Review article
The mitochondrial permeability transition: A current perspective on its identity and role in ischaemia/reperfusion injury

https://doi.org/10.1016/j.yjmcc.2014.08.018Get rights and content

Highlights

  • Proposals for the molecular identity of the MPTP are critically reviewed.

  • Contact sites (CS) between the inner and outer membranes regulate MPTP opening.

  • MPTP inhibition protects hearts from ischaemia/reperfusion (I/R) injury.

  • Hexokinase 2 (HK2), which stabilises CS, dissociates from mitochondria in ischaemia.

  • Preconditioning protects against I/R by preventing HK2 dissociation and MPTP opening.

Abstract

The mitochondrial permeability transition pore (MPTP) is a non-specific pore that opens in the inner mitochondrial membrane (IMM) when matrix [Ca2+] is high, especially when accompanied by oxidative stress, high [Pi] and adenine nucleotide depletion. Such conditions occur during ischaemia and subsequent reperfusion, when MPTP opening is known to occur and cause irreversible damage to the heart. Matrix cyclophilin D facilitates MPTP opening and is the target of its inhibition by cyclosporin A that is cardioprotective. Less certainty exists over the composition of the pore itself, with structural and/or regulatory roles proposed for the adenine nucleotide translocase, the phosphate carrier and the FoF1 ATP synthase. Here we critically review the supporting data for the role of each and suggest that they may interact with each other through their bound cardiolipin to form the ATP synthasome. We propose that under conditions favouring MPTP opening, calcium-triggered conformational changes in these proteins may perturb the interface between them generating the pore. Proteins associated with the outer mitochondrial membrane (OMM), such as members of the Bcl-2 family and hexokinase (HK), whilst not directly involved in pore formation, may regulate MPTP opening through interactions between OMM and IMM proteins at “contact sites”. Recent evidence suggests that cardioprotective protocols such as preconditioning inhibit MPTP opening at reperfusion by preventing the loss of mitochondrial bound HK2 that stabilises these contact sites. Contact site breakage both sensitises the MPTP to [Ca2+] and facilitates cytochrome c loss from the intermembrane space leading to greater ROS production and further MPTP opening. This article is part of a Special Issue entitled "Mitochondria: From Basic Mitochondrial Biology to Cardiovascular Disease".

Introduction

The major role of mitochondria in the heart is the provision of ATP by oxidative phosphorylation to drive the contractile cycle and maintain ionic homeostasis. Oxidative phosphorylation requires the permeability barrier of the inner mitochondrial membrane (IMM) to be maintained. However, mammalian mitochondria contain a latent non-specific pore within their inner membrane, known as the mitochondrial permeability transition pore (MPTP). Opening of the MPTP not only prevents mitochondria from synthesising ATP by oxidative phosphorylation, but also allows reversal of the FoF1 ATP synthase causing hydrolysis of the ATP produced by glycolysis or any remaining “healthy” mitochondria [1]. If this occurs for any length of time, cells become depleted of ATP and will eventually die by necrosis. In essence, MPTP opening converts mitochondria from ATP providers that energise the cell to agents of cell death, akin to the caring Dr Jekyll turning into the murderous Mr Hyde [2]. It is now widely accepted that the MPTP plays a major role in determining the extent of injury the heart suffers during reperfusion after a prolonged period of ischaemia; such ischaemia/reperfusion injury (I/R) is reflected in the size of the necrotic area or infarct (see [2], [3], [4]). In this article we will first review what is currently known about the mechanism and molecular identity of the MPTP, paying particular attention to significant new developments since our previous review in this journal [1]. We will then briefly summarise the evidence that MPTP opening is a key event in I/R injury and finally review how inhibiting MPTP opening during reperfusion is cardioprotective.

Section snippets

Historical perspective

It has been known for more than sixty years that mitochondria become leaky, uncoupled and massively swollen if they are exposed to high calcium concentrations, especially in the presence of phosphate and when accompanied by oxidative stress (see [2], [5]). This phenomenon became known as the permeability transition and was originally thought to reflect activation of endogenous phospholipase A2 leading to phospholipid breakdown within the IMM [6]. However, seminal studies in the late seventies

The molecular identity of the MPTP

Although the exact molecular identity of the MPTP is yet to be determined, several proteins have been implicated in its formation and regulation. Here we will critically review the evidence for the involvement of each of these proteins, focussing especially on recent papers proposing a role for the FoF1 ATP synthase [49], [50], [51], [52]. For a more detailed account of the evidence for the role of the adenine nucleotide translocase (ANT) and phosphate carrier (PiC) the reader is referred to

