Metformin prevents glucotoxicity by alleviating oxidative and ER stress–induced CD36 expression in pancreatic beta cells

https://doi.org/10.1016/j.jdiacomp.2016.09.001Get rights and content

Abstract

Aim/hypothesis

Cluster determinant 36 (CD36), a fatty acid transporter, was reported to have a pivotal role in glucotoxicity-induced beta cell dysfunction. However, little is known about how glucotoxicity influences CD36 expression, and it is unknown whether this action can be counteracted by metformin. In the present study, we showed that metformin counteracts glucotoxicity by alleviating oxidative and endoplasmic reticulum (ER) stress–induced CD36 expression.

Methods

We used primary rat islets as well as INS-1 cells for 72 h to 24 h with 30 mM glucose, respectively. Thapsigargin was used as strong ER stressor, and Sulfo-N-succinimidyl oleate (SSO) and RNA interference were chosen for CD36 inhibition. Free fatty acid uptake was measured by radioisotope tracing technique.

Results

Exposure of isolated rat islets to high glucose (HG) for 3 days decreased insulin and pancreatic duodenal homeobox1 (Pdx1) mRNA expression, with the suppression of glucose-stimulated insulin secretion (GSIS) along with elevation of reactive oxygen species (ROS) levels. Incubation with metformin restored insulin and Pdx1 mRNA expression with significant improvements in GSIS and decrease in ROS production. HG exposure in INS-1 cells increased free fatty acid uptake via induction of CD36 along with impaired insulin and Pdx1 mRNA expression. Moreover, thapsigargin also increased the induction of CD36 expression. Metformin blocked HG- and thapsigargin-induced CD36 expression. In addition, the simultaneous inhibition of intracellular ROS production by metformin or CD36 activation by SSO or CD36 siRNA significantly decreased the apoptotic response in HG-treated INS-1 cells.

Conclusion/interpretation

In conclusion, metformin conferred protection against HG-induced apoptosis of pancreatic beta cells, largely by interfering with ROS production, and inhibited the CD36-mediated free fatty acid influx. This report provides evidence that the inhibition of CD36 may have potential therapeutic effects against hyperglycemia-induced beta cell damage in diabetes.

Introduction

Hyperglycemia, an important pathologic characteristic of type 2 diabetes, primarily results from insulin resistance and beta cell dysfunction. The latter is essential for the development of diabetes because hyperglycemia does not develop until compensatory insulin secretion failure. Excessive nutrition and obesity increase the metabolic load in pancreatic beta cells, and this leads to pancreatic beta cell dysfunction and death (Han, 2016, Kim and Yoon, 2011, Poitout and Robertson, 2002, Poitout and Robertson, 2008, Robertson et al., 2004). Although, the molecular signals that trigger functional deterioration of the beta cell are unknown, several studies have provided evidence that the dysfunction occurs in the presence of elevated glucose levels in type 2 diabetes. Moreover, pancreatic islets are relatively vulnerable to oxidative stress due to a low capacity of antioxidant enzymes (Jung et al., 2014, Karunakaran and Park, 2013, Robertson et al., 2003). Thus, beta cell preservation is an essential component of type 2 diabetes management.

Metformin is currently recommended as first-line therapy for all newly diagnosed type 2 diabetes patients. Metformin regulates blood glucose levels primarily by decreasing hepatic glucose production and improving insulin sensitivity (Bailey and Turner, 1996, Prager and Schernthaner, 1983). Meanwhile, there have been controversies whether metformin directly has beneficial effects on beta cell function. In particular, metformin restores insulin secretion altered by chronic exposure to high glucose or high free fatty acid levels in vitro (Lupi et al., 1999, Patane et al., 2000) and ameliorates the deleterious effects of free fatty acid levels in pancreatic human islets (Lupi et al., 2002). Marchetti et al.(2004) showed that the beneficial effects of metformin on beta cell function may be mediated by the alleviation of oxidative stress. However, the exact mechanism of these effects has not been clarified. Recently, we reported that fatty acid translocase cluster determinant 36 (CD36), a membrane glycoprotein, influences the induction of glucotoxicity in pancreatic beta cells (Kim et al., 2012). Previous findings showed that CD36 is not only a fatty acid transporter but also related to a signaling cascade under metabolic stress conditions. Thus, CD36 would be an important therapeutic target in diabetic conditions (Karunakaran, Moon, Lee, & Won, 2015). In this study, we examined the effects of metformin on high glucose-induced CD36 expression and found that metformin decreased high glucose-induced CD36 expression via the inhibition of oxidative and endoplasmic reticulum(ER) stress signaling.

Section snippets

Chemicals

Sulfo-N-succinimidyl oleate (SSO) was obtained from Cayman Chemical (Ann Arbor, MI, USA). Antibodies against caspase-3, phosphorylated JNK, eIF2α, and PERK were obtained from Cell signaling Technology (Danvers, MA, USA). Antibody against CHOP was obtained from Santa Cruz (Dallas, TX, USA). CD36 antibody was obtained from Cayman Chemical (Ann Arbor, MI, USA) and antibody against actin was obtained from Abcam (Cambridge, UK).

