Elsevier

Cellular Signalling

Volume 21, Issue 1, January 2009, Pages 169-177
Cellular Signalling

Coupling mitochondrial dysfunction to endoplasmic reticulum stress response: A molecular mechanism leading to hepatic insulin resistance

https://doi.org/10.1016/j.cellsig.2008.10.004Get rights and content

Abstract

Mitochondrial dysfunction and endoplasmic reticulum (ER) stress are considered critical components in the development of insulin resistance and Type 2 diabetes. However, understanding the molecular mechanisms underlying these individual disorders and how they are linked has been challenging. Here, we provide evidence that elevated levels of cytosolic free Ca2+ due to mitochondrial dysfunction and concomitant activation of p38 mitogen activated protein kinase (MAPK) induce ER stress response in human liver sk-HepI cells. Blocking Ca2+ release from mitochondria or ER using ruthenium red or ryanodine ameliorated the increase in expression of gluconeogenic enzymes due to mitochondrial dysfunction. Disturbance in mitochondrial function results in the activation of p38 MAPK and related transcription factors that are directly responsible for increased phosphoenolpyruvate carboxykinase (PEPCK) expression. In addition, abnormal activation of c-Jun N-terminal kinase (JNK) influences the PEPCK expression by affecting insulin signaling and Forkhead box O (Foxo) 1 activity. Alleviation of ER stress response using a chemical chaperone reduces p38 MAPK activation, as well as PEPCK overexpression, indicating that ER stress response strengthens mitochondrial stress-induced abnormalities. Our results demonstrate that mitochondrial dysfunction is directly linked to the ER stress response, and together, cause aberrant insulin signaling and an abnormal increase of hepatic gluconeogenesis.

Introduction

Increasing evidence suggests that mitochondrial dysfunction and endoplasmic reticulum (ER) stress response are causative of insulin resistance and type 2 diabetes, the most prevalent and serious metabolic disease, and yet the molecular mechanisms of this association are poorly understood [1], [2], [3], [4]. One possible mechanism by which mitochondrial dysfunction induces insulin resistance is predicted in the skeletal muscle. Mitochondrial dysfunction is responsible for elevation of several kinase activities, such as protein kinase C (PKC) or c-Jun N-terminal kinase (JNK), which increases serine phosphorylation of insulin receptor substrate-1 (IRS-1) on specific sites and inhibits its function. This inhibition results in disturbance of insulin-stimulated glucose uptake, leading to insulin resistance [5]. In addition, impaired mitochondrial function reduces expression of IRS-1, which may promote long-term insulin resistance [6].

ER stress response, also known as the unfolded protein response (UPR), is triggered by accumulation of unfolded proteins in the ER lumen. The initial event in ER stress is adaptation to the stress and restoration of normal ER function. In this step, translation of newly synthesized proteins is prevented. At the same time, the production of ER chaperones, especially Grp78, increases. When stress response is prolonged, cell death mechanisms are induced. Increased production of Bcl-2 family protein, transcription factors including CCAAT/enhancer-binding protein homologous protein (CHOP), as well as activation of caspases participate in this ER-mediated apoptosis [7], [8].

Several pathologies, including cardiac disease, Alzheimer's disease, and diabetes are related closely to the disruption of Ca2+ homeostasis [9], [10], [11]. Mitochondria and the ER are major internal Ca2+ storage organelles, which act as a Ca2+ buffer that regulates Ca2+-dependent signaling in the cytosol. The ryanodine receptor (RyR) and inositol-1,4,5-triphosphate receptor (IP3R) are Ca2+ release channels located in the ER membrane. In contrast, sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) pumps Ca2+ into the ER lumen. In addition, Ca2+ uptake into the mitochondria is primarily achieved via the mitochondrial Ca2+ uniporter (MCU), and its movement is driven by consumption of mitochondrial membrane potential ΔΨm. The effluxes from mitochondria are mediated by a Na+/Ca2+ exchanger (NCX) or permeability transition pore (PTP). The release and uptake of Ca2+ in these organelles is precisely controlled by Ca2+ itself, or an expanding group of messengers, such as inositol-1,4,5-triphosphate (IP3) [12], [13]. Under physiological conditions, ER chaperones Grp78, calreticulin, calnexin, and calsequestrin, also participate in Ca2+ homeostasis in the ER lumen [14].

Mitochondria and the ER form endomembrane networks, and interact both physically and functionally. The linkages of these organelles, called mitochondria-associated ER membrane (MAM), are enriched in ER chaperones including Grp78. The mitochondrial voltage-dependent anion channel (VDAC) interacts with IP3R on the ER membrane, and Ca2+ moves directly from the ER to mitochondria through this connection [15], [16]. Recently, Hayashi and Su showed that another ER chaperone, sigma-1 receptor (Sig-1R), interacts with Grp78 at MAM, and regulates Ca2+ movement into mitochondria via IP3Rs [17]. However, little is known about how these organelles and Ca2+ homeostasis participate together in the development of insulin resistance and diabetes. Here we show that mitochondrial dysfunction induces ER stress responses, and they coordinately increase levels of gluconeogenic enzymes, as observed in many insulin resistance states, through p38 MAPK and JNK in a Ca2+-dependent manner.

