You are on page 1of 11

Mitochondrion 10 (2010) 8393

Contents lists available at ScienceDirect

Mitochondrion
journal homepage: www.elsevier.com/locate/mito

Review

Gases in the mitochondria


Pamela B.L. Pun 1, Jia Lu 2, Enci M. Kan 3, Shabbir Moochhala *
Combat Care Laboratory, Defence Medical and Environmental Research Institute, DSO National Laboratories, 27 Medical Drive, #12-00, Singapore 117510, Singapore

a r t i c l e

i n f o

a b s t r a c t
Gasomodulators nitric oxide, carbon monoxide and hydrogen sulphide are important physiological mediators that have been implicated in disorders such as neurodegeneration and sepsis. Some of their biological functions involve the mitochondria. In particular, their inhibition of cytochrome c oxidase has received much attention as this can cause energy depletion and cytotoxicity. However, reports that cellular energy production and cell survival are maintained even in the presence of gasomodulators are not uncommon. In both cases, modulation of mitochondrial targets by the gasomodulators appears to be an important event. We provide an overview of the effects of the gasomodulators on the mitochondria. 2009 Elsevier B.V. and Mitochondria Research Society. All rights reserved.

Article history: Received 3 July 2009 Received in revised form 3 November 2009 Accepted 7 December 2009 Available online 22 December 2009 Keywords: Nitric oxide Carbon monoxide Hydrogen sulphide Gasomodulators Mitochondria

1. Introduction Gasomodulators, also known as gasotransmitters, have emerged over the last decade as important physiological mediators involved in various organs such as the nervous and cardiovascular systems (Moore et al., 2003; Wu and Wang, 2005; Olson and Donald, 2009). To qualify as gasomodulators, gases should: (a) be able to diffuse through biological membranes, (b) be produced endogenously and this production should be regulated, (c) mediate biological processes at physiological concentrations, and (d) have specic biological targets (Wang, 2002). Other properties proposed to be common to gasomodulators include short half-lives and their propensity to cause damage when present in excess (Kasparek et al., 2007). Numerous gases such as ammonia, nitrous oxide and sulphur dioxide are speculated to be of physiological signicance (Li and Moore, 2007). However, only three gases have thus far been denitively established as gasomodulators, namely nitric oxide (NO), carbon monoxide (CO) and hydrogen sulphide (H2S) (Wang,
Abbreviations: ATA, atmospheres absolute; CBS, cystathionine-b-synthase; CO, carbon monoxide; COX, cytochrome c oxidase; CSE, cystathionine-c-lyase; ETC, electron transport chain; GI, gastrointestinal; H2O2, hydrogen peroxide; H2S, hydrogen sulphide; HBOT, hyperbaric oxygen therapy; HO, heme oxygenase; MAO, monoamine oxidase; mPT, mitochondrial permeability transition; mtDNA, Mitochondrial DNA; mtNOS, Mitochondrial nitric oxide synthase; NO, Nitric oxide; ONOO-, Peroxynitrite; ROS, Reactive oxygen species ; SQR, Sulphide-quino-oxidoreductase; UCP, Uncoupling protein. * Corresponding author. Tel.: +65 6485 7201; fax: +65 6485 7226. E-mail addresses: u0402652@alumni.nus.edu.sg (P.B.L. Pun), ljia@dso.org.sg (J. Lu), kenci@dso.org.sg (E.M. Kan), mshabbir@dso.org.sg (S. Moochhala). 1 Tel.: +65 6485 7217; fax: +65 6485 7226. 2 Tel.: +65 6485 7204; fax: +65 6485 7226. 3 Tel.: +65 6485 7212; fax: +65 6485 7226.

2002). The relevance of gasomodulators in biology has previously been reviewed in terms of their involvements in different organs systems and/or diseases (Wang et al., 2006; Fukuto and Collins, 2007; Kasparek et al., 2007; Li and Moore, 2007). Briey, at physiological concentrations, all three gases participate in cell signalling and regulate tissue function. For instance, in the gastrointestinal (GI) tract, NO, CO and H2S all act on the GI smooth muscle cells and inhibit GI contraction (Kasparek et al., 2007). Similarly, in the cardiovascular system, all three gases promote vasodilation and angiogenesis, resulting in cardioprotection (Li et al., 2009). Within the context of diseases, the gasomodulators have attracted signicant attention particularly in the eld of sepsis in which the gases are thought to exert protective effects by variously acting as antioxidants, decreasing inammation or even inducing suspended animation (Baumgart et al., 2009). Unfortunately, gasomodulators could be detrimental when present in excess. NO, for instance, is neurotoxic at high concentrations and is believed to contribute to the pathogenesis of neurodegenerative disorders such as Parkinsons disease and Alzheimers disease (Molina et al., 1998; Calabrese et al., 2007). Likewise, excessive CO could cause brain damage by binding to, and thus excluding oxygen from, haemoglobin (Prockop and Chichkova, 2007). Supra-physiological levels of H2S, meanwhile, may participate in the pathogenesis of a variety of conditions including inammation, sepsis and stroke (Lowicka and Beltowski, 2007). Interestingly, a common feature of many of these conditions is the mitochondria (Beal, 2007; Exline and Crouser, 2008). It appears plausible then that one mechanism by which gasomodulators exert biological functions involves mitochondrial targets. The purpose of this review, therefore, is to provide a brief but concise overview on the effects of gasomodulators in the mitochondria. We discuss

1567-7249/$ - see front matter 2009 Elsevier B.V. and Mitochondria Research Society. All rights reserved. doi:10.1016/j.mito.2009.12.142

84

P.B.L. Pun et al. / Mitochondrion 10 (2010) 8393

these effects independent of any disease and organ system, for which the reader is referred to other excellent reviews (Moore et al., 2003; Wu and Wang, 2005; Wang et al., 2006; Fukuto and Collins, 2007; Kasparek et al., 2007; Li and Moore, 2007; Olson and Donald, 2009). Our review focuses on the three established gasomodulators, NO, CO and H2S. 2. Nitric oxide The discovery that the endothelium-derived relaxing factor is NO in 1987 (Palmer et al., 1987) led to a burgeoning research eld investigating the biological signicance of NO. In particular, the effect of NO on the mitochondria is one relevant research eld to reactive oxygen species biology. NO synthesis is catalysed by NO synthase (NOS) of which the neuronal NOS (nNOS), endothelial NOS (eNOS) and inducible NOS (iNOS) are the most well known with other known spliced and post-translationally modied variants. However, the source of NO in the mitochondria is still debatable, with many citing the presence of a putative mitochondrial NOS (mtNOS) as a possible source. The rst reports of mtNOS came in 1995 where Bates et al. demonstrated the immunocytochemical localization of NOS in the rat mitochondria. Progressively, several other groups also reported NOS function in the mitochondria (Ghafourifar and Richter, 1997; Giulivi et al., 1998). Typically, NOS isoforms are encoded by nuclear DNA rather than mtDNA. It has thus been postulated that mtNOS may simply be a variant of the more established NOS isoforms (nNOS, eNOS, or iNOS) that is synthesized in and translocated from the cytosol into the mitochondria (Finocchietto et al., 2009). Initially, eNOS was touted as the cytosolic candidate (Kobzik et al., 1995), followed by iNOS (Elfering et al., 2002). However, recent data has indicated strong evidence that a modied nNOS may be the primary candidate for mtNOS. Kanai et al. (2001) reported that nNOS knockout in mice abolished calcium-induced mitochondrial release of NO. Further evidence showed that the mechanism via which nNOS translocates into mitochondria is through the processing of a PDZ domain from 157 kDa to 144 kDa (Carreras et al., 2008). The presence of iNOS in mitochondria could be due to a pathologic condition as iNOS was shown to be present in mitochondria during endotoxemia brought about in experimental sepsis models (Lisdero et al., 2004; Lopez et al., 2006). Interestingly, another novel family of putative NOS, Arabidopsis thaliana NOS1 (AtNOS1) which does not shown any homology with existing animal NOSs was found to be associated with NO production (Guo et al., 2003). The mammalian homolog of AtNOS1, the mouse mAtNOS1, has also been characterized and is found to be localized to the inner mitochondrial membrane of mouse broblasts (Zemojtel et al., 2006). Although NO from AtNOS1 has been reported to protect against oxidative damage in plants (Guo and Crawford, 2005), the functions of mATNOS1 in mammalians remain unclear and research is still in the in vitro stage which may not reect accurately the effects in in vivo physiological conditions. As the community continues to determine whether eNOS, nNOS, iNOS or even whether mAtNOS1 makes up the genuine mtNOS, the levels of mtNOS and consequently, NO production have been reported to be modulated by hormones (Carreras et al., 2001), development and aging (Riobo et al., 2002; Valdez et al., 2004) as well as environmental changes (Peralta et al., 1993). The effects of different NO levels on mitochondria function especially in relation to its inhibition of cytochrome c oxidase (COX) will be discussed in the following paragraphs. NO is an inhibitor of the electron transport chain (ETC), with the propensity to affect the activities of possibly all complexes of the respiratory chain. Most attention has focused on its inuence on COX activity. COX is the nal complex in the ETC and mediates the reduction of oxygen to water. This complex contains a heme

a3CuB binuclear centre to which oxygen binds when both metals are in the reduced state (i.e. Fe2+ and Cu+) (Tsukihara et al., 1996). However, NO can compete with oxygen for binding to the same site, resulting in competitive but reversible inhibition of COX (Brown and Cooper, 1994; Cleeter et al., 1994). It has been suggested that NO is not a typical competitive inhibitor which functions simply by binding reversibly to COX (Pearce et al., 2003). This stems from observations that the inhibitory effects on COX of NO and cyanide (another COX inhibitor) are not additive (Pearce et al., 2003). The authors of that study proposed that NO rst binds to the heme a3 site of COX. In the meantime, superoxide is formed at the CuB site when Cu+ donates an electron to oxygen. The resulting superoxide reacts with NO to form peroxynitrite (ONOO). The subsequent reduction of ONOO by COX then generates nitrite and water. Should this hypothesis be true, one may expect that the formation of ONOO at the binuclear centre of COX should render the enzyme highly vulnerable to nitrosative damage. However, the group has indicated the conversion of ONOO to nitrite and water to be quick and facile, suggesting that ONOO will be so rapidly converted to nitrite that it will not react with COX to cause damage. Furthermore, studies by another group have found ONOO to have little effect on COX activity in isolated rat heart mitochondria (Radi et al., 1994; Cassina and Radi, 1996). However, the propensity of ONOO to inhibit COX appears to be dependent both on cell type and ONOO concentration (Bolanos et al., 1995). Nonetheless, it remains plausible that even if ONOO is not quickly reacted away, that it is unlikely to cause severe irreversible damage to the COX protein. NO could also participate in uncompetitive inhibition by binding to the oxidized form of the enzyme, forming nitrite in the process and decreasing COX activity (Cooper et al., 1997). These two modes of inhibition can be distinguished based on their sensitivity to oxygen and light, with competitive inhibition being responsive and uncompetitive inhibition being resistant to changes in either parameter (Sarti et al., 2000; Mason et al., 2006). It is also apparent from these unique characteristics that the two forms of inhibition dominate under different conditions, with competitive inhibition likely to predominate when oxygen levels are low and COX is, for the most part, in the reduced state. Conversely, uncompetitive inhibition is more likely to occur under the opposite conditions (Sarti et al., 2000; Mason et al., 2006). While oxygen can modulate the inhibitory effects of NO by competing with it for binding to COX, its effects on this process are likely to be manifold as oxygen can inuence NO synthesis and breakdown, thus regulating levels of NO and its downstream activities, a prospect that has been previously discussed (Cooper and Giulivi, 2007). Fig. 1 summarizes the inhibition of COX by NO under low and high oxygen conditions. Based upon studies in isolated mitochondria, the IC50 of NO has been determined to increase in direct proportion to the square of the oxygen tension (Kovisto et al., 1997). As the typical oxygen concentration in mammalian tissues is 30 lM, it appears that physiological levels of NO (1200 nM) are sufcient to signicantly inhibit respiration (Brown, 1999) and be of cellular relevance. Besides COX, NO and its derivative, ONOO, could also inhibit the other complexes in the ETC (Bolanos et al., 1996; Cassina and Radi, 1996; Lizasoain et al., 1996; Stadler et al., 1991a,b). The precise pathway by which such inhibition is achieved remains poorly dened although there are candidate mechanisms including tyrosine nitration, S-nitrosation and damage to the ironsulphur (Fe S) clusters of the complex (Brown, 1999; Brown and Borutaite, 2004; Welter et al., 1996). In addition, ONOO can decrease aconitase activity (Castro et al., 1994), thus reducing the availability of substrates required for oxidative phosphorylation. Since NO reduces electron ux through the respiratory chain, it follows then that NO must increase reactive oxygen species (ROS) production by the ETC, and indeed, evidence for this is plentiful.

