Estrogen regulation of cardiac cAMP-L-type Ca2+ channel pathway modulates sex differences in basal contraction and responses to β2AR-mediated stress in left ventricular apical myocytes

Cell Commun Signal. 2019 Apr 15;17(1):34. doi: 10.1186/s12964-019-0346-2.

Abstract

Backgrounds/aim: Male and female hearts have many structural and functional differences. Here, we investigated the role of estrogen (E2) in the mechanisms of sex differences in contraction through the cAMP-L-type Ca2+channel pathway in adult mice left ventricular (LV) apical myocytes at basal and stress state.

Methods: Isolated LV apical myocytes from male, female (Sham) and ovariectomised mice (OVX) were used to investigate contractility, Ca2+ transients and L-type Ca2+ channel (LTCC) function. The levels of β2AR, intracellular cAMP, phosphodiesterase (PDE 3 and PDE 4), RyR2, PLB, SLN, and SERCA2a were compared among the experimental groups.

Results: We found that (1) intracellular cAMP, ICaL density, contraction and Ca2+ transient amplitudes were larger in Sham and OVX + E2 myocytes compared to male and OVX. (2) The mRNA expression of PDE 3 and 4 were lower in Sham and OVX + E2 groups compared with male and OVX groups. Treatment of myocytes with IBMX (100 μM) increased contraction and Ca2+ transient amplitude in both sexes and canceled differences between them. (3) β2AR-mediated stress decreased cAMP concentration and peak contraction and Ca2+ transient amplitude only in male and OVX groups but not in Sham or OVX + E2 groups suggesting a cardioprotective role of E2 in female mice. (4) Pretreatment of OVX myocytes with GPR30 antagonist G15 (100 nM) abolished the effects of E2, but ERα and ERβ antagonist ICI 182,780 (1 μM) did not. Moreover, activation of GPR30 with G1 (100 nM) replicated the effects of E2 on cAMP, contraction and Ca2+ transient amplitudes suggesting that the acute effects of E2 were mediated by GPR30 via non-genomic signaling. (5) mRNA expression of RyR2 was higher in myocytes from Sham than those of male while PLB and SLN were higher in male than Sham but no sex differences were observed in the mRNA of SERCA2a.

Conclusion: Collectively, these results demonstrate that E2 modulates the expression of genes related to the cAMP-LTCC pathway and contributes to sex differences in cardiac contraction and responses to stress. We also show that estrogen confers cardioprotection against cardiac stress by non-genomic acute signaling via GPR30.

Keywords: 17β-estradiol; Ca2+ transients; Cardiac stress; L-type Ca2+ channel; Phosphodiesterase; Sex differences in contractility.

Publication types

  • Comparative Study
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Calcium Channels, L-Type / metabolism*
  • Calcium Channels, T-Type / metabolism
  • Calcium Signaling
  • Cardiotonic Agents / pharmacology
  • Cyclic AMP / metabolism*
  • Cyclic Nucleotide Phosphodiesterases, Type 3 / metabolism
  • Cyclic Nucleotide Phosphodiesterases, Type 4 / metabolism
  • Estradiol / pharmacology
  • Estradiol / physiology*
  • Female
  • Gene Expression Regulation
  • Male
  • Mice
  • Myocardial Contraction / drug effects
  • Myocardial Contraction / physiology*
  • Myocytes, Cardiac / drug effects
  • Myocytes, Cardiac / physiology*
  • Receptors, Adrenergic, beta-1 / metabolism
  • Receptors, Adrenergic, beta-2 / metabolism*
  • Receptors, Estrogen / metabolism
  • Receptors, G-Protein-Coupled / agonists
  • Receptors, G-Protein-Coupled / metabolism
  • Sex Characteristics*
  • Ventricular Function, Left / drug effects
  • Ventricular Function, Left / physiology*

Substances

  • ADRB2 protein, mouse
  • Adrb1 protein, mouse
  • Calcium Channels, L-Type
  • Calcium Channels, T-Type
  • Cardiotonic Agents
  • GPER1 protein, mouse
  • Receptors, Adrenergic, beta-1
  • Receptors, Adrenergic, beta-2
  • Receptors, Estrogen
  • Receptors, G-Protein-Coupled
  • Estradiol
  • Cyclic AMP
  • Cyclic Nucleotide Phosphodiesterases, Type 3
  • Cyclic Nucleotide Phosphodiesterases, Type 4