Introduction

Hydrogen sulfide (H2S), a colorless, flammable and water-soluble gas with the characteristic odor of rotten eggs, has been known for decades because of its toxicity and as an environmental hazard [1, 2]. Inhibition of mitochondrial respiration – more potent than that of cyanide [3] – resulting from blockade of cytochrome c oxidase is the main mechanism of H2S toxicity [4, 5]. During recent years, however, H2S has been recognized as an important signaling molecule of the cardiovascular system, the inflammatory system and the nervous system. Alongside nitric oxide (NO) and carbon monoxide, therefore, H2S is now known as the third endogenous gaseotransmitter [1, 6].

Since H2S is a small ubiquitous gaseous diffusible molecule, its putative interest for intensive care research is obvious. Consequently, inhibitors of its endogenous production as well as compounds that donate H2S have been studied in various models of shock resulting from hemorrhage [79], ischemia/reperfusion [1018], endotoxemia [1921], bacterial sepsis [2225] and nonmicrobial inflammation [2629] – which, however, yielded rather controversial data with respect to the proinflammatory or anti-inflammatory properties of H2S. The present article reviews the current literature on the therapeutic potential of H2S, with a special focus on clinically relevant studies in – if available – large animal models.

Biological chemistry

In mammals, H2S is synthesized from the sulfur-containing amino acid L-cysteine by either cystathionine-β-synthase or cystathionine-γ-lyase, both using pyridoxal 5'-phosphate (vitamin B6) as a cofactor [3032]. This synthesis results in low micromolar H2S levels in the extracellular space, which can be rapidly consumed and degraded by various tissues. Similarly to NO and carbon monoxide, H2S is a lipophilic compound that easily permeates cell membranes without using specific transporters. Via direct inhibition, NO as well as carbon monoxide are involved in the regulation of cystathionine-β-synthase, but not cystathionine-γ-lyase, which can be activated by lipopolysaccharide (LPS) [1, 6].

There are three known pathways of H2S degradation: mitochondrial oxidation to thiosulfate, which is further converted to sulfite and sulfate; cytosolic methylation to dimethylsulfide; and sulfhemoglobin formation after binding to hemoglobin [6]. Similar to NO and carbon monoxide, H2S can also bind to hemoglobin – which was therefore termed the common sink for the three gaseous transmitters [33]. Consequently, saturation with one of these gases might lead to enhanced plasma concentrations and, subsequently, to biological effects of the other gases [1]. Table 1 summarizes the physicochemistry of H2S in mammalian tissues.

Table 1 Physicochemistry and biology of hydrogen sulfide

Mechanisms of H2S

H2S exerts its effects in biological systems through a variety of interrelated mechanisms (for a review see [1]). Our current knowledge of the biology of H2S predominantly stems from in vitro studies in various cell and isolated organ systems, either using cystathionine-γ-lyase inhibitors such as D,L-propargylglycine (PAG) and β-cyanoalanine, or administration of H2S gas or H2S donors such as sodium disulfide (Na2S) and sodium hydrogen sulfide (NaHS). While high (high micromolar to millimolar) levels are invariably accompanied with cytotoxic effects [34] – which result from free radical generation, glutathione deletion, intracellular iron release and pro-apoptotic action through both the death receptor and mitochondrial pathways [35] – lower (low micromolar) levels have been shown to exert either cytoprotective (antinecrotic or antiapoptotic) effects [1013, 36] or proapoptotic properties [3739], depending on the cell type and on the experimental conditions.

Cytochrome c oxidase, a component of the oxidative phosphorylation machinery within the mitochondrium, is one intracellular target of H2S [4, 5]. Both the toxic effects of H2S as well as the induction of a so-called "suspended animation" [40, 41] are referred to in this inhibition of mitochondrial respiration [42, 43], and thus may represent a possible mechanism for the regulation of cellular oxygen consumption [44].

Activation of potassium-dependent ATP channels is another major mechanism of H2S, which in turn causes vasodilation, preconditioning against ischemia/reperfusion injury and myocardial protection [45]. Various findings support this concept [1, 6, 46]: potassium-dependent ATP channel blockers (sulfonylurea derivates – for example, glibenclamide) attenuated the H2S-induced vasodilation both in vivo and in vitro [47, 48], and stimulation of potassium-dependent ATP channels was demonstrated in the myocardium, pancreatic β cells, neurons and the carotid sinus [6]. Moreover, glibenclamide reversed the otherwise marked Na2S-related increase of the hepatic arterial buffer response capacity that counteracts reduction of portal venous flow, whereas PAG decreased this compensatory mechanism [49].