Cell culture and primary islet isolation

The INS-1 rat insulinoma cell line was cultured in 5% CO2/95% air at 37 

Effects of metformin on high glucose and thapsigargin-induced cytotoxicity

As shown in Fig. 1A, the viability of INS-1 cells significantly decreased under high glucose (30 mM) treatment. However, the decrease in cell viability was prevented by metformin treatment. ER stress can mediate the cytotoxic effects of high glucose, and we also checked whether metformin is effective against thapsigargin-induced cytotoxicity. Thapsigargin (0.5 μM) decreased the cell viability of INS-1 cells, which was blocked by metformin pretreatment (Fig. 1B). In agreement with this, metformin

Discussion

Our previous study showed that hyperglycemia increases CD36 expression and decreases insulin secretion, accompanied by increased apoptosis of INS-1 cells (Kim et al., 2012). However, the underlying mechanism of CD36 induction by hyperglycemia remains unclear. In the present study, we demonstrate that metformin prevents hyperglycemia-induced beta-cell apoptosis by suppressing ROS production, thereby decreasing CD36 expression.

Glucotoxicity is an important factor that contributes to pancreatic

Conclusion

In conclusion, our results show that metformin treatment ameliorated high glucose-induced beta cell dysfunction by decreasing intracellular ROS production and suppressed CD36 expression and free fatty acid uptake (Fig. 8).

Funding

This study was supported by a grant of Yeungnam University Medical Center (2014).

Author contributions

JSM and UK contributed to the conception and design of the study, the acquisition, analysis and interpretation of the data, and drafting of the article. SE contributed to the experiment and interpretation of the data. IKL, HWL, KCW and YWK contributed to the conception and design of the study, the analysis and interpretation of the data, and the revision of the article.

Acknowledgments

Authors thank to Ms. Ye-Jin Seo (Yeungnam University College of Medicine, Daegu, Korea) for technical assistance.

References (35)

  • G Da Silva Xavier et al.

    Role of AMP-activated protein kinase in the regulation by glucose of islet beta cell gene expression

    Proceedings of the National Academy of Sciences of the United States of America

    (2000)
  • H Farhangkhoee et al.

    Glucose-induced up-regulation of CD36 mediates oxidative stress and microvascular endothelial cell dysfunction

    Diabetologia

    (2005)
  • CY Han

    Roles of reactive oxygen species on insulin resistance in adipose tissue

    Diabetes and Metabolism Journal

    (2016)
  • Y Ihara et al.

    Hyperglycemia causes oxidative stress in pancreatic beta-cells of GK rats, a model of type 2 diabetes

    Diabetes

    (1999)
  • M Janabi et al.

    Oxidized LDL-induced NF-kappa B activation and subsequent expression of proinflammatory genes are defective in monocyte-derived macrophages from CD36-deficient patients

    Arteriosclerosis, Thrombosis, and Vascular Biology

    (2000)
  • KY Jung et al.

    Therapeutic approaches for preserving or restoring pancreatic β-cell function and mass

    Diabetes and Metabolism Journal

    (2014)
  • U Karunakaran et al.

    A systematic review of oxidative stress and safety of antioxidants in diabetes: Focus on islets and their defense

    Diabetes and Metabolism Journal

    (2013)
  • Cited by (53)

    • Metformin inhibits MAPK signaling and rescues pancreatic aquaporin 7 expression to induce insulin secretion in type 2 diabetes mellitus

      2021, Journal of Biological Chemistry
      Citation Excerpt :

      Some studies showed that metformin ameliorates the inhibition of insulin secretion from INS-1 cells under glucotoxic conditions by suppressing the ATP-conducting mitochondrial outer membrane voltage-dependent anion channel-1 (VDAC1) (33) or activating AMPK/sirtuin 1 (SIRT1)/peroxisome proliferator-activated receptor gamma coactivators-1α (PGC-1α) signal pathway (34). Some other studies showed that metformin prevents high-glucose-induced Rac1 [GTP-bound conformation] activation, nuclear translocation, cluster determinant 36 (CD36) expression, and this ultimately prevents high-glucose-induced β-cell dysfunction (35, 36). Whether metformin regulates AQP7 expression through VDAC1, AMPK/SIRT1/PGC-1α, Rac1, CD36, or some other pathway or whether these pathways synergy with the MAPK signal in pancreas, it needs further studies.

    • Ischemia-reperfusion injury

      2021, Organ Repair and Regeneration: Preserving Organs in the Regenerative Medicine Era
    • Role of CD36 in central nervous system diseases

      2024, Neural Regeneration Research
    View all citing articles on Scopus

    Conflicts of Interest: There are no conflicts of interest to disclose.

    1

    Jun. Sung Moon and Udayakumar Karunakaran equally contributed as first authors.

    2

    Yong-Woon Kim and Kyu Chang Won have contributed as corresponding authors.

    View full text