Contribution of glucocorticoid–mineralocorticoid receptor pathway on the obesity-related adipocyte dysfunction

https://doi.org/10.1016/j.bbrc.2012.01.139Get rights and content

Abstract

Aims

Mineralocorticoid receptor (MR) blockade ameliorated insulin resistance with improvements in adipocytokine dysregulation, inflammation, and excess of reactive oxygen species (ROS) in obese adipose tissue and adipocytes, but its mechanism has not been clarified. The 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), producing active glucocorticoids, is highly expressed in adipocytes and glucocorticoids bind to MR with higher affinity than to glucocorticoid receptor (GR). We investigated whether glucocorticoids effect on adipocytokines and ROS through MR in adipocytes. In addition, fat distributions of MR and GR were investigated in human subjects.

Methods and Results

Corticoid receptors and their target genes were examined in adipose tissue of obese db/db mice. 3T3-L1 adipocytes were treated with glucocorticoids, H2O2, MR antagonist eplerenone (EP), GR antagonist RU486 (RU), MR-siRNA, and/or N-acetylcysteine. Human adipose tissues were obtained from seven patients who underwent abdominal surgery. The mRNA levels of MR and its target gene were higher in db/db mice than in control db/m + mice. In 3T3-L1 adipocytes, glucocorticoids, similar to H2O2, caused the dysregulation of mRNA levels of various genes related to adipocytokines and the increase of intracellular ROS. Such changes were rectified by MR blockade, not by GR antagonist. In human fat, MR mRNA level was increased in parallel with the increase of body mass index (BMI) and its increase was more significant in visceral fat, while there were no apparent correlations of GR mRNA level to BMI or fat distribution.

Conclusion

Glucocorticoid-MR pathway may contribute to the obesity-related adipocytokine dysregulation and adipose ROS.

Highlights

Glucocorticoids induced adipocytokine dysregulation and ROS in adipocytes. ► Such changes were rectified by MR blockade, not by GR antagonist. ► In human, adipose MR was increased in parallel with body mass index. ► Glucocorticoid-MR pathway may contribute to the obesity-related disorders.

Introduction

Accumulating clinical evidence indicates that excessive accumulation of visceral fat is closely associated with obesity-related metabolic disorders [1]. Our group demonstrated that overproduction of reactive oxygen species (ROS) in obese fat tissue leads to the dysregulation of adipocytokines and accelerates the development of metabolic disorder [2].

Several studies demonstrated that activation of MR promotes inflammation, proliferation, and fibrosis via ROS generation. As we and other group previously indicated [3], [4], mineralocorticoid receptor (MR) blockade by MR antagonist eplerenone (EP) ameliorated insulin resistance and adipocytokine dysregulation, and reduced ROS and macrophage infiltration in adipose tissue of obese mice. In adipocytes, MR activation caused the increase of ROS and the adipocytokine dysregulation, while MR blockade improved such changes [3]. Collectively, adipose MR may play an important role in metabolic disorder in the development of obesity.

The MR binds not only to aldosterone but also to glucocorticoid with equal affinity [5]. In addition, the affinities of glucocorticoids (cortisol in humans, corticosterone in rodents) for MR are 10-fold higher than those for glucocorticoid receptor (GR) [6]. 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), which converts cortisone (an inactive corticoid) into cortisol (an active corticoid), is highly expressed in adipocytes [7]. These evidences suggest that glucocorticoid action could be exhibited mainly through MR in adipocytes, but adipose glucocorticoid-MR pathway has not been fully elucidated. We here tested the glucocorticoid action in adipocytes by using antagonists for MR and GR. The effect of glucocorticoid on adipocytokines and adipose ROS was also examined. In addition, mRNA expressions for important proteins, including MR, GR, PPARγ2, NADPH oxidase subunit p22, adiponectin, and 11β-HSD1, were directly compared in subcutaneous and visceral fat of human subjects, according to their BMI.

