Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Oct 1.
Published in final edited form as: Pharmacol Res. 2010 Jun 11;62(4):289–297. doi: 10.1016/j.phrs.2010.06.002

Hydrogen Sulfide and Ischemia - Reperfusion Injury

Chad K Nicholson 1, John W Calvert 1
PMCID: PMC2917489  NIHMSID: NIHMS213935  PMID: 20542117

Abstract

Gasotransmitters are lipid soluble, endogenously produced gaseous signaling molecules that freely permeate the plasma membrane of a cell to directly activate intracellular targets, thus alleviating the need for membrane-bound receptors. The gasotransmitter family consists of three members: nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S). H2S is the latest gasotransmitter to be identified and characterized and like the other members of the gasotransmitter family, H2S was historically considered to be a toxic gas and an environmental/occupational hazard. However with the discovery of its presence and enzymatic production in mammalian tissues, H2S has gained much attention as a physiological signaling molecule. Also, much like NO and CO, H2S’s role in ischemia/reperfusion (I/R) injury has recently begun to be elucidated. As such, modulation of endogenous H2S and administration of exogenous H2S has now been demonstrated to be cytoprotective in various organ systems through diverse signaling mechanisms. This review will provide a detailed description of the role H2S plays in different model systems of I/R injury and will also detail some of the mechanisms involved with its cytoprotection.

1. Overview of Gasotransmitters

Cellular signaling often involves complex systems, whereby interactions between membrane-bound proteins and signaling molecules lead to the activation of intracellular molecules. These intracellular molecules act as secondary messengers, which then relay a signal to a specific destination. A set of endogenous gaseous molecules called gasotransmitters possesses similar signaling capabilities as other signaling molecules but does not require the regular string of regulatory mechanisms to transmit a signal [1]. Gasotransmitters are lipid soluble, endogenously produced, and freely permeate the plasma membrane of a cell to pass the message directly to an intracellular target [2]. Nitric oxide (NO) was the first gasotransmitter to be recognized as a signaling molecule, when it was identified as both a smooth muscle relaxer through the actions of acetylcholine [3] and an activator of macrophages [4]. NO is synthesized by the enzyme nitric oxide synthase (NOS) from the oxidation of the guanidine group of L-arginine [5]. There are three known isoforms of NOS that have been characterized, purified and cloned: neuronal nitric oxide synthase (nNOS), involved with neuronal signal transmission [6]; inducible nitric oxide synthase (iNOS), responsible for macrophage activation [7]; and endothelial nitric oxide synthase (eNOS), plays a role in vasorelaxation [8]. Calcium-calmodulin pathways regulate the eNOS and nNOS isoforms whereas iNOS is independent of these actions [9]. When NO is produced it diffuses through the endothelial cell membrane into smooth muscle cells (SMCs) where it activates guanylyl cyclase to produce cyclic guanosine monophospate (cGMP) [10]. Since these discoveries it has been shown that increasing NO by the administration of NO donors [11], inhaled gas therapy [12], or overexpression of eNOS protects against ischemic injury (heart, brain, liver, etc.). Preconditioning with NO has also been shown to be protective in cerebral [13], intestinal [14], and hepatic [15] ischemia.

Following the discovery of NO as a gasotransmitter, carbon monoxide (CO) was found to have similar roles. CO is formed endogenously by the enzyme heme oxygenase (HO) through the degradation of heme. There are three known isoforms of HO: HO-1, inducible under cellular stress; HO-2, homeostatic form; and HO-3, recently found in rat brain but no gene for HO-3 has yet to been found [16]. HO-1 is responsible for degrading heme into biliverdin and CO. Biliverdin is quickly reduced in the cell to bilirubin, which is a very important antioxidant. Induction of HO-1 through pharmacological agents has been shown to significantly reduce myocardial infarct size in vivo [17]. Yet et al. [18] demonstrated that a cardiac specific HO-1 transgenic mouse is also protected in myocardial ischemia/reperfusion (MI/R) injury [18]. CO, like NO, is also a known vasodilator through the activation of guanylyl cyclase [19]. However, some evidence suggests calcium-activated potassium channels are targets of CO in the context of vasorelaxation [20]. The cytoprotective effects of CO are also not limited to the heart, as it has been shown that CO contributes to central nervous system mediated blood pressure regulation [21], protects against pulmonary [22] and renovascular hypertension [23], modulates atherosclerosis [24], improves both allograft and xenograft survival following organ transplantation [25], and exerts a restorative effect on the pathologic remodeling response after balloon angioplasty [26].

2. Emergence of H2S as the Third Gasotransmitter

The third member of the gasotransmitter family to be identified was hydrogen sulfide (H2S), a fetid smelling molecule long thought of as a toxic gas. Warenycia et al [27] first reported that H2S was produced in very low concentrations endogenously when they were investigating acute H2S poisoning in the brain. Soon after this discovery, evidence of a physiological role of H2S began to unravel. Skrajny et al [28] found increased levels of serotonin and reduced levels of norepinephrine in the frontal cortex of a rat when chronically exposed to 20 ppm of H2S and in 1996, Abe et al [29] suggested that H2S was an endogenous neuromodulator, as they showed that physiological concentrations of H2S enhanced NMDA receptor-mediated responses and aided in the induction of hippocampal long-term potentiation. Shortly after, Hosoki et al [30] reported that an enzyme, which produces H2S, is present in the ileum, portal vein, and thoracic aorta and proposed that H2S may be an endogenous smooth muscle relaxant. Accompanying these discoveries was an interest in the physiological role of H2S in biological systems.

2.1 Physiological Role of H2S in Biological Systems

There are three known enzymes that produce H2S endogenously in mammalian tissue: cystathionine β-synthase (CBS), cystathionine γ-lyase (CGL or CSE), and 3-mercaptopyruvate sulfur transferase (3MST). In most tissues, CBS and CSE are responsible for catalyzing the production of H2S. CBS and CSE are both pyridoxal-5’-phosphate-dependent enzymes that utilize L-cysteine and homocysteine as substrates to liberate ammonium, pyruvate, and H2S [1]. It was originally believed that CBS was responsible for H2S production in the brain through the activation of the Ca2+/calmodulin pathway [31], but the discovery that approximately 90% of H2S in the brain is produced by 3MST [32] has changed the perceived role of CBS in H2S production. H2S is produced by 3MST from L-cysteine and α-ketoglutarate through the metabolism with cysteine aminotransferase (CAT) [32]. Although 3MST is responsible for the majority of H2S production in the brain, it is localized mainly in neurons, whereas CBS is located in astrocytes, suggesting that some H2S signaling in the brain may require CBS.

Under physiological conditions, two thirds of H2S is dissociated into H+ and HS and the remaining one third is in its undissociated form (H2S ⇔ HS + H+, pKa = 6.9) [33]. There are three major fates of H2S in the body. First, most of the H2S produced in the body is oxidized in the mitochondria to an end product of sulfate [34]. The remaining H2S is either methylated by thiol S-methyltransferase (TSMT) to methanethiol and dimethylsulfide [35] or binds to methemoglobin to form sulfhemoglobin [36]. Sulfate and thiosulfate are the major end product of H2S and are excreted in urine [34].

3. Cytoprotective Effects of Endogenous/Exogenous H2S

The physiological actions of H2S make this gas ideally suited to protect the heart, brain, liver, kidney, and lungs against injury during ischemia/reperfusion (I/R). In recent years, the cytoprotective effects of endogenous and exogenous H2S have been investigated in models of in vitro [37,38] and in vivo [3945] ischemic injury (summarized in the Table). The effects of endogenous H2S have primarily been studied by pharmacologically inhibiting CGL and by genetically targeting CGL in mice, whereas the effects of exogenous H2S have been studied through the administration of H2S in the form of sodium hydrosulfide (NaHS), sodium sulfide (Na2S), or H2S gas.

Table.

