Alterations in sarcomeric protein expression, phosphorylation and contractile function in hypertrophic cardiomyopathy patients carrying a founder mutation in myosin binding protein C

2008 ◽  
Vol 7 ◽  
pp. 30-31
Author(s):  
S VANDIJK ◽  
D DOOIJES ◽  
D DEKKERS ◽  
J LAMERS ◽  
F TENCATE ◽  
...  
Open Heart ◽  
2020 ◽  
Vol 7 (1) ◽  
pp. e001220
Author(s):  
Berglind Adalsteinsdottir ◽  
Michael Burke ◽  
Barry J Maron ◽  
Ragnar Danielsen ◽  
Begoña Lopez ◽  
...  

ObjectiveThe myosin-binding protein C (MYBPC3) c.927-2A>G founder mutation accounts for >90% of sarcomeric hypertrophic cardiomyopathy (HCM) in Iceland. This cross-sectional observational study explored the penetrance and phenotypic burden among carriers of this single, prevalent founder mutation.MethodsWe studied 60 probands with HCM caused by MYBPC3 c.927-2A>G and 225 first-degree relatives. All participants underwent comprehensive clinical evaluation and relatives were genotyped.ResultsGenetic and clinical evaluation of relatives identified 49 genotype-positive (G+) relatives with left ventricular hypertrophy (G+/LVH+), 59 G+without LVH (G+/LVH−) and 117 genotype-negative relatives (unaffected). Compared with HCM probands, G+/LVH+ relatives were older at HCM diagnosis, had less LVH, a less prevalent diastolic dysfunction, fewer ECG abnormalities, lower serum N-terminal pro-B-type natriuretic peptide (NT-proBNP) and high-sensitivity cardiac troponin I levels, and fewer symptoms. The penetrance of HCM was influenced by age and sex; specifically, LVH was present in 39% of G+males but only 9% of G+females under age 40 years (p=0.015), versus 86% and 83%, respectively, after age 60 (p=0.89). G+/LVH− subjects had normal wall thicknesses, diastolic function and NT-proBNP levels, but subtle changes in LV geometry and more ECG abnormalities than their unaffected relatives.ConclusionsPhenotypic expression of the Icelandic MYBPC3 founder mutation varies by age, sex and proband status. Men are more likely to have LVH at a younger age, and disease manifestations were more prominent in probands than in relatives identified via family screening. G+/LVH− individuals had subtle clinical differences from unaffected relatives well into adulthood, indicating subclinical phenotypic expression of the pathogenic mutation.


2012 ◽  
Vol 111 (suppl_1) ◽  
Author(s):  
Md. Abdur Razzaque

Background: Myosin binding protein C (MyBP-C) is a thick filament protein consisting of 1274 amino acid residues (149kD) and mutations in the cardiac isoform (cardiac MyBP-C; cMyBP-C) are responsible for a substantial proportion (20-35%) of identified cases of familial hypertrophic cardiomyopathy (FHC). Recently we found a 40kD fragment is produced from cMyBP-C when the heart is stressed, using a stimulus such as ischemia reperfusion injury. This fragment can be detected in both the mouse and human heart and appears to be stable. Its ability to interfere with normal cardiac function is unexplored. Methods and Results: To understand the potential pathogenicity of the 40kd fragment in vivo, we generated cardiac myocyte-specific transgenic mice (TG) using a Tet-Off inducible system to permit controlled expression in cardiomyocytes. When 40kD protein expression is induced by crossing the responder animals with tetracycline transactivator (tTA) mice, the double TG mice show protein expression and, subsequently, sarcomere dysgenesis and altered cardiac geometry. The double transgenic heart fails between 3 to 17 weeks of age. Expression, the fragment in cardiomyocytes led to development of significant cardiac hypertrophy with myofibrillar disarray and fibrosis. Subsequent analyses showed that MEK-ERK hypertrophic signaling pathways were activated. To determine the role of this pathway in the pathogenic response being generated, we subjected an experimental cohort of animals to treatment with the MAPK/ERK kinase inhibitor U0126 during pregnancy. The drug effectively improved heart function and prolonged survival as compared to the untreated control cohort. Conclusions: The data show that a 40kD fragment of cMyBP-C, which is generated during the development of heart disease in both the mouse and human, is a pathogenic fragment whose presence leads to hypertrophic cardiomyopathy and heart failure. Blockade of the MEK-ERK pathway was effective therapeutically in decreasing morbidity and increasing lifespan in the face of continued synthesis of the fragment.


Circulation ◽  
1999 ◽  
Vol 100 (4) ◽  
pp. 446-449 ◽  
Author(s):  
Yoshinori L. Doi ◽  
Hiroaki Kitaoka ◽  
Nobuhiko Hitomi ◽  
Manatsu Satoh ◽  
Akinori Kimura

2010 ◽  
Vol 56 (1) ◽  
pp. 59-65 ◽  
Author(s):  
Takayoshi Hirota ◽  
Toru Kubo ◽  
Hiroaki Kitaoka ◽  
Tomoyuki Hamada ◽  
Yuichi Baba ◽  
...  

2019 ◽  
Vol 73 (9) ◽  
pp. 932
Author(s):  
Berglind Adalsteinsdottir ◽  
Michael Burke ◽  
Carolyn Ho ◽  
Barry Maron ◽  
Ragnar Danielsen ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document