Histone deacetylase 3-selective inhibitor RGFP966 ameliorates impaired glucose tolerance through β-cell protection

https://doi.org/10.1016/j.taap.2020.115189Get rights and content

Highights

  • RGFP966 ameliorated blood glucose level in low-dose of STZ-induced pre-diabetic mice.

  • RGFP966 improved β cell function in low-dose of STZ-induced pre-diabetic mice.

  • RGFP966 promoted insulin secretion in vivo and in vitro.

  • RGFP966 partially attenuated apoptosis induced by PA via HDAC3/Erk1/2 /caspase 3 in vitro.

Abstract

The potential therapeutic effect of histone deacetylase 3 (HDAC3) pharmacologic inhibition on diabetes has been focused recently. RGFP966, as a highly-selective HDAC3 inhibitor, its possible roles and underlying mechanism in the treatment of diabetes needs to be clarified. In this study, low-dose streptozotocin (STZ)-induced pre-diabetic mice were used to test the regulatory ability of RGFP966 in blood glucose and insulin. We isolated the islets both from normal C57BL/6 J mice and KKAy mice with spontaneous type 2 diabetes to determine the potency of RGFP966 on glucose-stimulated insulin secretion. NIT-1 pancreatic β-cells induced by sodium palmitate (PA) were applied to identify the protective effects of RGFP966 against β-cell apoptosis. The results showed that administration of RGFP966 in the pre-diabetic mice not only significantly reduced hyperglycemia, promoted phase I insulin secretion, improved morphology of islets, but also increased glucose infusion rate (GIR) during hyperglycemic clamp test. When treated in vitro, RGFP966 enhanced insulin secretion and synthesis in islets of normal C57BL/6J mice and diabetic KKAy mice. In addition, it partially attenuated PA-induced apoptosis in NIT-1 cells. Therefore, our research suggests that RGFP966, probably through selective inhibition of HDAC3, might serve as a novel potential preventive and therapeutic candidate for diabetes.

Introduction

Diabetes, as a chronic disease that causes hyperglycemia, poses a great threat to human health. β cells in the islets of the pancreas are the only type of cells that downregulate blood glucose levels by secreting insulin hormone. β-cell dysfunction is the main cause of onset and/or development in both type 1 and type 2 diabetes. In the stage of pre-diabetes, also known as impaired glucose tolerance, β-cell dysfunction is also present (Prentki and Nolan, 2006). However, to date, most small molecule anti-diabetic drugs have little effect on restoring the function of β cells.

Histone deacetylase 3 (HDAC3) is a member of the class I HDACs, which also includes HDAC1, HDAC2, and HDAC8. Histone deacetylases regulate gene expression through deacetylation of histone proteins, which modulate chromatin structure (Haberland et al., 2009; Verdin and Ott, 2015). As a class I HDAC, HDAC3 is the predominant HDAC associated with nuclear receptor co-repressors, silencing mediator of retinoic acid and thyroid hormone receptor (SMRT or NCoR2), and nuclear receptor co-repressor 1 (NCoR1 or NCoR). It is also the only class I HDAC found in endogenous SMRT and NCoR complexes (Guenther et al., 2000; Li et al., 2000; Wen et al., 2000). In addition, its catalytic function requires a physical interaction with the deacetylase-activating domain, a conserved domain in SMRT and NCoR proteins (Watson et al., 2012; You et al., 2013). In recent years, multiple studies have shown that HDAC3 inhibitors not only serve as anti-cancer agents, but can also treat metabolic disorders (Emmett et al., 2017; West and Johnstone, 2014). One study showed that BRD3308, a specific HDAC3 inhibitor, improved glycemia and insulin secretion in obese diabetic rats (Lundh et al., 2015). Deletion of HDAC3 in adult β cells can promote glucose tolerance via increasing insulin secretion (Remsberg et al., 2017).

In the past several years, the HDAC3-specific inhibitor RGFP966 has been shown to have multiple effects. It causes decreased cell growth in cutaneous T cell lymphoma by disrupting the DNA replication of rapidly cycling tumor cells (Wells et al., 2013); enhances extinction of drug-seeking behavior by influencing gene expression within the infralimbic cortex, hippocampus, and nucleus accumbens (Malvaez et al., 2013); may have an effective role in developing therapeutics for inflammatory lung diseases because of its anti-inflammatory properties (Leus et al., 2016); can prevent retinal ganglion cell loss following optic nerve crush (Schmitt et al., 2018); and can also prevent diabetes-induced aortic pathologies by regulating relative gene expression in the liver (Zhang et al., 2018a).