Summary of H2S in Ischemia/Reperfusion Injury

Organ Experimental Model* Treatment** Effects Ref.
Heart Langendorff hanging
heart model (30 min
I/2 hr R)
NaHS (1 (µM) in
perfusate 10 min
prior to R
20% reduction in infarct size 46
Perfused rat heart
(30 min I/90 min R)
NaHS (1 µM) at
onset of R
Significant decrease in
myocardial infarct size
50
Langendorff hanging
heart model (30 min
I/10 min R)
NaHS (100 µM) in
perfusate prior to I
Decreased duration and
severity of I/R-induced
arrhythmias
37
Isolated rat cardiac
myocytes
NaHS (10–100 µM)
with simulated I
solution
Increased myocyte viability
and shape
37
Langendorff hanging
heart model (40 min
I/2 hr R)
PAG-treated (prior
to I)
38% increase in myocardial
infarct size
41
in vivo (25 min I/2 hr
R)
NaHS (3 mg/kg,
r.j.v.) 15 min prior to
I
26% reduction in myocardial
infarct size
40
in vivo (30 min I/24 hr
R)
Na2S (50 µg/kg, i.c.)
at R
72% reduction in infarct size 39
in vivo (45 min I/24 hr
R)
Na2S (100 µg/kg,
i.v.) 24 hr before I
46% reduction in infarct;
reduction of oxidative stress;
decreased anti-apoptotic
signaling
47
in vivo (60 min I/2 hr
R)
Na2S (100 µg/kg,
i.v.) 10 min before R
2.3-fold reduction in infarct
size in porcine model
48
in vivo (60 min I/2 hr
R)
Na2S (2 mg/kg per
hr, i.v. infusion) at I
significantly decreased the
area of necrosis; higher
expression of cell survival
proteins; decrease apoptosis
49
Brain Primary cultures of
cortical neurons
NaHS (100 µM) with
glutamate
Protects neurons from
glutamate toxicity
38
Cultured
hippocampal HT22
cells
NaHS (300 µM) with
glutamate
Improved survival of HT22
cells
55
Cultured brain
endothelial cells
NaHS (0.05 and 0.1
mM) with
methionine
Attenuated cell death and
radical formation
56
in vivo (24 hr I of
MCA)
NaHS (0.18
mmol/kg, i.p.) 10
min prior I
Increased infarct volume
150%
58
in vivo (90 min I of
MCA)
2 days of H2S gas
(80 ppm)
Reduced infarct size by 50% 42
in utero (5 min I/24 hr
R of BUA)
NaHS (O.4375
µmol/kg, i.p.)
Protects fetal brains by
reinstating the GSH levels
decreased by in utero I/R
59
in vivo (8 min CA
followed by CPR)
Na2S (0.55 mg/kg
f.v.) 1 min before
CPR
Improved neurological
function; decreased
apoptotic proteins and
activation of anti-apoptotic
proteins
60
Liver in vivo (60 min I/5 hr
R)
Na2S (1 mg/kg, i.v.)
5 min prior to R
Reduced AST and ALT
levels, LPO levels, increase
in antioxidant signaling and
decrease in anti-apoptotic
signaling
43
Kidney in vivo (45 min I/72 hr
R)
PAG (1 ml/kg, i.p.)
NaHS (100 µmol/kg)
topically on kidney
PAG decreased renal
function; NaHS treatment
increased renal function
44
in vivo (45 min I/6 hr
R)
-- Ischemia decreased CBS
activity and H2S levels
45
Lung in vivo (45 min I/ 45
min R of PA)
H2S gas (50, 100
µmol/l) 5 min prior to
I
Pulmonary protecion;
decreased histological injury;
increased perfusion flow rate;
lowered lung wet/dry ratio;
improved lung compliance;
lowered MDA levels
62
*

I = Ischemia; R = Reperfusion; CA = Cardiac Arrest; CPR = Cardiopulmonary Resuscitation; MCA = Middle Cerebral Artery; BUA = Bilateral Uteroovarian Artery; PA = Pulmonary Artery; LV = left ventricle;

**

r.j.v. = right jugular vein; i.c. = intracardiac; i.v. = intravenous; i.p. = intraperitoneal; f.v. = femoral venous

3.1 Cardioprotective Effects of H2S

A number of studies have demonstrated the cytoprotective effects of H2S in myocardial I/R (MI/R) injury [37,3941,4650]. The first study to show cytoprotection against MI/R injury was Johansen et al [46] who examined the hypothesis that the protective actions of H2S are mediated by ATP-sensitive potassium channel (KATP) opening. In this study, the authors performed a dose response study using the Lagendorff hanging heart model with rat hearts. They observed an approximate 20% reduction in infarct size when treatment with NaHS (1 µM) was started 10 minutes prior to reperfusion and maintained until 10 min of reperfusion. Using KATP blockers they also provide evidence to support the involvement of KATP channel opening as the mechanism of action. Another study executed by Bian et al [37] examined the effect of endogenous H2S and exogenous application of NaHS on cardiac rhythm in isolated rat hearts subjected to low-flow ischemia. In this study, NaHS was administered prior to ischemia at a concentration of 100 µM in the perfusate (3 min/each cycle separated by 5 min of recovery). The hearts were then subjected to 30 min of low-flow ischemia followed by 10 min of reperfusion. They found that the NaHS-treated group had a significantly decreased duration and severity of I/R-induced arrhythmias. Isolated rat cardiac myocytes were also investigated using a simulated ischemia solution [i.e., glucose-free Krebs buffer containing 10 mM 2-deoxy-D-glucose (2-DOG) and 10 mM sodium dithionite (Na2S2O4)] and were found to have a significantly increased viability and cell shape when treated with NaHS (10–100 µM). Bliksoen et al. [41] examined the protective effects of H2S in global I/R in the heart, the mechanistic role of H2S in ischemic preconditioning, and the involvement of protein kinase phosphorylation with H2S. In this study, the authors blocked H2S production from CSE by using D,L-propargylglycine (PAG) in a Lagendorff perfusion model. A 38% increase in myocardial infarct size was observed after 40 min of low flow ischemia in the PAG-treated (prior to ischemia) hearts when compared to the control hearts. The dramatic increase in infarct size indicates that endogenous H2S plays a key role in protecting the heart from MI/R injury. Evidence supporting cardioprotection of both exogenous and endogenous H2S has also been observed in vivo. Sivarajah et al. [40] were the first to show that endogenous H2S could be protective in myocardial injury. In this study mice were subjected to 25 min of regional myocardial ischemia followed by 2 hr of reperfusion. H2S was administered as NaHS (3 mg/kg) 15 min prior to ischemia. A 26% reduction in infarct size was observed in the treated group when compared to the vehicle. Shortly after, Elrod et al. [39] investigated the potential of H2S as a cardioprotective agent when given at the time of reperfusion. In this study mice subjected to 30 min of left coronary artery (LCA) ischemia and 24 hr reperfusion displayed a 72% reduction in infarct size when administered H2S in the form of Na2S (50 µg/kg) at the time of reperfusion. Elrod et al. also tested the cardioprotective effects of endogenous H2S by subjecting mice with a cardiac-specific over expression of CGL (about a 2 fold increase in myocardial H2S) to MI/R. After 45 min of left coronary artery occlusion and 72 hr reperfusion, the CGL transgenic mice displayed a 47% reduction in infarct size. This implies that increasing the production of endogenous H2S can have similar cardioprotective effects as exogenous H2S. Intriguingly inhibition of the endogenous production of H2S has also been shown to cause hypertension and diminish endothelium-dependent vasodilatation [51] as well as exacerbates MI/R injury in mice [52]. These in vitro, ex vivo, and in vivo studies clearly show that exogenous and endogenous H2S have cardioprotective effects and provide evidence that H2S may be a potential therapeutic agent for the treatment of cardiovascular disease.

3.2 Neuroprotective Effects of H2S

It has been reported that oxidative stress associated with I/R injury causes major damage to neurons [53]. An in vitro study of the efficacy of H2S protection against oxidative stress in neurons was performed using glutamate (1mM) and demonstrated that NaHS (100 µM) protects neurons from glutamate toxicity [38]. Kimura et al [54] also suggested that H2S may not itself function as an antioxidant in neuronal cells, but instead induces the production of the potent antioxidant glutathione (GSH). Another study on the neuroprotective role of H2S was completed on a clonal hippocampal nerve cell line, HT22, using a model of oxidative stress [55]. HT22 cells exposed to glutamate (5 mM) that were treated with NaHS (10–300 µM) showed improved survival over untreated cells. Furthermore, Tyagi et al [56] investigated the effects of H2S on methionine (Met)-induced oxidative stress in mouse brain endothelial cells (bEnd3). They found that pre-treatment with NaHS (0.05 and 0.1mM) attenuated cell death as well as peroxynitrite and superoxide anion formation in cells treated with Met (1.14 mM). Further in vivo studies have revealed additional roles of H2S in the brain. For example, an elevated level of one of the cerebral substrates of H2S, plasma homocysteine (Hcy), is a known risk factor for acute stroke [57]. Qu et al. [58] tested the effect of H2S on the development of infarction in rats using a focal cerebral ischemic stroke model by inducing a permanent unilateral occlusion of the left middle cerebral artery (MCA) and found that administration of NaHS (0.18 mmol/kg) 10 min prior to the occlusion increased the infarct volume by approximately 150% compared to the control. Additionally, inhibition of the H2S producing enzymes CBS and CSE using pharmacological inhibitors (in order of potency: aminooxyacetic acid [AOAA], hydroxylamine [HA], PAG, β-cyanoalanine [β-CNA]) significantly reduced infarct volume. However more recent studies have shown a protective role for H2S in vivo at much lower concentrations. Florian et al [42] examined the effects of H2S administered after cerebral ischemia by exposing rats to 2 days of H2S gas (80 ppm H2S in 19.5% O2). The exposure of H2S increased the concentration of H2S in the brain from 1.12 ± 4.6 µg/g to 2.70 ± 5.3 µg/g, induced a state of hypothermia (30.8 ± 0.7 °C) and reduced infarct size by 50%. Kimura et al [59] studied the effect of H2S on GSH levels in the brains of fetal mice using an intrauterine I/R (5 min/24 hr) model. Intrauterine I/R caused a 24% decrease in GSH levels. However, mice treated with NaHS (0.4375 µmol/kg, i.p.) 15 min before fetal ischemia showed improved fetal brain levels of GSH compared to the untreated mice, suggesting that H2S protects by reinstating GSH levels that were lowered during intrauterine I/R. Minamishima et al [60] used a murine model to study the neuroprotective effects of H2S (Na2S, 0.55 mg/kg) after cardiac arrest/cardiopulmonary resuscitation (CA/CPR). They observed improved neurological function, decreased activation of the apoptotic protein caspase-3 in the hippocampus, and enhanced phosphorylation (activation) of the anti-apoptotic protein GSK-3β in the brain cortex in mice injected with Na2S via a femoral venous line 1 min prior to CPR.