P.B.L. Pun et al. / Mitochondrion 10 (2010) 8393

85

H2O2
Low [NO]

.-

Low Oxygen

CuBI heme a3II

O2

.-

B. ROS
production
High [NO]

heme aII

CuAI

O2 NO

4eONOO-

2H+
fast

NO2+ H2O

C. Rapid conversion

COX

A. Competitive
Inhibition

terminates NO inhibitive effects

Fig. 1. Summary of effects of NO inhibition on COX during low oxygen condition. (A) NO competes with O2 and binds reversibly to COX, leaving O2 to be reduced by ). (B) ROS production is propagated as superoxide is either COX to superoxide (O2 converted to H2O2 (at low [NO]) or peroxynitrite (at high [NO]). (C) Rapid conversion of peroxynitrite to H2O and NO3 terminates by-product effects of NO inhibition on COX.

For instance, superoxide and hydrogen peroxide (H2O2) levels are elevated in cells exposed to NO (Ponderoso et al., 1996; Wei et al., 2000; Sarkela et al., 2001). NO and superoxide can further react to form ONOO, while hydrogen peroxide may go onto form the highly reactive and damaging hydroxyl radical. This formation of a cascade of reactive species could be detrimental and promote cell death by causing oxidative/nitrosative damage to cellular components (Therond, 2006). For instance, peroxidative damage of cardiolipin disrupts the cardiolipincytochrome c complex, leading to the release of cytochrome c (Ushmorov et al., 1999; Vlasova et al., 2006). Similarly, oxidation of protein thiols in the mitochondrial permeability transition (mPT) pore leads to pore opening, consequently causing a drop in mitochondrial membrane potential, mitochondrial swelling and further release of cytochrome c (Hortelano et al., 1997; Bal-Price and Brown, 2000; Saviani et al., 2002). Additionally, ROS can activate signalling pathways such as that involving p38 MAPK, leading to the up-regulation and translocation of pro-apoptotic proteins like Bax to the mitochondria (Ghatan et al., 2000; Cheng et al., 2001). It should be noted, however, that the ability of NO to induce apoptosis is concentration-dependent, and that NO at low concentrations is anti-apoptotic rather than pro-apoptotic (Choi et al., 2002; Yoshioka et al., 2006). However, when present in greater abundance, and if ATP is available, NO can cause apoptosis by the pathways mentioned above. Given that NO inhibits respiration, ATP levels can be expected to drop as a result. However, cells may be more resilient to NO-induced declines in energy production and even apoptosis than would be expected in theory. The two main reasons for such resistance lie in increased mitochondrial biogenesis and up-regulation of glycolysis and glucose uptake. Firstly, NO, by activating guanylate cyclase and increasing cGMP levels, activates mitochondrial biogenesis in a PFC-1a and Tfam-dependent manner (Nisoli et al., 2003; Clementi and Nisoli, 2005). With more mitochondria, the energy production capacity of the cell, or tissue as a whole, is increased, thus providing a buffer against any drop in ATP production due to ETC inhibition predicted per mitochondrion. Secondly, while high levels of ROS can be detrimental, more moderate levels are important in cell signalling. In this context, ROS generated by NO-mediated inhibition of the respiratory complexes can participate in signalling pathways such as those involving Nrf2 and AMPK, leading to cytoprotection. For instance, Nrf2 activation induces the expression of antioxidant genes, thus protecting

against oxidative damage and consequently apoptosis (Dhakshinamoorthy and Porter, 2004). The NO-derived ONOO may also exert an indirect antioxidant effect via the pentose phosphate shunt which generates NADPH, a substrate crucial for the regeneration of the antioxidant, glutathione (Garcia-Nogales et al., 2003). Activation of AMPK also protects cells by up-regulating glycolysis and glucose uptake (Cidad et al., 2004; Almeida et al., 2005) to compensate for any drop in ATP production due to respiratory inhibition. Because ATP production via glycolysis is far less efcient than that by oxidative phosphorylation (in terms of number of ATP molecules produced per glucose molecule utilized), the adequacy of such compensation is questionable. Nonetheless, the maintenance of ATP production further protects the cell from apoptosis as it makes possible the preservation of the mitochondrial membrane potential by allowing the reversal of the F0F1-ATPase (Almeida et al., 2001). Taken together, these events minimize oxidative damage, allow for a compensatory increase in ATP production, and reduce the likelihood of apoptosis occurring, thereby protecting the cell. Because they may occur to varying extents in different cell types, some cells such as neuronal cells may be more susceptible to NO toxicity relative to others like the glial cells (Almeida et al., 2001; Bolanos and Almeida, 2006). As a result, cell types with less capacity for glycolytic ATP production are more likely to undergo necrosis than those with greater propensity for glycolysis (Borutaite and Brown, 2003). Considering the above points, it is apparent that NO could be detrimental or protective to the cell, depending on factors such as cell type and NO levels. To maintain such a delicate balance in NO levels requires a homeostatic pathway governing NO levels within mitochondria. One such candidate pathway involves mitochondrial nitric oxide synthase (mtNOS; a proposed NOS isoform that exists in the mitochondria in close association with COX of the ETC) (Ghafourifar and Richter, 1997; Persichini et al., 2005) and mitochondrial calcium concentration ([Ca2+]m). High [Ca2+]m increases mtNOS activity, generating more NO within the mitochondria. NO, in turn, opens the mPT pore, decreasing mitochondrial membrane potential. As a result, Ca2+ is released from the mitochondria. NO also reduces Ca2+ uptake into the mitochondria, further decreasing [Ca2+]m. As [Ca2+]m drops, mtNOS activity correspondingly decreases. NO levels thus decrease and [Ca2+]m subsequently increases, repeating the cycle again (Ghafourifar and Richter, 1999). 3. Carbon monoxide CO is produced endogenously as a by-product of heme breakdown in a process catalysed by heme oxygenase (HO)-1 and 2 (Yoshida and Kikuchi, 1974). Although heme catabolism by HO also generates biliverdin and free iron, each of which has its own physiological roles (Elbirt and Bonkovsky, 1999; Foresti et al., 2004), the effects of HO-1 on the mitochondria appears to be due predominantly to CO production (DAmico et al., 2006). In the mitochondria, CO has been shown to inhibit COX (Petersen, 1977; Pankow and Ponsold, 1984; Miro et al., 1998; Alonso et al., 2003; Iheagwara et al., 2007; Zuckerbraun et al., 2007). Like NO, CO achieves this inhibitory effect by competing with oxygen for binding to the reduced form of the enzyme. Unlike NO, this is the only mode of inhibition inhibition of COX by CO. That is, CO is unable to bind to the oxidized form of COX and therefore, its inhibition is exclusively competitive (Petersen, 1977). CO itself is converted to carbon dioxide by COX after binding (Young and Caughey, 1986). It is thought that under physiological conditions, CO-mediated inhibition of COX is unlikely to be signicant because CO concentration is probably very low due to its relatively high afnity for haemoglobin and myoglobin, meaning that any available CO is far more likely to bind to these proteins than to COX

86

P.B.L. Pun et al. / Mitochondrion 10 (2010) 8393

(Cooper and Brown, 2008). However, the veracity of this statement remains to be conrmed. Afterall, it appears that a mitochondrial isoform of HO-1 exists (Converso et al., 2006; Slebos et al., 2007). If this was true, the local concentration of CO at the site of COX within the mitochondria could be sufciently high for signicant inhibition of COX by CO. In addition, interactions between CO and NO (as will be discussed later in this review) could modulate the inuence of CO on COX as well. Even if endogenous CO production is indeed too low to produce signicant COX inhibition, the pathological relevance of such inhibition cannot be ignored especially since exogenous CO from conditions such as smoke exposure during res can greatly elevate CO levels within the body (Varon et al., 1999; Alarie, 2002). Therefore, in such situations, the CO concentration in blood and tissues could increase so much as to cause signicant inhibition in vivo. COX inhibition by CO (Fig. 2) causes electrons to accumulate within the ETC, resulting in increased ROS generation by the mitochondria. A study by Zuckerbraun et al. (2007) found that treatment of cells with CO generated higher levels of ROS than similar treatment with antimycin A alone. Application of both CO and antimycin A together led to ROS production that was increased relative to baseline (non-treated cells) but which was comparable to that generated by antimycin A treatment alone and signicantly less than that due to CO by itself. The authors of that study thus concluded that the site of CO-induced ROS generation was complex III. The amount of ROS generated is dependent on the concentration of CO. The exposure of cultured cells to low levels of exogenous CO (<100 ppm) failed to generate any signicant change in ROS production. However, at higher CO concentrations (1000 and 10,000 ppm), ROS production was clearly elevated (Deng et al., 2008). This dependency on CO concentration is not unexpected given how CO inhibition of COX forms the basis for increased ROS production in this context, and how this inhibition is competitive, meaning that the higher the levels of CO, the greater the inhibitory effect. One study has further suggested that the amount of oxygen exposure following CO treatment is another determinant of the levels of ROS production (Piantadosi et al., 1995). In this study, rats were exposed to 10, 000 ppm CO for 30 min, followed by hyperbaric oxygen therapy (HBOT) for 2 h at either 1.5 or 2.5 atmospheres absolute (ATA). It was found that ROS levels were decreased by HBOT at 1.5 ATA, but increased at 2.5 ATA. The authors suggested that these contrasting effects could be due to

B. Modulate COX
expression

CO

Low Oxygen

translation of COX

mitochondrial DNA

CuBI heme a3II O2 CO A. Competitive COX


Inhibition

Complex III eeC. ROS e- e- production + CO2 +

CuAI heme aII

O2

.-

H2O2

.-

Fig. 2. Summary of effects of CO inhibition on COX at low oxygen condition. (A) CO competes with O2 and binds reversibly to COX. (B) CO inhibits COX1 subunit biogenesis by preventing translation despite increasing mtDNA copy number. (C) CO is converted to CO2 with concomitant release of electrons that is fed to complex III to produce ROS.