However, it remains unknown if RGFP966 has similar roles in β-cell function as other HDAC3 inhibitors such as BRD3308. The results of this study showed that in β cells, the HDAC3 specific-inhibitor RGFP966 not only promotes insulin secretion, but also prevents apoptosis induced by sodium palmitate (PA). Besides, it protects β cells function in pre-diabetic stage.

Section snippets

Materials

Streptozotocin (STZ) was product of Sigma Aldrich (S0130; St. Louis, MO, USA). RGFP966 was purchased from Selleck Chemicals (S7229; Houston, TX, USA).

Ethics

The experiments were conducted in accordance with the Standards for Laboratory Animals (GB14925-2001) and the Guidelines on the Human Treatment of Laboratory Animals (MOST 2006a) established by the People's Republic of China.

Pre-diabetic mouse model

A pre-diabetic mouse model induced by low-dose STZ (STZ pre-diabetic mice) was described as following. Normal ICR mice

HDAC3 expression increases in β cells treated with STZ or PA

PA treatment induced HDAC3 expression in NIT-1 cells, which could be reversed by RGFP966 (Supplemental Fig. 2A). Likewise, STZ treatment increased HDAC3 expression in both mouse islets and NIT-1 cells (Supplemental Fig. 2B and C), and this inducement could be significantly alleviated by RGFP966 (Supplemental Fig. 2C).

HDAC3-selective inhibitor RGFP966 decreases blood glucose level in STZ pre-diabetic mice

After 5 weeks of treatment, the body weight of mice was not influenced by RGFP966 (Fig. 1A). However, non-fasting blood glucose level of mice in the STZ + RGFP966 group

Discussion

STZ, as a selective islet β-cell cytotoxic agent, is typically used to cause complete β-cell necrosis and establish type 1 diabetes animal models (Furman, 2015). In our study, to produce a model of insulin-deficient, but not insulin-resistant pre-type 2 diabetes, a low dose of STZ was given once to ICR mice. In these STZ mice, glucose tolerance was impaired, but the fasting glucose level was normal, associated with about a 40% loss of β cells. Although research reports are increasingly

Authors' contribution

Lei L carried out the western-blot, immunohistochemistry and TUNEL assay and interpreted data and wrote the manuscripts. Bai GL did experiments in vivo, glucose-stimulated insulin assay, caspases 3/9 activity assay and Q-PCR. Wang X did the animal experiments. Liu SN did the hyperglycemic clamp test. Xia J and Wu S did experiments related to structure and chemical function of compounds. Huan Y and Shen ZF conceived the study, participated in its design and coordination. All the authors listed

Declaration of Competing Interest

The authors declare that there is no conflict of interest.

Acknowledgments

The project was supported by CAMS Innovation Fund for Medical Sciences (2016-I2M-4-001, 2017-I2M-1-010), the Drug Innovation Major Project (2018ZX09711001-003-005), National Natural Science Foundation of China (81603027, 81973379, 81803597) and Natural Science Foundation of Beijing Municipality (7202137).

We thank LetPub (www.letpub.com) for its linguistic assistance during the preparation of this manuscript.

References (36)

  • A.L. Feng et al.

    Paracrine GABA and insulin regulate pancreatic alpha cell proliferation in a mouse model of type 1 diabetes

    Diabetologia

    (2017)
  • B.L. Furman

    Streptozotocin-induced diabetic models in mice and rats

    Curr. Protoc. Pharmacol.

    (2015)
  • M.G. Guenther et al.

    A core SMRT corepressor complex containing HDAC3 and TBL1, a WD40-repeat protein linked to deafness

    Genes Dev.

    (2000)
  • M. Haberland et al.

    The many roles of histone deacetylases in development and physiology: implications for disease and therapy

    Nat. Rev. Genet.

    (2009)
  • K. Hayashi et al.

    Strain differences in the diabetogenic activity of streptozotocin in mice

    Biol. Pharm. Bull.

    (2006)
  • M.L. Jacobsen et al.

    IL-1beta-induced chemokine and Fas expression are inhibited by suppressor of cytokine signalling-3 in insulin-producing cells

    Diabetologia

    (2009)
  • H. Jin et al.

    Paeonol attenuates isoflurane anesthesia-induced hippocampal neurotoxicity via modulation of JNK/ERK/P38MAPK pathway and regulates histone acetylation in neonatal rat

    J. Matern. Fetal Neonatal Med.

    (2020)
  • A.E. Karlsen et al.

    Suppressor of cytokine signalling (SOCS)-3 protects beta cells against IL-1beta-mediated toxicity through inhibition of multiple nuclear factor-kappaB-regulated proapoptotic pathways

    Diabetologia

    (2004)
  • 1

    Lei Lei and Guoliang Bai contributed equally to this work.

    2

    Yi Huan and Zhufang Shen was the co-corresponding authors.

    View full text