The above studies reveal a variety of neuroprotective roles of H2S. In vitro and in vivo studies demonstrate that low concentrations of H2S can mediate oxidative stress by restoring GSH levels in the brain, whereas higher concentrations of H2S have been shown to have effects that range from magnifying cerebral damage in the ischemic stoke model to inducing a state of neuroprotective. Studies aimed at investigating the role of 3MST in cerebral I/R injury could potentially elucidate the role H2S plays in modulating cerebral ischemia.

3.3 H2S and Hepatoprotection

The cytoprotective effects of H2S can also be seen in the liver. Jha et al [43] tested the hepatoprotective effects of H2S in a murine model of hepatic I/R by subjecting mice to 60 min of ischemia followed by 5 hr of reperfusion. Na2S was administered intravenously 5 min before reperfusion and serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels were measured to quantify liver injury. Na2S treatments reduced AST levels by 71% and ALT levels by 69% suggesting that H2S attenuates hepatic I/R injury. The authors also demonstrated that the hepatoprotective effects of H2S were mediated by a decrease in lipid peroxidation levels, an increase in antioxidant signaling, and an increase in anti-apoptotic signaling. While this study provides evidence for exogenous H2S therapy it does not address the ability of endogenous H2S to affect hepatic injury. Therefore additional studies are warranted to fully elucidate the hepatoprotective effects of H2S.

3.4 H2S and Renal Ischemia

There has been a collection of studies that suggest exogenous and endogenous H2S is protective against renal I/R injury. Tripatara et al [44] investigated the effects of endogenous and exogenous H2S in renal I/R. They treated rats with PAG (1ml/kg, i.p.) 1 hr before ischemia to inhibit CSE and found that animals displayed increased serum creatine levels signifying a decrease in renal function. Next they performed a bilateral renal occlusion, this time treating the mice with NaHS topically on the kidneys 15 prior to ischemia. A significant increase in renal function was observed, indicating that H2S plays a key role in protection against renal ischemia. Xu et al. [45] examined if renal injury was caused by the reduction in activity of CBS in the kidney during I/R by measuring H2S and creatine levels after 45 min of renal ischemia and 6 hr of reperfusion. A significant decrease of H2S and an increase in plasma creatine levels were observed in the rats subjected to the ischemia, indicating that during ischemia, H2S levels drop along with kidney function. The activity of CBS and CGL were also measured in the rats after ischemia; CBS activity levels were significantly decreased while CGL activity was not significantly changed. These data suggest that not only does H2S play a role in renal ischemia, but also a major enzyme responsible for H2S production is inhibited. Moreover, because CBS is being inhibited, there is an accumulation of Hcy causing a worsening in renal I/R injury [61].

3.5 H2S and Pulmonary Ischemia

Only a small number of studies have investigated the role of H2S in pulmonary I/R injury. Fu et al [62] explored the potential of H2S as a lung protective agent. They measured endogenous H2S generation in the rat lung under I/R injury and found that ischemia caused a 46% increase in CSE activity resulting in a 54% increase in H2S levels. They also perfused the rat lungs with H2S gas (50 µmol/l, 100 µmol/l) 5 min prior to ischemia and found that the pretreated mice displayed pulmonary protection as assessed by lung morphology. H2S also decreased I/R-induced lung histological injury, increased lung perfusion flow rate, lowered lung wet/dry weight ratio, and improved lung compliance. Malondialdehyde (MDA) levels were also measured to determine the amount of oxidation in the cells and it was found that H2S had significantly lowered MDA levels, suggesting that H2S attenuated oxidative stress in the lung.

3.6 Limitations of Pharmacological Inhibitors of H2S Producing Enzymes

Although multiple pharmacological inhibitors of H2S biosynthesis are available, the potency, selectivity, and permeability of the compounds could be challenging when using these inhibitors to suppress a particular H2S producing enzyme. PAG and β-CNA are commonly used to inhibit CSE. Unfortunately, they have low potency (25–100 mg/kg) and selectivity in most organs with limited cell membrane permeability [63]. Also, AOAA and HA frequently used inhibitors of CBS, target the pyridoxal-binding site of the enzyme and therefore have the potential of affecting other enzymes in biological systems [64]. Lastly, hydrogen peroxide and tetrathionate are used to inhibit 3MST by interfering with its catalytic cysteine residue. However, many enzymes use cysteines in the catalytic sites and would thus be inhibited by these chemicals [65]. Therefore, there is much room for the development of more specific and potent inhibitors of H2S producing enzymes in which could aid in expanding the field’s understanding of the endogenous role of H2S.

4. Mechanisms of Action and the Diverse Physiological Profile of H2S

It is shown above that H2S is protective in many biological systems. The mechanisms through which H2S can protect these systems are important to understand in order to utilize its effectiveness as a cytoprotection agent. H2S has a wide range of physiological roles in mammalian tissue. In the nervous system, H2S has been shown to function as a neuromodulator [29], modulate NMDA receptors (NMDAR) by inducing the production of cyclic-adenosine monophosphate (cAMP) [66], and act as an inhibitor of peroxynitrite (ONOO) [67]. In the cardiovascular system, numerous roles for H2S have been identified: vasorelaxant [68] and antiapoptotic [69] properties by opening of KATP channels, proangiogenic factor through the phosphorylation of Akt [70], modulator of leukocyte-mediated inflammation [71], upregulator of antioxidant signaling [43], and involved in cytoprotection through the preservation of mitochondrial function [39].

4.1 Katp Channel – Vasodilation or Calcium Handling

KATP channels are located on the sarcolemma [72], the inner mitochondrial membrane [73], and nuclear membrane [74] and play an important role in glucose metabolism in the cell by membrane hyperpolarization. KATP channels are composed of two types of subunits: four inwardly rectifying potassium channels Kir6 (Kir6.1, Kir6.2) and four sulfonylurea receptors (SUR1, SUR2A, SUR2B) [75]. Kir6 subunits form the pore of the channel and are capable of opening and closing to regulate ion flow [73]. SUR receptors are a high affinity receptor sensitive to [ATP/ADP] that facilitates the opening/closing of the Kir6 subunits [76] and are the target of many pharmacological compounds. It has been proposed that H2S-stimulated vasorelaxation occurs through the opening of the KATP channel [68]. Zhao et al [68] attempted to assess the physiological role of H2S in the regulation of vascular contractility, the modulation of H2S production in vascular tissues, and the underlying mechanism involved. To achieve this, Zhao et al. gave an intravenous bolus injection of NaHS (2.8 and 14 µmol/kg) to rats and observed a decrease in mean arterial blood pressure similar to an injection of pinacidil (2.8 µmol/kg), a known KATP channel opener. Supporting this hypothesis, H2S has also been shown to increase KATP channel currents in isolated smooth muscle cells [77]. Cardioprotection, however, may be limited only to specific KATP channels. In an in vitro study, Bian et al. [37] used the mitochondrial KATP blocker 5-hydroxydeconoate (5-HD) to demonstrate that mitochondrial KATP channels were not responsible for the cardioprotective effects of H2S. However, they found that inhibition of the sarcolemmal KATP channels took away the cardioprotective effects of H2S. Sivarajah et al. [69] recently performed an in vivo MI/R study to investigate the effect NaHS on infarct size and apoptosis caused by ischemia (25 min) and reperfusion (2 hr). Sivarajah et al. inhibited the mitochondrial KATP channel with 5-HD and found, contradictory to Bian et al., that H2S did not protect the heart when the mitochondrial channel blocker was applied intravenously. These results suggests a new method in studying mitochondrial KATP channels must be developed to obtain more consistent results before any conclusions can be taken from these experiments.

In MI/R models, Ca2+ handling is critical in controlling cellular damage. During ischemia, excess Ca2+ and Na+ builds up in the cytosol of the cell [78]. Due to the large [Ca2+] in the cell at the onset of reperfusion, the reoxygenation of the cell causes myofibrils to generate a large amount of force, called hypercontraction [79]. The excessive force irreparably damages the cytoskeleton. Therefore, Ca2+ handling is essential in controlling reperfusion damage in the cell. Protein kinase C (PKC) has been shown to be involved with Ca2+ handling and protects against I/R damage. There are three isoforms of PKC in the heart [80]: PKCα, PKCε, and PKCδ. Recently, Pan et al. [81] examined the regulatory effect of PKC on intracellular Ca2+ handling in H2S-preconditioning and demonstrated that H2S preconditioning in rat myocytes activates all three isoforms of PKC, lowers intracellular [Ca2+] and protects the cardiomyocytes from damage. However an in vivo model of H2S and PKC activation during I/R should be completed to solidify their roles in mediating intracellular [Ca2+].