The role of the MPTP in ischaemia/reperfusion injury

It is now widely accepted that mitochondrial dysfunction, and particularly MPTP opening, plays a major role in determining the extent of injury the heart suffers during reperfusion after a prolonged period of ischaemia. This has been well reviewed elsewhere [2], [4], [21], [125] and only a brief summary will be given here. First, the conditions that occur following ischaemia and reperfusion are exactly those that would induce MPTP opening. In particular, the heart experiences calcium overload,

Inhibiting the MPTP is cardioprotective

As noted above, inhibitors of MPTP opening such as CsA and SfA provide protection from reperfusion injury in a variety of experimental models including those relevant to the clinical setting. Thus, using the Langendorff perfused rat heart, Hausenloy and colleagues demonstrated that reduction of infarct size is observed when CsA is added during the first 15 min of reperfusion [138]. Protection has also been observed using in vivo mouse and rabbit models of reperfusion injury [130], [131]

Conclusions and future directions

The central role of MPTP opening in causing reperfusion injury appears well established as is the cardioprotection offered by its inhibition. Indeed, pharmacological interventions targeting CyP-D have been proven effective in a wide range of models [2], [21], [194], [195] including a small proof of principle clinical trial [140]. Nevertheless, the effects are modest and not observed in all species [139], [194]. One reason for this may be that CyP-D only facilitates MPTP opening which can occur

Disclosures

None.

Acknowledgements

We would like to thank the many colleagues who have contributed to the research we have presented in this article that was performed in our own laboratory over many years. We are also extremely grateful for continuous funding of our research by the British Heart Foundation.

References (198)

  • P. Bernardi et al.

    Modulation of the mitochondrial permeability transition pore — effect of protons and divalent cations

    J Biol Chem

    (1992)
  • S.A. Novgorodov et al.

    Magnesium ion modulates the sensitivity of the mitochondrial permeability transition pore to cyclosporin A and ADP

    Arch Biochem Biophys

    (1994)
  • I. Szabo et al.

    Modulation of the mitochondrial megachannel by divalent cations and protons

    J Biol Chem

    (1992)
  • P. Bernardi et al.

    Modulation of the mitochondrial cyclosporin A-sensitive permeability transition pore. 1. Evidence for 2 separate Me2+ binding sites with opposing effects on the pore open probability

    J Biol Chem

    (1993)
  • S.J. Clarke et al.

    Sanglifehrin A acts as a potent inhibitor of the mitochondrial permeability transition and reperfusion injury of the heart by binding to cyclophilin-D at a different site from cyclosporin A

    J Biol Chem

    (2002)
  • A.W.C. Leung et al.

    The mitochondrial phosphate carrier interacts with cyclophilin D and may play a key role in the permeability transition

    J Biol Chem

    (2008)
  • A.P. Halestrap et al.

    Oxidative stress, thiol reagents, and membrane potential modulate the mitochondrial permeability transition by affecting nucleotide binding to the adenine nucleotide translocase

    J Biol Chem

    (1997)
  • F. Di Lisa et al.

    A CaPful of mechanisms regulating the mitochondrial permeability transition

    J Mol Cell Cardiol

    (2009)
  • P. Bernardi

    Modulation of the mitochondrial cyclosporin-A-sensitive permeability transition pore by the proton electrochemical gradient — evidence that the pore can be opened by membrane depolarization

    J Biol Chem

    (1992)
  • L. Scorrano et al.

    On the voltage dependence of the mitochondrial permeability transition pore — a critical appraisal

    J Biol Chem

    (1997)
  • F. Ichas et al.

    Mitochondria are excitable organelles capable of generating and conveying electrical and calcium signals

    Cell

    (1997)
  • F. Ichas et al.

    From calcium signaling to cell death: two conformations for the mitochondrial permeability transition pore. Switching from low- to high-conductance state

    Biochim Biophys Acta

    (1998)
  • V Petronilli et al.

    Transient and long-lasting openings of the mitochondrial permeability transition pore can be monitored directly in intact cells by changes in mitochondrial calcein fluorescence

    Biophys J

    (1999)
  • P. Korge et al.

    Protective role of transient pore openings in calcium handling by cardiac mitochondria

    J Biol Chem

    (2011)
  • S. Menazza et al.

    CypD(−/−) hearts have altered levels of proteins involved in Krebs cycle, branch chain amino acid degradation and pyruvate metabolism

    J Mol Cell Cardiol

    (2013)
  • M Carraro et al.

