin, Yong-Ri
Ryzhov, Sergey
Yin, Haifeng
Gomez-Cabrero, David
Lindner, Volkhard
Lara-Astiaso, David
Prósper, Felipe
Source :
Circulation
Publication Year :
2020

Abstract

BACKGROUND: Cardiac fibroblasts (CF) have a central role in the ventricular remodeling process associated with different types of fibrosis. Recent studies have shown that fibroblasts do not respond homogeneously to heart injury. Due to the limited set of bona fide fibroblast markers, a proper characterization of fibroblast population heterogeneity in response to cardiac damage is still missing. The purpose of this study was to define the CF heterogeneity during ventricular remodeling and the underlying mechanisms that regulate their function. METHODS: Collagen1α1-GFP(+) CF were characterized after myocardial infarction (MI) by single-cell and bulk RNA-seq, ATAC-seq and functional assays. Swine and patient samples were studied using bulk RNA-seq. RESULTS: We identified and characterized a unique CF subpopulation that emerges after MI in mice. These activated fibroblasts exhibit a clear pro-fibrotic signature, express high levels of Collagen Triple Helix Repeat Containing 1 (Cthrc1) and localize into the scar. Non-canonical TGF-β signaling and different transcription factors including SOX9 are important regulators mediating their response to cardiac injury. Moreover, the absence of CTHRC1 results in pronounced lethality due to ventricular rupture. Finally, a population of CF with a similar transcriptome was identified in a swine model of MI and in heart tissue from patients with MI and dilated cardiomyopathy. CONCLUSIONS: We report CF heterogeneity, their dynamics during the course of MI and redefine the CF that respond to cardiac injury and participate in myocardial remodeling. Our study identifies Cthrc1 as a novel regulator of the healing scar process, and as a target for future translational studies.

Details

Language :
English
Database :
OpenAIRE
Journal :
Circulation
Accession number :
edsair.pmid..........b4603694c375a5c82980cf7a9fae0cf1