4.2 H2S and Apoptosis

The mechanism behind the cardioprotective effects of H2S is not limited to modulation of KATP channels and Ca2+ handling. There is much evidence to suggest that H2S also has anti-apoptotic roles in the cell during M/IR. The activation of two important cell survival pathways, extracellular signal-regulated kinase (ERK1/2)/mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI-3-kinase), can inactivate pro-apoptotic pathways through interaction with PKCε [82] or inhibition of Bad and caspase-9 [83], respectively. The activation of the ERK1/2 and PI-3-kinase pathways has been found to be influenced by H2S [84,85]. Additionally, activation of the PI-3-kinase/Akt pathway increases the production of the cardioprotective gasotransmitter NO [86]. In an attempt to elucidate the signaling mechanism of H2S in the ERK1/2 pathway during cardioprotection, Hu et al [85] found that preconditioning rat myocytes with NaHS (1–100 µM) not only increased cell viability, but that the blockade of ERK1/2 or Akt during preconditioning or ischemia significantly decreased the cardioprotection of H2S. Western blot analysis also revealed that H2S preconditioning increased the phosphorylation status of ERK1/2 and Akt, further activating the protein’s pro-survival mechanisms. Elrod et al [39] performed an in vitro experiment with isolated adult myocytes, which were subjected to 6 hr of hypoxia and 12 hr reoxygenation to further examine the role of H2S on apoptotic pathways. They found that myocytes treated with NaHS displayed a decreased activation of caspase-3 and a decrease in the number of nuclei with fragmented DNA (a result of apoptotic signaling cascades). These data suggest that H2S inhibited the progression of apoptosis after MI/R injury. Additionally, Sivarajah et al [69] demonstrated that regional MI/R on rat hearts increased the phosphorylation of p38, MAPK, and JNK1/2, pro-apoptotic proteins in the heart, but the administration of NaHS 15 min prior to ischemia significantly reduced the phosphorylation of p38, MAPK, and JNK caused by I/R injury. Cytochrome C, another pro-apoptotic protein, is an essential part of the electron transport chain, but when subjected to pro-apoptotic stimuli, it transports out of the mitochondria into the cytosol. There it activates other apoptotic proteins, including caspase-3, an activator of apoptosis [87]. To determine the effect of H2S on the cytochrome C apoptotic pathway during I/R injury, Calvert et al [47] performed a study where mice were given either saline (vehicle) or Na2S 24 hr prior to ischemia. After 45 min of ischemia and 24 hr of reperfusion, the tissue isolated from the vehicle treated mice hearts, when compared to sham operated mice, had a significant decrease in the expression of uncleaved caspase-3 (inactive) and a significant increase in the expression of cleaved caspase-3 (active), as well as an induced translocation of cytochrome C from the mitochondria to the cytosol. The mice treated with Na2S exhibited a preservation of uncleaved caspase-3, a reduction in cleaved caspase-3, and a reduction in the translocation of cytochrome C to the cytosol (compared to the vehicle treated mice). Therefore, Na2S protects the heart tissue from undergoing apoptotic signaling during MI/R.

The Signal Transducers and Activators of Transcription (STAT) pathway has also been shown to be an important part of the myocardium response to myocardial infarction [88] and an overexpression of the STAT isoform STAT-3 has been shown to be cardioprotective [89]. Inhibition of STAT-3 has also been shown to increase caspase-3 expression in the heart, thereby increasing I/R injury [90]. Calvert et al demonstrated in vivo that H2S activates PKCε and STAT-3 by altering their phosphorylation, inhibiting the proapoptotic factor Bad, upregulating pro-survival factors Bcl-2 and Bcl-xL, and upregulating heat shock proteins (HSPs) [47]. Therefore, evidence suggests H2S has many anti-apoptotic roles in MI/R.

4.3 H2S and Mitochondrial Protection

The life of a cell is dependent on the degree of mitochondrial functionality. During I/R, mitochondria are subjected to oxygen deprivation, reactive oxygen species (ROS) overproduction, and mitochondrial membrane potential (ΔΨm) depolarization [91]. Roth et al [92] have shown that H2S at high levels (80 ppm) can induce a state of hypothermia in mice by inhibiting cytochrome c oxidase, in turn decreasing their metabolic rate and core body temperature. They also showed that inducing this “suspended animation” state can prevent ischemic damage to cells. Elrod et al [39] evaluated potential mechanisms of H2S-mediated mitochondrial preservation using an in vitro model of Na2S treatment. They found that isolated mitochondria, after 30 min of hypoxia, treated with Na2S (10 µM) had a greater recovery of posthypoxic respiration rate. Similarly mice that were given Na2S at reperfusion displayed a reduction in mitochondrial swelling and an increase in matrix density suggesting preservation in mitochondrial function.

Under myocardial ischemia, the production of ROS is accelerated and all of the cell’s antioxidants become depleted. H2S is a cytochrome C oxidase inhibitor and therefore inhibits respiration [93]. Inhibition of respiration has been shown to decrease the production of ROS thereby maintaining ΔΨm [94]. Thus H2S, at low concentrations, can decrease the production of ROS and preserve mitochondrial function [52,95]. Another factor concerning cellular injury is Ca2+ influx. The Ca2+ uniporter is the main pathway for Ca2+ influx [96] during myocardial ischemia. Pan et al [81] demonstrated that H2S activates PKC and lowers intracellular [Ca2+] in cardiomyocytes protecting the cells from injury. H2S also has the ability to activate other pathways. The PI3K-Akt and Erk 1/2 pro-survival kinase cascades (Reperfusion Injury Salvage Kinase, RISK pathway) are activated during I/R and launches anti-apoptotic responses [97]. A major target of the RISK pathway is the mitochondrial permeability transition pore (mPTP) [97]. Opening of the mPTP occurs during reperfusion and is a terminal cellular event contributing to myocardial injury [96]. H2S can overactivate Akt, a major kinase in the RISK pathway [85]. Akt then activates eNOS which evidence suggests inhibits the opening of mPTP [97]. Therefore, H2S acts to preserve mitochondrial function, thereby imparting cytoprotection.

4.4 H2S and Inflammation

Inflammatory responses are an important part of early I/R injury [98]. Li et al [99] investigated the effects of a H2S donor, S-diclofenac, on anti-inflammatory activity by administering the proinflammatory bacterial endotoxin lipopolysaccharide (LPS, 10 mg/kg intraperitoneally) to rats, with a group being treated with S-diclofenac (47.2 µmol/kg, i.p.) 60 min prior to LPS injection. They found that the H2S donor significantly reduced the rise in plasma concentrations of proinflammatory interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) while also augmenting the rise in plasma concentrations of the anti-inflammatory cytokine interleukin-10 (IL-10). Other studies have investigated the pathways that may be involved in the anti-inflammatory effects of H2S. After I/R, early inflammatory effects are derived from leukocyte adhesion to endothelial cells [100]. Zanardo et al [71] examined H2S as an endogenous regulator of leukocyte-endothelial interactions. They pretreated rats intragastrically with Na2S (1–100 µmol/kg) or NaHS (100 µmol/kg) 30 min before administering the leukocyte adhering aspirin and Formyl-Methionyl-Leucyl-Phenylalanine (fMLP). They found that rats treated with H2S dose-dependly decreased leukocyte adherence to endothelial cells. Additionally, Zanardo et al revealed that the rats pretreated with the KATP channel antagonist glibenclamide did not have a decrease in leukocyte adherence suggesting that the anti-inflammatory effects of H2S was through KATP channel activation. Yusof et al [101] studied the effects of preconditioning with H2S on leukocyte rolling (2nd step in leukocyte-endothelial interactions during ischemia [102]) and adhesion induced by ischemia. They injected NaHS (14 µmol/kg, i.p.) 24 hr before ischemia, 1 hr before ischemia, and at reperfusion in mice receiving 45 min of superior mesenteric artery ischemia and 60 min reperfusion. They observed that H2S initiated late-phase preconditioning (tolerance to ischemia 12–24 hr after preconditioning stimulus) and through Western blot analysis revealed a significant increase in phosphorylation (activation) of eNOS and p38 MAPK. These data suggests that the anti-inflammatory effects of preconditioning with H2S are eNOS and p38 MAPK pathway dependent since the protective roles of eNOS and p38 MAPK have already been established [103,104]. More mechanistic studies of H2S in inflammation could provide insight into preventing inflammatory tissue damage after I/R.

5. Summary and Perspective concerning H2S in Ischemic Disorders

Whether it is endogenous or exogenous, H2S has a wide range of protective functions after I/R injury throughout the body. Mechanisms of protection include control of intracellular [Ca2+] by stimulating KATP channel opening, anti-apoptotic pathway (ERK1/2/MAPK, PI-3 kinase/Akt, JAK-STAT) activation, mitochondrial protection through preservation of ΔΨm and inhibition of mPTP opening, and antiinflammatory effects by activation of eNOS and p38 MAPK. Since the discovery of the cytoprotective effects of H2S, developing methods to up-regulate endogenous H2S production or provide efficacious treatments of H2S donors is becoming a focus of scientists and clinicians alike. Ikaria Holdings Incorporated has two clinical trials beginning based on an H2S drug, IK-1001 (Na2S). The first ongoing study is the reduction of I/R mediated cardiac injury in patients undergoing coronary artery bypass graft surgery (CABG) to evaluate the capability of IK-1001 as a robust treatment for abbreviating surgical injury. Patients undergoing surgery will receive intravenous infusion for six hours while having the CABG surgery and will be evaluated six months later. The other clinical trial by Ikaria Holdings Inc. that is beginning is a pharmokinetic assessment of Na2S in patients with impaired renal function. In this study patients with healthy, mild, and moderately impaired renal function will be injected intravenously with IK-1001 (1.5 mg/kg/hr) for 3 hours while patients with severely impaired renal function will receive IK-1001 (1.0 mg/kg/hr) for 3 hours intravenously. The patients will then be followed over a seven-day period to investigate any restoration in renal function.