An endothelium-dependent effect seems to contribute to these vasodilatory properties: in human endothelial cells, H2S caused direct inhibition of the angiotensin-converting enzyme [50], and, finally, H2S can enhance the vasorelaxation induced by NO [51, 52]. The interaction between H2S and NO with respect to vascular actions is, however, fairly complex: low H2S concentrations may cause vasoconstriction as a result of an attenuated vasorelaxant effect of NO due to scavenging of endothelial NO and formation of an inactive nitrosothiol [5254]. The local oxygen concentration apparently assumes importance for the vasomotor properties of H2S as well [55]: while H2S had vasodilator properties at 40 μM oxygen concentration (that is, an oxygen partial pressure of approximately 30 mmHg), it exerted vaso-constrictor effects at a 200 μM oxygen concentration (that is, aan oxygen partial pressure of approximately 150 mmHg) [56]. Finally, the H2S-related inhibition of oxidative phosphorylation also contributes to the vasodilatation [57].

Owing to its SH group that allows reduction of disulfide bonds and radical scavenging, H2S also exerts biological effects as an antioxidant [9], in particular as an endogenous peroxynitrite scavenger [58], which is consistent with its cytoprotective effects in various cell-based experiments [59, 60]. In this context the effect of H2S on intracellular signal pathways assumes particular importance: in LPS-stimulated macrophages, pretreatment with physically dissolved gaseous H2S or the H2S-donor NaHS was affiliated with diminished activation of the nuclear transcription factor NF-κB and inhibition of the inducible isoform of the NO synthase. This effect coincided with increased expression of heme oxygenase-1, and co-incubation with carbon monoxide mimicked the cytoprotection exerted by H2S [61].

Conflicting data are available on the effects of H2S on other intracellular signal transduction pathways; for example, the mitogen-activated protein kinase pathway and the phosphatidyinositol-3-kinase/Akt pathway [20, 6165]. Depending on the cell lines used, both inhibitory [20] and activating [36, 61, 64] effects on p38 mitogen-activated protein kinase were reported, whereas H2S seems not to affect the stress-activated protein kinase c-Jun N-terminal kinase [61, 65]. In contrast, activation of the extracellular signal-regulated kinase 1/2 pathway has been implicated in the H2S-related ischemic preconditioning [48], both its proinflammatory [63, 65] and anti-inflammatory [20, 61] effects, as well as in the induction of apoptosis [62]. While the influence of H2S on extracellular signal-regulated kinase seems to be rather comprehensible [25], studies exploring the effect on downstream pathways result in conflictive statements.

Jeong and colleagues reported that H2S enhances NO production and inducible NO synthase expression by potentiating IL-1β-induced NF-κB in vascular smooth muscle cells [63], which is consistent with the H2S-induced NF-κB activation and subsequent proinflammatory cytokine production in IFNγ-primed monocytes [65]. Nevertheless, any H2S effect on NF-κB and its transcription-regulated mediators (for example, inducible NO synthase, cytokines and apoptotic factors) may be cell-type dependent and stimulus dependent. In fact, in addition to the above-mentioned decreased NF-κB activation and inducible NO synthase expression in LPS-stimulated macrophages [61], H2S administration also attenuated inducible NO synthase expression, NO production, as well as TNFα secretion in microglia exposed to LPS [20].

In the context of these contradictory findings, the doses of the H2S donors administered may assume particular importance. Even the physiologically relevant concentrations [36, 64] might have to be reconsidered due to overestimation of basal H2S levels: murine plasma sulfide levels are reported between 10 and 34 μM [21, 22], and are increased up to 20 to 65 μM after endotoxin injection [21] or cecal ligation and puncture [22]. A reduction of plasma sulfide concentration from 50 μM to ~25 μM, finally, was reported in patients with coronary heart disease [1], whereas plasma sulfide levels increased from 44 to 150 μM in patients with sepsis [21]. It should be noted, however, that the distinct techniques used by various groups to determine sulfide levels may account for the marked variability in the baseline values reported. The various derivatization methods, which are inherent to the analytic procedures, are likely to liberate sulfide from its bound forms so that the exact amount of free and bioavailable sulfide may be lower than frequently reported [66]. In fact, Mitsuhashi and colleagues reported that the blood sulfite concentrations (that is, the product of mitochondrial sulfide oxidation) were 3.75 ± 0.88 μM only in patients with pneumonia (versus 1.23 ± 0.48 μM in healthy control individuals) [67]. Infusing 2.4 and 4.8 mg/kg/hour in anesthetized and mechanically ventilated pigs over 8 hours resulted in maximum blood sulfide levels of 2.0 and 3.5 μM, respectively (baseline levels 0.5 to 1.2 μM) in our experiments [16].

