4.7 Article

Oestrogen receptor α in T cells controls the T cell immune profile and glucose metabolism in mouse models of gestational diabetes mellitus

期刊

DIABETOLOGIA
卷 64, 期 7, 页码 1660-1673

出版社

SPRINGER
DOI: 10.1007/s00125-021-05447-x

关键词

Gestational diabetes mellites; Hepatokines; Insulin secretion; Interleukin 17; Maternal beta cell adaptation; Oestrogen receptor a; Pancreatic islet; Th17; Treg

资金

  1. Japan Society for the Promotion of Science (JSPS KAKENHI) [JP15K09410, JP15K15599, JP18K08469, JP19H05011, JP19K08997]
  2. Grant for Research Activities of the First Bank of Toyama Scholarship Foundation
  3. Mitsubishi Tanabe Pharma Corporation

向作者/读者索取更多资源

This study using a mouse model demonstrates that deletion of ER alpha in T cells results in impaired maternal adaptation of insulin secretion, changes in hepatokine profiles, and enhanced chronic inflammation in gWAT, shedding light on the pathogenesis of GDM.
Aims/hypothesis The imbalance between maternal insulin resistance and a relative lack of insulin secretion underlies the pathogenesis of gestational diabetes mellitus (GDM). Alterations in T cell subtypes and increased levels of circulating proinflammatory cytokines have been proposed as potential mechanisms underlying the pathophysiology of insulin resistance in GDM. Since oestrogen modulates T cell immunity, we hypothesised that oestrogen plays a homeostatic role in visceral adipose tissue by coordinating T cell immunity through oestrogen receptor alpha (ER alpha) in T cells to prevent GDM. Methods Female CD4-cre ER alpha(fl/fl) (KO) mice on a C57BL/6 background with ER alpha ablation specifically in T cells, and ER alpha(fl/fl) (ER alpha-floxed [FL]) mice were fed 60 kJ% high-fat diet (HFD) for 4 weeks. Female mice mated with male BALB/c mice to achieve allogenic pregnancy and were maintained on an HFD to generate the GDM model. Mice were divided into four experimental groups: non-pregnant FL, non-pregnant KO, pregnant FL (FL-GDM) and pregnant KO (KO-GDM). GTTs and ITTs were performed on day 12.5 or 13.5 and 16.5 after breeding, respectively. On day 18.5 after breeding, mice were killed and T cell subsets in the gonadal white adipose tissue (gWAT) and spleen were analysed using flow cytometry. Histological examination was also conducted and proinflammatory gene expression in gWAT and the liver was evaluated. Results KO mice that mated with BALB/c mice showed normal fertility rates and fetal weights as compared with FL mice. Body and tissue weights were similar between FL and KO mice. When compared with FL-GDM mice, KO-GDM mice showed decreased insulin secretion (serum insulin concentration 15 min after glucose loading: 137.3 +/- 18.3 pmol/l and 40.1 +/- 36.5 pmol/l, respectively; p < 0.05), impaired glucose tolerance (glucose AUC in GTT: 2308.3 +/- 54.0 mmol/l x min and 2620.9 +/- 122.1 mmol/l x min, respectively; p < 0.05) and increased numbers of T helper (Th)17 cells in gWAT (0.4 +/- 0.0% vs 0.8 +/- 0.1%; p < 0.05). However, the contents of Th1 and regulatory T cells (Tregs) in gWAT remained similar between FL-GDM and KO-GDM. Glucose-stimulated insulin secretion was similar between isolated islets derived from FL and KO mice, but was reduced by IL-17A treatment. Moreover, the levels of proinflammatory gene expression, including expression of Emr1 and Tnfa in gWAT, were significantly higher in KO-GDM mice than in FL-GDM mice (5.1-fold and 2.7-fold, respectively; p < 0.01 for both). Furthermore, KO-GDM mice showed increased expression of genes encoding hepatokines, Ahsg and Fgf21 (both were 2.4-fold higher vs FL-GDM mice; p < 0.05 and p = 0.09, respectively), with no changes in inflammatory gene expression (e.g., Tnfa and Ifng) in the liver compared with FL-GDM mice. Conclusions/interpretation Deletion of ER alpha in T cells caused impaired maternal adaptation of insulin secretion, changes in hepatokine profiles, and enhanced chronic inflammation in gWAT alongside an abnormal increase in Th17 cells. These results suggest that the ER alpha-mediated oestrogen signalling effects in T cells regulate T cell immunity and contribute to glucose homeostasis in pregnancy.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据