4.3 Article

Mono- and Biallelic Protein-Truncating Variants in Alpha-Actinin 2 Cause Cardiomyopathy Through Distinct Mechanisms

Journal

Publisher

LIPPINCOTT WILLIAMS & WILKINS
DOI: 10.1161/CIRCGEN.121.003419

Keywords

actinin; cardiomyopathies; hypertrophy; mass spectrometry; mutation; sarcomeres

Funding

  1. Knut and Alice Wallenberg Foundation
  2. National Institutes of Health [R01HL105993]
  3. ARRA Award from the National Center for Research Resources (NCRR) [1S10RR026780-01]

Ask authors/readers for more resources

The study revealed characteristics of hypertrophy, structural disarray, impaired contractility, and aberrant Ca2+ signaling in patient-derived iPSC-cardiomyocytes with ACTN2 protein-truncating variants. The loss of ACTN2's C-terminus disrupts interactions with other actinins and sarcolemma-associated proteins, potentially leading to arrhythmic and relaxation defects. This data advances understanding of ACTN2's role in the human heart and establishes recessive inheritance of ACTN2 truncation as causative of disease.
Background: ACTN2 (alpha-actinin 2) anchors actin within cardiac sarcomeres. The mechanisms linking ACTN2 mutations to myocardial disease phenotypes are unknown. Here, we characterize patients with novel ACTN2 mutations to reveal insights into the physiological function of ACTN2. Methods: Patients harboring ACTN2 protein-truncating variants were identified using a custom mutation pipeline. In patient-derived iPSC-cardiomyocytes, we investigated transcriptional profiles using RNA sequencing, contractile properties using video-based edge detection, and cellular hypertrophy using immunohistochemistry. Structural changes were analyzed through electron microscopy. For mechanistic studies, we used co-immunoprecipitation for ACTN2, followed by mass-spectrometry to investigate protein-protein interaction, and protein tagging followed by confocal microscopy to investigate introduction of truncated ACTN2 into the sarcomeres. Results: Patient-derived iPSC-cardiomyocytes were hypertrophic, displayed sarcomeric structural disarray, impaired contractility, and aberrant Ca2+-signaling. In heterozygous indel cells, the truncated protein incorporates into cardiac sarcomeres, leading to aberrant Z-disc ultrastructure. In homozygous stop-gain cells, affinity-purification mass-spectrometry reveals an intricate ACTN2 interactome with sarcomere and sarcolemma-associated proteins. Loss of the C-terminus of ACTN2 disrupts interaction with ACTN1 (alpha-actinin 1) and GJA1 (gap junction protein alpha 1), 2 sarcolemma-associated proteins, which may contribute to the clinical arrhythmic and relaxation defects. The causality of the stop-gain mutation was verified using CRISPR-Cas9 gene editing. Conclusions: Together, these data advance our understanding of the role of ACTN2 in the human heart and establish recessive inheritance of ACTN2 truncation as causative of disease.

Authors

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

Reviews

Primary Rating

4.3
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available