the effects of H2O2 production by the avoenzyme monoamine oxidase (MAO) in the presence of high oxygen concentrations. It has been reported that aside from normal mitochrondrial processes, MAO-mediated oxidative deamination of biogenic amines provides a substantial source of H2O2 (Di Lisa et al., 2009). It is therefore plausible that at high oxygen tensions, as occurs in HBOT, increased oxygen availability to MAO facilitates ROS production, a prospect that has previously been discussed in a review by Murphy (2009) with respect to mitochondrial electron carriers. In light of this possibility, it is puzzling then that ROS levels should be decreased at 1.5ATA and increased at 2.5ATA. Afterall, under both conditions, oxygen tension still is high relative to normal atmospheric conditions. We suggest that a threshold could possibly exist, below which the elevated oxygen tension promotes antioxidant activities, thus reducing ROS levels, and above which the antioxidant defences are overwhelmed, leading to a rise in ROS levels. We emphasize, however, that this remains to be veried experimentally. Nonetheless, what the above example demonstrates is the inuence of high oxygen exposure in exacerbating the effects of CO. It is also noteworthy that upper limits should be set for the administration of HBOT to address oxidative stress issues and prevent HBOT from becoming a double-edged sword. Intriguingly, there have been reports suggesting that CO actually decreases ROS production by inhibiting NADPH oxidase, reducing NO production by iNOS, and binding of free heme, thereby limiting ROS generation by the Fenton reaction (Srisook et al., 2006). However, the relevance of these events within the mitochondria is unclear as NADPH oxidase, iNOS and free heme are localized largely in the cytosol or extracellularly. Therefore, unless ROS produced by these enzymes diffuse into the mitochondria, the effects of CO on mitochondrial ROS levels can be expected to be mostly modulated by its effects on the ETC. The importance of such compartmentalization has been clearly demonstrated by one group which showed that CO uptake by brain cells inhibits cerebral energy metabolism in rats even after its elimination from the blood (Brown and Piantadosi, 1992). Besides, CO can also displace NO from heme sites, thereby increasing NO levels (Piantadosi, 2002). The interaction of competing pathways can therefore further modulate ROS generation induced by CO. It is noteworthy that not only does CO inhibit COX competitively, but it also decreases protein levels of its subunit, COX1 (Iheagwara et al., 2007) (Fig. 2). This is somewhat surprising given how CO increases mitochondrial DNA (mtDNA) copy number (Lancel et al., 2009). Since COX1 is a mitochondrially encoded gene (Mason and Schatz, 1973), the increase in mtDNA copy number suggests that COX1 expression should also increase. However, COX1 protein levels were decreased, while no effect was observed on its transcript levels (Iheagwara et al., 2007). This suggests that CO affects translation of the COX1 mRNA transcript into the protein. The exact mechanism by which this is achieved is unclear. It is possible that CO interferes with the translation process by interacting with proteins such as Pet309 and Mss51, which are involved in COX1 biosynthesis (Decoster et al., 1990; Manthey and McEwen, 1995). Such interference may possibly be mediated directly by CO or indirectly via CO-induced ROS. However, this hypothesis has yet to be proven experimentally and is purely speculative. Alternatively, as the authors of the study suggested, elevated ROS could cause oxidative damage to the COX1 protein, resulting in its faster degradation (Iheagwara et al., 2007). Besides modulating COX1 expression at the protein level, it appears that CO could also achieve likewise at the transcript level. As mentioned above, there is an increase in the copy number of mtDNA, which encodes COX1. Therefore, assuming a constant rate of transcription, one would expect there to be higher levels of mRNA transcripts. However, while COX1 mRNA levels did not decrease, it did not increase either. The failure to observe increased mRNA lev-

P.B.L. Pun et al. / Mitochondrion 10 (2010) 8393

87

els is consistent with studies from Kristal et al. (1997), and Crawford et al. (1997) investigating ROS-mediated inhibition of mRNA transcription and increased degradation respectively. Although no direct cause-effect relationship has been demonstrated between CO and transcription of COX1, there does appear to be a potential for such a causative relationship to exist. Given that CO inhibits COX of the ETC, it is reasonable to predict that ATP levels should consequently decrease. In addition, CO exposure causes mitochondrial membrane hyperpolarisation. As this can be prevented by oligomycin (an inhibitor of the F0F1-ATPase), it appears that this hyperpolarisation is due to reversal of the F0F1-ATPase (Zuckerbraun et al., 2007). That is, protons are extruded at the expense of ATP hydrolysis. Yet despite this, several studies have found ATP levels to be maintained or even increased by CO treatment (Piantadosi et al., 1988; Brown and Piantadosi, 1992; Matsuoka et al., 1993; Lavitrano et al., 2004; Tsui et al., 2005; Zuckerbraun et al., 2007). Even where a drop in ATP levels was reported, the decrease appeared to be short-term (Sokal et al., 1982). Prolonged CO exposure led instead to an elevation of ATP content of tissues (Sokal et al., 1982). These ndings suggest that CO exposure conditions cells such that compensatory responses are invoked which maintain ATP levels. Similarly, despite CO increasing ROS production and thus causing oxidative damage (Piantadosi et al., 2006; Taskiran et al., 2007; Kim et al., 2008; Scragg et al., 2008; Lancel et al., 2009), cells that had received prior treatment with CO are generally more resistant to subsequent oxidative stress and are less likely to undergo apoptosis as a result (Kim et al., 2006; Li et al., 2006). Besides, while CO induces mPT in the short-term after initial exposure, there is recovery by day 7 even in the continued presence of CO (Piantadosi et al., 2006). Again, these observations support the idea of a CO-inducible pre-conditioning of cells. In both situations, the pre-conditioning appears to be ROS-mediated. The rapid elevation of ROS levels induced by CO leads to the expression of antioxidants such as MnSOD (Frankel et al., 2000; Thom et al., 2000; Liu et al., 2006; Piantadosi et al., 2008). This enzyme catalyses the dismutation of superoxide to H2O2, thereby increasing the mitochondrial H2O2 leak .Among others, mitochondrial H2O2 can then activate PKB. PKB in turn deactivates GSK3b. This process releases Nrf2 which binds the ARE within the nucleus and induces Nrf1 expression (Piantadosi et al., 2008). Both Nrf1 and Nrf2 subsequently trigger mitochondrial biogenesis in a PGC1a-Tfam-dependent pathway. In fact, as low as picomolar levels of CO can induce mitochondrial biogenesis. Although CO could also activate PGC1a via guanylate cyclase, this is unlikely to be an important means in cells as CO has far lower afnity for guanylate cyclase than NO (Suliman et al., 2007). CO can further exert antiinammatory and anti-apoptotic effects via molecules such as PPARc (Bilban et al., 2006), NFjB (Brouard et al., 2002; Kim et al., 2006), STAT (Zhang et al., 2005) and p38MAPK (Silva et al., 2006; Kohmoto et al., 2007). The precise mechanism by which CO regulates these molecules has yet to be dened. It has, however, been suggested that there exist transcription factors that are regulated by gases such as CO although it remained to be seen which signalling pathways are activated downstream of these genes (Bilban et al., 2008). CO could further protect against apoptosis by binding to cytochrome c of the cytochrome ccardiolipin complex, thus inhibiting its peroxidise activity and preventing cell death (Kapetanaki et al., 2009). The activation of these manifold pathways collectively have the following effects: (i) The induction of antioxidants in response to the initial rise in ROS production and ARE binding by Nrf2 conditions cells to be more resistant to oxidative stress; (ii) Mitochondrial biogenesis can be expected to help compensate for any decrease in ATP production by each mitochondrion. (iii) The expression of genes like TGFb downstream of the major transcription factors e.g. NFjB, together with the mainte-

nance of cytochrome ccardiolipin integrity, inhibits apoptosis and inammation. One should note, however, that at very high levels of CO, excessive ROS production may overwhelm even these compensatory mechanisms and cause severe cell damage, leading to cell death either by apoptosis (Piantadosi et al., 1997; Uemura et al., 2001; Toghi et al., 2006) or necrosis (Wang et al., 1990; Piantadosi et al., 1997; Uemura et al., 2001). 4. Hydrogen sulphide H2S is produced from L-cysteine in a process catalysed by the enzymes cystathionine-c-lyase (CSE) and cystathionine-b-synthase (CBS) (Stipanuk and Beck, 1982). Although the mitochondria are a rich source of acid-labile sulphur, it is unlikely to be a major source of sulphide in cells as the mitochondrial pH is not usually acidic. Rather it appears that under physiological conditions, intracellular H2S largely originates from bound sulphur released upon alkalinisation of the cytoplasm e.g. during neuronal excitation (Ishigami et al., 2009). While this contradicts earlier ndings that as much as a quarter of endogenous sulphide in rat brains is found in the mitochondria (Warenycia et al., 1989), it should be noted that experimental conditions differed between the two studies, with the earlier one having been conducted at acidic pH and the more recent study at alkaline pH. Besides, even assuming the mitochondria to contain most of a cells sulphur content, it does not necessarily mean that the mitochondria is the main source of endogenous and active sulphide because that would require the sulphide to be released from its stores and near-neutral pH conditions within the mitochondria makes this unlikely. Like NO and CO, H2S is known to affect the activity of the respiratory chain. To be more specic, H2S is both a substrate and inhibitor of the ETC (Nicholls and Kim, 1982). At low concentrations (<20 lM), H2S is primarily a substrate, donating electrons to the ETC at the level of ubiquinone (Volkel and Grieshaber, 1996; Goubern et al., 2007). The electron is then passed onto complex III and COX, and nally to oxygen (Volkel and Grieshaber, 1996; Goubern et al., 2007). This electron for oxidative phosphorylation thus originates from sulphide oxidation in a process catalysed by sulphide:quino-oxidoreductase (SQR), an enzyme found on the inner mitochondrial membrane (Theissen and Martin, 2008). Therefore, at low concentrations, H2S actually stimulates oxidative phosphorylation and increases ATP production. At higher concentrations, however, the inhibitory effect of H2S on COX becomes dominant, reaching complete inhibition at approximately 50 lM (Volkel and Grieshaber, 1996). Three molecules of sulde are required for the inhibition of each COX molecule (Nicholls and Kim, 1982). Unlike CO and NO, H2S inhibition of COX is non-competitive with respect to oxygen (Petersen, 1977), with H2S binding to the oxidized form of the enzyme (Wever et al., 1975). Under conditions of complete COX inhibition by H2S, electrons from sulphide oxidation can no longer be passed onto complex III and COX. Rather, in the lugworm, Arenicola marina, electrons from sulphide oxidation are transferred to an alternative oxidase, which in turn, passes on the electron directly to oxygen, producing water without concomitant ATP generation (as would have occurred if the electron was used in oxidative phosphorylation instead) (Volkel and Grieshaber, 1996). There is no evidence as yet for the presence of an alternative oxidase in human cells. However, studies in human colonic adenocarcinoma cells have demonstrated that under the same conditions (i.e. complete inhibition of COX by H2S), electrons are channelled through complex II instead, reversing the direction of the reaction it catalyses, resulting in the reduction of fumarate to form succinate (Goubern et al., 2007). The physiological signicance of such alternative pathways is unclear although conceivably, the channelling of electrons from sulphide oxidation elsewhere (either to complex II or to an alternative oxi-

