P-343 Potential Role of the Ubiquitin proteasome System in Hypertrophic Cardiomyopathy Caused by Mutant Cardiac Myosin-Binding Protein C

2009 ◽  
Vol 4 ◽  
pp. S149
Author(s):  
Udin Bahrudin ◽  
Hiroko Morisaki ◽  
Takayuki Morisaki ◽  
Osamu Igawa ◽  
Seiji Takashima ◽  
...  
2019 ◽  
Vol 116 (3) ◽  
pp. 466a-467a
Author(s):  
Darshan V. Trivedi ◽  
Saswata S. Sarkar ◽  
Arjun S. Adhikari ◽  
Makenna M. Morck ◽  
Kristina B. Kooiker ◽  
...  

Circulation ◽  
2007 ◽  
Vol 116 (suppl_16) ◽  
Author(s):  
Giulia Mearini ◽  
Christina Gedicke ◽  
Elisabeth Krämer ◽  
Friedrich Buck ◽  
Peirang Cao ◽  
...  

Background. Familial hypertrophic cardiomyopathy (FHC) is a sarcomeropathy frequently associated with cardiac myosin-binding protein C (cMyBP-C) gene mutations. Most of them result in a frameshift and C-terminal truncated proteins. However, truncated mutants were not detected in myocardial tissue of FHC patients. A recent study showed that a truncated cMyBP-C (M7t) is rapidly and quantitatively degraded by the ubiquitin-proteasome system (UPS) after adenoviral gene transfer in neonatal rat cardiac myocytes (NRCM). Since the diversity and specificity of UPS regulation lies in E3 ubiquitin ligases, which specifically target proteins and direct the ubiquitination process, the present study investigated whether the sarcomere-specific E3 ligases, atrogin-1 and MuRF1, mediate degradation of wild-type (WT) and truncated M7t cMyBP-C. Methods and Results. NRCM were co-infected with atrogin-1 and WT or M7t cMyBP-C adenoviruses. Both overexpressed WT and M7t were co-immunoprecipitated with atrogin-1 suggesting that both forms of cMyBP-C interact with atrogin-1. However, whereas the amount of overexpressed WT cMyBP-C did not change, the M7t protein level was 80% lower in NRCM infected with atrogin-1 than without. This suggests that atrogin-1 is the E3 ligase mediating proteasome degradation of truncated M7t cMyBP-C only. We then investigated whether MuRF1 could regulate the level of endogenous cMyBP-C in MuRF1-KO mice and compared to WT. The heart to body weight ratio and the chymotrypsine-like activity of the proteasome were similar in KO and WT mice. The mRNA levels of cMyBP-C, and other E3 ligases, atrogin-1 and MuRF3, were similar between KO and WT ventricles. Strikingly, the protein level of cMyBP-C was 150% and 30% higher in neonatal and 24 wks-old KO compared to age-matched WT mice. Conclusion . These data suggest that atrogin-1, located at the Z-band, mediates the proteasome degradation of truncated M7t cMyBP-C resulting from a FHC mutation, the lack of MuRF1 is not compensated by other E3 ligases and is not associated with hypertrophy in KO mice, and most importantly, MuRF1, mainly located at the M-band, specifically regulates the protein level of cMyBP-C.


2017 ◽  
Vol 121 (suppl_1) ◽  
Author(s):  
Amelia A Glazier ◽  
Adam Helms ◽  
Neha Hafeez ◽  
Srisha Kotlo ◽  
Jaime Yob ◽  
...  

The most commonly mutated gene in hypertrophic cardiomyopathy (HCM) is cardiac myosin binding protein C (MYBPC3). Over 90% of MYBPC3 mutations are nonsense, but whether these mutations manifest in loss- or gain-of-function is unresolved. Evidence suggests MYBPC3 mutants impact protein quality control mechanisms. The objective of this study was to evaluate interactions of MYBPC3 with proteostatic systems and test the hypothesis that these interactions affect protein homeostasis in cardiomyocytes. WT and mutant MYBPC3 constructs were expressed in neonatal rat ventricular myocytes (NRVMs) via adenovirus. Mutant MYBPC3 induced ubiquitin proteasome system reporter GFPu accumulation (fold increase in GFPu-positive cells vs control: WT 138±14.0%, mutant 198±27.2%, mean±SEM, p<0.05 vs control and WT), indicating proteasome dysfunction. Affinity purification/mass spectrometry identified molecular chaperones Hsp70 and Hsc70 as prominent interactors with MYBPC3. We observed MYBPC3 degradation by cycloheximide chase in response to Hsc70 siRNA knockdown or pharmacological treatment with Hsp70 activator YM-1. Hsc70 knockdown slowed degradation of WT and mutant MYBPC3 (WT control t ½ =5.47±0.70 hr, WT Hsc70 knockdown t ½ =13.5±1.62 hr; mutant control t ½ =3.42±0.61 hr, mutant Hsc70 knockdown t ½ =9.87±0.95 hr), while YM-1 treatment accelerated degradation (WT DMSO t ½ =10.2±3.28 hr, WT YM-1 t ½ =3.16±0.61 hr; mutant DMSO t ½ =11.7±2.67 hr, mutant YM-1 t ½ =1.37±0.16 hr). We then evaluated whether transferrin uptake via clathrin mediated endocytosis, a critical Hsc70-dependent activity, was affected by mutant MYBPC3. Transferrin uptake was significantly decreased in NRVMs expressing mutant MYBPC3 compared to WT and untreated controls (transferrin-positive cells: control 22.93±3.34%, WT 17.47±0.70%, mutant 9.30±1.63%, mean±SEM, p<0.05 vs control and WT). In conclusion, we have demonstrated that Hsp70 chaperones interact with MYBPC3 in cardiomyocytes and affect MYBPC3 degradation, suggesting MYBPC3 is a client of Hsp70 and Hsc70. Additionally, expression of mutant MYBPC3 causes ubiquitin proteasome impairment and interferes with normal Hsc70 function. These results support our hypothesis that mutant MYBPC3 affects protein homeostasis in HCM.


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

Sign in / Sign up

Export Citation Format

Share Document