Metabolic effects of H2S: induction of suspended animation

Suspended animation is a hibernation-like metabolic status characterized by a marked yet reversible reduction of energy expenditure, which allows nonhibernating species to sustain environmental stress, such as extreme changes in temperature or oxygen deprivation [41, 68].

In landmark work, the Roth's group provided evidence that inhaled H2S can induce such a suspended animation [40, 41]: in awake mice, breathing 80 ppm H2S caused a dose-dependent reduction of both the respiratory rate and the heart rate as well as of oxygen uptake and carbon dioxide production, which was ultimately associated with a drop in body core temperature to levels ~2°C above ambient temperature [40]. All these effects were completely reversible after H2S washout, and thereafter animals presented with a totally normal behavior. A follow-up study confirmed these observations, and the authors demonstrated using telemetry and echocardiography that the bradycardia-related fall in cardiac output coincided with an unchanged stroke volume and blood pressure. These physiologic effects of inhaled H2S were present regardless of the body core temperature investigated (27°C and 35°C) [69].

It is noteworthy that anesthesia may at least partially blunt the myocardial effect of inhaled H2S. In mechanically ventilated mice instrumented with left ventricular pressure volume conductance catheters and assigned to 100 ppm inhaled H2S, we found that hypothermia alone (27°C) but not normothermic H2S inhalation (38°C) decreased the cardiac output due to a fall in heart rate, whereas both the stroke volume as well as the parameters of systolic and diastolic function remained unaffected (Table 2) [70]. Interestingly, inhaled H2S in combination with hypothermia, however, was concomitant with the least stimulation of oxygen flux induced by addition of cytochrome c during state 3 respiration with combined complex I and complex II substrates (Figure 1) [71]. Since stimulation by cytochrome c should not occur in intact mitochondria, this finding suggests better preservation of mitochondrial integrity under these conditions [72].

Figure 1
figure 1

Cytochrome c-stimulated mitochondrial oxygen flux in livers from anesthetized and mechanically ventilated mice. Ratio of mitochondrial oxygen flux in homogenized livers from anesthetized and mechanically ventilated mice after addition in relation to before addition of cytochrome c. Since stimulation by cytochrome c should not occur in intact mitochondria, the smallest value (that is, a ratio close to 1.00) suggests preservation of mitochondrial integrity. Animals were subjected to inhaled hydrogen sulfide (H2S) (100 ppm over 5 hours) or vehicle gas during normothermia (38°C) and hypothermia (27°C) [63]. Data presented as mean ± standard deviation, n = 8 in each group. #P < 0.05 versus control, 38°C.

Table 2 Cardiac effects of inhaled H2S in anesthetized and mechanically ventilated mice during normothermia and hypothermia

In good agreement with the concept that a controlled reduction in cellular energetic expenditure would allow maintenance of ATP homoeostasis [41] and thus of improving outcome during shock states due to preserved mitochondrial function [73, 74], the group of Roth and colleagues subsequently demonstrated that pretreatment with inhaled H2S (150 ppm) for only 20 minutes markedly prolonged survival without any apparent detrimental effects for mice exposed to otherwise lethal hypoxia (5% oxygen) [75] and for rats undergoing lethal hemorrhage (60% of the calculated blood volume over 40 minutes) [8]. It is noteworthy that in the latter study the protective effect was comparable when using either inhaled H2S or a single intravenous bolus of Na2S [75]: parenteral sulfide administration has a number of practical advantages (ease of administration, no need for inhalation delivery systems, no risk of exposure to personnel, no issues related of the characteristic odor of H2S gas) and, in particular, avoids the pulmonary irritant effects of inhaled H2S, which can be apparent even at low inspiratory gaseous concentrations [76]. Finally, it is noteworthy that hypothermia is not a prerequisite of H2S-related cytoprotection during hemorrhage: the H2S donor NaHS improved hemodynamics, attenuated metabolic acidosis, and reduced oxidative and nitrosative stress in rats subjected to controlled hemorrhage at a mean blood pressure of 40 mmHg (Figure 2) [9].

