Triiodothyronine maintains cardiac transverse-tubule structure and function

https://doi.org/10.1016/j.yjmcc.2021.06.010Get rights and content
Under a Creative Commons license
open access

Highlights

  • Thyroid hormone deficient hearts have disorganized T-tubules, impaired EC coupling

  • Thyroid deficiency causes decreased rates of Ca2+ release/re-uptake, reduced contractility

  • STORM imaging shows reduced RyR2 cluster number and size in T3-deficient myocytes

  • T3 induces Jph2 expression, improves T-tubule organization, increases RyR clusters

Abstract

Subclinical hypothyroidism and low T3 syndrome are commonly associated with an increased risk of cardiovascular disease (CVD) and mortality. We examined effects of T3 on T-tubule (TT) structures, Ca2+ mobilization and contractility, and clustering of dyadic proteins.

Thyroid hormone (TH) deficiency was induced in adult female rats by propyl-thiouracil (PTU; 0.025%) treatment for 8 weeks. Rats were then randomized to continued PTU or triiodo-L-thyronine (T3; 10 μg/kg/d) treatment for 2 weeks (PTU + T3). After in vivo echocardiographic and hemodynamic recordings, cardiomyocytes (CM) were isolated to record Ca2+ transients and contractility. TT organization was assessed by confocal microscopy, and STORM images were captured to measure ryanodine receptor (RyR2) cluster number and size, and L-type Ca2+ channel (LTCC, Cav1.2) co-localization. Expressed genes including two integral TT proteins, junctophilin-2 (Jph-2) and bridging integrator-1 (BIN1), were analyzed in left ventricular (LV) tissues and cultured CM using qPCR and RNA sequencing.

The T3 dosage used normalized serum T3, and reversed adverse effects of TH deficiency on in vivo measures of cardiac function. Recordings of isolated CM indicated that T3 increased rates of Ca2+ release and re-uptake, resulting in increased velocities of sarcomere shortening and re-lengthening. TT periodicity was significantly decreased, with reduced transverse tubules but increased longitudinal tubules in TH-deficient CMs and LV tissue, and these structures were normalized by T3 treatment. Analysis of STORM data of PTU myocytes showed decreased RyR2 cluster numbers and RyR localizations within each cluster without significant changes in Cav1.2 localizations within RyR clusters. T3 treatment normalized RyR2 cluster size and number. qPCR and RNAseq analyses of LV and cultured CM showed that Jph2 expression was T3-responsive, and its increase with treatment may explain improved TT organization and RyR-LTCC coupling.

Keywords

Transverse-tubules
STORM
Ca2+
Junctophilin-2
Thyroid hormone
Ryanodine receptors

Abbreviations

BIN1
Bridging integrator-1 or amphiphysin 2
Jph2
Junctophilin 2
BDM
2,3-butanedione monoxime
DAPI
4′,6-diamidino-2-phenylindole
ECC
Excitation contraction coupling, EC coupling
CVD
Cardiovascular diseases
HF
Heart failure
LV
Left ventricle
Cav1.2
Calcium channel voltage-dependent L-type alpha 1C subunit
LTCC
L-type calcium channels
RyR2
Ryanodine receptor 2
SR
Sarco-(endo)plasmic reticulum
SERCA
Sarcoplasmic reticulum calcium-ATPase
LT3S
Low T3-syndrome
T3
Triiodo-L-thyronine
T4
Thyroxine
TT or T-tubules
Transverse-tubules
WGA
Wheat germ agglutinin

Cited by (0)

1

Present address: Jiangsu Hengrui Medicine Co. Ltd., China

2

Present address: Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Key Laboratory of Molecular Cardiovascular Science, Beijing, China.