Review
Role of oxidative stress, endoplasmic reticulum stress, and c-Jun N-terminal kinase in pancreatic β-cell dysfunction and insulin resistance

https://doi.org/10.1016/j.biocel.2005.04.003Get rights and content

Abstract

Type 2 diabetes is the most prevalent and serious metabolic disease affecting people all over the world. Pancreatic β-cell dysfunction and insulin resistance are the hallmark of type 2 diabetes. Normal β-cells can compensate for insulin resistance by increasing insulin secretion and/or β-cell mass, but insufficient compensation leads to the onset of glucose intolerance. Once hyperglycemia becomes apparent, β-cell function gradually deteriorates and insulin resistance aggravates. Under diabetic conditions, oxidative stress and endoplasmic reticulum stress are induced in various tissues, leading to activation of the c-Jun N-terminal kinase pathway. The activation of c-Jun N-terminal kinase suppresses insulin biosynthesis and interferes with insulin action. Indeed, suppression of c-Jun N-terminal kinase in diabetic mice improves insulin resistance and ameliorates glucose tolerance. Thus, the c-Jun N-terminal kinase pathway plays a central role in pathogenesis of type 2 diabetes and could be a potential target for diabetes therapy.

Introduction

The number of patients with type 2 diabetes is markedly increasing worldwide, and nowadays type 2 diabetes is recognized as the most prevalent and serious metabolic disease. In addition, type 2 diabetes is predicted to be an increasing economic and healthcare burden in the near future. Therefore, it is very important to examine a molecular mechanism for pathogenesis of type 2 diabetes and to explore a therapeutic target for diabetes. The development of type 2 diabetes is associated with a combination of pancreatic β-cell dysfunction and insulin resistance. Normal β-cells can compensate for insulin resistance by increasing insulin secretion or β-cell mass, but insufficient compensation leads to the onset of glucose intolerance. Chronic hyperglycemia is a cause of impairment of insulin biosynthesis and secretion; once hyperglycemia becomes apparent, β-cell function gradually deteriorates and insulin resistance aggravates (Jonas et al., 1999, Moran et al., 1997, Poitout and Robertson, 2002; Weir, Laybutt, Kaneto, Bonner-Weir, & Sharma, 2001). This process is called “glucotoxicity” (Fig. 1). Similar to the paradoxically deleterious effects of chronic hyperglycemia, free fatty acids, which are essential fuels in the normal state, become toxic when they are chronically present in excessive levels (Unger, 1995). This process is called “lipotoxicity” (Fig. 1). Under such conditions, oxidative stress and endoplasmic reticulum (ER) stress are provoked and the c-Jun N-terminal kinase (JNK) pathway is activated in various tissues. Here we show a role of oxidative stress, ER stress, and the JNK pathway in pancreatic β-cell dysfunction and insulin resistance; the JNK activation suppresses insulin biosynthesis and interferes with insulin action and, in reverse, suppression of the JNK pathway in diabetic mice improves insulin resistance and ameliorates glucose tolerance.

Section snippets

Oxidative stress and pancreatic β-cell dysfunction

Chronic hyperglycemia is a cause of impairment of insulin biosynthesis and secretion; once hyperglycemia becomes apparent, β-cell function gradually deteriorates (Jonas et al., 1999, Moran et al., 1997, Poitout and Robertson, 2002, Weir et al., 2001). This process is called “glucotoxicity” (Fig. 1). The mechanism for glucotoxicity is, at least in part, mediated by overloads of reactive oxygen species. Similar to the paradoxically deleterious effects of chronic hyperglycemia, free fatty acids,

Oxidative stress and insulin resistance

The hallmark of the disease is insulin resistance as well as pancreatic β-cell dysfunction. Under diabetic conditions, various insulin target tissues such as liver, muscle, and fat become resistant to insulin. The pathophysiology of insulin resistance involves a complex network of insulin signaling pathways. After insulin binds to insulin receptor on cell surface, insulin receptor and its substrates are phosphorylated, which leads to activation of various insulin signaling pathways. It has been

Conclusion

The JNK activation is involved in the progression of insulin resistance as well as deterioration of pancreatic β-cell function. Indeed, suppression of the JNK pathway in obese diabetic mice markedly improves insulin resistance and β-cell function and ameliorates glucose tolerance. Taken together, the JNK pathway plays a crucial role in the progression of insulin resistance as well as β-cell dysfunction and thus could be a potential therapeutic target for diabetes (Fig. 4).