Section snippets

Materials

Oligomycin and the somatic cell ATP assay kit were obtained from Sigma (St Louis, MO, USA). BAPTA-AM was purchased from Molecular Probes (Eugene, OR, USA). The SB203580 (SB), SP600125 (SP) and 4-phenyl butyric acid (PBA) were from Calbiochem (San Diego, CA, USA). Ryanodine and ruthenium red were obtained from Tocris (Tocris Cookson Ltd., Bristol, UK). Antibodies against IRS-1, phosphor IRS-1(Tyr632), phosphoenolpyruvate carboxykinase (PEPCK), PGC1α, Grp78, and CHOP were purchased from Santa

Abnormal activation of hepatic gluconeogenesis due to mitochondrial dysfunction

The liver is an important organ for maintaining blood glucose homeostasis, and increased hepatic gluconeogenesis is an important contributor to the fasting hyperglycemia found in patients with type 2 diabetes. We therefore established a cell model system to investigate the role of mitochondrial dysfunction in the development of insulin resistance, particularly the increase in hepatic gluconeogenesis. In oligomycin-treated sk-HepI human liver cells, ΔΨm was dramatically reduced (Fig. 1A), with a

Discussion

Increased hepatic gluconeogenesis is a hallmark of type 2 diabetes. In this study, we investigated the molecular mechanism that leads to this rise. Mitochondrial function is impaired by genetic or environmental stresses, and their defects are believed to disturb insulin signaling [6], [35]. However, little is known about the influence of mitochondrial dysfunction on hepatic gluconeogenesis. We generated a cell model using human liver cell lines, sk-HepI, whose mitochondrial function was

Conclusions

We demonstrated here that mitochondrial dysfunction triggers an ER stress response in a Ca2+- and p38 MAPK-dependent manner, thereby inducing aberrant insulin signaling and hepatic gluconeogenesis, as observed in insulin resistance and type 2 diabetes. Additionally, the ER stress response induced by mitochondrial dysfunction increases cellular steady-state Ca2+ levels by releasing Ca2+ through their channels and facilitates p38 MAPK activation, which together creates the vicious circle that

Acknowledgment

This study was supported by research grants from the Korean National Institute of Health (4845-300-210-13).

References (43)

  • Y. Nakatani et al.

    J. Biol. Chem.

    (2005)
  • D.T. Rutkowski et al.

    Trends in Cell Biol.

    (2004)
  • P. Pizzo et al.

    Trends Cell Biol.

    (2007)
  • T. Hayashi et al.

    Cell

    (2007)
  • A. Barthel et al.

    Trends Endocrinol. Metab.

    (2005)
  • J. Cheong et al.

    J. Biol.Chem.

    (1998)
  • L. Qiao et al.

    J. Biol.Chem.

    (2006)
  • H.Y. Liu et al.

    J. Biol. Chem.

    (2007)
  • D.A. Salih et al.

    Curr. Opin. Cell Biol.

    (2008)
  • M.S. Kim et al.

    Cytokine

    (2005)
  • R. Parish et al.

    Curr. Diabetes Rep.

    (2005)
  • K.T. Petersen et al.

    N. Engl. J. Med.

    (2004)
  • U. Özcan et al.

    Science

    (2004)
  • B.B. Lowell et al.

    Science

    (2005)
  • J.H. Lim et al.

    Diabetologia

    (2006)
  • C. Xu et al.

    J. Clin. Invest.

    (2005)
  • N. Demaurex et al.

    Science

    (2003)
  • L. Walter et al.

    J. Bioenerg. Biomembranes

    (2005)
  • R. Rizzuto et al.

    Nat. Genet.

    (2003)
  • M.J. Berridge et al.

    Nat. Rev., Mol. Cell Biol.

    (2003)
  • P.S. Brookes et al.

    Am. J. Physiol. Cell Physiol.

    (2004)
  • Cited by (131)

    • LncEDCH1 improves mitochondrial function to reduce muscle atrophy by interacting with SERCA2

      2022, Molecular Therapy Nucleic Acids
      Citation Excerpt :

      Numerous studies have shown that mitochondria and ER are physically and functionally interconnected to maintain the homeostasis of cytosolic calcium.20,21 ER stress can induce mitochondrial dysfunction, leading to insulin resistance.22 To investigate the role of LncEDCH1 in mitochondria, we measured mitochondrial content and function after LncEDCH1 inhibition and overexpression in CPMs.

    View all citing articles on Scopus
    1

    Present address: School of Oriental medicine, Pusan National University, 30 Jangjeon-dong, Geumjeong-gu, Busan, 609-735, South Korea.

    View full text