Though H2S shows therapeutic potential in I/R injury, an underlying problem with the H2S field is that the conventional H2S donors, NaHS and Na2S, are short acting and do not give a continuous release of H2S. Recent studies are suggesting that garlic derivatives could provide an answer to this problem. Garlic has been used for prevention and treatment of atherosclerosis, hyperlipidemia, thrombosis, hypertension, and diabetes [105]. Garlic contains a number of sulfur rich chemical compounds including diallyl disulfide (DADS) and diallyl trisulfide (DATS). Benavides et al [106] conducted a study that demonstrated that DADS and DATS induced H2S production and are responsible for the vasoactivity of garlic. However, exhaustive studies demonstrating the protective effects of DADS and DATS in I/R injury have yet to be published.

Although the physiological and cytoprotective effects of H2S have been examined, the mechanisms involved are not fully understood. In the past 20 years, H2S has come from a reputation as a toxic gas to potentially a therapeutic drug for the treatment I/R injury. Recent research has revealed many beneficial roles for H2S in ischemia for a variety of tissues. For example, H2S has been shown to be cardioprotective through both endogenous and exogenous applications at or prior to ischemia, neuroprotective via H2S-induced hypothermia, hepatoprotective by decreasing lipid peroxidation, increasing antioxidant and anti-apoptotic signaling, renal protective through the actions of CBS, and protective against pulmonary ischemia injury by the activity of CSE. Although much progress has been made in the H2S field regarding its physiological roles in different organ systems, there is room still available for more advancement to be made. First, the enzymes responsible for the endogenous production of H2S (CBS, CGL, and 3MST) need to be investigated further for other functions and regulatory mechanisms they may have inside biological systems. Knowing an enzyme’s specific role and localization in each individual organ system will allow for the targeting of that enzyme to modify endogenous concentrations of H2S suitable for the treatment of I/R injury in that tissue. Second, improvement in H2S donors could allow for treatments to mimic the endogenous protective effects that H2S has in the body. As a result, traditional treatments aimed at combating I/R injury could be challenged with the development of new therapeutic strategies designed capitalize on the potential protective effects of endogenous and exogenous H2S.

Acknowledgement

Supported by grants from the American Diabetes Association (7-09-BS-26) and the National Institutes of Health National Heart Lung and Blood Institute (NHLBI) 1R01HL098481-01 to J.W.C. This work was also supported by funding from the Carlyle Fraser Heart Center (CFHC) of Emory University Hospital Midtown.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Author’s Disclosure