References (99)

  • M.E. Horb et al.

    Experimental conversion of liver to pancreas

    Curr Biol

    (2003)
  • H. Inoue et al.

    A gene encoding a transmembrane protein is mutated in patients with diabetes mellitus and optic atrophy (Wolfram syndrome)

    Nat Genet

    (1998)
  • S. Jacob et al.

    Oral administration of RAC-alpha-lipoic acid modulates insulin sensitivity in patients with type-2 diabetes mellitus: A placebo-controlled pilot trial

    Free Rad Biol Med

    (1999)
  • S. Jacqueminet et al.

    Inhibition of insulin gene expression by long-term exposure of pancreatic β-cells to palmitate is dependent upon the presence of a stimulatory glucose concentration

    Metabolism

    (2000)
  • J.-C. Jonas et al.

    Chronic hyperglycemia triggers loss of pancreatic β cell differentiation in an animal model of diabetes

    J Biol Chem

    (1999)
  • H. Kaneto et al.

    Activation of the hexosamine pathway leads to deterioration of pancreatic β-cell function by provoking oxidative stress

    J Biol Chem

    (2001)
  • H. Kaneto et al.

    Involvement of c-Jun N-terminal kinase in oxidative stress-mediated suppression of insulin gene expression

    J Biol Chem

    (2002)
  • K. Kuwabara et al.

    Purification and characterization of a novel stress protein, the 150-kDa oxygen-regulated protein (ORP150), from cultured rat astrocytes and its expression in ischemic mouse brain

    J Biol Chem

    (1996)
  • S. Lenzen et al.

    Low antioxidant enzyme gene expression in pancreatic islets compared with various other mouse tissues

    Free Rad Biol Med

    (1996)
  • P. Maechler et al.

    Hydrogen peroxide alters mitochondrial activation and insulin secretion in pancreatic beta cells

    J Biol Chem

    (1999)
  • T. Miyatsuka et al.

    Ectopically expressed PDX-1 in liver initiates endocrine and exocrine pancreas differentiation but causes dysmorphogenesis

    Biochem Biophys Res Commun

    (2003)
  • Y. Nakatani et al.

    Modulation of the JNK pathway in liver affects insulin resistance status

    J Biol Chem

    (2004)
  • Y. Nakatani et al.

    Involvement of ER stress in insulin resistance and diabetes

    J Biol Chem

    (2005)
  • R.P. Robertson

    Chronic oxidative stress as a central mechanism for glucose toxicity in pancreatic islet beta cells in diabetes

    J Biol Chem

    (2004)
  • K. Sakai et al.

    Mitochondrial reactive oxygen species reduce insulin secretion by pancreatic β-cells

    Biochem Biophys Res Commun

    (2003)
  • A. Tirosh et al.

    Oxidative stress disrupts insulin-induced cellular redistribution of insulin receptor substrate-1 and phosphatidylinositol 3-kinase in 3T3-L1 adipocytes: A putative cellular mechanism for impaired protein kinase B activation and GLUT4 translocation

    J Biol Chem

    (1999)
  • H. Wang et al.

    Pdx1 level defines pancreatic gene expression pattern and cell lineage differentiation

    J Biol Chem

    (2001)
  • U. Ahlgren et al.

    β-Cell-specific inactivation of the mouse Ipf1/Pdx1 gene results in loss of the β-cell phenotype and maturity onset diabetes

    Genes Dev

    (1998)
  • A. Ammendrup et al.

    The c-Jun amino-terminal kinase pathway is preferentially activated by interleukin-1 and controls apoptosis in differentiating pancreatic β-cells

    Diabetes

    (2000)
  • M. Aridor et al.

    Integration of endoplasmic reticulum signaling in health and disease

    Nat Med

    (1999)
  • J.W. Baynes et al.

    Role of oxidative stress in diabetic complications: A new perspective on an old paradigm

    Diabetes

    (1999)
  • L.C. Bollheimer et al.

    Chronic exposure to free fatty acid reduces pancreatic β cell insulin content by increasing basal insulin secretion that is not compensated for by a corresponding increase in proinsulin biosynthesis translation

    J Clin Invest

    (1998)
  • C. Bonny et al.

    Cell-permeable peptide inhibitors of JNK: Novel blockers of β-cell death

    Diabetes

    (2001)
  • M. Brissova et al.

