Abstract 262: Gene Expression Profiling of Hypertrophic and Failing Cardiomyocytes Identifies New Players Involved in Heart Failure

2019 ◽  
Vol 125 (Suppl_1) ◽  
Author(s):  
Marta Vigil-Garcia ◽  
Charlotte J Demkes ◽  
Joep E Eding ◽  
Danielle Versteeg ◽  
Hesther de Ruiter ◽  
...  
2021 ◽  
Author(s):  
Michael G. Levin ◽  
Noah L. Tsao ◽  
Tiffany R. Bellomo ◽  
William Paul Bone ◽  
Krishna G. Aragam ◽  
...  

Heart failure (HF) is a leading cause of cardiovascular morbidity and mortality, yet the contribution of common genetic variation to HF risk has not been fully elucidated, particularly in comparison to other common cardiometabolic traits. We conducted a multi-ancestry genome-wide association study (GWAS) meta-analysis of all-cause HF including up to 56,722 HF cases and 1,133,054 controls, identifying 4 novel loci. We then performed a multi-ancestry multivariate association study of HF and related cardiac imaging endophenotypes, identifying 71 conditionally-independent variants, including 16 novel loci. Secondary colocalization and transcriptome-wide association analyses identified known and novel candidate cardiomyopathy genes, which were validated in gene-expression profiling of failing and healthy human hearts. Colocalization, gene expression profiling, and Mendelian randomization provided convergent evidence for the roles of BCKDHA and circulating branch-chain amino acids in heart failure and cardiac structure. Finally, proteome-wide Mendelian randomization revealed 11 circulating proteins associated with HF or quantitative imaging traits. These analyses highlight similarities and differences among heart failure and associated cardiovascular imaging endophenotypes, implicate novel common genetic variation in the pathogenesis of HF, and identify circulating proteins that may represent novel cardiomyopathy treatment targets.


2015 ◽  
Vol 7 (1) ◽  
Author(s):  
Agata Maciejak ◽  
Marek Kiliszek ◽  
Marcin Michalak ◽  
Dorota Tulacz ◽  
Grzegorz Opolski ◽  
...  

2010 ◽  
Vol 51 (3) ◽  
pp. 353-368 ◽  
Author(s):  
S. Szmit ◽  
M. Jank ◽  
H. Maciejewski ◽  
M. Grabowski ◽  
R. Glowczynska ◽  
...  

2004 ◽  
Vol 99 (3) ◽  
pp. 230-238 ◽  
Author(s):  
J�rg Stypmann ◽  
G�nter Breithardt ◽  
Eric Schulze-Bahr ◽  
Petra Ursula Seiler

Author(s):  
Naoko Yamaguchi ◽  
Junhua Xiao ◽  
Deven Narke ◽  
Devin Shaheen ◽  
Xianming Lin ◽  
...  

Background: Elevated intracardiac pressure due to heart failure induces electrical and structural remodeling in the left atrium (LA) that begets atrial myopathy and arrhythmias. The underlying molecular pathways that drive atrial remodeling during cardiac pressure overload are poorly defined. The purpose of this study is to characterize the response of the ETV1 signaling axis in the LA during cardiac pressure overload in humans and mouse models and explore the role of ETV1 in atrial electrical and structural remodeling. Methods: We performed gene expression profiling in 265 left atrial samples from patients who underwent cardiac surgery. Comparative gene expression profiling was performed between two murine models of cardiac pressure overload, transverse aortic constriction (TAC) banding and Angiotensin II (AngII) infusion, and a genetic model of Etv1 cardiomyocyte-selective knockout ( Etv1 f/f Mlc2a Cre/+ ). Results: Using the Cleveland Clinic biobank of human LA specimens, we found that ETV1 expression is decreased in patients with reduced ejection fraction. Consistent with its role as an important mediator of the Neuregulin-1 (NRG1) signaling pathway and activator of rapid conduction gene programming, we identified a direct correlation between ETV1 expression level and NRG1, ERBB4, SCN5A , and GJA5 levels in human LA samples. In a similar fashion to heart failure patients, we showed that left atrial ETV1 expression is downregulated at the RNA and protein levels in murine pressure overload models. Comparative analysis of LA RNA-seq datasets from TAC and AngII treated mice showed a high Pearson correlation, reflecting a highly ordered process by which the LA undergoes electrical and structural remodeling. Cardiac pressure overload produced a consistent downregulation of ErbB4, Etv1, Scn5a, and Gja5 and upregulation of profibrotic gene programming, which includes Tgfbr1/2, Igf1, and numerous collagen genes. Etv1 f/f Mlc2a Cre/+ mice displayed atrial conduction disease and arrhythmias. Correspondingly, the LA from Etv1 f/f Mlc2a Cre/+ mice showed downregulation of rapid conduction genes and upregulation of profibrotic gene programming, whereas analysis of a gain-of-function ETV1 RNA-seq dataset from neonatal rat ventricular myocytes transduced with Etv1 showed reciprocal changes. Conclusions: ETV1 is downregulated in the LA during cardiac pressure overload, contributing to both electrical and structural remodeling.


Sign in / Sign up

Export Citation Format

Share Document