88

P.B.L. Pun et al. / Mitochondrion 10 (2010) 8393

dase) prevents their accumulation in the ETC and, hence, reduces ROS generation. In addition, it is possible that the reversal of reactions at complex II prevents the build-up of substrates intended for oxidative phosphorylation, thereby allowing the substrates to be similarly channelled elsewhere e.g. to glycolysis. It has been suggested, however, that since ATP generation by glycolysis is far less productive than that by oxidative phosphorylation, that there is still likely to be a shortfall in ATP production (Leschelle et al., 2005). Indeed, ATP levels have been found to decrease at high H2S concentrations (Nicholson et al., 1998; Eghbal et al., 2004; Hildebrandt and Grieshaber, 2008), indicating that the demand for ATP outstrips supply. Besides simple inhibition of COX enzyme activity, H2S can further decrease ATP production by up-regulating uncoupling protein (UCP)-2 and down-regulating the protein expressions of subunits I and II of COX (Leschelle et al., 2005). The authors suggested these events to be part of an adaptation process to H2S. For example, increasing UCP-2 expression, though decreasing ATP production, also reduces ROS generation by the ETC, thus protecting the cell from oxidative stress. The reduction in expression of the COX subunits, on the other hand, may be a means to conserve metabolic expenses. This argument was based on the belief that at high H2S concentrations, COX would be inhibited anyhow. Thus it would be of little use to synthesize more COX. Down-regulating COX expression would therefore minimize wastage of and allow resources to be put to better use. Fig. 3 summarizes the inhibition of COX by H2S under high oxygen conditions. Other evidence that cells appear to be able to adapt to H2S and/ or undergo pre-conditioning in its presence is also available. For instance, in lung mitochondria from H2S-treated rats, COX activity decreased initially, then recovered to baseline by 24 h post-exposure (Khan et al., 1990). Similarly, while single acute exposure to H2S signicantly inhibits COX activity in rat nasal respiratory epithelium, prolonged exposure produced no apparent change. This lack of effect was concomitant to a regeneration of the nasal respiratory mucosa, suggesting that the regenerated mucosa is conditioned to withstand the effects of H2S (Dorman et al., 2002). Furthermore, H2S increases lactate release without any effect on its oxidation, pointing towards an up-regulation of glycolysis which may compensate for any declines in ATP production due to COX inhibition (Leschelle et al., 2005). Such compensation, unfortunately, is unlikely to be sufcient as has been mentioned above. In addition, cells pre-treated with H2S are more resistant

Complex II
Succinate

H+ eee- e-

O2

High Oxygen

Electrons from

sulphide oxidation
Fumarate

CuBII heme a3III

H2S C. Modulate COX

B. Reversal of catalytic
reactions at Complex II

H2S A. Non-Competitive
Inhibition of COX inhibits ETC at complex IV

heme

CuAII

expression

aIII

Decreased expression of COX subunits

COX

Fig. 3. Summary of effects of H2S inhibition on COX at high oxygen condition. (A) H2S binds non-competitively to COX. (B) Electrons from sulphide oxidation are channelled through complex II, causing a reversal of its catalytic reaction (i.e. producing succinate from fumarate) when H2S concentrations are high. (C) H2S down-regulates expression of COX subunits.

to oxidative stress, likely because of an increase in GSH synthesis triggered by elevated ROS levels (Kimura and Kimura, 2004; Kimura et al., 2006). These observations collectively indicate a compensatory response to H2S, which would suggest that cells should have greater resilience to subsequent H2S treatment. However, this may not always hold true as one study has reported that pre-treated cells are actually more vulnerable than cells without prior H2S exposure to COX inhibition by a second low dose of H2S (Leschelle et al., 2005). The authors attributed their observations to the presence of an excess capacity for oxidative phosphorylation in non-pre-treated cells which was lost during the rst exposure period in pre-treated cells in which COX expression was found to be down-regulated. Alternatively, it is also possible that vulnerability to H2S may be cell-specic. Afterall, while lung cells appear to be susceptible to H2S-mediated COX inhibition, liver cells seem more resistant, with no COX inhibition recorded even at levels as high as 400 ppm H2S (Dorman et al., 2002). The reason for such disparities is unclear, although it may likewise be due to differences in buffering capacity against COX inhibition. A third possibility is that the model system in which the effects of H2S are investigated could itself inuence outcome. For example, while the above study, which was conducted in rats, found the liver to be resistant to H2S-induced COX inhibition (Dorman et al., 2002), a separate study using isolated rat hepatocytes in culture reported a signicant decrease in COX activity following H2S exposure (Eghbal et al., 2004). Again, there is no proven explanation as yet for such discordance. We speculate that the compensatory responses invoked in whole animals may involve multiple organ systems and, therefore, differ from that seen in isolated systems such as cell cultures, resulting in varying extents of protection against the negative effects of H2S. Not surprisingly then, depending on the study model and the concentration of H2S used, ROS levels may or may not be signicantly altered by H2S treatment. Increased ROS production can lead to apoptosis by inducing mPT (Eghbal et al., 2004). H2S exposure further promotes apoptosis by inducing cytochrome c release from the mitochondria, triggering Bax translocation to the mitochondria, stabilizing pro-apoptotic molecules like p21 and down-regulating anti-apoptotic proteins like Bcl-2 (Baskar et al., 2007; Adhikari and Bhatia, 2008). However, despite appearing to be pro-apoptotic, a recent study reported the contrary with H2S mitigating rotenone-induced apoptosis in neuroblastoma cells (Hu et al., 2009). In that study, H2S was found to, among other things, prevent rotenone-induced mitochondrial membrane depolarization, cytochrome c release and decrease in Bcl-2:Bax ratio. Intriguingly, H2S itself is known to cause loss of mitochondrial membrane potential, cytochrome c release and dampening of the Bcl-2:Bax ratio (Eghbal et al., 2004; Baskar et al., 2007; Adhikari and Bhatia, 2008). The authors suggested that H2S attenuated rotenone-induced apoptosis by inhibiting the activation of the pro-apoptotic MAPK pathway in a process mediated by mitochondrial ATP-sensitive potassium (mitoKATP) channels (Hu et al., 2009). This stemmed from observations that blockage of these channels prevented H2S-mediated protection. That H2S activation of the mitoKATP channels is cytoprotective has also been suggested in a separate study on cortical neuronal cell cultures (Kimura et al., 2006). We suggest that H2S further affords protection against rotenone by increasing glutathione biosynthesis, thus improving the antioxidant to oxidant status of cells. However, it should be noted that no measures of glutathione was done in the study involving rotenone and our suggestion remains purely speculative. Even assuming these explanations to be wholly accurate, it remains difcult to reconcile the apparently dual and opposite effects of H2S on apoptosis for the following reasons: for ROS to induce apoptosis, ROS levels must be so high as to cause signicant

P.B.L. Pun et al. / Mitochondrion 10 (2010) 8393

89

cell damage. The effects of H2S on ROS production are dose-dependent (Eghbal et al., 2004). Therefore, H2S levels must be sufciently high to be pro-apoptotic. However, a similar argument is valid when considering the anti-apoptotic effects of H2S. It appears that such cytoprotection is dependent on the opening of the mitoKATP channel. For these channels to open, ATP levels must drop signicantly. This too only occurs at high H2S concentration. Based on these arguments, both detrimental and protective effects would take place in situations where H2S levels are high. However, either one or the other has to dominate in the outcome, and what inuences the balance between the two processes has yet to be dened. It is possible that there is a threshold level of H2S below which the anti-apoptotic events dominate, and above which cell death occurs. However, no study is known to us that has clearly demonstrated and dened the existence of such a threshold and further investigation is required to determine the veracity of our hypothesis. 5. Interactions between the gasomodulators It is apparent from available literature that NO, CO and H2S share several similarities (Fig. 4). To put it very generally, all three: 1. are gasomodulators, 2. can inhibit COX and, in the broader perspective, reduce oxidative phosphorylation, 3. have the potential to increase ROS production, induce apoptosis and cause mitochondrial dysfunction, 4. yet, at the same time, also induce compensatory responses such as increased mitochondrial biogenesis, possibly via signalling pathways involving, for example, Nrf2 and various kinases, 5. in fact, at low gas concentrations, these events may even be cytoprotective. Given such similarities, NO, CO and H2S are naturally expected to interact with and modulate each other. There are multiple pieces of evidence supporting this idea, especially pertaining to NO and CO. For instance, both NO and CO can bind guanylate cyclase and thereby increase cGMP levels, triggering downstream events like mitochondrial biogenesis (Nisoli et al., 2003; Clementi and Nisoli, 2005; Suliman et al., 2007; Vieira et al., 2008). As NO has higher afnity for guanylate cyclase than CO, it is likely that its binding to guanylate cyclase pre-dominates. However, in conditions when NO production is low, it is also possible that CO becomes the principle binding ligand for guanylate cyclase

(Piantadosi et al., 2008). Recent evidence suggests that H2S could modulate cGMP signalling by NO, providing another point of interaction between the gases (Pong and Eldred, 2009). In another example, both CO and NO prevent HIF1a stabilization (Huang et al., 1999), plausibly by creating metabolic hypoxia (Xu et al., 2005; Zuckerbraun et al., 2007). This is a situation in which oxygen supply to the cells is low, but cells fail to perceive this drop in oxygen levels because CO and NO inhibit oxidative phosphorylation, thereby decreasing oxygen consumption and increasing the availability of oxygen to other enzymes that regulate HIF1a stability (Xu et al., 2005). A third example of a common outcome between CO and NO is the induction of mitochondrial membrane hyperpolarisation, conceivably involving the reversal of the F0F1-ATPase (Almeida et al., 2001; Zuckerbraun et al., 2007). Besides additive effects, the interactions could also be independent of each other or be antagonistic in nature. For instance, CO induces mitochondrial biogenesis even in eNOS or iNOS knockout mice, suggesting that its effects are independent of NO (Suliman et al., 2007). In terms of antagonism, studies have demonstrated that the combination of CO and NO produces less inhibition of COX activity than CO alone (Pearce et al., 2008), and that CO prevents apoptosis induced by the NO derivative, ONOO (Li et al., 2006). While most of the above quoted studies focused on CO and NO, there is also evidence, albeit less plentiful, of an interaction of these gases with H2S. Unfortunately though, very few studies have focused specically on their interaction within the mitochondria. For illustration purposes, we briey discuss potential interactions between the three gases with respect to one mitochondria-related parameter and one general (non-mitochondria-specic) aspect, namely COX inhibition and gas biosynthesis. With respect to COX inhibition, there appears to be a potential three-way interaction between NO, CO and H2S. CO, for instance, could, in principle, displace NO from COX, thereby increasing the levels of free NO, which would in turn lead to a rise in ONOO formation and elevation of mitochondrial oxidative/nitrosative damage (Piantadosi, 2008). Supercially, this appears to contradict observations that COX inhibition is greater when only CO is present, as opposed to when both CO and NO are available (Pearce et al., 2008). Afterall, if CO indeed displaces NO from COX, then inhibition of COX by CO should not be affected by the presence of NO. However, assuming the hypothesis that NO is not a typical reversibly bound competitive inhibitor holds true, such observations may not be surprising. According to the hypothesis by Pearce et al. (2008), COX is converted to the oxidized state after NO inhibition. While CO dissociates more slowly from COX than does NO,

Fig. 4. Summary of effects of the three gasomodulators NO, CO and H2S on the mitochondria. All three gases modulate a ne balance between cytotoxic and cytoprotective events by mediating processes such as ATP production, apoptosis and oxidative stress.

