Login / Signup

Hepatic thyroid hormone signalling modulates glucose homeostasis through the regulation of GLP-1 production via bile acid-mediated FXR antagonism.

Ying YanZhoumin NiuChao SunPeng LiSiyi ShenShengnan LiuYuting WuChuyu YunTingying JiaoSheng JiaYuying LiZhong-Ze FangLin ZhaoJiqiu WangCarol XieChang-Tao JiangYuying LiXiaoyun FengCheng HuJing-Jing JiangHao Ying
Published in: Nature communications (2022)
Thyroid hormones (TH) regulate systemic glucose metabolism through incompletely understood mechanisms. Here, we show that improved glucose metabolism in hypothyroid mice after T3 treatment is accompanied with increased glucagon-like peptide-1 (GLP-1) production and insulin secretion, while co-treatment with a GLP-1 receptor antagonist attenuates the effects of T3 on insulin and glucose levels. By using mice lacking hepatic TH receptor β (TRβ) and a liver-specific TRβ-selective agonist, we demonstrate that TRβ-mediated hepatic TH signalling is required for both the regulation of GLP-1 production and the insulinotropic and glucose-lowering effects of T3. Moreover, administration of a liver-targeted TRβ-selective agonist increases GLP-1 and insulin levels and alleviates hyperglycemia in diet-induced obesity. Mechanistically, T3 suppresses Cyp8b1 expression, resulting in increased the levels of Farnesoid X receptor (FXR)-antagonistic bile acids, thereby potentiating GLP-1 production and insulin secretion by repressing intestinal FXR signalling. T3 correlates with both plasma GLP-1 and fecal FXR-antagonistic bile acid levels in people with normal thyroid function. Thus, our study reveals a role for hepatic TH signalling in glucose homeostasis through the regulation of GLP-1 production via bile acid-mediated FXR antagonism.
Keyphrases
  • type diabetes
  • blood glucose
  • high fat diet induced
  • metabolic syndrome
  • poor prognosis
  • blood pressure
  • drug delivery
  • mouse model
  • long non coding rna
  • body mass index