None

References

  • 1.Wang R. Two's company, three's a crowd: Can h2s be the third endogenous gaseous transmitter? FASEB J. 2002;16:1792–1798. doi: 10.1096/fj.02-0211hyp. [DOI] [PubMed] [Google Scholar]
  • 2.Wang R. The gasotransmitter role of hydrogen sulfide. Antioxid Redox Signal. 2003;5:493–501. doi: 10.1089/152308603768295249. [DOI] [PubMed] [Google Scholar]
  • 3.Furchgott RF, Zawadzki JV. The obligatory role of endothelial cells in the relaxation of arterial smooth muscle by acetylcholine. Nature. 1980;288:373–376. doi: 10.1038/288373a0. [DOI] [PubMed] [Google Scholar]
  • 4.Hibbs JB, Jr, Vavrin Z, Taintor RR. L-arginine is required for expression of the activated macrophage effector mechanism causing selective metabolic inhibition in target cells. J Immunol. 1987;138:550–565. [PubMed] [Google Scholar]
  • 5.Michel T, Feron O. Nitric oxide synthases: Which, where, how, and why? J Clin Invest. 1997;100:2146–2152. doi: 10.1172/JCI119750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Stagliano NE, Dietrich WD, Prado R, Green EJ, Busto R. The role of nitric oxide in the pathophysiology of thromboembolic stroke in the rat. Brain Res. 1997;759:32–40. doi: 10.1016/s0006-8993(97)00200-x. [DOI] [PubMed] [Google Scholar]
  • 7.Xie QW, Cho HJ, Calaycay J, Mumford RA, Swiderek KM, Lee TD, Ding A, Troso T, Nathan C. Cloning and characterization of inducible nitric oxide synthase from mouse macrophages. Science. 1992;256:225–228. doi: 10.1126/science.1373522. [DOI] [PubMed] [Google Scholar]
  • 8.Lamas S, Marsden PA, Li GK, Tempst P, Michel T. Endothelial nitric oxide synthase: Molecular cloning and characterization of a distinct constitutive enzyme isoform. Proc Natl Acad Sci U S A. 1992;89:6348–6352. doi: 10.1073/pnas.89.14.6348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hurt KJ, Musicki B, Palese MA, Crone JK, Becker RE, Moriarity JL, Snyder SH, Burnett AL. Akt-dependent phosphorylation of endothelial nitric-oxide synthase mediates penile erection. Proc Natl Acad Sci U S A. 2002;99:4061–4066. doi: 10.1073/pnas.052712499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Dudzinski DM, Igarashi J, Greif D, Michel T. The regulation and pharmacology of endothelial nitric oxide synthase. Annu Rev Pharmacol Toxicol. 2006;46:235–276. doi: 10.1146/annurev.pharmtox.44.101802.121844. [DOI] [PubMed] [Google Scholar]
  • 11.Siegfried MR, Carey C, Ma XL, Lefer AM. Beneficial effects of spm-5185, a cysteine-containing no donor in myocardial ischemia-reperfusion. Am J Physiol. 1992;263:H771–H777. doi: 10.1152/ajpheart.1992.263.3.H771. [DOI] [PubMed] [Google Scholar]
  • 12.Hataishi R, Rodrigues AC, Neilan TG, Morgan JG, Buys E, Shiva S, Tambouret R, Jassal DS, Raher MJ, Furutani E, Ichinose F, Gladwin MT, Rosenzweig A, Zapol WM, Picard MH, Bloch KD, Scherrer-Crosbie M. Inhaled nitric oxide decreases infarction size and improves left ventricular function in a murine model of myocardial ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. 2006;291:H379–H384. doi: 10.1152/ajpheart.01172.2005. [DOI] [PubMed] [Google Scholar]
  • 13.Heurteaux C, Lauritzen I, Widmann C, Lazdunski M. Essential role of adenosine, adenosine a1 receptors, and atp-sensitive k+ channels in cerebral ischemic preconditioning. Proc Natl Acad Sci U S A. 1995;92:4666–4670. doi: 10.1073/pnas.92.10.4666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Hotter G, Closa D, Prados M, Fernandez-Cruz L, Prats N, Gelpi E, Rosello-Catafau J. Intestinal preconditioning is mediated by a transient increase in nitric oxide. Biochem Biophys Res Commun. 1996;222:27–32. doi: 10.1006/bbrc.1996.0692. [DOI] [PubMed] [Google Scholar]
  • 15.Peralta C, Hotter G, Closa D, Gelpi E, Bulbena O, Rosello-Catafau J. Protective effect of preconditioning on the injury associated to hepatic ischemia-reperfusion in the rat: Role of nitric oxide and adenosine. Hepatology. 1997;25:934–937. doi: 10.1002/hep.510250424. [DOI] [PubMed] [Google Scholar]
  • 16.Kikuchi G, Yoshida T, Noguchi M. Heme oxygenase and heme degradation. Biochem Biophys Res Commun. 2005;338:558–567. doi: 10.1016/j.bbrc.2005.08.020. [DOI] [PubMed] [Google Scholar]
  • 17.Hangaishi M, Ishizaka N, Aizawa T, Kurihara Y, Taguchi J, Nagai R, Kimura S, Ohno M. Induction of heme oxygenase-1 can act protectively against cardiac ischemia/reperfusion in vivo. Biochem Biophys Res Commun. 2000;279:582–588. doi: 10.1006/bbrc.2000.3973. [DOI] [PubMed] [Google Scholar]
  • 18.Yet SF, Tian R, Layne MD, Wang ZY, Maemura K, Solovyeva M, Ith B, Melo LG, Zhang L, Ingwall JS, Dzau VJ, Lee ME, Perrella MA. Cardiac-specific expression of heme oxygenase-1 protects against ischemia and reperfusion injury in transgenic mice. Circ Res. 2001;89:168–173. doi: 10.1161/hh1401.093314. [DOI] [PubMed] [Google Scholar]
  • 19.Kharitonov VG, Sharma VS, Pilz RB, Magde D, Koesling D. Basis of guanylate cyclase activation by carbon monoxide. Proc Natl Acad Sci U S A. 1995;92:2568–2571. doi: 10.1073/pnas.92.7.2568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Wang R, Wu L. The chemical modification of kca channels by carbon monoxide in vascular smooth muscle cells. J Biol Chem. 1997;272:8222–8226. doi: 10.1074/jbc.272.13.8222. [DOI] [PubMed] [Google Scholar]
  • 21.Johnson EE, Srikanth CV, Sandgren A, Harrington L, Trebicka E, Wang L, Borregaard N, Murray M, Cherayil BJ. Siderocalin inhibits the intracellular replication of mycobacterium tuberculosis in macrophages. FEMS Immunol Med Microbiol. 2010;58:138–145. doi: 10.1111/j.1574-695X.2009.00622.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Christou H, Morita T, Hsieh CM, Koike H, Arkonac B, Perrella MA, Kourembanas S. Prevention of hypoxia-induced pulmonary hypertension by enhancement of endogenous heme oxygenase-1 in the rat. Circ Res. 2000;86:1224–1229. doi: 10.1161/01.res.86.12.1224. [DOI] [PubMed] [Google Scholar]
  • 23.Wiesel P, Patel AP, Carvajal IM, Wang ZY, Pellacani A, Maemura K, DiFonzo N, Rennke HG, Layne MD, Yet SF, Lee ME, Perrella MA. Exacerbation of chronic renovascular hypertension and acute renal failure in heme oxygenase-1-deficient mice. Circ Res. 2001;88:1088–1094. doi: 10.1161/hh1001.091521. [DOI] [PubMed] [Google Scholar]
  • 24.Ishikawa K, Sugawara D, Wang X, Suzuki K, Itabe H, Maruyama Y, Lusis AJ. Heme oxygenase-1 inhibits atherosclerotic lesion formation in ldl-receptor knockout mice. Circ Res. 2001;88:506–512. doi: 10.1161/01.res.88.5.506. [DOI] [PubMed] [Google Scholar]
  • 25.Sato K, Balla J, Otterbein L, Smith RN, Brouard S, Lin Y, Csizmadia E, Sevigny J, Robson SC, Vercellotti G, Choi AM, Bach FH, Soares MP. Carbon monoxide generated by heme oxygenase-1 suppresses the rejection of mouse-to-rat cardiac transplants. J Immunol. 2001;166:4185–4194. doi: 10.4049/jimmunol.166.6.4185. [DOI] [PubMed] [Google Scholar]
  • 26.Tulis DA, Durante W, Peyton KJ, Evans AJ, Schafer AI. Heme oxygenase-1 attenuates vascular remodeling following balloon injury in rat carotid arteries. Atherosclerosis. 2001;155:113–122. doi: 10.1016/s0021-9150(00)00552-9. [DOI] [PubMed] [Google Scholar]
  • 27.Warenycia MW, Goodwin LR, Benishin CG, Reiffenstein RJ, Francom DM, Taylor JD, Dieken FP. Acute hydrogen sulfide poisoning Demonstration of selective uptake of sulfide by the brainstem by measurement of brain sulfide levels. Biochem Pharmacol. 1989;38:973–981. doi: 10.1016/0006-2952(89)90288-8. [DOI] [PubMed] [Google Scholar]
  • 28.Skrajny B, Hannah RS, Roth SH. Low concentrations of hydrogen sulphide alter monoamine levels in the developing rat central nervous system. Can J Physiol Pharmacol. 1992;70:1515–1518. doi: 10.1139/y92-215. [DOI] [PubMed] [Google Scholar]
  • 29.Abe K, Kimura H. The possible role of hydrogen sulfide as an endogenous neuromodulator. J Neurosci. 1996;16:1066–1071. doi: 10.1523/JNEUROSCI.16-03-01066.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hosoki R, Matsuki N, Kimura H. The possible role of hydrogen sulfide as an endogenous smooth muscle relaxant in synergy with nitric oxide. Biochem Biophys Res Commun. 1997;237:527–531. doi: 10.1006/bbrc.1997.6878. [DOI] [PubMed] [Google Scholar]
  • 31.Eto K, Ogasawara M, Umemura K, Nagai Y, Kimura H. Hydrogen sulfide is produced in response to neuronal excitation. J Neurosci. 2002;22:3386–3391. doi: 10.1523/JNEUROSCI.22-09-03386.2002. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 32.Shibuya N, Tanaka M, Yoshida M, Ogasawara Y, Togawa T, Ishii K, Kimura H. 3-mercaptopyruvate sulfurtransferase produces hydrogen sulfide and bound sulfane sulfur in the brain. Antioxid Redox Signal. 2009;11:703–714. doi: 10.1089/ars.2008.2253. [DOI] [PubMed] [Google Scholar]
  • 33.Lowicka E, Beltowski J. Hydrogen sulfide (h2s) - the third gas of interest for pharmacologists. Pharmacol Rep. 2007;59:4–24. [PubMed] [Google Scholar]
  • 34.Belardinelli MC, Chabli A, Chadefaux-Vekemans B, Kamoun P. Urinary sulfur compounds in down syndrome. Clin Chem. 2001;47:1500–1501. [PubMed] [Google Scholar]
  • 35.Furne J, Springfield J, Koenig T, DeMaster E, Levitt MD. Oxidation of hydrogen sulfide and methanethiol to thiosulfate by rat tissues: A specialized function of the colonic mucosa. Biochem Pharmacol. 2001;62:255–259. doi: 10.1016/s0006-2952(01)00657-8. [DOI] [PubMed] [Google Scholar]
