Elsevier

Biochimie

Volume 84, Issues 2–3, February–March 2002, Pages 131-141
Biochimie

Mitochondrial reactive oxygen species in cell death signaling

https://doi.org/10.1016/S0300-9084(02)01369-XGet rights and content

Abstract

During apoptosis, mitochondrial membrane permeability (MMP) increases and the release into the cytosol of pro-apoptotic factors (procaspases, caspase activators and caspase-independent factors such as apoptosis-inducing factor (AIF)) leads to the apoptotic phenotype. Apart from this pivotal role of mitochondria during the execution phase of apoptosis (documented in other reviews of this issue), it appears that reactive oxygen species (ROS) produced by the mitochondria can be involved in cell death. These toxic compounds are normally detoxified by the cells, failing which oxidative stress occurs. However, ROS are not only dangerous molecules for the cell, but they also display a physiological role, as mediators in signal transduction pathways. ROS participate in early and late steps of the regulation of apoptosis, according to different possible molecular mechanisms. In agreement with this role of ROS in apoptosis signaling, inhibition of apoptosis by anti-apoptotic Bcl-2 and Bcl-xL is associated with a protection against ROS and/or a shift of the cellular redox potential to a more reduced state. Furthermore, the fact that active forms of cell death in yeast and plants also involve ROS suggests the existence of an ancestral redox-sensitive death signaling pathway that has been independent of caspases and Bcl-2.

Introduction

The Bcl-2/Ced-9 family of proteins is involved in positive and negative regulation of apoptotic cell death (review in 〚1〛). Among the anti-apoptotic members, Bcl-2 and Bcl-xL are negative regulators of cell death, preventing cells from undergoing apoptosis induced by various stimuli in a wide variety of cell types 〚2〛, 〚3〛, whereas others, such as Bax and Bid promote or accelerate cell death. In mammals, most of our understanding of Bcl-2 regulation of apoptosis involves its mitochondrial membrane association. Although for a long time apoptosis was considered to be under the control of nuclear events, mitochondria appear today as the central control point of programmed cell death (PCD) 〚4〛, 〚5〛, 〚6〛, 〚7〛. It is now clear that mitochondrial membrane-associated Bcl-2 acts by regulating the release in the cytosol of pro-apoptotic factors usually sequestered in the mitochondrial intermembrane space (reviewed in other articles of this special issue of Biochimie). Moreover, inhibition of apoptosis by anti-apoptotic Bcl-2 and Bcl-xL is associated with a protection against oxidants and/or a shift of the cellular redox potential to a more reduced state. These results have suggested that reactive oxygen species (ROS) could be mediators of apoptosis. However, other data have shown that apoptosis can occur in very low oxygen environments 〚8〛, 〚9〛, which has cast doubt on this interpretation. A growing number of data show now that ROS, mainly produced by the mitochondria, can be involved in cell death. On the one hand, they can produce an oxidative stress leading to cell destruction, as observed during necrosis or the so-called ‘post-mitochondrial’ phase of apoptosis. On the other hand, in several instances, ROS derived from mitochondria are also involved in the initiation phase of apoptosis contributing to cell death signaling. The present review will focus on the role of mitochondrial ROS in cell death signaling and the possible molecular mechanisms.

Section snippets

Mitochondria are the major site of ROS production

ROS, such as superoxide anion, radicals, hydrogen and organic peroxides, are generated by all aerobic cells as by-products of a number of metabolite reactions and in response to various stimuli 〚10〛. Mitochondria are believed to be a major site of ROS production, according to an endogenous and continuous physiological process under aerobic conditions (Fig. 1). However, endoplasmic reticulum and nuclear membranes also contain electron (e) transport chains that can lose e and generate

Oxidative stress

An excess of ROS in mitochondria causes an oxidative stress, with an enhanced activity of the antioxidant defense system and mitochondrial damage. Several situations, such as dysfunctional complex I 〚23〛, chemical poisoning and ischaemia followed by reperfusion, may subject tissues and cells to oxidative stress The main targets of ROS in mitochondria are the protein components of the membranes and the polyunsaturated fatty acids. In membranes, ROS affect cysteine residues (sulfhydryl groups),

Protection of the cell

To prevent oxidative damage, mammalian cells have developed a complex antioxidant defense system that includes non-enzymatic antioxidants (e.g. glutathione, thioredoxine) as well as enzymatic activities (e.g. catalase, SOD) 〚25〛.

