In order to bring you the best possible user experience, this site uses Javascript. If you are seeing this message, it is likely that the Javascript option in your browser is disabled. For optimal viewing of this site, please ensure that Javascript is enabled for your browser.
Did you know that your browser is out of date? To get the best experience using our website we recommend that you upgrade to a newer version. Learn more.

A novel etiology of Left Ventricular Non-Compaction

Commented by the ESC WG on Development, Anatomy & Pathology

ESC Working Groups
Basic Science - Cardiac Diseases - Cardiomyopathies

Left ventricular non-compaction (LVNC) is a heterogenous cardiac disorder characterized by excessive trabeculations and deep trabecular recesses in the left ventricle, with clinical outcomes that range from asymptomatic to heart failure, arrhythmias, and sudden cardiac death1. LVNC is considered as a primary genetic cardiomyopathy according to the American Heart Association2, but it is still “unclassified” in the most recent Statement of the European Society of Cardiology3, illustrating the need for better understanding of the origins of this disease. Its prevalence is growing and it is considered as the third most common cardiomyopathy.

LVNC is thought to arise from an arrest of normal compaction of the extensive mesh of trabeculae present in the embryonic ventricles during development4. However, despite its importance in the formation of an efficient ventricular myocardium, ventricular compaction is poorly understood. Recent findings have shown that a non-compaction phenotype arises when proliferation of compact, but not trabecular, cardiomyocytes was inhibited5. Moreover, Kristy Red-Horse’s lab has shown that endothelial-cardiomyocyte crosstalk is also required, suggesting that, beside cardiomyocytes, other cell types are involved in the etiology of this cardiomyopathy6.

In this paper published in European Heart Journal, Rhee et al. identified secreted molecules from endothelial or endocardial cells that drive cardiomyocyte proliferation and maturation7. They used single-cell RNA sequencing to characterize and compare the molecular signatures of individual cell types from control and non-compacted fetal mouse hearts. They identified a specific cluster of proliferating endocardial cells in non-compacted hearts. Regarding cardiomyocytes, they found differential molecular signatures between control and mutant cells, with an increase in genes involved in glycolysis, hypoxia and fatty acid metabolism. They also found that known genes associated with LVNC in human are found dysregulated in cell types other than cardiomyocytes, demonstrating the importance of the cross talk between these different cell types during ventricular development. Most importantly, they found Col15a1 to be downregulated in mutant and a rescue experiment shows that adding Col15a1 to primary culture of non-compacted cardiomyocytes improves their proliferation capacity. Moreover, they identified upregulation of four secreted factors from non-compacted endocardium that inhibit cardiomyocyte proliferation and stimulate maturation. Finally, they showed that blocking coronary angiogenesis results in a non-compaction phenotype.

This study shows the importance of vascular-derived factors, or angiocrines, that can influence cardiomyocytes behavior by modulating their proliferation and maturation - key cellular processes in ventricular compaction. The findings point to a novel etiology of LVNC, by which dysregulation of paracrine signaling from endothelial cells can reduce cardiomyocyte proliferation and induce a non-compaction phenotype.

References


1. Towbin JA, Lorts A, Jefferies JL. Left ventricular non-compaction cardiomyopathy. Lancet 2015, 386, 813-825.
2. Maron BJ, Towbin JA, Thiene G, Antzelevitch C, Corrado D, Arnett D, Moss AJ, Seidman CE, Young JB. Contemporary definitions and classification of the cardiomyopathies: An american heart association scientific statement from the council on clinical cardiology, heart failure and transplantation committee, quality of care and outcomes research and functional genomics and translational biology interdisciplinary working groups, and council on epidemiology and prevention. Circulation 2006, 113, 1807-1816.
3. Elliott P, Andersson B, Arbustini E, Bilinska Z, Cecchi F, Charron P, Dubourg O, Kuhl U, Maisch B, McKenna WJ, et al. Classification of the cardiomyopathies: A position statement from the European Society of Cardiology working group on myocardial and pericardial diseases. Eur Heart J 2008, 29, 270-276.
4. Freedom RM, Yoo SJ, Perrin D, Taylor G, Petersen S, Anderson RH. The morphological spectrum of ventricular noncompaction. Cardiol Young 2005, 15, 345-364.
5. Tian X, Li Y, He L, Zhang H, Huang X, Liu Q, Pu W, Zhang L, Li Y, Zhao H, et al. Identification of a hybrid myocardial zone in the mammalian heart after birth. Nat Commun 2017, 8, 87.
6. Rhee S, Chung JI, King DA, D'Amato G, Paik DT, Duan A, Chang A, Nagelberg D, Sharma B, Jeong Y, et al. Endothelial deletion of ino80 disrupts coronary angiogenesis and causes congenital heart disease. Nat Commun 2018, 9, 368.
7. Rhee S, Paik DT, Yang JY, Nagelberg D, Williams I, Tian L, Roth R, Chandy M, Ban J, Belbachir N, et al. Endocardial/endothelial angiocrines regulate cardiomyocyte development and maturation and induce features of ventricular non-compaction. Eur Heart J 2021, ehab298.

The content of this article reflects the personal opinion of the author/s and is not necessarily the official position of the European Society of Cardiology.

Contact us

ESC Working Group on Development, Anatomy & Pathology

European Society of Cardiology

European Heart House
Les Templiers
2035 Route des Colles
CS 80179 Biot

06903, Sophia Antipolis, FR

Tel: +33.4.92.94.76.00