Review article
What is the mitochondrial permeability transition pore?

https://doi.org/10.1016/j.yjmcc.2009.02.021Get rights and content

Abstract

Under conditions of mitochondrial calcium overload, especially when accompanied by oxidative stress, elevated phosphate concentrations and adenine nucleotide depletion, a non-specific pore, the mitochondrial permeability transition pore (MPTP), opens in the inner mitochondrial membrane. MPTP opening enables free passage into the mitochondria of molecules of < 1.5 kDa including protons. The resulting uncoupling of oxidative phosphorylation leads to ATP depletion and necrotic cell death and it is now widely recognised that MPTP opening is a major cause of reperfusion injury and an effective target for cardioprotection. The properties of the MPTP are well defined, but despite extensive research in many laboratories, its exact molecular identity remains uncertain. Knockout studies have confirmed a role for cyclophilin-D (CyP-D), probably mediated by its peptidyl-prolyl cis–trans isomerase activity facilitating a conformational change of an inner membrane protein. However, the identity of the membrane component(s) remains controversial. Knockout studies have eliminated an essential role for either the voltage dependent anion channel (VDAC) or the adenine nucleotide translocase (ANT), although a regulatory role for the ANT was confirmed. Our own studies implicate the mitochondrial phosphate carrier (PiC) in MPTP formation and are consistent with a calcium-triggered conformational change of the PiC, facilitated by CyP-D, inducing pore opening. We propose that this is enhanced by an association of the PiC with the “c” conformation of the ANT. Agents that modulate pore opening may act on either or both the PiC and the ANT. However, knockdown and reconstitution studies are awaited to confirm or refute this model.

Introduction

It was recognised more than 40 years ago that energised mitochondria exposed to high loads of calcium can undergo massive swelling that is characterised by a large decrease in light scattering and can be partially reversed by calcium chelation [1], [2]. It was originally suggested that this swelling might represent a non-specific permeabilisation of the mitochondrial inner membrane through activation of Ca-sensitive phospholipases leading to the build up of fatty acids and lysophospholipids in the membrane (see [3]). However, pioneering studies by Haworth and Hunter [4], [5] and later by Crompton et al. [6] provided evidence that the increase in permeability was the result of the opening of a non-specific channel of defined diameter in the membrane that exhibited a molecular cut-off of about 1.5 kDa. This channel became known at the mitochondrial permeability transition pore (MPTP) and a wide range of activators and inhibitors of pore opening were identified. These are summarised in Table 1 and will be further discussed in Section 2. Of particular significance was the observation that opening of the MPTP is greatly enhanced by adenine nucleotide depletion, elevated phosphate and oxidative stress which are conditions known to accompany reperfusion of the heart following a period of ischemia (see [7], [8], [9]).

One major consequence of MPTP opening is that the inner mitochondrial membrane no longer maintains a barrier to protons which leads to dissipation of the proton motive force. The resulting uncoupling of oxidative phosphorylation not only prevents mitochondria from making ATP, but the proton-translocating ATPase goes into reverse. This means that within any cell the “open” mitochondria hydrolyse ATP generated by glycolysis and any “closed” mitochondria, leading to ATP depletion and bioenergetic failure of the myocyte (see [7], [8]). A second consequence of MPTP opening is that all small molecular weight molecules equilibrate across the inner membrane, including cofactors and ions. This will not only lead to the disruption of metabolic gradients between the mitochondria and cytosol, including the release of accumulated Ca2+, but will also cause mitochondrial swelling. This occurs because the increased permeability of the inner mitochondrial membrane to small molecules mediates equilibration of all low molecular weight osmolytes whilst retaining proteins within their respective compartments. Since the matrix protein concentration is higher than that in the cytosol and intermembrane space, it exerts a colloidal osmotic pressure leading to swelling of the matrix compartment (see [7], [8], [10]). Swelling can occur without inner membrane rupture because the cristae unfold, but as the matrix expands it exerts pressure on the outer membrane that eventually ruptures. This releases cytochrome c[11], [12], [13] and other pro-apoptotic proteins which have the potential to initiate apoptotic cell death (see [14], [15], [16]). However, only if the pores close again sufficiently to maintain ATP levels will apoptopic death predominate over necrotic cell death (see [8]).

It was first was recognised by Martin Crompton that opening of the pore might account for the damage experienced by the heart during reperfusion after a period of ischemia [6], [17]. Subsequent studies in his laboratory using isolated cardiac myocytes [18], [19] and this laboratory using Langendorf perfused hearts [20], [21] demonstrated directly that MPTP opening does occur in such reperfusion injury, and that preventing pore opening provides protection against reperfusion injury. These pioneering studies were slow to be appreciated. However, in recent years data have accumulated from many different laboratories that have confirmed the central role of MPTP opening in reperfusion injury and its importance as a pharmacological target for cardioprotection (see [8], [9], [22], [23], [24]).

