Show simple item record

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

dc.contributor.authorHelms, AS
dc.contributor.authorTang, VT
dc.contributor.authorO’Leary, TS
dc.contributor.authorFriedline, S
dc.contributor.authorWauchope, M
dc.contributor.authorArora, A
dc.contributor.authorWasserman, AH
dc.contributor.authorSmith, ED
dc.contributor.authorLee, LM
dc.contributor.authorWen, XW
dc.contributor.authorShavit, JA
dc.contributor.authorLiu, AP
dc.contributor.authorPrevis, MJ
dc.contributor.authorDay, SM
dc.coverage.spatialUnited States
dc.date.accessioned2022-12-29T17:47:32Z
dc.date.available2022-12-29T17:47:32Z
dc.date.issued2020-01-30
dc.identifier.issn2379-3708
dc.identifier.issn2379-3708
dc.identifier.urihttps://www.ncbi.nlm.nih.gov/pubmed/31877118
dc.identifier.urihttps://hdl.handle.net/2027.42/175345en
dc.description.abstractMutations 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.
dc.format.mediumElectronic
dc.languageeng
dc.publisherAmerican Society for Clinical Investigation
dc.relation.haspartARTN e133782
dc.subjectCardiology
dc.subjectCardiovascular disease
dc.subjectChaperones
dc.subjectMonogenic diseases
dc.subjectAlleles
dc.subjectCardiomyopathy, Hypertrophic
dc.subjectCarrier Proteins
dc.subjectCell Line
dc.subjectCodon, Nonsense
dc.subjectFrameshift Mutation
dc.subjectGene Editing
dc.subjectGenetic Predisposition to Disease
dc.subjectHeterozygote
dc.subjectHumans
dc.subjectMuscle Development
dc.subjectMutation
dc.subjectMyocytes, Cardiac
dc.subjectRNA, Messenger
dc.subjectSarcomeres
dc.subjectTranscriptome
dc.titleEffects of MYBPC3 loss-of-function mutations preceding hypertrophic cardiomyopathy
dc.typeArticle
dc.identifier.pmid31877118
dc.description.bitstreamurlhttp://deepblue.lib.umich.edu/bitstream/2027.42/175345/2/133782.2-20200120151342-covered-e0fd13ba177f913fd3156f593ead4cfd.pdf
dc.identifier.doi10.1172/jci.insight.133782
dc.identifier.doihttps://dx.doi.org/10.7302/6726
dc.identifier.sourceJCI Insight
dc.description.versionPublished version
dc.date.updated2022-12-29T17:47:06Z
dc.identifier.orcid0000-0002-0234-6430
dc.identifier.orcid0000-0002-5915-9492
dc.identifier.orcid0000-0001-8990-2737
dc.identifier.orcid0000-0002-0309-7018
dc.description.filedescriptionDescription of 133782.2-20200120151342-covered-e0fd13ba177f913fd3156f593ead4cfd.pdf : Published version
dc.identifier.volume5
dc.identifier.issue2
dc.identifier.startpage133782
dc.identifier.name-orcidHelms, AS; 0000-0002-0234-6430
dc.identifier.name-orcidTang, VT
dc.identifier.name-orcidO’Leary, TS
dc.identifier.name-orcidFriedline, S
dc.identifier.name-orcidWauchope, M
dc.identifier.name-orcidArora, A
dc.identifier.name-orcidWasserman, AH
dc.identifier.name-orcidSmith, ED; 0000-0002-5915-9492
dc.identifier.name-orcidLee, LM
dc.identifier.name-orcidWen, XW; 0000-0001-8990-2737
dc.identifier.name-orcidShavit, JA
dc.identifier.name-orcidLiu, AP; 0000-0002-0309-7018
dc.identifier.name-orcidPrevis, MJ
dc.identifier.name-orcidDay, SM
dc.working.doi10.7302/6726en
dc.owningcollnameInternal Medicine, Department of


Files in this item

Show simple item record

Remediation of Harmful Language

The University of Michigan Library aims to describe library materials in a way that respects the people and communities who create, use, and are represented in our collections. Report harmful or offensive language in catalog records, finding aids, or elsewhere in our collections anonymously through our metadata feedback form. More information at Remediation of Harmful Language.

Accessibility

If you are unable to use this file in its current format, please select the Contact Us link and we can modify it to make it more accessible to you.