90

P.B.L. Pun et al. / Mitochondrion 10 (2010) 8393

CO also has a far lower association constant for COX. Thus, when both CO and NO are present, NO is expected to be the dominant binding partner of COX. Although CO is, in theory, capable of displacing NO from COX, it should not be able to bind COX, which is in the oxidized state after NO binding. As a result, COX inhibition occurs to a lesser extent when both gases are present than when only CO is available. Staying on the issue of COX inhibition, Cooper and Brown have further suggested that since H2S acts as a substrate of the ETC, the inhibition of COX by either NO or CO could prevent H2S oxidation (Cooper and Brown, 2008). If alternative pathways to which electrons from H2S oxidation can be channelled are not available, this would result in an increase in free H2S which would further inhibit COX. Therefore, there could potentially be an additive effect of the three gasomodulators on COX inhibition. However, as we have discussed above, electrons may be passed onto either an alternative oxidase (Volkel and Grieshaber, 1996) or to complex II of the ETC (Goubern et al., 2007). It is uncertain then how relevant such an additive effect is both in vitro and in vivo. Nonetheless, it is also plausible that electron acceptance by these alternative pathways can become saturated, resulting in cumulative inhibition of COX as proposed. Pertaining to the issue of biosynthesis, one study conducted in macrophage cell cultures found that H2S reduces iNOS expression, and thus decrease NO generation, via the HO-1/CO system (Oh et al., 2006). In the absence of CO, such inhibition cannot occur. CO alone, however, is sufcient to reduce NO production in this system. A separate study in rats generated similar results, with higher H2S levels being associated with decreased NO and elevated CO levels (Li et al., 2007). Other studies have conversely found NO to modulate CO and H2S production. For instance, NO is known to increase CO generation by inducing HO-1 (Durante et al., 1997; Datta and Lianos, 1999; Bouton and Demple, 2000). As for H2S, studies in isolated CBS enzyme systems have found NO to inhibit this enzyme (Taoka and Banerjee, 2001), which presumably has the effect of decreasing H2S production. As CO also binds CBS, in fact doing so with higher afnity than does NO (Taoka and Banerjee, 2001), it is plausible that CO has a similar inuence over H2S biosynthesis although this remains to be conrmed experimentally. However, these effects again appear to be specic to the model system under study as a separate investigation in rat smooth muscle cells found the opposite effect, with NO donors increasing H2S production, plausibly, by inducing CSE expression (Zhao et al., 2001). Alternatively, the eventual outcome of NO on H2S generation could perhaps vary with time. Afterall, both CBS and CSE are involved in H2S biosynthesis, with CBS catalysing the formation of cystathionine from homocysteine and serine, and CSE catalysing the next step which generates L-cysteine and a-ketobutyrate from cystathionine. Both enzymes then participate in the conversion of a-ketobutyrate to pyruvate and eventually H2S (Li and Moore, 2007). Based on the results of the above studies, NO inhibits CBS, but induces CSE. In the short-term, up-regulation of CSE would increase H2S production. However, as the effects of upstream inhibition of CBS lter downstream overtime, levels of the CSE substrate, cystathionine, will drop. Without adequate substrate, H2S synthesis can be expected to decrease in the long run. In short, we postulate that NO may increase H2S production, then decrease it subsequently. Note, however, that our hypothesis remains to be experimentally veried. Besides the examples quoted above, there are many other plausible interactions between NO, CO and H2S. For instance, their manifold effects on apoptosis and signalling pathways such as those involving Nrf2 and MAPK suggest potential points of intersection. Unfortunately, studies specically designed to test these interactions are lacking, and such inter-relationships remain purely hypothetical.

6. Conclusion This review has highlighted the inuence of the three gasomodulators NO, CO and H2S on the mitochondria and its related functions such as apoptosis. Unfortunately, very few studies have investigated the interaction between these three gases within the mitochondria. As such, further work is needed to characterize cross-talk between the gasomodulators and dene their in vivo relevance. This would, among other things, require investigations into their relative in vivo concentration, especially within the mitochondria, and more in-depth functional studies. Since the mitochondria are implicated in numerous diseases, the regulatory functions of these gases in the mitochondria likely play a role in inuencing disease outcome. It is hopeful that with further studies, more light will be shed on the involvement of gasomodulators in mitochondrial function and diseases, which should map out novel directions for pharmacological therapies. References
Adhikari, S., Bhatia, M., 2008. H2S-induced pancreatic acinar cell apoptosis is mediated via JNK and p38 MAP kinase. J. Cell Mol. Med. 12, 13741383. Alarie, Y., 2002. Toxicity of re smoke. Crit. Rev. Toxicol. 32, 259289. Almeida, A., Almeida, J., Bolanos, J.P., Moncada, S., 2001. Different responses of astrocytes and neurons to nitric oxide: the role of glycolytically generated ATP in astrocyte protection. Proc. Natl. Acad. Sci. USA 98, 1529415299. Almeida, A., Cidad, P., Delgado-Esteban, M., Fernandez, E., Garcia-Nogales, P., Bolanos, J.P., 2005. Inhibition of mitochondrial respiration by nitric oxide: its role in glucose metabolism and neuroprotection. J. Neurosci. Res. 79, 166 171. Alonso, J.R., Cardellach, F., Lopez, S., Casademont, J., Miro, O., 2003. Carbon monoxide specically inhibits cytochrome c oxidase of human mitochondrial respiratory chain. Pharmacol. Toxicol. 93, 142146. Bal-Price, A., Brown, G.C., 2000. Nitric oxide-induced necrosis and apoptosis in PC12 cells mediated by mitochondria. J. Neurochem. 75, 14551464. Baskar, R., Li, L., Moore, P.K., 2007. Hydrogen sulphide-induces DNA damage and changes in apoptotic gene expression in human lung broblast cells. FASEB J. 21, 247255. Baumgart, K., Radermacher, P., Wagner, F., 2009. Applying gases for microcirculatory and cellular oxygenation in sepsis: effects of nitric oxide, carbon monoxide and hydrogen sulphide. Curr. Opin. Anaesthesiol. 22, 168 176. Beal, M.F., 2007. Mitochondria and neurodegeneration. Novartis Found. Symp. 287, 183192. Bilban, M., Bach, F.H., Otterbein, S.L., Ifedigbo, E., dAvila, J.C., Esterbauer, H., Chin, B.Y., Usheva, A., Robson, S.C., Wagner, O., Otterbein, L.E., 2006. Carbon monoxide orchestrates a protective response through PPARgamma. Immunity 24, 601 610. Bilban, M., Haschemi, A., Wegiel, B., Chin, B.Y., Wagner, O., Otterbein, L.E., 2008. Heme oxygenase and carbon monoxide initiate homeostatic signalling. J. Mol. Med. 86, 267279. Bolanos, J.P., Almeida, A., 2006. Modulation of astroglial energy metabolism by nitric oxide. Antioxid. Redox Signal. 8, 955965. Bolanos, J.P., Heales, S.J., Land, J.M., Clark, J.B., 1995. Effect of peroxynitrite on the mitochondrial respiratory chain: differential susceptibility of neurones and astrocytes in primary culture. J. Neurochem. 64, 19651972. Bolanos, J.P., Heales, S.J., Peuchen, S., Barker, J.E., Land, J.M., Clark, J.B., 1996. Nitric oxide-mediated mitochondrial damage: a potential neuroprotective role for glutathione. Free Radical Biol. Med. 21, 9951001. Borutaite, V., Brown, G.C., 2003. Nitric oxide induces apoptosis via hydrogen peroxide, but necrosis via energy and thiol depletion. Free Radical Biol. Med. 35, 14571468. Bouton, C., Demple, B., 2000. Nitric oxide-inducible expression of heme oxygenase-1 in human cells. Translation independent stabilization of the mRNA and evidence for direct action of nitric oxide. J. Biol. Chem. 275, 32688 32693. Brouard, S., Berberat, P.O., Tobiash, E., Seldon, M.P., Bach, F.H., Soares, M.P., 2002. Heme oxygenase-1-derived carbon monoxide requires the activation of transcription factor NF-kappa B to protect endothelial cells from tumour necrosis factor-alpha-mediated apoptosis. J. Biol. Chem. 277, 1795017961. Brown, G.C., 1999. Nitric oxide and mitochondrial respiration. Biochim. Biophys. Acta 1411, 351369. Brown, G.C., Borutaite, V., 2004. Inhibition of mitochondrial respiratory complex I by nitric oxide, peroxynitrite and S-nitrosothiols. Biochim. Biophys. Acta 1658, 4449. Brown, G.C., Cooper, C.E., 1994. Nanomolar concentrations of nitric oxide reversibly inhibit synaptosomal respiration by competing with oxygen at cytochrome oxidase. FEBS Lett. 356, 295298. Brown, S.D., Piantadosi, C.A., 1992. Recovery of energy metabolism in rat brain after carbon monoxide hypoxia. J. Clin. Invest. 89, 666672.