The role, if any, of the MPTP in healthy cells remains unclear since mice lacking cyclophilin-D (CyP-D), a component of the MPTP (see Section 3.1), appear normal but as predicted are protected against ischemic injury [25], [26], [27], [28]. However, a recent paper has reported that CyP-D knockout mice display greater anxiety with less tendency to explore and a facilitation of avoidance behaviour. They also exhibit an abnormal accumulation of white adipose tissue resulting in adult-onset obesity but whether or not the loss of MPTP opening accounts for these effects is not known [29]. Another suggested role for the MPTP in healthy cells is that it might provide a mechanism by which aging mitochondria are removed from the cell by autophagy [30]. Thus, as mitochondria age they are exposed to more oxidative stress and so become more susceptible to MPTP opening. Eventually, a time will come when the amount of oxidative stress in an older mitochondrion is sufficient to cause MPTP opening at resting calcium concentrations and this might enable it to be recognised by autophagic vacuoles and removed. Such a mechanism would prevent damaged mitochondria from proliferating whilst healthy mitochondria would continue to replicate. However, although this is an attractive hypothesis, the CyP-D knockout mice provide no direct evidence for it [29].

In view of its central role in reperfusion injury, the MPTP has become an obvious target for cardioprotection. Indeed, it has been demonstrated that a wide variety of cardioprotective protocols prevent MPTP opening during reperfusion. These include drugs that directly inhibit the MPTP such as sanglifehrin A (SfA), cyclosporin A (CsA) and its non-immunosuppressive analogues 6-MeAla-CsA, 4-methyl-val-CsA, N-methyl-4-isoleucine-CsA (NIM811) and d-3-MeAla-4-EtVal-CsA (Debio-025) [20], [21], [31], [32], [33] as well as protocols that inhibit MPTP opening indirectly through decreasing oxidative stress or pH. Such is the case for ischemic preconditioning [34] and post-conditioning [35], temperature preconditioning [36], urocortin [37], Na+/H+ exchanger inhibitors such as cariporide [38], antioxidants including pyruvate [39] and the anaesthetic propofol [40] and mitochondrial-targeted ubiquinone antioxidants [41].

A drug that inhibits the MPTP directly has the potential to be of great value in protecting the heart during cardiac surgery or the treatment of a coronary thrombosis. Indeed, proof of principal has been provided by preliminary studies demonstrating that CsA improved cardiac performance following treatment of coronary thrombosis with angioplasty [42]. However, CsA and Sfa are not ideal for cardioprotection for two reasons. First, they have the potential to exert unwanted side-effects through their interactions with other cyclophilins such as Cyp-A (see [8]). Second, their ability to inhibit MPTP opening is overcome as the magnitude of the stimulus responsible for MPTP opening ([Ca2+], oxidative stress or adenine nucleotide depletion) is increased [21], [31], [43], [44]. Thus it would be desirable to develop new drugs that target the MPTP whose potency is not constrained in this way. A major obstacle that stands in the way of achieving this goal is our uncertainty over the exact molecular composition and mechanism of the MPTP. In this review I will attempt to summarise what the current state of knowledge is.

Section snippets

MPTP opening is triggered by matrix calcium

The primary trigger for opening of the MPTP is matrix [Ca2+]. Thus, in vitro, the pore can be opened by calcium addition and then rapidly closed again by calcium chelation with the extent of pore opening being determined by the matrix [Ca2+] [4], [6], [17], [45]. However, the concentration of calcium required is highly dependent on the prevailing conditions [46], [47] as discussed further below (Section 3) and illustrated in Fig. 1. Thus opening can occur without calcium addition if a pore

Factors that regulate the MPTP

Increased mitochondrial matrix [Ca2+] alone may be inadequate to elicit MPTP opening and additional factors such as oxidative stress, adenine nucleotide depletion, elevated phosphate concentrations and mitochondrial depolarisation are thought to be critical. Indeed such factors, and especially oxidative stress, may be more important than increases in [Ca2+], in the MPTP opening seen under conditions such as ischemia/reperfusion (see [8], [48], [49], [62]). The regulatory factors that are most

The molecular identity of the MPTP

Work from this laboratory and many others has sought to elucidate the molecular mechanism of the MPTP. Although its exact composition remains uncertain [22], [92], [93], [94] several proteins have been implicated in either the structure or regulation of the MPTP with varying degrees of certainty. These are considered in turn below.

A working model for the MPTP

A working model for the MPTP that is consistent with our data is summarised in Fig. 3. It is proposed that dimers may form between that ANT and PiC in the inner mitochondrial membrane for which there is some direct evidence within the ‘ATP synthasome’ [142], [143]. Oxidative stress is suggested to enhance the interaction between these two proteins which is also greater when the ANT is in the ‘c’ conformation. Increased matrix [Ca2+] will trigger a conformational change in one or both of these

Acknowledgments

This work was supported by a Programme Grant (RG/03/002) and a research studentship (FS/04/043) from the British Heart Foundation. I am grateful to the many colleagues whose efforts over the years have contributed to our current, if flawed, understanding of the MPTP.

References (147)

  • 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)
  • L. Argaud et al.