Figure 2
figure 2

Hydrogen sulfide-related hemodynamic effects in rats subjected to hemorrhage and subsequent retransfusion. Time course of the difference in (a) mean blood pressure (ΔMAP) and (b) carotid blood flow (ΔCBF) in rats subjected to 60 minutes of hemorrhage (MAP 40 mmHg) and subsequent retransfusion of shed blood. Ten minutes prior to retransfusion, animals received vehicle (n = 11; open circles) or the hydrogen sulfide donor sodium hydrogen sulfide (bolus 0.2 mg/kg, n = 11; closed circles) [9]. Data presented as mean (standard deviation). #P < 0.05 versus controls.

The clinical relevance of murine models may be questioned because, due to their large surface area/mass ratio, rodents can rapidly drop their core temperature [77]. In fact, other authors failed to confirm the metabolic effect of inhaled H2S in anesthetized and mechanically ventilated piglets (body weight ~6 kg) or in H2S-sedated and spontaneously breathing sheep (body weight ~74 kg) exposed to up to 80 or 60 ppm H2S, respectively [78, 79]. These findings may be due to the dosing or timing of H2S, and are in contrast to recent data from our own group: in anesthetized and mechanically ventilated swine (body weight ~45 kg) that underwent transient thoracic aortic balloon occlusion, infusing the intravenous H2S donor Na2S over 10 hours reduced the heart rate and cardiac output without affecting the stroke volume, thereby reducing oxygen uptake and carbon dioxide production and, ultimately, core temperature [16]. The metabolic effect of H2S coincided with an attenuation of the early reperfusion-related hyperlactatemia – suggesting a reduced need for anaerobic ATP generation during the ischemia period – and an improved noradrenaline responsiveness, indicating both improved heart function and vasomotor response to catecholamine stimulation [16].

H2S-induced cytoprotection during ischemia–reperfusion

Deliberate hypothermia is a cornerstone of the standard procedures to facilitate neurological recovery after cardiac arrest and to improve postoperative organ function after cardiac and transplant surgery. Consequently, several authors investigated the therapeutic potential of H2S-induced suspended animation after ischemia–reperfusion injury – and H2S protected the lung [14], the liver [12], the kidney (Figure 3) [17, 80], and, in particular, the heart [10, 11, 13, 15, 18, 62, 8183]. H2S administered prior to reperfusion therefore limited the infarct size and preserved left ventricular function in mice [10] and in swine [11].

Figure 3
figure 3

Hydrogen sulfide attenuation of oxidative DNA damage in the kidney after organ ischemia–reperfusion. Oxidative DNA damage (tail moment in the alkaline version of the comet assay [89]) in kidney tissue biopsies prior to (left panel) and after 2 hours of organ ischemia and 8 hours of reperfusion (right panel) in control swine (n = 7; open box plots) and in animals treated with the hydrogen sulfide donor sodium disulfide (Na2S) (n = 8; grey box plots). Renal ischemia was induced by inflating the balloon of an intra-aortic catheter positioned at the renal artery orifices. Na2S infusion was infused before kidney ischemia (2 mg/kg/hour over 2 hours) as well as during the first 4 hours of reperfusion (1 mg/kg/hour) [72]. Data presented as median (quartiles, range). #P < 0.05 versus before ischemia, §P < 0.05 versus control.

While these findings were obtained without induction of hypothermia, preserved mitochondrial function documented by an increased complex I and complex II efficiency assumed major importance for the H2S-induced cytoprotection [10]. The important role of preserved mitochondrial integrity was further underscored by the fact that 5-hydroxydeconoate, which is referred to as a mitochondrial potassium-dependent ATP-channel blocker, abolished the anti-apoptotic effects of H2S [18]. Clearly, anti-inflammatory and anti-apoptotic effects also contributed to the improved postischemic myocardial function: treatment with H2S was associated with reduced myocardial myeloperoxidase activity and an absence of the increase in the IL-1β levels (that is, attenuated tissue inflammation [10, 18]), as well as complete inhibition of thrombin-induced leukocyte rolling, a parameter for leukocyte–endothelium interaction [10]. Moreover, the ischemia–reperfusion-induced activation of p38 mitogen-activated protein kinase, of c-Jun N-terminal kinase and of NF-κB was also attenuated by H2S [18]. Finally, H2S exerted anti-apoptotic effects as shown by reduced TUNEL staining [10, 11] and by expression of cleaved caspase-9 [18], caspase-3 [10, 11], poly-ADP-ribose-polymerase [11] and the cell death-inducing proto-oncogene c-fos [13].