Section snippets

Animals

Male BKS.Cg-m+/+Leprdb/J (db/db) obese mice were purchased from Charles River Laboratories (Charles River Japan Inc., Yokohama, Japan). Their respective lean control male db/m + heterozygous littermates were purchased from the same supplier. Mice were sacrificed at 10 weeks of age by intraperitoneous administration of 5% pentobarbital in one shot. The experimental protocol was approved by the Ethics Review Committee for Animal Experimentation of Osaka University School of Medicine. This study also

Expression of corticoid receptors and their target genes in obese model mice

The mRNA levels of corticoid receptors and their target genes were examined in white adipose tissue (WAT) of db/db and db/m + mice (Fig. 1). MR mRNA level of db/db mice was significantly higher than that of db/m + mice (Fig. 1A). In parallel with the change of MR, serum- and glucocorticoid-induced kinase 1 (Sgk1), a MR target gene [12], was also significantly increased in db/db mice (Fig. 1B). On the other hand, both GR and regulated in DNA damage and development 1 (REDD1), a GR target gene [13],

Discussion

The main findings of the present study were: (1) MR and its target gene mRNA levels were significantly increased in obese adipose tissues. (2) Physiological concentration of glucocorticoid caused the increase of adipose ROS and the adipocytokine dysregulation. (3) Glucocorticoid-induced ROS and adipocytokine dysregulation were significantly reversed by blockade of MR, not by GR antagonism. (4) In human visceral adipose tissue, MR mRNA level was increased as the BMI increased, while no apparent

Funding

This work was supported by Grants-in-Aid for Scientific Research (C) No. 22590979 (to N. M.), and Scientific Research on Innovative Areas No. 22126008 (to T. F.).

Acknowledgments

We are grateful to Pfizer Inc (New York, NY) for providing eplerenone. We thank Miyuki Nakamura for the excellent technical assistance. We also thank all members of the III laboratory (Adiposcience laboratory), Department of Metabolic Medicine, Osaka University for helpful discussions on the project.

References (24)

  • N. Maeda et al.

    PPAR gamma ligands increase expression and plasma concentrations of adiponectin, an adipose-derived protein

    Diabetes

    (2001)
  • H. Hitomi et al.

    Aldosterone suppresses insulin signaling via the downregulation of insulin receptor substrate-1 in vascular smooth muscle cells

    Hypertension

    (2007)
  • Cited by (60)

    • Oxidative Stress: A Unifying Paradigm in Hypertension

      2020, Canadian Journal of Cardiology
    • The beneficial effects of N-acetyl cysteine (NAC) against obesity associated complications: A systematic review of pre-clinical studies

      2019, Pharmacological Research
      Citation Excerpt :

      Data presented supported an equal or greater efficacy for NAC when compared to these antidiabetic drugs in reducing lipid accumulation, improving insulin sensitivity, and blocking ROS production in cultured adipocytes. Moreover, similar outcome was observed in other studies that tested the comparative effects of NAC with anti-inflammatory agents such as pyrrolidine [76], including corticosterone hormones [77]. These studies reported that NAC could limit lipopolysaccharide and TNFα induced nitric oxide production by inhibiting nitric oxide synthase and NF-κB, while reducing excess ROS production.

    • Obesity-Related Heart Failure With a Preserved Ejection Fraction: The Mechanistic Rationale for Combining Inhibitors of Aldosterone, Neprilysin, and Sodium-Glucose Cotransporter-2

      2018, JACC: Heart Failure
      Citation Excerpt :

      Hyperaldosteronism not only causes sodium retention, but also promotes the accumulation and inflammation of epicardial adipose tissue and the development of microvascular rarefaction and fibrosis in the underlying cardiac muscle (45–50). The transformation of perivisceral fat to a maladaptive proinflammatory phenotype may depend on mineralocorticoid receptor signaling (46–48). Mechanistic studies and clinical trials support a role for mineralocorticoid receptor antagonists in obesity-related HFpEF (Central Illustration).

    View all citing articles on Scopus
    View full text