Pathogenic properties of the N-terminal region of cardiac myosin binding protein-C in vitro

J Muscle Res Cell Motil. 2012 May;33(1):17-30. doi: 10.1007/s10974-012-9292-y. Epub 2012 Apr 17.

Abstract

Cardiac myosin binding protein-C (cMyBP-C) plays a role in sarcomeric structure and stability, as well as modulating heart muscle contraction. The 150 kDa full-length (FL) cMyBP-C has been shown to undergo proteolytic cleavage during ischemia-reperfusion injury, producing an N-terminal 40 kDa fragment (mass 29 kDa) that is predominantly associated with post-ischemic contractile dysfunction. Thus far, the pathogenic properties of such truncated cMyBP-C proteins have not been elucidated. In the present study, we hypothesized that the presence of these 40 kDa fragments is toxic to cardiomyocytes, compared to the 110 kDa C-terminal fragment and FL cMyBP-C. To test this hypothesis, we infected neonatal rat ventricular cardiomyocytes and adult rabbit ventricular cardiomyocytes with adenoviruses expressing the FL, 110 and 40 kDa fragments of cMyBP-C, and measured cytotoxicity, Ca(2+) transients, contractility, and protein-protein interactions. Here we show that expression of 40 kDa fragments in neonatal rat ventricular cardiomyocytes significantly increases LDH release and caspase 3 activity, significantly reduces cell viability, and impairs Ca(2+) handling. Adult cardiomyocytes expressing 40 kDa fragments exhibited similar impairment of Ca(2+) handling along with a significant reduction of sarcomere length shortening, relaxation velocity, and contraction velocity. Pull-down assays using recombinant proteins showed that the 40 kDa fragment binds significantly to sarcomeric actin, comparable to C0-C2 domains. In addition, we discovered several acetylation sites within the 40 kDa fragment that could potentially affect actomyosin function. Altogether, our data demonstrate that the 40 kDa cleavage fragments of cMyBP-C are toxic to cardiomyocytes and significantly impair contractility and Ca(2+) handling via inhibition of actomyosin function. By elucidating the deleterious effects of endogenously expressed cMyBP-C N-terminal fragments on sarcomere function, these data contribute to the understanding of contractile dysfunction following myocardial injury.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Acetylation
  • Actins / metabolism
  • Actomyosin / metabolism
  • Adenoviridae / genetics
  • Adenoviridae / metabolism
  • Amino Acid Sequence
  • Animals
  • Calcium / metabolism
  • Cardiac Myosins / metabolism
  • Carrier Proteins / metabolism*
  • Cell Death
  • Cells, Cultured
  • Heart Ventricles / metabolism
  • Heart Ventricles / pathology
  • Immunoprecipitation
  • L-Lactate Dehydrogenase / metabolism
  • Mice
  • Molecular Sequence Data
  • Molecular Weight
  • Muscle Contraction
  • Myocardium / metabolism
  • Myocardium / pathology*
  • Myocytes, Cardiac / metabolism
  • Myocytes, Cardiac / pathology*
  • Peptide Fragments / metabolism
  • Protein Binding
  • Protein Interaction Mapping
  • Proteolysis
  • Rabbits
  • Rats
  • Rats, Sprague-Dawley
  • Reperfusion Injury / metabolism
  • Reperfusion Injury / pathology
  • Sarcomeres / metabolism
  • Sarcomeres / pathology

Substances

  • Actins
  • Carrier Proteins
  • Peptide Fragments
  • myosin-binding protein C
  • Actomyosin
  • L-Lactate Dehydrogenase
  • Cardiac Myosins
  • Calcium