Mitochondria possess an antioxidant system with the superoxide dismutase (SOD), NADH and a complete glutathione redox system, which is formed of glutathione reductase, reduced glutathione (GSH), and glutathione peroxidase (Fig. 1). This system allows the reduction of

Physiological role

Cells possess multiple sites for ROS production and a few mechanisms for their degradation. ROS are also involved in different physiological processes, as mediators in signal transduction pathway, activating proteins such as tyrosine kinase, mitogen-activated protein kinase system or small Ras proteins. Current evidence indicates that different stimuli use ROS as signaling messengers to activate transcription factors, such as AP-1 and NF-κB, and induce gene expression 〚31〛. Thus, they are

ROS participate in early and late steps of the regulation of apoptosis

Since the discovery of ROS involvement in TNF-α-induced killing, their contribution to the activation of the execution machinery was extended to PCD triggered by a wide range of apoptosis inducers 〚62〛. Notably, ROS accumulation preceded mitochondrial membrane alterations, nuclear condensation and other typical apoptotic events. This point can be illustrated by TNF-α-induced apoptosis. Indeed, experiments using antioxidants show that ROS act upstream of mitochondrial membrane depolarization 〚63〛

Origin of apoptosis-mediated ROS

Although fatty acid metabolites, such as those produced from arachidonic acid by the lipoxygenase pathway, may be mediators of apoptosis 〚71〛, it was established that both ROS accumulation and the programmed cell death process require the presence of a functional mitochondrial respiratory chain in most ROS-dependent cell death systems 〚40〛, 〚51〛, 〚63〛, 〚72〛. Indeed, it was shown that an upstream inhibition, with chemical compounds acting on complex I 〚39〛, 〚51〛, or an elimination of the

Mechanisms of ROS signaling

The involvement of mitochondrial ROS in some cell death transduction pathways leads to fundamental questions concerning, on the one hand, the causal event of the increased ROS generation and, on the other hand, the molecular mechanisms underlying the ROS signaling. Two viewpoints must first be considered to address the question of the origin of ROS accumulation, which can indeed result from an increased production or from a reduced scavenging by the cellular detoxifying systems. Much of the

Inhibition of apoptosis by Bcl-2 and Bcl-xL is associated with a protection against ROS and/or a shift of the cellular redox potential to a more reduced state

Several lines of evidence support the idea that Bcl-2, in addition to its role in the regulation of the permeability of the outer mitochondrial membrane, might act as an antioxidant partner to block a putative ROS-mediated step in the cascade of events required for apoptosis. For example, during TNF-α-induced apoptosis, which involves ROS signaling 〚63〛, Bcl-2, as antioxidants do, prevents ROS accumulation and the subsequent events (mitochondrial membrane depolarization, Bax relocalization,

ROS, plants PCD and Yeast pseudo-apoptosis

In plants, ROS are also involved in some PCD pathways, such as the synchronous programmed death of petal cells 〚89〛, the hypersensitive response (HR) of plants resistant to microbial pathogens 〚90〛, or the hormonally regulated cell death pathway in barley aleurone cells 〚91〛. More surprising is the occurrence of a ROS-associated apoptosis-like process in unicellular eukaryotic cells like yeasts 〚92〛. Saccharomyces cerevisiae and Schizosaccharomyces pombe yeast species do not contain endogenous

Conclusion

In conclusion, mitochondria are involved in the decision of cells to survive or not at several levels (Fig. 4). First, mitochondria can activate the cell death machinery by releasing in the cytosol pro-apoptotic factors such as procaspases, caspase activators (i.e. cytochrome c and Smac/Diablo) or caspase-independent factors such as AIF. Furthermore, the importance of mitochondria in apoptosis has been reinforced by studies showing the contribution of reactive oxygen species in cell death

References (113)

  • R. Pinkus et al.

    Role of oxidants and antioxidants in the induction of AP-1, NF-kB, and glutathione S-transferase gene expression

    J. Biol. Chem.

    (1996)
  • A.S. Lundberg et al.

    Genes involved in senescence and immortalization

    Curr. Opin. Cell Biol.

    (2000)
  • S. Pervaiz et al.

    Superoxide anion inhibits drug-induced tumor cell death

    FEBS Lett. 459

    (1999)
  • R.H. Burdon

    Superoxide and hydrogen peroxide in relation to mammalian cell proliferation

    Free Radic. Biol. M

    (1995)
  • J.R. Lancaster et al.

    Inhibition of target cell mitochondrial electron transfer by tumor necrosis factor

    FEBS Lett.

    (1989)
  • K. Schulze-Osthoff et al.

    Cytotoxic activity of tumor necrosis factor is mediated by early damage of mitochondrial functions. Evidence for the involvement of mitochondrial radical generation

    J. Biol. Chem.

    (1992)
  • R.J. Carmody et al.

    Reactive oxygen species as mediators of photoreceptor apoptosis in vitro

    Exp. Cell Res.

    (1999)
  • L.J.S. Greenlund et al.

    Superoxide dismutase delays neuronal apoptosis: a role for reactive oxygen species in programmed neuronal death

    Neuron

    (1995)
  • P. Mehlen et al.

    Small stress proteins as novel regulators of apoptosis

    J. Biol. Chem.

    (1996)
  • A. Quillet-Mary et al.

    Implication of mitochondrial hydrogen peroxide generation in ceramide- induced apoptosis

    J. Biol. Chem.

    (1997)
  • P.F. Li et al.

