4.8 Article

Sequential Defects in Cardiac Lineage Commitment and Maturation Cause Hypoplastic Left Heart Syndrome

Journal

CIRCULATION
Volume 144, Issue 17, Pages 1409-1428

Publisher

LIPPINCOTT WILLIAMS & WILKINS
DOI: 10.1161/CIRCULATIONAHA.121.056198

Keywords

autophagy; cell cycle; heart defects; congenital; hypoplastic left heart syndrome; induced pluripotent stem cells; unfolded protein response; whole exome sequencing

Funding

  1. European Research Council [788381, 261053]
  2. German Research Foundation [KR3770/7-3, KR3770/11-1, KR3770/14-1]
  3. Transregio Research Unit [267]
  4. German Center for Cardiovascular Research [FKZ 81Z0600601]
  5. Fondazione Umberto Veronesi
  6. European Research Council (ERC) [261053, 788381] Funding Source: European Research Council (ERC)

Ask authors/readers for more resources

The study revealed disruptions in gene programs and cellular processes in patients with hypoplastic left heart syndrome (HLHS), particularly in key aspects of fetal ventricular cardiogenesis such as the cell cycle and cardiomyocyte maturation. These abnormalities result in defects in early differentiation of cardiac progenitors, leading to left ventricular hypoplasia.
Background: Complex molecular programs in specific cell lineages govern human heart development. Hypoplastic left heart syndrome (HLHS) is the most common and severe manifestation within the spectrum of left ventricular outflow tract obstruction defects occurring in association with ventricular hypoplasia. The pathogenesis of HLHS is unknown, but hemodynamic disturbances are assumed to play a prominent role. Methods: To identify perturbations in gene programs controlling ventricular muscle lineage development in HLHS, we performed whole-exome sequencing of 87 HLHS parent-offspring trios, nuclear transcriptomics of cardiomyocytes from ventricles of 4 patients with HLHS and 15 controls at different stages of heart development, single cell RNA sequencing, and 3D modeling in induced pluripotent stem cells from 3 patients with HLHS and 3 controls. Results: Gene set enrichment and protein network analyses of damaging de novo mutations and dysregulated genes from ventricles of patients with HLHS suggested alterations in specific gene programs and cellular processes critical during fetal ventricular cardiogenesis, including cell cycle and cardiomyocyte maturation. Single-cell and 3D modeling with induced pluripotent stem cells demonstrated intrinsic defects in the cell cycle/unfolded protein response/autophagy hub resulting in disrupted differentiation of early cardiac progenitor lineages leading to defective cardiomyocyte subtype differentiation/maturation in HLHS. Premature cell cycle exit of ventricular cardiomyocytes from patients with HLHS prevented normal tissue responses to developmental signals for growth, leading to multinucleation/polyploidy, accumulation of DNA damage, and exacerbated apoptosis, all potential drivers of left ventricular hypoplasia in absence of hemodynamic cues. Conclusions: Our results highlight that despite genetic heterogeneity in HLHS, many mutations converge on sequential cellular processes primarily driving cardiac myogenesis, suggesting novel therapeutic approaches.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available