Controversial role of H2S in animal models of inflammation

Despite the promising data mentioned above, it is still a matter of debate whether H2S is a metabolic mediator or a toxic gas [84] – particularly given the rather controversial findings on the immune function reported in various models of systemic inflammation. In fact, H2S exerted both marked proinflammatory effects [19, 2125, 27, 85] and anti-inflammatory effects [9, 10, 18, 20, 2830]. Studies using inhibitors of endogenous H2S production such as PAG demonstrated pro-nounced proinflammatory effects of H2S: PAG attenuated organ injury, blunted the increase of the proinflammatory cytokine and chemokine levels as well as the myeloperoxidase activity in the lung and liver, and abolished leukocyte activation and trafficking in LPS-induced endotoxemia [19, 21] or cecal ligation and puncture-induced sepsis [2225, 86]. In good agreement with these findings, the H2S donor NaHS significantly aggravated this systemic inflammation [2125, 86]. Although similar results were found during caerulin-induced pancreatitis [27, 87], the role of H2S during systemic inflammatory diseases is still a matter of debate. Zanardo and colleagues reported reduced leukocyte infiltration and edema formation using the air pouch and carrageenan-induced hindpaw edema model in rats injected with the H2S donors NaHS and Na2S [30]. Moreover, in mice with acute lung injury induced by combined burn and smoke levels, inhalation, a single Na2S bolus decreased tissue IL-1β increased IL-10 levels, and attenuated protein oxidation in the lung, which ultimately resulted in markedly prolonged survival [28].

Variable dosing and timing make it difficult to definitely conclude on the proinflammatory and/or anti-inflammatory effects of H2S: while the median sulfide lethal dose in rats has been described to be approximately 3 mg/kg intravenously [1], studies in the literature report on doses ranging from 0.05 to 5 mg/kg. In addition, there are only a small number of reports on continuous intravenous infusion rather than bolus administration. Finally, the role of the suspended animation-related hypothermia per se remains a matter of debate. While some studies report that spontanoues hypothermia and/or control of fever may worsen the outcome [88], other authors describe decreased inflammation [89] and improved survival after inducing hypothermia in sepsis [90].

We found in anesthetized and mechanically ventilated mice undergoing sham operation for surgical instrumentation that normothermic H2S (100 ppm) inhalation (38°C) over 5 hours and hypothermia (27°C) alone comparably attenuated the inflammatory chemokine release (monocyte chemotactic protein-1, macrophage inflammatory protein-2 and growth-related oncogen/keratinocyte-derived chemokine) in the lung tissue. While H2S did not affect the tissue concentrations of TNFα, combining hypothermia and inhaled H2S significantly decreased tissue IL-6 expression (Table 3) [91].

Table 3 Lung tissue concentrations of inflammatory chemokines after inhaling H2S during normothermia or hypothermia

Conclusion

Based on the concept that multiorgan failure secondary to shock, inflammation and sepsis may actually be an adaptive hypometabolic reponse to preserve ATP homoeostasis [92] – such as has been demonstrated for the septic heart [93] – and thus represent one of the organism's strategies to survive under stress conditions, the interest of inducing a hibernation-like suspended animation with H2S is obvious. Investigations have currently progressed most for the treatment of myocardial ischemia [94]. It must be underscored, however, that only a relatively small proportion of the published studies was conducted in clinically relevant large animal models [11, 16, 95], and, furthermore, that the findings reported are controversial [16, 78, 79].

Moreover, several crucial issues warrant further investigation before the clinical application of this concept. First, the role of hypothermia for any suspended animation-related organ protection is well established [96], but its impact remains a matter of debate for H2S-related organ protection. Clearly, in the rodent studies [10, 12, 18, 28], any cytoprotective effect was apparent without a change in core body temperature, but localized metabolic effects cannot be excluded [10]. In addition, the role of any H2S-related hypothermia remains controversial in the context of systemic inflammation [88]. Second, similar to the friend and foe character of NO, no definitive conclusions can be made as to whether H2S exerts proinflammatory or anti-inflammatory properties [1, 6, 85]. Finally, in addition to the question of dosing and timing (for example, bolus administration versus continuous intravenous infusion), the preferred route of H2S administration remains to be settled: while inhaling gaseous H2S probably allows easily titrating target blood concentrations, it is well established that this method can also directly cause airway irritation [76].

While H2S-induced suspended animation in humans to date may still be referred to as science fiction, there are ample promising preclinical data that this approach is a fascinating new therapeutic perspective for the management of shock states that merits further investigation.

Note

This article is part of a review series on Gaseous Mediators, edited by Peter Radermacher.

Other articles in the series can be found online at http://www.ccforum.com/series/gaseous_mediators