  • 36.Beauchamp RO, Jr, Bus JS, Popp JA, Boreiko CJ, Andjelkovich DA. A critical review of the literature on hydrogen sulfide toxicity. Crit Rev Toxicol. 1984;13:25–97. doi: 10.3109/10408448409029321. [DOI] [PubMed] [Google Scholar]
  • 37.Bian JS, Yong QC, Pan TT, Feng ZN, Ali MY, Zhou S, Moore PK. Role of hydrogen sulfide in the cardioprotection caused by ischemic preconditioning in the rat heart and cardiac myocytes. J Pharmacol Exp Ther. 2006;316:670–678. doi: 10.1124/jpet.105.092023. [DOI] [PubMed] [Google Scholar]
  • 38.Kimura Y, Kimura H. Hydrogen sulfide protects neurons from oxidative stress. FASEB J. 2004;18:1165–1167. doi: 10.1096/fj.04-1815fje. [DOI] [PubMed] [Google Scholar]
  • 39.Elrod JW, Calvert JW, Morrison J, Doeller JE, Kraus DW, Tao L, Jiao X, Scalia R, Kiss L, Szabo C, Kimura H, Chow CW, Lefer DJ. Hydrogen sulfide attenuates myocardial ischemia-reperfusion injury by preservation of mitochondrial function. Proc Natl Acad Sci U S A. 2007;104:15560–15565. doi: 10.1073/pnas.0705891104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Sivarajah A, McDonald MC, Thiemermann C. The production of hydrogen sulfide limits myocardial ischemia and reperfusion injury and contributes to the cardioprotective effects of preconditioning with endotoxin, but not ischemia in the rat. Shock. 2006;26:154–161. doi: 10.1097/01.shk.0000225722.56681.64. [DOI] [PubMed] [Google Scholar]
  • 41.Bliksoen M, Kaljusto ML, Vaage J, Stenslokken KO. Effects of hydrogen sulphide on ischaemia-reperfusion injury and ischaemic preconditioning in the isolated, perfused rat heart. Eur J Cardiothorac Surg. 2008;34:344–349. doi: 10.1016/j.ejcts.2008.03.017. [DOI] [PubMed] [Google Scholar]
  • 42.Florian B, Vintilescu R, Balseanu AT, Buga AM, Grisk O, Walker LC, Kessler C, Popa-Wagner A. Long-term hypothermia reduces infarct volume in aged rats after focal ischemia. Neurosci Lett. 2008;438:180–185. doi: 10.1016/j.neulet.2008.04.020. [DOI] [PubMed] [Google Scholar]
  • 43.Jha S, Calvert JW, Duranski MR, Ramachandran A, Lefer DJ. Hydrogen sulfide attenuates hepatic ischemia-reperfusion injury: Role of antioxidant and antiapoptotic signaling. Am J Physiol Heart Circ Physiol. 2008;295:H801–H806. doi: 10.1152/ajpheart.00377.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Tripatara P, Patel NS, Collino M, Gallicchio M, Kieswich J, Castiglia S, Benetti E, Stewart KN, Brown PA, Yaqoob MM, Fantozzi R, Thiemermann C. Generation of endogenous hydrogen sulfide by cystathionine gamma-lyase limits renal ischemia/reperfusion injury and dysfunction. Lab Invest. 2008;88:1038–1048. doi: 10.1038/labinvest.2008.73. [DOI] [PubMed] [Google Scholar]
  • 45.Xu Z, Prathapasinghe G, Wu N, Hwang SY, Siow YL, O K. Ischemia-reperfusion reduces cystathionine-beta-synthase-mediated hydrogen sulfide generation in the kidney. Am J Physiol Renal Physiol. 2009;297:F27–F35. doi: 10.1152/ajprenal.00096.2009. [DOI] [PubMed] [Google Scholar]
  • 46.Johansen D, Ytrehus K, Baxter GF. Exogenous hydrogen sulfide (h2s) protects against regional myocardial ischemia-reperfusion injury--evidence for a role of k atp channels. Basic Res Cardiol. 2006;101:53–60. doi: 10.1007/s00395-005-0569-9. [DOI] [PubMed] [Google Scholar]
  • 47.Calvert JW, Jha S, Gundewar S, Elrod JW, Ramachandran A, Pattillo CB, Kevil CG, Lefer DJ. Hydrogen sulfide mediates cardioprotection through nrf2 signaling. Circ Res. 2009;105:365–374. doi: 10.1161/CIRCRESAHA.109.199919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Sodha NR, Clements RT, Feng J, Liu Y, Bianchi C, Horvath EM, Szabo C, Sellke FW. The effects of therapeutic sulfide on myocardial apoptosis in response to ischemia-reperfusion injury. Eur J Cardiothorac Surg. 2008;33:906–913. doi: 10.1016/j.ejcts.2008.01.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Osipov RM, Robich MP, Feng J, Liu Y, Clements RT, Glazer HP, Sodha NR, Szabo C, Bianchi C, Sellke FW. Effect of hydrogen sulfide in a porcine model of myocardial ischemia-reperfusion: Comparison of different administration regimens and characterization of the cellular mechanisms of protection. J Cardiovasc Pharmacol. 2009;54:287–297. doi: 10.1097/FJC.0b013e3181b2b72b. [DOI] [PubMed] [Google Scholar]
  • 50.Ji Y, Pang QF, Xu G, Wang L, Wang JK, Zeng YM. Exogenous hydrogen sulfide postconditioning protects isolated rat hearts against ischemia-reperfusion injury. Eur J Pharmacol. 2008;587:1–7. doi: 10.1016/j.ejphar.2008.03.044. [DOI] [PubMed] [Google Scholar]
  • 51.Yang G, Wu L, Jiang B, Yang W, Qi J, Cao K, Meng Q, Mustafa AK, Mu W, Zhang S, Snyder SH, Wang R. H2s as a physiologic vasorelaxant: Hypertension in mice with deletion of cystathionine gamma-lyase. Science. 2008;322:587–590. doi: 10.1126/science.1162667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Calvert JW, Coetzee WA, Lefer DJ. Novel insights into hydrogen sulfide-mediated cytoprotection. Antioxid Redox Signal. 2009 doi: 10.1089/ars.2009.2882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Coyle JT, Puttfarcken P. Oxidative stress, glutamate, and neurodegenerative disorders. Science. 1993;262:689–695. doi: 10.1126/science.7901908. [DOI] [PubMed] [Google Scholar]
  • 54.Kimura H, Nagai Y, Umemura K, Kimura Y. Physiological roles of hydrogen sulfide: Synaptic modulation, neuroprotection, and smooth muscle relaxation. Antioxid Redox Signal. 2005;7:795–803. doi: 10.1089/ars.2005.7.795. [DOI] [PubMed] [Google Scholar]
  • 55.Kimura Y, Dargusch R, Schubert D, Kimura H. Hydrogen sulfide protects ht22 neuronal cells from oxidative stress. Antioxid Redox Signal. 2006;8:661–670. doi: 10.1089/ars.2006.8.661. [DOI] [PubMed] [Google Scholar]
  • 56.Tyagi N, Moshal KS, Sen U, Vacek TP, Kumar M, Hughes WM, Jr, Kundu S, Tyagi SC. H2s protects against methionine-induced oxidative stress in brain endothelial cells. Antioxid Redox Signal. 2009;11:25–33. doi: 10.1089/ars.2008.2073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Boysen G, Brander T, Christensen H, Gideon R, Truelsen T. Homocysteine and risk of recurrent stroke. Stroke. 2003;34:1258–1261. doi: 10.1161/01.STR.0000069017.78624.37. [DOI] [PubMed] [Google Scholar]
  • 58.Qu K, Chen CP, Halliwell B, Moore PK, Wong PT. Hydrogen sulfide is a mediator of cerebral ischemic damage. Stroke. 2006;37:889–893. doi: 10.1161/01.STR.0000204184.34946.41. [DOI] [PubMed] [Google Scholar]
  • 59.Kimura Y, Goto Y, Kimura H. Hydrogen sulfide increases glutathione production and suppresses oxidative stress in mitochondria. Antioxid Redox Signal. 2010;12:1–13. doi: 10.1089/ars.2008.2282. [DOI] [PubMed] [Google Scholar]
  • 60.Minamishima S, Bougaki M, Sips PY, Yu JD, Minamishima YA, Elrod JW, Lefer DJ, Bloch KD, Ichinose F. Hydrogen sulfide improves survival after cardiac arrest and cardiopulmonary resuscitation via a nitric oxide synthase 3-dependent mechanism in mice. Circulation. 2009;120:888–896. doi: 10.1161/CIRCULATIONAHA.108.833491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Prathapasinghe GA, Siow YL, O K. Detrimental role of homocysteine in renal ischemia-reperfusion injury. Am J Physiol Renal Physiol. 2007;292:F1354–F1363. doi: 10.1152/ajprenal.00301.2006. [DOI] [PubMed] [Google Scholar]
  • 62.Fu Z, Liu X, Geng B, Fang L, Tang C. Hydrogen sulfide protects rat lung from ischemia-reperfusion injury. Life Sci. 2008;82:1196–1202. doi: 10.1016/j.lfs.2008.04.005. [DOI] [PubMed] [Google Scholar]
  • 63.Szabo C. Hydrogen sulphide and its therapeutic potential. Nat Rev Drug Discov. 2007;6:917–935. doi: 10.1038/nrd2425. [DOI] [PubMed] [Google Scholar]
  • 64.Li L, Moore PK. Putative biological roles of hydrogen sulfide in health and disease: A breath of not so fresh air? Trends Pharmacol Sci. 2008;29:84–90. doi: 10.1016/j.tips.2007.11.003. [DOI] [PubMed] [Google Scholar]
  • 65.Linden DR, Levitt MD, Farrugia G, Szurszewski JH. Endogenous production of h2s in the gastrointestinal tract: Still in search of a physiologic function. Antioxid Redox Signal. 2010;12:1135–1146. doi: 10.1089/ars.2009.2885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kimura H. Hydrogen sulfide induces cyclic amp and modulates the nmda receptor. Biochem Biophys Res Commun. 2000;267:129–133. doi: 10.1006/bbrc.1999.1915. [DOI] [PubMed] [Google Scholar]
  • 67.Whiteman M, Armstrong JS, Chu SH, Jia-Ling S, Wong BS, Cheung NS, Halliwell B, Moore PK. The novel neuromodulator hydrogen sulfide: An endogenous peroxynitrite 'scavenger'? J Neurochem. 2004;90:765–768. doi: 10.1111/j.1471-4159.2004.02617.x. [DOI] [PubMed] [Google Scholar]
  • 68.Zhao W, Zhang J, Lu Y, Wang R. The vasorelaxant effect of h(2)s as a novel endogenous gaseous k(atp) channel opener. EMBO J. 2001;20:6008–6016. doi: 10.1093/emboj/20.21.6008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Sivarajah A, Collino M, Yasin M, Benetti E, Gallicchio M, Mazzon E, Cuzzocrea S, Fantozzi R, Thiemermann C. Anti-apoptotic and anti-inflammatory effects of hydrogen sulfide in a rat model of regional myocardial i/r. Shock. 2009;31:267–274. doi: 10.1097/SHK.0b013e318180ff89. [DOI] [PubMed] [Google Scholar]
  • 70.Cai WJ, Wang MJ, Moore PK, Jin HM, Yao T, Zhu YC. The novel proangiogenic effect of hydrogen sulfide is dependent on akt phosphorylation. Cardiovasc Res. 2007;76:29–40. doi: 10.1016/j.cardiores.2007.05.026. [DOI] [PubMed] [Google Scholar]