P.B.L. Pun et al. / Mitochondrion 10 (2010) 8393 Calabrese, V., Mancuso, C., Calvani, M., Rizzarelli, E., Buttereld, D.A., Stella, A.M., 2007. Nitric oxide in the central nervous system: neuroprotection versus neurotoxicity. Nat. Rev. Neurosci. 8, 766775. Carreras, M.C., Peralta, J.G., Converso, D.P., Finocchietto, P.V., Rebagliati, I., Zaninovich, A.A., Poderoso, J.J., 2001. Modulation of liver mitochondrial NOS is implicated in thyroid-dependent regulation of O(2) uptake. Am. J. Physiol. Heart Circ. Physiol. 281, H2282H2288. Carreras, M.C., Franco, M.C., Molinas, M.F., Gonzalez, A.S., Serra, M.P., Finocchietto, P.V., Converso, D.P., Poderoso, J.J., 2008. O18 Neuronal nitric oxide synthase trafcking into mitochondria involves the processing of the PDZ domain. Nitric Oxide 19, 24. Cassina, A., Radi, R., 1996. Differential inhibitory action of nitric oxide and peroxynitrite on mitochondrial electron transport. Arch. Biochem. Biophys. 328, 309316. Castro, L., Rodriguez, M., Radi, R., 1994. Aconitase is readily inactivated by peroxynitrite, but not by its precursor, nitric oxide. J. Biol. Chem. 269, 29409 29415. Cheng, A., Chan, S.L., Milhavet, O., Wang, S., Mattson, M.P., 2001. P38 MAP kinase mediates nitric oxide-induced apoptosis of neural progenitor cells. J. Biol. Chem. 276, 4332043327. Choi, B.M., Pae, H.O., Jang, S.I., Kim, Y.M., Chung, H.T., 2002. Nitric oxide as a proapoptotic as well as anti-apoptotic modulator. J. Biochem. Mol. Biol. 35, 116 126. Cidad, P., Almeida, A., Bolanos, J.P., 2004. Inhibition of mitochondrial respiration by nitric oxide rapidly stimulates cytoprotective GLUT3-mediated glucose uptake through 50 -AMP-activated protein kinase. Biochem. J. 384, 629636. Cleeter, M.W., Cooper, J.M., Darley-Usmar, V.M., Moncada, S., Schapira, A.H., 1994. Reversible inhibition of cytochrome c oxidase, the terminal enzyme of the mitochondrial respiratory chain, by nitric oxide. Implications for neurodegenerative diseases. FEBS Lett. 345, 5054. Clementi, E., Nisoli, E., 2005. Nitric oxide and mitochondrial biogenesis: a key to long-term regulation of cellular metabolism. Comp. Biochem. Physiol. 142, 102 110. Converso, D.P., Taille, C., Carreras, M.C., Jaitovich, A., Ponderoso, J.J., Boczkowski, J., 2006. HO-1 is located in liver mitochondria and modulates mitochondrial heme content and metabolism. FASEB J. 20, 12361238. Cooper, C.E., Brown, G.C., 2008. The inhibition of mitochondrial cytochrome oxidase by the gases carbon monoxide, nitric oxide, hydrogen cyanide and hydrogen sulphide: chemical mechanism and physiological signicance. J. Bioenerg. Biomembr. 40, 533539. Cooper, C.E., Giulivi, C., 2007. Nitric oxide regulation of mitochondrial oxygen consumption II: molecular mechanism and tissue physiology. Am. J. Physiol. Cell Physiol. 292, C1993C2003. Cooper, C.E., Torres, J., Sharpe, M.A., Wilson, M.T., 1997. Nitric oxide ejects electrons from the binuclear centre of cytochrome c oxidase by reacting with oxidized copper: a general mechanism for the interaction of copper proteins with nitric oxide? FEBS Lett. 414, 281284. Crawford, D.R., Wang, Y., Schools, G.P., Kochheiser, J., Davies, K.J.A., 1997. Downregulation of mammalian mitochondrial RNAs during oxidative stress. Free Radical Biol. Med. 22, 551559. DAmico, G., Lam, F., Hagan, T., Moncada, S., 2006. Inhibition of cellular respiration by endogenously produced carbon monoxide. J. Cell Sci. 119, 22912298. Datta, P.K., Lianos, E.A., 1999. Nitric oxide induces heme oxygenase-1 gene expression in mesangial cells. Kidney Int. 55, 17341739. Decoster, E., Simon, M., Hatat, D., Faye, G., 1990. The MSS51 gene product is required for the translation of the COX1 mRNA in yeast mitochondria. Mol. Gen. Genet. 224, 111118. Deng, M., Zhao, J.Y., Fan, D.S., 2008. Effects of exogenous carbon monoxide on neurocytes: experiment in vitro. Zhonghua Yi Xue Za Zhi 88, 30853089. Dhakshinamoorthy, S., Porter, A.G., 2004. Nitric oxide-induced transcriptional upregulation of protective genes by Nrf2 via the antioxidant response element counteracts apoptosis of neuroblastoma cells. J. Biol. Chem. 279, 2009620107. Di Lisa, F., Kaludercic, N., Carpi, A., Menabo, R., Giorgio, M., 2009. Mitochondrial pathways for ROS formation, myocardial injury: the relevance of p66(Shc), monoamine oxidase. Basic Res. Cardiol. 104, 131139. Dorman, D.C., Moulin, F.J.M., McManus, B.E., Mahle, K.C., James, R.A., Struve, M.F., 2002. Cytochrome oxidase inhibition induced by acute hydrogen sulphide inhalation: correlation with tissue sulphide concentrations in the rat brain, liver, lung and nasal epithelium. Toxicol. Sci. 65, 1825. Durante, W., Kroll, M.H., Christodoulides, N., Peyton, K.J., Scahfer, A.I., 1997. Nitric oxide induces heme oxygenase-1 gene expression and carbon monoxide production in vascular smooth muscle cells. Circ. Res. 80, 557564. Eghbal, M.A., Pennefather, P.S., OBrien, P.J., 2004. H2S cytotoxicity mechanism involves reactive oxygen species formation and mitochondrial depolarisation. Toxicology 203, 6976. Elbirt, K.K., Bonkovsky, H.L., 1999. Heme oxygenase: recent advances in understanding its regulation and role. Proc. Assoc. Am. Physicians 111, 438 447. Elfering, S.L., Sarkela, T.M., Giulivi, C., 2002. Biochemistry of mitochondrial nitricoxide synthase. J. Biol. Chem. 277, 3807938086. Exline, M.C., Crouser, E.D., 2008. Mitochondrial mechanisms of sepsis-induced organ failure. Front Biosci. 13, 50305041. Finocchietto, P.V., Franco, M.C., Holod, S., Gonzalez, A.S., Converso, D.P., Arciuch, V.G.A., Serra, M.P., Ponderoso, J.J., Carreras, M.C., 2009. Mitochodnrial nitric oxide synthase: a masterpiece of metabolic adaptation, cell growth, transformation, and death. Exp. Biol. Med. 234, 10201028.

91

Foresti, R., Green, C.J., Motterlini, R., 2004. Generation of bile pigments by haem oxygenase: a rened cellular strategy in response to stressful insults. Biochem. Soc. Symp. 71, 177192. Frankel, D., Mehindate, K., Schipper, H.M., 2000. Role of heme oxygenase-1 in the regulation of manganese superoxide dismutase gene expression in oxidativelychallenged astroglia. J. Cell Physiol. 185, 8086. Fukuto, J.M., Collins, M.D., 2007. Interactive endogenous small molecule (gaseous) signalling: implications for teratogenesis. Curr. Pharm. Des. 13, 29522978. Garcia-Nogales, P., Almeida, A., Bolanos, J.P., 2003. Peroxynitrite protects neurons against nitric oxide-mediated apoptosis. A key role for glucose-6-phosphate dehydrogenase activity in neuroprotection. J. Biol. Chem. 278, 864874. Ghafourifar, P., Richter, C., 1997. Nitric oxide synthase activity in mitochondria. FEBS Lett. 418, 291296. Ghafourifar, P., Richter, C., 1999. Mitochondrial nitric oxide synthase regulates mitochondrial matrix pH. Biol. Chem. 380, 10251028. Ghatan, S., Larner, S., Kinoshita, Y., Hetman, M., Patel, L., Xia, Z., Youle, R.J., Morrison, R.S., 2000. P38 MAP kinase mediates bax translocation in nitric oxide-induced apoptosis in neurons. J. Cell Biol. 150, 335347. Giulivi, C., Poderoso, J.J., Boveris, A., 1998. Production of nitric oxide by mitochondria. J. Biol. Chem. 273, 1103811043. Goubern, M., Andriamihaja, M., Nubel, T., Blachier, F., Bouillaud, F., 2007. Sulde, the rst inorganic substrate for human cells. FASEB J. 21, 16991706. Guo, F.Q., Crawford, N.M., 2005. Arabidopsis nitric oxide synthase1 is targeted to mitochondria and protects against oxidative damage and dark-induced senescence. Plant Cell. 17, 34363450. Guo, F.Q., Okamoto, M., Crawford, N.M., 2003. Identication of a plant nitric oxide synthase gene involved in hormonal signaling. Science 302, 100103. Hildebrandt, T.M., Grieshaber, M.K., 2008. Redox regulation of mitochondrial sulphide oxidation in the lugworm, Arenicola marina. J. Exp. Biol. 211, 2617 2623. Hortelano, S., Dallaporta, B., Zamzami, N., Hirsch, T., Susin, S.A., Marzo, I., Bosca, L., Kroemer, G., 1997. Nitric oxide induces apoptosis via triggering mitochondrial permeability transition. FEBS Lett. 410, 373377. Hu, L.F., Lu, M., Wu, Z.Y., Wong, P.T., Bian, J.S., 2009. Hydrogen sulphide inhibits rotenone-induced apoptosis via preservation of mitochondrial function. Mol. Pharmacol. 75, 2734. Huang, L.E., Willmore, W.G., Gu, J., Goldberg, M.A., Bunn, H.F., 1999. Inhibition of hypoxia-inducible factor 1 activation by carbon monoxide and nitric oxide: implications for oxygen sensing and signalling. J. Biol. Chem. 274, 90389044. Iheagwara, K.N., Thom, S.R., Deutschman, C.S., Levy, R.J., 2007. Myocardial cytochrome oxidase activity is decreased following carbon monoxide exposure. Biochim. Biophys. Acta 1772, 11121116. Ishigami, M., Hiraki, K., Umemura, K., Ogasawara, Y., Ishii, K., Kimura, H., 2009. A source of hydrogen sulphide and a mechanism of its release in the brain. Antioxid. Redox Signal. 11, 205214. Kanai, A.J., Pearce, L.L., Clemens, P.R., Birder, L.A., VanBibber, M.M., Choi, S.Y., de Groat, W.C., Peterson, J., 2001. Identication of a neuronal nitric oxide synthase in isolated cardiac mitochondria using electrochemical detection. Proc. Natl. Acad. Sci. USA 98, 1412614131. Kapetanaki, S.M., Silkstone, G., Husu, I., Liebl, U., Wilson, M.T., Vos, M.H., 2009. Interaction of carbon monoxide with the apoptosis-inducing cytochrome c cardiolipin complex. Biochemistry 48, 16131619. Kasparek, M.S., Linden, D.R., Kreis, M.E., Sarr, M.G., 2007. Gasotransmitters in the gastrointestinal tract. Surgery 143, 455459. Khan, A.A., Schuler, M.M., Prior, M.G., Yong, S., Coppock, R.W., Florence, L.Z., Lillie, L.E., 1990. Effects of hydrogen sulphide exposure on lung mitochondrial respiratory chain enzymes in rats. Toxicol. Appl. Pharmacol. 103, 482490. Kim, H.S., Loughran, P.A., Kim, P.K., Billiar, T.R., Zuckerbraun, B.S., 2006. Carbon monoxide protects hepatocytes from TNF-alpha/actinomycin D by inhibition of the caspase-8-mediated apoptotic pathway. Biochem. Biophys. Res. Commun. 344, 11721178. Kim, H.S., Loughran, P.A., Rao, J., Billiar, T.R., Zuckerbraun, B.S., 2008. Carbon monoxide activates NF-kappa B via ROS generation and Akt pathways to protect against cell death of hepatocytes. Am. J. Physiol. Gastrointest. Liver Physiol. 295, G146G152. Kimura, Y., Kimura, H., 2004. Hydrogen sulphide protects neurons from oxidative stress. FASEB J. 18, 11651167. Kimura, Y., Dargusch, R., Schubert, D., Kimura, H., 2006. Hydrogen sulphide protects HT22 neuronal cells from oxidative stress. Antioxid. Redox Signal. 8, 661670. Kobzik, L., Stringer, B., Balligand, J.L., Reid, M.B., Stamler, J.S., 1995. Endothelial type nitric oxide synthase in skeletal muscle bers: mitochondrial relationships. Biochem. Biophys. Res. Commun. 211, 375381. Kohmoto, J., Nakao, A., Stolz, D.B., Kaizu, T., Tsung, A., Ikeda, A., Shimizu, H., Takahashi, T., Tomiyama, K., Sugimoto, R., Choi, A.M., Billiar, T.R., Murase, N., McCurry, K.R., 2007. Carbon monoxide protects rat lung transplants from ischemia-reperfusion injury via a mechanism involving p38 MAPK pathway. Am. J. Transplant. 7, 22792290. Kovisto, A., Matthias, A., Bronnikov, G., Nedergaard, J., 1997. Kinetics of the inhibition of mitochondrial respiration by NO. FEBS Lett. 417, 7580. Kristal, B.S., Koopmans, S.J., Jackson, C.T., Ikeno, Y., Park, P.-J., Yu, B.P., 1997. Oxidantmediated repression of mitochondrial transcription in diabetic rats. Free Radical Biol. Med. 22, 813822. Lancel, S., Hassoun, S.M., Favory, R., Decoster, B., Motterlini, R., Neviere, R., 2009. Carbon monoxide rescues mice from lethal sepsis by supporting mitochondrial energetic metabolism and activating mitochondrial biogenesis. J. Pharmacol. Exp. Ther. 329, 641648.