    Channel formation by yeast F-ATP synthase and the role of dimerization in the mitochondrial permeability transition

    J Biol Chem

    (2014)
  • L. He et al.

    Regulated and unregulated mitochondrial permeability transition pores: a new paradigm of pore structure and function?

    FEBS Lett

    (2002)
  • M. Gutierrez-Aguilar et al.

    Genetic manipulation of the cardiac mitochondrial phosphate carrier does not affect permeability transition

    J Mol Cell Cardiol

    (2014)
  • V Giorgio et al.

    Cyclophilin D modulates mitochondrial F0F1-ATP synthase by interacting with the lateral stalk of the complex

    J Biol Chem

    (2009)
  • J. Kallen et al.

    Structure of human cyclophilin A in complex with the novel immunosuppressant sanglifehrin A at 1.6 angstrom resolution

    J Biol Chem

    (2005)
  • E. Basso et al.

    Properties of the permeability transition pore in mitochondria devoid of cyclophilin D

    J Biol Chem

    (2005)
  • T.T. Nguyen et al.

    Cysteine 203 of cyclophilin D is critical for cyclophilin D activation of the mitochondrial permeability transition pore

    J Biol Chem

    (2011)
  • A. Ruck et al.

    Reconstituted adenine nucleotide translocase forms a channel for small molecules comparable to the mitochondrial permeability transition pore

    FEBS Lett

    (1998)
  • Z Hu et al.

    Down-regulation of adenine nucleotide translocase 3 and its role in camptothecin-induced apoptosis in human hepatoma QGY7703 cells

    FEBS Lett

    (2009)
  • S. Da Cruz et al.

    Proteomic analysis of the mouse liver mitochondrial inner membrane

    J Biol Chem

    (2003)
  • CR Pestana et al.

    Ca(2+) binding to c-state of adenine nucleotide translocase (ANT)-surrounding cardiolipins enhances (ANT)-Cys(56) relative mobility: a computational-based mitochondrial permeability transition study

    Biochim Biophys Acta

    (2009)
  • G. Petrosillo et al.

    Interaction of peroxidized cardiolipin with rat-heart mitochondrial membranes: induction of permeability transition and cytochrome c release

    FEBS Lett

    (2006)
  • S. Chalmers et al.

    The relationship between free and total calcium concentrations in the matrix of liver and brain mitochondria

    J Biol Chem

    (2003)
  • E. Basso et al.

    Phosphate is essential for inhibition of the mitochondrial permeability transition pore by cyclosporin A and by cyclophilin D ablation

    J Biol Chem

    (2008)
  • P. Varanyuwatana et al.

    The roles of phosphate and the mitochondrial phosphate carrier in the mechanism of the permeability transition

    Mitochondrion

    (2012)
  • B. Kadenbach et al.

    The mitochondrial phosphate carrier has an essential requirement for cardiolipin

    FEBS Lett

    (1982)
  • D. Cheneval et al.

    The mitochondrial phosphate carrier reconstituted in liposomes is inhibited by doxorubicin

    FEBS Lett

    (1983)
  • A. Ortiz et al.

    Membrane fusion and the lamellar-to-inverted-hexagonal phase transition in cardiolipin vesicle systems induced by divalent cations

    Biophys J

    (1999)
  • A.P. Halestrap

    A pore way to die: the role of mitochondria in reperfusion injury and cardioprotection

    Biochem Soc Trans

    (2010)
  • D.J. Hausenloy et al.

    The mitochondrial permeability transition pore as a target for preconditioning and postconditioning

    Basic Res Cardiol

    (2009)
  • P. Bernardi

    The mitochondrial permeability transition pore: a mystery solved?

    Front Physiol

    (2013)
  • T.E. Gunter et al.

    Mechanisms by which mitochondria transport calcium

    Am J Physiol

    (1990)
  • I. Al-Nasser et al.

    The reversible Ca2+-induced permeabilisation of rat liver mitochondria

    Biochem J

    (1986)
  • M. Crompton et al.

    Inhibition by cyclosporin A of a Ca2+-dependent pore in heart mitochondria activated by inorganic phosphate and oxidative stress

    Biochem J

    (1988)
  • A.P. Halestrap et al.

    Inhibition of Ca2+-induced large amplitude swelling of liver and heart mitochondria by Cyclosporin A is probably caused by the inhibitor binding to mitochondrial matrix peptidyl-prolyl cis–trans isomerase and preventing it interacting with the adenine nucleotide translocase

    Biochem J

    (1990)
  • Cited by (349)

    View all citing articles on Scopus
    View full text