    Superoxide induces apoptosis in cardiomyocytes, but proliferation and expression of transforming growth factor-beta1 in cardiac fibroblasts

    FEBS Lett.

    (1999)
  • S.K. Manna et al.

    Overexpression of manganese superoxide dismutase suppresses tumor necrosis factor-induced apoptosis and activation of nuclear transcription factor-kappaB and activated protein-1

    J. Biol. Chem.

    (1998)
  • H.J. Majima et al.

    Prevention of mitochondrial injury by manganese superoxide dismutase reveals a primary mechanism for alkaline-induced cell death

    J. Biol. Chem.

    (1998)
  • K. Nomura et al.

    Mitochondrial phospholipid hydroperoxide glutathione peroxidase suppresses apoptosis mediated by a mitochondrial death pathway

    J. Biol. Chem.

    (1999)
  • M. Degli Esposti et al.

    Mitochondria and cells produce reactive oxygen species in virtual anaerobiosis: relevance to ceramide-induced apoptosis

    FEBS Lett.

    (1998)
  • M.D. Jacobson

    Reactive oxygen species and programmed cell death

    Trends Biochem. Sci.

    (1996)
  • N. Maulik et al.

    Oxidative stress developed during the reperfusion of ischemic myocardium induces apoptosis

    Free Radic. Biol. M

    (1998)
  • S. Tada-Oikawa et al.

    Generation of hydrogen peroxide precedes loss of mitochondrial membrane potential during DNA alkylation-induced apoptosis

    FEBS Lett.

    (1999)
  • J. Cai et al.

    Superoxide in apoptosis. Mitochondrial generation triggered by cytochrome c loss

    J. Biol. Chem.

    (1998)
  • T.I. Gudz et al.

    Direct inhibition of mitochondrial respiratory cahin complex III by cell-permeable ceramide

    J. Biol. Chem.

    (1997)
  • D.J. van den Dobbelsteen et al.

    Rapid and specific efflux of reduced glutathione during apoptosis induced by anti-Fas/APO-1 antibody

    J. Biol. Chem.

    (1996)
  • G. Kroemer et al.

    Mitochondrial control of apoptosis

    Immunol. Today

    (1997)
  • Y.T. Hsu et al.

    Bax in murine thymus is a soluble monomeric protein that displays differential detergent-induced conformations

    J. Biol. Chem.

    (1998)
  • D.M. Hockenbery et al.

    Bcl-2 functions in an antioxidant pathway to prevent apoptosis

    Cell

    (1993)
  • Y.Y. Tyurina et al.

    Direct evidence for antioxidant effect of Bcl-2 in PC12 rat pheochromocytoma cells

    Arch. Biochem. Biophys.

    (1997)
  • H.M. Steinman

    The Bcl-2 oncoprotein functions as a pro-oxidant

    J. Biol. Chem.

    (1995)
  • W. Greenhalf et al.

    Role of mitochondria and C-terminal membrane anchor Bcl-2 in Bax induced growth arrest and mortality in Saccharomyces cerevisiae

    FEBS Lett.

    (1996)
  • M. Hanada et al.

    Structure-function analysis of Bcl-2 protein. Identification of conserved domains important for homodimerization with Bcl-2 and heterodimerization with Bax

    J. Biol. Chem.

    (1995)
  • C. James et al.

    CED-4 induces chromatin condensation in Schizosaccharomyces pombe and is inhibited by direct physical association with CED-9

    Curr. Biol.

    (1997)
  • A. Gross et al.

    BCL-2 family members and the mitochondria in apoptosis

    Genes Dev.

    (1999)
  • L.T. Zhong et al.

    Bredesen D.E., bcl-2 inhibits death of central neural cells induced by multiple agents

    Proc. Natl. Acad. Sci. USA

    (1993)
  • B. Mignotte et al.

    Mitochondria and apoptosis

    Eur. J. Biochem.

    (1998)
  • N. Zamzami et al.

    The mitochondrion in apoptosis: how Pandora's box opens

    Nat. Rev. Mol. Cell Biol.

    (2001)
  • K.F. Ferri et al.

    Mitochondria: the suicide organelles

    Bioessays

    (2001)
  • M.D. Jacobson et al.

    Programmed cell death and Bcl-2 protection in very low oxygen

    Nature

    (1995)
  • S. Shimizu et al.

    Prevention of hypoxia-induced cell death by Bcl-2 and Bcl-XL

    Nature

    (1995)
  • I. Fridovich

    The biology of oxygen radicals

    Science

    (1978)
  • S. Papa et al.

    Reactive oxygen species, mitochondria, apoptosis and aging

    Mol. Cell. Biochem.

    (1997)
  • K. Takeshige et al.

    NADH- and NADPH-dependent formation of superoxide anions by bovine heart submitochondrial particles and NADH-ubiquinone reductase preparation

    Biochem. J.

    (1979)
  • A. Boveris et al.

    Role of ubiquinone in the mitochondrial generation of hydrogen peroxide

    Biochem. J.

    (1976)
  • Cited by (0)

    View full text