92

P.B.L. Pun et al. / Mitochondrion 10 (2010) 8393 Pearce, L.L., Lopez, M.E., Martinez-Bosch, S., Peterson, J., 2008. Antagonism of nitric oxide toward the inhibition of cytochrome c oxidase by carbon monoxide and cyanide. Chem. Res. Toxicol. 21, 20732081. Peralta, J.G., Llesuy, S., Evelson, P., Carreras, M.C., Flecha, B.G., Poderoso, J.J., 1993. Oxidative stress in skeletal muscle during sepsis in rats. Circ. Shock. 39, 153 159. Persichini, T., Mazzone, V., Polticelli, F., Moreno, S., Venturini, G., Clementi, E., Colasanti, M., 2005. Mitochondrial type I nitric oxide synthase physically interacts with cytochrome c oxidase. Neurosci. Lett. 384, 254259. Petersen, L.C., 1977. The effect of inhibitors on the oxygen kinetics of cytochrome c oxidase. Biochim. Biophys. Acta 460, 299307. Piantadosi, C.A., 2002. Biological chemistry of carbon monoxide. Antioxid. Redox Signal. 4, 259270. Piantadosi, C.A., 2008. Carbon monoxide, reactive oxygen signalling and oxidative stress. Free Radical Biol. Med. 45, 562569. Piantadosi, C.A., Lee, P.A., Sylvia, A.L., 1988. Direct effects of CO on cerebral energy metabolism in bloodless rats. J. Appl. Physiol. 65, 878887. Piantadosi, C.A., Tatro, L., Zhang, J., 1995. Hydroxyl radical production in the brain after CO hypoxia in rats. Free Radical Biol. Med. 18, 603609. Piantadosi, C.A., Zhang, J., Levin, E.D., Folz, R.J., Schmechel, D.E., 1997. Apoptosis and delayed neuronal damage after carbon monoxide poisoning in the rat. Exp. Neurol. 147, 103114. Piantadosi, C.A., Carraway, M.S., Suliman, H.B., 2006. Carbon monoxide, oxidative stress, and mitochondrial permeability pore transition. Free Radical Biol. Med. 40, 13321339. Piantadosi, C.A., Carraway, M.S., Babikr, A., Suliman, H.B., 2008. Heme oxygenase-1 regulates cardiac mitochondrial biogenesis via Nrf2-mediated transcriptional control of nuclear respiratory factor-1. Circ. Res. 103, 12321240. Ponderoso, J.J., Carreras, M.C., Lisdero, C., Riobo, N., Schopfer, F., Boveris, A., 1996. Nitric oxide inhibits electron transfer and increases superoxide radical production in rat heart mitochondria and submitochondrial particles. Arch. Biochem. Biophys. 328, 8592. Pong, W.W., Eldred, W.D., 2009. Interactions of the gaseous neuromodulators nitric oxide, carbon monoxide, and hydrogen sulphide in the salamander retina. J. Neurosci. Res.. doi:10.1002/jnr.22042. Prockop, L.D., Chichkova, R.I., 2007. Carbon monoxide intoxication: an updated review. J. Neurol. Sci. 262, 122130. Radi, R., Rodriguez, M., Castro, L., Telleri, R., 1994. Inhibition of mitochondrial electron transport by peroxynitrite. Arch. Biochem. Biophys. 308, 8995. Riobo, N.A., Melani, M., Sanjuan, N., Fiszman, M.L., Gravielle, M.C., Carreras, M.C., Cadenas, E., Poderoso, J.J., 2002. The modulation of mitochondrial nitric-oxide synthase activity in rat brain development. J. Biol. Chem. 277, 4244742455. Sarkela, T.M., Berthiaume, J., Elfering, S., Gybina, A.A., Giulivi, C., 2001. The modulation of oxygen radical production by nitric oxide in mitochondria. J. Biol. Chem. 276, 69456949. Sarti, P., Giuffre, A., Forte, E., Mastronicola, D., Barone, M.C., Brunori, M., 2000. Nitric oxide and cytochrome c oxidase: mechanisms of inhibition and NO degradation. Biochem. Biophys. Res. Commun. 274, 183187. Saviani, E.E., Orsi, C.H., Oliveira, J.F., Pinto-Maglio, C.A., Salgado, I., 2002. Participation of the mitochondrial permeability transition pore in nitric oxide-induced plant cell death. FEBS Lett. 510, 136140. Scragg, J.L., Dallas, M.L., Wilkinson, J.A., Varadi, G., Peers, C., 2008. Carbon monoxide inhibits L-type Ca2+ channels via redox modulation of key cysteine residues by mitochondrial reactive oxygen species J. Biol. Chem. 283, 2441224419. Silva, G., Cunha, A., Gregoire, I.P., Seldon, M.P., Soares, M.P., 2006. The antiapoptotic effect of heme oxygenase-1 in endothelial cells involves the degradation of p38 alpha MAPK isoform. J. Immunol. 177, 18941903. Slebos, D.J., Ryter, S.W., van der Toorn, M., Liu, F., Guo, F., Baty, C.J., Karlsson, J.M., Watkins, S.C., Kim, H.P., Wang, X., Lee, J.S., Postma, D.S., Kauffman, H.F., Choi, A.M., 2007. Mitochondrial localization and function of heme oxygenase-1 in cigarette smoke-induced cell death. Am. J. Respir. Cell Mol. Biol. 36, 409417. Sokal, J.A., Opacka, J., Gorny, R., Kolakowski, J., 1982. Effect of different conditions of acute exposure to carbon monoxide on the cerebral high-energy phosphates and ultrastructure of brain mitochondria in rats. Toxicol. Lett. 11, 213219. Srisook, K., Han, S.S., Choi, H.S., Li, M.H., Ueda, H., Kim, C., Cha, Y.N., 2006. CO from enhanced HO activity or from CORM-2 inhibits both O2- and NO production and downregulates HO-1 expression in LPS-stimulated macrophages. Biochem. Pharmacol. 71, 307318. Stadler, J., Billiar, T.R., Curran, R.D., Stuehr, D.J., Ochoa, J.B., Simmons, R.L., 1991a. Effect of exogenous and endogenous nitric oxide on mitochondrial respiration of rat hepatocytes. Am. J. Physiol. 260, C910C916. Stadler, J., Curran, R.D., Ochoa, J.B., Harbrecht, B.G., Hoffman, R.A., Simmons, R.L., Billiar, T.R., 1991b. Effect of endogenous nitric oxide on mitochondrial respiration of rat hepatocytes in vitro and in vivo. Arch. Surg. 126, 186191. Stipanuk, M.H., Beck, P.W., 1982. Characterization of the enzymic capacity for cysteine desulphhydration in liver and kidney of the rat. Biochem. J. 206, 267 277. Suliman, H.B., Carraway, M.S., Tatro, L.G., Piantadosi, C.A., 2007. A new activating role for CO in cardiac mitochondrial biogenesis. J. Cell Sci. 120, 299308. Taoka, S., Banerjee, R., 2001. Characterization of NO binding to human cystathionine beta synthase: possible implications of the effects of CO and NO binding to the human enzyme. J. Inorg. Biochem. 87, 245251. Taskiran, D., Nesil, T., Alkan, K., 2007. Mitochondrial oxidative stress in female and male rat brain after ex vivo carbon monoxide treatment. Toxicology 26, 645 651.

