Nitric Oxide Synthase 1 Modulates Basal and β-Adrenergic-Stimulated Contractility by Rapid and Reversible Redox-Dependent S-Nitrosylation of the Heart

PLoS One. 2016 Aug 16;11(8):e0160813. doi: 10.1371/journal.pone.0160813. eCollection 2016.

Abstract

S-nitrosylation of several Ca2+ regulating proteins in response to β-adrenergic stimulation was recently described in the heart; however the specific nitric oxide synthase (NOS) isoform and signaling pathways responsible for this modification have not been elucidated. NOS-1 activity increases inotropism, therefore, we tested whether β-adrenergic stimulation induces NOS-1-dependent S-nitrosylation of total proteins, the ryanodine receptor (RyR2), SERCA2 and the L-Type Ca2+ channel (LTCC). In the isolated rat heart, isoproterenol (10 nM, 3-min) increased S-nitrosylation of total cardiac proteins (+46±14%) and RyR2 (+146±77%), without affecting S-nitrosylation of SERCA2 and LTCC. Selective NOS-1 blockade with S-methyl-L-thiocitrulline (SMTC) and Nω-propyl-l-arginine decreased basal contractility and relaxation (-25-30%) and basal S-nitrosylation of total proteins (-25-60%), RyR2, SERCA2 and LTCC (-60-75%). NOS-1 inhibition reduced (-25-40%) the inotropic response and protein S-nitrosylation induced by isoproterenol, particularly that of RyR2 (-85±7%). Tempol, a superoxide scavenger, mimicked the effects of NOS-1 inhibition on inotropism and protein S-nitrosylation; whereas selective NOS-3 inhibitor L-N5-(1-Iminoethyl)ornithine had no effect. Inhibition of NOS-1 did not affect phospholamban phosphorylation, but reduced its oligomerization. Attenuation of contractility was abolished by PKA blockade and unaffected by guanylate cyclase inhibition. Additionally, in isolated mouse cardiomyocytes, NOS-1 inhibition or removal reduced the Ca2+-transient amplitude and sarcomere shortening induced by isoproterenol or by direct PKA activation. We conclude that 1) normal cardiac performance requires basal NOS-1 activity and S-nitrosylation of the calcium-cycling machinery; 2) β-adrenergic stimulation induces rapid and reversible NOS-1 dependent, PKA and ROS-dependent, S-nitrosylation of RyR2 and other proteins, accounting for about one third of its inotropic effect.

MeSH terms

  • Animals
  • Calcium / metabolism
  • Calcium-Binding Proteins / chemistry
  • Calcium-Binding Proteins / metabolism
  • Heart / drug effects
  • Heart / physiology*
  • Isoproterenol / pharmacology
  • Male
  • Myocardial Contraction* / drug effects
  • Myocardium / cytology
  • Myocardium / metabolism*
  • Nitric Oxide Synthase Type I / metabolism*
  • Oxidation-Reduction / drug effects
  • Phosphorylation / drug effects
  • Protein Multimerization / drug effects
  • Protein Processing, Post-Translational / drug effects
  • Protein Structure, Quaternary
  • Rats
  • Rats, Sprague-Dawley
  • Reactive Oxygen Species / metabolism
  • Receptors, Adrenergic, beta / metabolism*
  • S-Nitrosothiols / metabolism*

Substances

  • Calcium-Binding Proteins
  • Reactive Oxygen Species
  • Receptors, Adrenergic, beta
  • S-Nitrosothiols
  • phospholamban
  • Nitric Oxide Synthase Type I
  • Isoproterenol
  • Calcium

Grants and funding

Supported in part by Grants 1090757 (M.P. Boric), 1120595 and 1150662 (D.R. Gonzalez) and Anillos Act71 from Fondecyt (Chilean National Fund for Science and Technology).