    Specific inhibition of the mitochondrial permeability transition prevents lethal reperfusion injury

    J. Mol. Cell. Cardiol.

    (2005)
  • E. Basso et al.

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

    J. Biol. Chem.

    (2005)
  • 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)
  • 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)
  • T.E. Gunter et al.

    Mitochondrial calcium transport: mechanisms and functions

    Cell Calcium

    (2000)
  • I. Szabo et al.

    The giant channel of the inner mitochondrial membrane is inhibited by cyclosporin-A

    J. Biol. Chem.

    (1991)
  • T.A. Lohret et al.

    Activity of the mitochondrial multiple conductance channel is independent of the adenine nucleotide translocator

    J. Biol. Chem.

    (1996)
  • 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)
  • F. Ichas et al.

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

    Cell

    (1997)
  • 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)
  • A.P. Halestrap et al.

    The permeability transition pore complex: another view

    Biochimie

    (2002)
  • 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)
  • V. Petronilli et al.

    Modulation of the mitochondrial cyclosporin A-sensitive permeability transition pore .2. The minimal requirements for pore induction underscore a key role for transmembrane electrical potential, matrix pH, and matrix Ca2+

    J. Biol. Chem.

    (1993)
  • 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)
  • 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)
  • 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)
  • S.A. Novgorodov et al.

    The nonspecific inner membrane pore of liver mitochondria — modulation of cyclosporin sensitivity by ADP at carboxyatractyloside-sensitive and insensitive sites

    Biochem. Biophys. Res. Commun.

    (1991)
  • M. Klingenberg

    The ADP and ATP transport in mitochondria and its carrier

    Biochim. Biophys. Acta

    (2008)
  • M.C. Beatrice et al.

    The role of glutathione in the retention of Ca2+ by liver mitochondria

    J. Biol. Chem.

    (1984)
  • V. Petronilli et al.

    Physiological effectors modify voltage sensing by the cyclosporin A-sensitive permeability transition pore of mitochondria

    J. Biol. Chem.

    (1993)
  • V. Petronilli et al.

    The voltage sensor of the mitochondrial permeability transition pore is tuned by the oxidation-reduction state of vicinal thiols — increase of the gating potential by oxidants and its reversal by reducing agents

    J. Biol. Chem.

    (1994)
  • F. DiLisa et al.

    Negative contrast imaging of mitochondria by confocal microscopy — response to J. J. Lemasters et al

    Biophys. J.

    (1999)
  • P. Costantini et al.

    Modulation of the mitochondrial permeability transition pore by pyridine nucleotides and dithiol oxidation at two separate sites

    J. Biol. Chem.

    (1996)
  • M.P. Rigobello et al.

    Inhibition of rat liver mitochondrial permeability transition by respiratory substrates

    Arch. Biochem. Biophys.

    (1995)
  • A. Bindoli et al.

    Influence of the redox state of pyridine nucleotides on mitochondrial sulfhydryl groups and permeability transition

    Arch. Biochem. Biophys.

    (1997)
  • C. Salet et al.

    Singlet oxygen produced by photodynamic action causes inactivation of the mitochondrial permeability transition pore

    J. Biol. Chem.

    (1997)
  • J. Wudarczyk et al.

    Relation between the activities reducing disulfides and the protection against membrane permeability transition in rat liver mitochondria

    Arch. Biochem. Biophys.

    (1996)
  • R. Krämer

    Interaction of membrane surface charges with the reconstituted ADP/ATP-carrier from mitochondria

    Biochim. Biophys. Acta

    (1983)
  • E. Fontaine et al.

    A ubiquinone-binding site regulates the mitochondrial permeability transition pore

    J. Biol. Chem.

    (1998)
  • L. Walter et al.

    Three classes of ubiquinone analogs regulate the mitochondrial permeability transition pore through a common site

    J. Biol. Chem.

    (2000)
  • E. Fontaine et al.

    Regulation of the permeability transition pore in skeletal muscle mitochondria — modulation by electron flow through the respiratory chain complex

    J. Biol. Chem.

    (1998)
  • A.W. Leung et al.

    Recent progress in elucidating the molecular mechanism of the mitochondrial permeability transition pore

    Biochim. Biophys. Acta

    (2008)
  • S.L. Schreiber et al.

    The mechanism of action of cyclosporin-A and FK506

    Immunol. Today

    (1992)
  • A.R. Crofts et al.

    Calcium ion accumulation and volume changes of isolated liver mitochondria: reversal of calcium ion-induced swelling

    Biochem. J.

    (1965)
  • J.B. Chappell et al.

    Calcium ion accumulation and volume changes of isolated liver mitochondria: calcium ion-induced swelling

    Biochem. J.

    (1965)
  • T.E. Gunter et al.

    Mechanisms by which mitochondria transport calcium

    Am. J. Physiol.

    (1990)
  • M. Crompton et al.

    Evidence for the presence of a reversible Ca2+-dependent pore activated by oxidative stress in heart mitochondria

    Biochem. J.

    (1987)
  • Cited by (772)

    View all citing articles on Scopus
    View full text