Lavitrano, M., Smolenski, R.T., Musumeci, A., Maccherini, M., Slominska, E., Di Florio, E., Bracco, A., Mancini, A., Stassi, G., Patti, M., Giovannoni, R., Froio, A., Simeone, F., Forni, M., Bacci, M.L., DAlise, G., Cozzi, E., Otterbein, L.E., Yacoub, M.H., Bach, F.H., Cliase, F., 2004. Carbon monoxide improves cardiac energetics and safeguards the heart during reperfusion after cardiopulmonary bypass in pigs. FASEB J. 18, 10931095. Leschelle, X., Goubern, M., Andriamihaja, M., Blottiere, H.M., Couplan, E., GonzalezBarroso, M., Petit, C., Pagniez, A., Chaumontet, C., Mignotte, B., Bouillaud, F., Blachier, F., 2005. Adaptative metabolic response of human colonic epithelial cells to the adverse effects of the luminal compound sulphide. Biochim. Biophys. Acta 1725, 201212. Li, L., Moore, P.K., 2007. An overview of the biological signicance of endogenous gases: new roles for old molecules. Biochem. Soc. Trans. 35, 11381141. Li, M.H., Cha, Y.N., Surh, Y.J., 2006. Carbon monoxide protects PC12 cells from peroxynitrite-induced apoptotic death by preventing the depolarization of mitochondrial transmembrane potential. Biochem. Biophys. Res. Commun. 342, 984990. Li, X., Du, J., Jin, H., Tang, X., Bu, D., Tang, C., 2007. The regulatory effect of endogenous hydrogen sulphide on pulmonary vascular structure and gasotransmitters in rats with high pulmonary blood ow. Life Sci. 81, 841849. Li, L., Hsu, A., Moore, P.K., 2009. Actions and interactions of nitric oxide, carbon monoxide and hydrogen sulphide in the cardiovascular system and in inammation a tale of three gases! Pharmacol. Ther.. doi:10.1016/ j.pharmathera.2009.05.005. Lisdero, C.L., Carreras, M.C., Meulemans, A., Melani, M., Aubier, M., Boczkowski, J., Poderoso, J.J., 2004. The mitochondrial interplay of ubiquinol and nitric oxide in endotoxemia. Methods Enzymol. 382, 6781. Liu, S.H., Ma, K., Xu, B., Xu, X.R., 2006. The mechanisms of carbon monoxide inhalation on the apoptosis of pulmonary cells in rats with acute lung injury induced by lipopolysaccharide. Zhonghua Jie He He Hu Xi Za Zhi 29, 329332. Lizasoain, I., Moro, M.A., Knowles, R.G., Darley-Usmar, V., Moncada, S., 1996. Nitric oxide and peroxynitrite exert distinct effects on mitochondrial respiration which are differentially blocked by glutathione or glucose. Biochem. J. 314, 877880. Lopez, L.C., Escames, G., Tapias, V., Utrilla, P., Leon, J., Acuna-Castroviejo, D., 2006. Identication of an inducible nitric oxide synthase in diaphragm mitochondria from septic mice: its relation with mitochondrial dysfunction and prevention by melatonin. Int. J. Biochem. Cell Biol. 38, 267278. Lowicka, E., Beltowski, J., 2007. Hydrogen sulphide (H2S): the third gas of interest for pharmacologists. Pharmacol. Rep. 59, 424. Manthey, G.M., McEwen, J.E., 1995. The product of the nuclear gene PET309 is required for translation of mature mRNA and stability or production of introncontaining RNAs derived from the mitochondrial COX1 locus of Saccharomyces cerevisiae. EMBO J. 14, 40314043. Mason, T.L., Schatz, G., 1973. Cytochrome c oxidase from bakers yeast. II. Site of translation of the protein components. J. Biol. Chem. 248, 13551360. Mason, M.G., Nicholls, P., Wilson, M.T., Cooper, C.E., 2006. Nitric oxide inhibition of respiration involves both competitive (heme) and non-competitive (copper) binding to cytochrome c oxidase. Proc. Natl. Acad. Sci. USA 103, 708713. Matsuoka, M., Igisu, H., Tanaka, I., Hori, H., Koga, M., 1993. Brain energy metabolites in mice after an acute exposure to carbon monoxide. Res. Commun. Chem. Pathol. Pharmacol. 81, 1520. Miro, O., Casademont, J., Barrientos, A., Urbano-Marquez, A., Cardellach, F., 1998. Mitochondrial cytochrome c oxidase inhibition during acute carbon monoxide poisoning. Pharmacol. Toxicol. 82, 199202. Molina, J.A., Jimenez-Jimenez, F.J., Orti-Pareja, M., Navarro, J.A., 1998. The role of nitric oxide in neurodegeneration. Potential for pharmacological intervention. Drugs Aging 12, 251259. Moore, P.K., Bhatia, M., Moochhala, S., 2003. Hydrogen sulphide: from the smell of the past to the mediator of the future? Trends Pharmacol. Sci. 24, 609611. Murphy, M.P., 2009. How mitochondria produce reactive oxygen species. Biochem. J. 417, 113. Nicholls, P., Kim, J.K., 1982. Sulphide as an inhibitor and electron donor for the cytochrome c oxidase system. Can. J. Biochem. 60, 613623. Nicholson, R.A., Roth, S.H., Zhang, A., Zheng, J., Brookes, J., Skrainy, B., Bennington, R., 1998. Inhibition of respiratory and bioenergetic mechanisms by hydrogen sulphide in mammalian brain. J. Toxicol. Environ. Health A 54, 491507. Nisoli, E., Clementi, E., Paolucci, C., Cozzi, V., Tonello, C., Sciorati, C., Bracale, R., Valerio, A., Francolini, M., Moncada, S., Carruba, M.O., 2003. Mitochondrial biogenesis in mammals: the role of endogenous nitric oxide. Science 299, 896 899. Oh, G.S., Pae, H.O., Lee, B.S., Kim, B.N., Kim, J.M., Kim, H.R., Jeon, S.B., Jeon, W.K., Chae, H.J., Chung, H.T., 2006. Hydrogen sulphide inhibits nitric oxide production and nuclear factor-kappa B via heme oxygenase-1 expression in RAW264.7 macrophages stimulated with lipopolysaccharide. Free Radical Biol. Med. 41, 106119. Olson, K.R., Donald, J.A., 2009. Nervous control of circulationthe role of gasotransmitters, NO, CO and H2S. Acta Histochem. 111, 244256. Palmer, R.M., Ferrige, A.G., Moncada, S., 1987. Nitric oxide release accounts for the biological activity of endothelium-derived relaxing factor. Nature 327, 524 526. Pankow, D., Ponsold, W., 1984. Effect of carbon monoxide exposure on heart cytochrome c oxidase activity of rats. Biomed. Biochim. Acta 43, 11851189. Pearce, L.L., Bominaar, E.L., Hill, B.C., Peterson, J., 2003. Reversal of cyanide inhibition of cytochrome c oxidase by the auxiliary substrate nitric oxide. J. Biol. Chem. 278, 5213952145.

P.B.L. Pun et al. / Mitochondrion 10 (2010) 8393 Theissen, U., Martin, W., 2008. Sulde:quinone oxidoreductase (SQR) from the lugworm Arenicola marina shows cyanide- and thioredoxin-dependent activity. FEBS J. 275, 11311139. Therond, P., 2006. Oxidative stress and damages to biomolecules (lipids, proteins, DNA). Ann. Pharm. Fr. 64, 383389. Thom, S.R., Fisher, D., Xu, Y.A., Notarfrancesco, K., Ischiropoulos, H., 2000. Adaptive responses and apoptosis in endothelial cells exposed to carbon monoxide. Proc. Natl. Acad. Sci. USA 97, 13051310. Toghi, R., Tillmark, N., Dare, E., Aberg, A.M., Larsson, J.E., Ceccatelli, S., 2006. Hypoxia-independent apoptosis in neural cells exposed to carbon monoxide in vitro. Brain Res. 1098, 18. Tsui, T.Y., Siu, Y.T., Scholitt, H.J., Fan, S.T., 2005. Heme oxygenase-1-derived carbon monoxide stimulates adenosine triphosphate generation in human hepatocytes. Biochem. Biophys. Res. Commun. 336, 898902. Tsukihara, T., Aoyama, H., Yamashita, E., Tomizaki, T., Yamaguchi, H., Shinzawa, I.K., Nakashima, R., Yaono, R., Yoshikawa, S., 1996. The whole structure of the 13subunit oxidized cytochrome c oxidase at 2.8 angstrom. Science 272, 1136 1144. Uemura, K., Harada, K., Sadamitsu, D., Tsuruta, R., Takahashi, M., Aki, T., Yasuhara, M., Maekawa, T., Yoshida, K., 2001. Apoptotic and necrotic brain lesions in a fatal case of carbon monoxide poisoning. Forensic Sci. Int. 116, 213219. Ushmorov, A., Ratter, F., Lehmann, V., Droge, W., Schirrmacher, V., Umansky, V., 1999. Nitric oxide-induced apoptosis in human leukemic lines requires mitochondrial lipid degradation and cytochrome c release. Blood 93, 2342 2352. Valdez, L.B., Zaobornyj, T., Alvarez, S., Bustamante, J., Costa, L.E., Boveris, A., 2004. Heart mitochondrial nitric oxide synthase. Effects of hypoxia and aging. Mol. Aspects Med. 25, 4959. Varon, J., Marik, P.E., Fromm, R.E.J., Gueler, A., 1999. Carbon monoxide poisoning: a review for clinicians. J. Emerg. Med. 17, 8793. Vieira, H.L., Queiroga, C.S., Alves, P.M., 2008. Pre-conditioning induced by carbon monoxide provides neuronal protection against apoptosis. J. Neurochem. 107, 375384. Vlasova, I.I., Tyurin, V.A., Kapralov, A.A., Krunikov, I.V., Osipov, A.N., Potapovich, M.V., Stovanovsky, D.A., Kagan, V.E., 2006. Nitric oxide inhibits peroxidise activity of cytochrome ccardiolipin complex and blocks cardiolipin oxidation. J. Biol. Chem. 281, 45544562. Volkel, S., Grieshaber, M.K., 1996. Mitochondrial sulphide oxidation in Arenicola marina. Evidence for alternative electron pathways. Eur. J. Biochem. 235, 231 237. Wang, R., 2002. Twos company, threes a crowd: can H2S be the third endogenous gaseous transmitter? FASEB J. 16, 17921798.

93

Wang, C.Z., Li, A., Yang, Z.C., 1990. The pathophysiology of carbon monoxide poisoning and acute respiratory failure in a sheep model with smoke inhalation injury. Chest 97, 736742. Wang, Y.F., Jin, H.F., Tang, C.S., Du, J.B., 2006. Role of gasotransmitters in the pathogenesis of pulmonary hypertension. Beijing Da Xue Xue Bao 38, 326330. Warenycia, M.W., Goodwin, L.R., Benishin, C.G., Reiffenstein, R.J., Francom, D.M., Taylor, J.D., Dieken, F.P., 1989. Acute hydrogen sulphide poisoning. Demonstration of selective uptake of sulphide by the brainstem by measurement of brain sulphide levels. Biochem. Pharmacol. 38, 973981. Wei, T., Chen, C., Hou, J., Xin, W., Mori, A., 2000. Nitric oxide induces oxidative stress and apoptosis in neuronal cells. Biochim. Biophys. Acta 1498, 7279. Welter, R., Yu, L., Yu, C.A., 1996. The effects of nitric oxide on electron transport complexes. Arch. Biochem. Biophys. 331, 914. Wever, R., van Gelder, B.F., Dervartanian, D.V., 1975. Biochemical and biophysical studies on cytochrome c oxidase. XX. Reaction with sulphide. Biochim. Biophys. Acta 387, 189193. Wu, L., Wang, R., 2005. Carbon monoxide: endogenous production, physiological functions, and pharmacological applications. Pharmacol. Rev. 57, 585630. Xu, W., Charles, I.G., Moncada, S., 2005. Nitric oxide: orchestrating hypoxia regulation through mitochondrial respiration and the endoplasmic reticulum stress response. Cell Res. 15, 6365. Yoshida, T., Kikuchi, G., 1974. Sequence of the reaction of heme catabolism catalysed by the microsomal heme oxygenase system. FEBS Lett. 48, 256261. Yoshioka, Y., Yamamuro, A., Maeda, S., 2006. Nitric oxide/cGMP signalling pathway protects RAW264 cells against nitric oxide-induced apoptosis by inhibiting the activation of p38 mitogen-activated protein kinase. J. Pharmacol. Sci. 101, 126 134. Young, L.J., Caughey, W.S., 1986. Oxygenation of carbon monoxide by bovine heart cytochrome c oxidase. Biochemistry 25, 152161. Zemojtel, T., Kolanczyk, M., Kossler, N., Stricker, S., Lurz, R., Mikula, I., Duchniewicz, M., Schuelke, M., Ghafourifar, P., Martasek, P., Vingron, M., Mundlos, S., 2006. Mammalian mitochondrial nitric oxide synthase: characterization of a novel candidate. FEBS Lett. 580, 455462. Zhang, X., Shan, P., Alam, J., Fu, X.Y., Lee, P.J., 2005. Carbon monoxide differentially modulates STAT1 and STAT3 and inhibits apoptosis via a phosphatidylinositol 3-kinase/Akt and p38 kinase-dependent STAT3 pathway during anoxiareoxygenation injury. J. Biol. Chem. 280, 87148721. Zhao, W., Zhang, J., Lu, Y., Wang, R., 2001. The vasorelaxant effect of H2S as a novel endogenous gaseous KATP channel opener. EMBO J. 20, 60086016. Zuckerbraun, B.S., Chin, B.Y., Bilban, M., Costa dAvila, J., Rao, J., Billiar, T.R., Ottebein, L.E., 2007. Carbon monoxide signals via inhibition of cytochrome c oxidase and generation of mitochondrial reactive oxygen species. FASEB J. 21, 10991106.

You might also like