Effects of MYBPC3 loss-of-function mutations preceding hypertrophic cardiomyopathy

التفاصيل البيبلوغرافية
العنوان: Effects of MYBPC3 loss-of-function mutations preceding hypertrophic cardiomyopathy
المؤلفون: Mick Wauchope, Vi T. Tang, Allen P. Liu, Eric D. Smith, Jordan A Shavit, Xiaoquan Wen, Michael J. Previs, Aaron H Wasserman, Adam S. Helms, Sharlene M. Day, Lap Man Lee, Akul Arora, Sabrina Friedline, Thomas S. O’Leary
المصدر: JCI Insight. 5
بيانات النشر: American Society for Clinical Investigation, 2020.
سنة النشر: 2020
مصطلحات موضوعية: Sarcomeres, 0301 basic medicine, Heterozygote, Mutant, Biology, Muscle Development, medicine.disease_cause, Cell Line, Frameshift mutation, 03 medical and health sciences, 0302 clinical medicine, Protein biosynthesis, medicine, Humans, Genetic Predisposition to Disease, Myocytes, Cardiac, RNA, Messenger, Frameshift Mutation, Gene, Alleles, Loss function, Gene Editing, Messenger RNA, Mutation, Binding protein, General Medicine, Cardiomyopathy, Hypertrophic, Cell biology, 030104 developmental biology, Codon, Nonsense, 030220 oncology & carcinogenesis, Carrier Proteins, Transcriptome, Research Article
الوصف: Mutations in cardiac myosin binding protein C (MyBP-C, encoded by MYBPC3) are the most common cause of hypertrophic cardiomyopathy (HCM). Most MYBPC3 mutations result in premature termination codons (PTCs) that cause RNA degradation and a reduction of MyBP-C in HCM patient hearts. However, a reduction in MyBP-C has not been consistently observed in MYBPC3-mutant induced pluripotent stem cell cardiomyocytes (iPSCMs). To determine early MYBPC3 mutation effects, we used patient and genome-engineered iPSCMs. iPSCMs with frameshift mutations were compared with iPSCMs with MYBPC3 promoter and translational start site deletions, revealing that allelic loss of function is the primary inciting consequence of mutations causing PTCs. Despite a reduction in wild-type mRNA in all heterozygous iPSCMs, no reduction in MyBP-C protein was observed, indicating protein-level compensation through what we believe is a previously uncharacterized mechanism. Although homozygous mutant iPSCMs exhibited contractile dysregulation, heterozygous mutant iPSCMs had normal contractile function in the context of compensated MyBP-C levels. Agnostic RNA-Seq analysis revealed differential expression in genes involved in protein folding as the only dysregulated gene set. To determine how MYBPC3-mutant iPSCMs achieve compensated MyBP-C levels, sarcomeric protein synthesis and degradation were measured with stable isotope labeling. Heterozygous mutant iPSCMs showed reduced MyBP-C synthesis rates but a slower rate of MyBP-C degradation. These findings indicate that cardiomyocytes have an innate capacity to attain normal MyBP-C stoichiometry despite MYBPC3 allelic loss of function due to truncating mutations. Modulating MyBP-C degradation to maintain MyBP-C protein levels may be a novel treatment approach upstream of contractile dysfunction for HCM.
تدمد: 2379-3708
URL الوصول: https://explore.openaire.eu/search/publication?articleId=doi_dedup___::34145f3300bda4a414bef62333ee83ad
https://doi.org/10.1172/jci.insight.133782
حقوق: OPEN
رقم الأكسشن: edsair.doi.dedup.....34145f3300bda4a414bef62333ee83ad
قاعدة البيانات: OpenAIRE