    Reduction in pancreatic transcription factor PDX-1 impairs glucose-stimulated insulin secretion

    J Biol Chem

    (2002)
  • M. Brownlee

    Biochemistry and molecular cell biology of diabetic complications

    Nature

    (2001)
  • L.-Z. Cao et al.

    High glucose is necessary for complete maturation of Pdx1-VP16-expressing hepatic cells into functional insulin-producing cells

    Diabetes

    (2004)
  • L. Chang et al.

    Mammalian MAP kinase signalling cascades

    Nature

    (2001)
  • J.A. Corbett et al.

    Interleukin 1β induces the formation of nitric oxide by β-cells purified from rodent islets of Langerhans: Evidence for the β-cell as a source and site of action of nitric oxide

    J Clin Invest

    (1992)
  • S. Dutta et al.

    Regulatory factor linked to late-onset diabetes?

    Nature

    (1998)
  • D.L. Eizirik et al.

    Cytokines suppress human islet function irrespective of their effects on nitric oxide generation

    J Clin Invest

    (1994)
  • J.L. Evans et al.

    Oxidative stress and stress-activated signaling pathways: A unifying hypothesis of type 2 diabetes

    Endocr Rev

    (2002)
  • S. Ferber et al.

    Pancreatic and duodenal homeobox gene 1 induces expression of insulin genes in liver and ameliorates streptozotocin-induced hyperglycemia

    Nat Med

    (2000)
  • C.A. Haber et al.

    N-acetylcysteine and taurine prevent hyperglycemia-induced insulin resistance in vivo: Possible role of oxidative stress

    Am J Physiol Endocrinol Metab

    (2003)
  • H.P. Harding et al.

    Protein translation and folding are coupled by an endoplasmic-reticulum-resident kinase

    Nature

    (1999)
  • H.P. Harding et al.

    Endoplasmic reticulum stress and the development of diabetes

    Mol Cell

    (2000)
  • H.P. Harding et al.

    Endoplasmic reticulum stress and the development of diabetes: A review

    Diabetes

    (2002)
  • M. Hibi et al.

    Identification of an oncoprotein- and UV-responsive protein kinase that binds and potentiates the c-Jun activation domain

    Genes Dev

    (1993)
  • J. Hirosumi et al.

    A central role for JNK in obesity and insulin resistance

    Nature

    (2002)
  • A.M. Holland et al.

    Experimental control of pancreatic development and maintenance

    Proc Natl Acad Sci USA

    (2002)
  • Cited by (83)

    • Changes in the histopathology and in the proteins related to the MAPK pathway in the brains of rats exposed to pre and postnatal radiofrequency radiation over four generations

      2022, Journal of Chemical Neuroanatomy
      Citation Excerpt :

      ERK1/2 is critical for neuronal differentiation and plasticity and may also modulate neuronal survival” (Hetman and Gozdz (2004); Alonso et al., 2004). Some researchers defined that “the c-Jun NH2-terminal kinases (JNK) played an important role in cell regulation, contributing to functions, such as gene expression, cell proliferation, and programmed cell death as a member of the MAPK super-family (Kaneto et al., 2005; Abdelli et al., 2007; Weston and Davis (2007)) and JNK pathways are closely associated with several diseases, including neurodegeneration, tumors, and diabetes” (Nateri et al., 2005; Tuncman et al., 2006; Wilhelm et al., 2007). However, the results of some experiments have indicated that “JNK signaling plays a key role an important role in tau protein phosphorylation in neuronal tissues” (Reynolds et al., 1997; Yoshida et al., 2004; Schechter et al., 2005).

    • Hesperidin ameliorates pancreatic β-cell dysfunction and apoptosis in streptozotocin-induced diabetic rat model

      2019, Life Sciences
      Citation Excerpt :

      Glucose stimulates the translocation of PDX-1 into the nucleus that further promotes the transcription of insulin gene, and insulin secretion-associated genes such as glucokinase (GCK) and Glut-2 [33]. With a diabetes condition, sustained high glucose suppresses the PDX-1 gene, and activates PDX-1 translocation to the cytosol, which results in reducing the binding of PDX-1 and insulin gene promotor, thereby inhibiting insulin synthesis and secretion [34]. Numerous studies report that PDX-1, GCK, and Glut-2 protein expressions were downregulated in diabetic rats, causing impaired expression and secretion of insulin [35,36].

    View all citing articles on Scopus
    View full text