  • 71.Zanardo RC, Brancaleone V, Distrutti E, Fiorucci S, Cirino G, Wallace JL. Hydrogen sulfide is an endogenous modulator of leukocyte-mediated inflammation. FASEB J. 2006;20:2118–2120. doi: 10.1096/fj.06-6270fje. [DOI] [PubMed] [Google Scholar]
  • 72.Seino S, Miki T. Physiological and pathophysiological roles of atp-sensitive k+ channels. Prog Biophys Mol Biol. 2003;81:133–176. doi: 10.1016/s0079-6107(02)00053-6. [DOI] [PubMed] [Google Scholar]
  • 73.Inagaki N, Gonoi T, Clement JPt, Namba N, Inazawa J, Gonzalez G, Aguilar-Bryan L, Seino S, Bryan J. Reconstitution of ikatp: An inward rectifier subunit plus the sulfonylurea receptor. Science. 1995;270:1166–1170. doi: 10.1126/science.270.5239.1166. [DOI] [PubMed] [Google Scholar]
  • 74.Quesada I, Rovira JM, Martin F, Roche E, Nadal A, Soria B. Nuclear katp channels trigger nuclear ca(2+) transients that modulate nuclear function. Proc Natl Acad Sci U S A. 2002;99:9544–9549. doi: 10.1073/pnas.142039299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Trube G, Hescheler J. Inward-rectifying channels in isolated patches of the heart cell membrane: Atp-dependence and comparison with cell-attached patches. Pflugers Arch. 1984;401:178–184. doi: 10.1007/BF00583879. [DOI] [PubMed] [Google Scholar]
  • 76.Campbell JD, Sansom MS, Ashcroft FM. Potassium channel regulation. EMBO Rep. 2003;4:1038–1042. doi: 10.1038/sj.embor.7400003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Cheng Y, Ndisang JF, Tang G, Cao K, Wang R. Hydrogen sulfide-induced relaxation of resistance mesenteric artery beds of rats. Am J Physiol Heart Circ Physiol. 2004;287:H2316–H2323. doi: 10.1152/ajpheart.00331.2004. [DOI] [PubMed] [Google Scholar]
  • 78.Piper HM, Siegmund B, Ladilov Yu V, Schluter KD. Calcium and sodium control in hypoxic-reoxygenated cardiomyocytes. Basic Res Cardiol. 1993;88:471–482. doi: 10.1007/BF00795413. [DOI] [PubMed] [Google Scholar]
  • 79.Piper HM, Garcia-Dorado D, Ovize M. A fresh look at reperfusion injury. Cardiovasc Res. 1998;38:291–300. doi: 10.1016/s0008-6363(98)00033-9. [DOI] [PubMed] [Google Scholar]
  • 80.Rybin VO, Steinberg SF. Protein kinase c isoform expression and regulation in the developing rat heart. Circ Res. 1994;74:299–309. doi: 10.1161/01.res.74.2.299. [DOI] [PubMed] [Google Scholar]
  • 81.Pan TT, Neo KL, Hu LF, Yong QC, Bian JS. H2s preconditioning-induced pkc activation regulates intracellular calcium handling in rat cardiomyocytes. Am J Physiol Cell Physiol. 2008;294:C169–C177. doi: 10.1152/ajpcell.00282.2007. [DOI] [PubMed] [Google Scholar]
  • 82.Baines CP, Zhang J, Wang GW, Zheng YT, Xiu JX, Cardwell EM, Bolli R, Ping P. Mitochondrial pkcepsilon and mapk form signaling modules in the murine heart: Enhanced mitochondrial pkcepsilon-mapk interactions and differential mapk activation in pkcepsilon-induced cardioprotection. Circ Res. 2002;90:390–397. doi: 10.1161/01.res.0000012702.90501.8d. [DOI] [PubMed] [Google Scholar]
  • 83.Cantley LC. The phosphoinositide 3-kinase pathway. Science. 2002;296:1655–1657. doi: 10.1126/science.296.5573.1655. [DOI] [PubMed] [Google Scholar]
  • 84.Hausenloy DJ, Yellon DM. Survival kinases in ischemic preconditioning and postconditioning. Cardiovasc Res. 2006;70:240–253. doi: 10.1016/j.cardiores.2006.01.017. [DOI] [PubMed] [Google Scholar]
  • 85.Hu Y, Chen X, Pan TT, Neo KL, Lee SW, Khin ES, Moore PK, Bian JS. Cardioprotection induced by hydrogen sulfide preconditioning involves activation of erk and pi3k/akt pathways. Pflugers Arch. 2008;455:607–616. doi: 10.1007/s00424-007-0321-4. [DOI] [PubMed] [Google Scholar]
  • 86.Dimmeler S, Fleming I, Fisslthaler B, Hermann C, Busse R, Zeiher AM. Activation of nitric oxide synthase in endothelial cells by akt-dependent phosphorylation. Nature. 1999;399:601–605. doi: 10.1038/21224. [DOI] [PubMed] [Google Scholar]
  • 87.Skulachev VP. Cytochrome c in the apoptotic and antioxidant cascades. FEBS Lett. 1998;423:275–280. doi: 10.1016/s0014-5793(98)00061-1. [DOI] [PubMed] [Google Scholar]
  • 88.Barry SP, Townsend PA, Latchman DS, Stephanou A. Role of the jak-stat pathway in myocardial injury. Trends Mol Med. 2007;13:82–89. doi: 10.1016/j.molmed.2006.12.002. [DOI] [PubMed] [Google Scholar]
  • 89.Kunisada K, Negoro S, Tone E, Funamoto M, Osugi T, Yamada S, Okabe M, Kishimoto T, Yamauchi-Takihara K. Signal transducer and activator of transcription 3 in the heart transduces not only a hypertrophic signal but a protective signal against doxorubicin-induced cardiomyopathy. Proc Natl Acad Sci U S A. 2000;97:315–319. doi: 10.1073/pnas.97.1.315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Wang M, Wang Y, Abarbanell A, Tan J, Weil B, Herrmann J, Meldrum DR. Both endogenous and exogenous testosterone decrease myocardial stat3 activation and socs3 expression after acute ischemia and reperfusion. Surgery. 2009;146:138–144. doi: 10.1016/j.surg.2009.03.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Kevin LG, Camara AK, Riess ML, Novalija E, Stowe DF. Ischemic preconditioning alters real-time measure of o2 radicals in intact hearts with ischemia and reperfusion. Am J Physiol Heart Circ Physiol. 2003;284:H566–H574. doi: 10.1152/ajpheart.00711.2002. [DOI] [PubMed] [Google Scholar]
  • 92.Roth MB, Nystul T. Buying time in suspended animation. Sci Am. 2005;292:48–55. doi: 10.1038/scientificamerican0605-48. [DOI] [PubMed] [Google Scholar]
  • 93.Hill BC, Woon TC, Nicholls P, Peterson J, Greenwood C, Thomson AJ. Interactions of sulphide other ligands with cytochrome c oxidase An electron-paramagnetic-resonance study. Biochem J. 1984;224:591–600. doi: 10.1042/bj2240591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Aon MA, Cortassa S, O'Rourke B. Percolation and criticality in a mitochondrial network. Proc Natl Acad Sci U S A. 2004;101:4447–4452. doi: 10.1073/pnas.0307156101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Chen Q, Moghaddas S, Hoppel CL, Lesnefsky EJ. Reversible blockade of electron transport during ischemia protects mitochondria and decreases myocardial injury following reperfusion. J Pharmacol Exp Ther. 2006;319:1405–1412. doi: 10.1124/jpet.106.110262. [DOI] [PubMed] [Google Scholar]
  • 96.Aon MA, Cortassa S, Akar FG, O'Rourke B. Mitochondrial criticality: A new concept at the turning point of life or death. Biochim Biophys Acta. 2006;1762:232–240. doi: 10.1016/j.bbadis.2005.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Hausenloy DJ, Yellon DM. New directions for protecting the heart against ischaemia-reperfusion injury: Targeting the reperfusion injury salvage kinase (risk)-pathway. Cardiovasc Res. 2004;61:448–460. doi: 10.1016/j.cardiores.2003.09.024. [DOI] [PubMed] [Google Scholar]
  • 98.Entman ML, Michael L, Rossen RD, Dreyer WJ, Anderson DC, Taylor AA, Smith CW. Inflammation in the course of early myocardial ischemia. FASEB J. 1991;5:2529–2537. doi: 10.1096/fasebj.5.11.1868978. [DOI] [PubMed] [Google Scholar]
  • 99.Li L, Rossoni G, Sparatore A, Lee LC, Del Soldato P, Moore PK. Anti-inflammatory and gastrointestinal effects of a novel diclofenac derivative. Free Radic Biol Med. 2007;42:706–719. doi: 10.1016/j.freeradbiomed.2006.12.011. [DOI] [PubMed] [Google Scholar]
  • 100.Granger DN, Benoit JN, Suzuki M, Grisham MB. Leukocyte adherence to venular endothelium during ischemia-reperfusion. Am J Physiol. 1989;257:G683–G688. doi: 10.1152/ajpgi.1989.257.5.G683. [DOI] [PubMed] [Google Scholar]
  • 101.Yusof M, Kamada K, Kalogeris T, Gaskin FS, Korthuis RJ. Hydrogen sulfide triggers late-phase preconditioning in postischemic small intestine by an no- and p38 mapk-dependent mechanism. Am J Physiol Heart Circ Physiol. 2009;296:H868–H876. doi: 10.1152/ajpheart.01111.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Chang KC, Tees DF, Hammer DA. The state diagram for cell adhesion under flow: Leukocyte rolling and firm adhesion. Proc Natl Acad Sci U S A. 2000;97:11262–11267. doi: 10.1073/pnas.200240897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Yamaguchi T, Kamada K, Dayton C, Gaskin FS, Yusof M, Yoshikawa T, Carter P, Korthuis RJ. Role of enos-derived no in the postischemic anti-inflammatory effects of antecedent ethanol ingestion in murine small intestine. Am J Physiol Heart Circ Physiol. 2007;292:H1435–H1442. doi: 10.1152/ajpheart.00282.2006. [DOI] [PubMed] [Google Scholar]
  • 104.Kaiser RA, Bueno OF, Lips DJ, Doevendans PA, Jones F, Kimball TF, Molkentin JD. Targeted inhibition of p38 mitogen-activated protein kinase antagonizes cardiac injury and cell death following ischemia-reperfusion in vivo. J Biol Chem. 2004;279:15524–15530. doi: 10.1074/jbc.M313717200. [DOI] [PubMed] [Google Scholar]
  • 105.Banerjee SK, Maulik SK. Effect of garlic on cardiovascular disorders: A review. Nutr J. 2002;1:4. doi: 10.1186/1475-2891-1-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Benavides GA, Squadrito GL, Mills RW, Patel HD, Isbell TS, Patel RP, Darley-Usmar VM, Doeller JE, Kraus DW. Hydrogen sulfide mediates the vasoactivity of garlic. Proc Natl Acad Sci U S A. 2007;104:17977–17982. doi: 10.1073/pnas.0705710104. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES