CBI 7027

No. of Pages 8, Model 5G

19 April 2014 Chemico-Biological Interactions xxx (2014) xxx–xxx 1

Contents lists available at ScienceDirect

Chemico-Biological Interactions journal homepage: www.elsevier.com/locate/chembioint

2

Mini-review

5 4 6

Hydrogen sulfide in hemostasis: Friend or foe?

7 8 9 10 1 4 2 2 13 14 15 16 17 18 19 20 21 22 23

Q1

Beata Olas ⇑ Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland

a r t i c l e

i n f o

Article history: Received 12 November 2013 Received in revised form 2 April 2014 Accepted 6 April 2014 Available online xxxx Keywords: Hydrogen sulfide Cardiovascular system Hemostasis Blood platelet activation

a b s t r a c t Hydrogen sulfide (H2S) is a well known toxic gas that is synthesized from the amino acids: cysteine (Cys) and homocysteine (Hcy) by three enzymes: cystathionine-b-synthase (CBS), cystathionine-c-lyase (CSE) and mercaptopyruvate sulfurtransferase (3-MST). Hydrogen sulfide, like carbon monoxide (CO) or nitric oxide (NO) is a signaling molecule in different biological systems, including the cardiovascular systems. Moreover, hydrogen sulfide plays a role in the pathogenesis of various cardiovascular diseases. It modulates different elements of hemostasis (activation of blood platelet, and coagulation process) as well as proliferation and apoptosis of vascular smooth muscle cells. However, the biological role and the therapeutic potential of H2S is not clear. This review summarizes the different functions of hydrogen sulfide in hemostasis. Ó 2014 Elsevier Ireland Ltd. All rights reserved.

25 26 27 28 29 30 31 32 33 34 35 36

37 38 39 40 41 42 43 44 45 46 47 48

Contents 1. 2. 3. 4. 5. 6.

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Metabolism of endogenous hydrogen sulfide. . . . Biological properties of hydrogen sulfide . . . . . . . Regulation of hemostasis by hydrogen sulfide . . . Effects of hydrogen sulfide on the cardiovascular Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Conflict of Interest . . . . . . . . . . . . . . . . . . . . . . . . . Transparency Document . . . . . . . . . . . . . . . . . . . . Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

....... ....... ....... ....... system . ....... ....... ....... ....... .......

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

. . . . . . . . . .

00 00 00 00 00 00 00 00 00 00

49 50 51

1. Introduction

52

Hydrogen sulfide (H2S), noted for its smell of rotten eggs, is traditionally known as an air pollutant and a poisonous gas [1,2]. It is moderately soluble in water. Aqueous solutions of hydrogen sulfide have a slightly acidic pH, because hydrogen sulfide dissociates in two-step process into H+ and hydrogen sulfide anion (HS) and then into sulfide (S2). In physiological pH (pH = 7.4, at 37 °C) less than 1/5 of H2S is undissociated, and the remaining 4/5 are mainly HS (pKa1 = 6.98, at 25 °C) with a small admixture of sulfide ions (pKa2 = 19 ± 2) [3–6]. The physiological relevance of hydrogen sulfide gained more attention as it was detected in different mammalian tissues such

53 54 55 56 57 58 59 60 61 62

⇑ Tel.: +48 (42) 665 58 08; fax: +48 (42) 635 44 84.

as: brain, liver, kidney, heart, aorta and also lungs, mediating pleiotropic effects [7,8]. Endogenous concentrations of H2S in human plasma are described with range from 34 lM to 65 lM [9]. Its physiological level in the brain is up to threefold higher than in serum [10,11]. However, H2S concentration in human tissues depends on the method used for measurement and the donor’s age [9,12]. Moreover, most methods, like the spectrophotometric measurement of hydrogen sulfide with the use N0 N-dimethyl-p-phenylenediamine, monitor not only hydrogen sulfide, but also other species as S2 and HS. Endogenous concentration of H2S is regulated by many metabolic pathways, such as mitochondrial oxidation and cytosine methylation. Hydrogen sulfide can be scavenged by methemoglobin or metallo- or disulfide-containing molecules, i.e. oxidized glutathione – GSSG [13–16].

E-mail address: [email protected] http://dx.doi.org/10.1016/j.cbi.2014.04.006 0009-2797/Ó 2014 Elsevier Ireland Ltd. All rights reserved.

Please cite this article in press as: B. Olas, Hydrogen sulfide in hemostasis: Friend or foe?, Chemico-Biological Interactions (2014), http://dx.doi.org/ 10.1016/j.cbi.2014.04.006

63 64 65 66 67 68 69 70 71 72 73 74 75 76 77

CBI 7027

No. of Pages 8, Model 5G

19 April 2014 2

B. Olas / Chemico-Biological Interactions xxx (2014) xxx–xxx

92

Therapeutic and toxic effects of hydrogen sulfide depend on the concentration. Exposure to H2S (>700 ppm) causes loss of consciousness (syncope), paralysis of respiratory system and may also lead to death. In concentration 300–700 ppm hydrogen sulfide causes syncope and can sometimes lead to death if the exposition of the H2S is longer than 30 min. A few breaths of air containing high levels of >H2S (200–300 ppm) and an exposition of more than 1 h cause to the paralysis respiratory system. Hydrogen sulfide is present in effluent from hydrothermal vents and sulfur springs (at very low concentrations), which have been proposed to act as ‘‘pores’’ in the Earth surface, providing a source of energy in the form of reducing equivalents and of iron–sulfur centers [17]. H2S has special therapeutic effects in treatment of some diseases: arteriosclerosis, arterial hypertension, chronic heavy metal toxicity, skin diseases or rheumatism [7].

93

2. Metabolism of endogenous hydrogen sulfide

94

Hydrogen sulfide is the final product of amino acids metabolism, which contain sulfur (homocysteine (Hcy) and cysteine (Cys), which is synthesized from L-methionine through the transsulfuration pathway with Hcy as an intermediate, or liberated from endogenous proteins). The substrate for the generation of endogenous H2S is L-cysteine. However, 3-mercaptopyruvate sulfurtransferase together with D-amino acid oxidase emerged as the fourth way for endogenous hydrogen sulfide production from D-cysteine, predominantly in the kidney and the cerebellum [18]. There are two main pathways of L-cysteine catabolism: (1) thiol group of Cys is oxidized by cysteine dioxygenase (CDO) to cysteine sulfinate, which may be decarboxylated to taurine or converted to pyruvate and sulfite, afterward oxidized to sulfate by sulfite oxidase (SO); (2) cysteine sulfur atom is removed without its oxidation which results in H2S production – process of ‘‘desulfhydration’’. Desulfuration may be catalyzed by the two enzymes, which take part in Hcy transsulfuration pathway: cystathionine b-synthase (CBS) and cysthationine c-lyase (CSE). CBS and CSE are vitamin B6-dependent (pyridoxal 50 -phosphate-dependent). H2S production by CBS involves the condensation of Hcy with Cys to yield cysthationine; and during this reaction the H2S is liberated [19]. CSE catalyzes the conversion of L-cysteine to pyruvate, thiocysteine and ammonia. Then, thiocysteine nonenzymatically decomposes to cysteine and hydrogen sulfide. Recently it has been shown that 3-mercaptopyruvate sulfurtransferase (3-MST) also takes part in endogenous H2S generation. 3-MST produces H2S in conjunction with cysteine aminotransferase (CAT) from L-cysteine in the presence of a-ketoglutarate. CAT catalyses the transformation between L-cysteine and a-ketoglutarate. The products of this reaction are 3-mercaptopyruvate and L-glutamate. 3-MST transfers sulfur from 3-mercaptopyruvate into sulphurous acid and in the reaction are produced pyruvate and thiosulfate, and the later is reduced to hydrogen sulfide in the presence of reduced form of glutathione (GSH) [17,20]. The CBS pathway is reported to contribute to endogenous H2S production in the central nervous system, CSE is a major H2S-producing enzyme in the cardiovascular system and 3-MST in the myocardium [21,22]. During the enzymatic pathway, H2S can be immediately released or stored in the cells. Pathways of synthesis of endogenous hydrogen sulfide are shown on Fig. 1. There are two mechanisms for the release of hydrogen sulfide. First, H2S is immediately released after its enzymatical formation. Second mechanism is that the produced H2S is stored intracellular in a sulfane sulfur bond and is released in response to a physiological signal [23,24]. Sulfane sulfur has regulatory effects in biological systems. These functions include activation or inactivation of enzymes, posttranscriptional modification of transfer RNA and synthesis of the sulfur-containing cofactors or vitamins [25–27].

78 79 80 81 82 83 84 85 86 87 88 89 90 91

95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110

111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141

3. Biological properties of hydrogen sulfide

142

The physiological effects of endogenous H2S may be multifaceted. In different animal disease models, hydrogen sulfide exerts a classical pleiotropic profile with anti-inflammatory, cytoprotective, and anti-apoptotic action [28–31]. Other experiments have reported that hydrogen sulfide inhibits smooth muscle cell proliferation by promoting apoptosis, i.e. in human aortic smooth muscle. H2S (in a concentration-dependent manner) induces apoptosis through activation of the mitogen activated protein kinase (MAPK) pathway [32]. Moreover, H2S stimulates the expression of heme oxygenase 1 (HO-1) through a signaling pathway depending on extracellular signal-regulated kinase (ERK) (Fig. 2). In addition, HO-1 reduces production of NO by inhibiting the expression of inducible nitric oxide synthase (iNOS). H2S prevents phosphorylation of IjB and subsequent degradation. As a consequence the NF-jB mediated transcription is decreased. H2S not only activates ERK 1/2, but also stimulates phosphorylation p-38 MAPK (mitogen activated kinase) [33–35]. Hydrogen sulfide plays also a role in vascular homeostasis [11,29], because it has anti-adhesive and anti-inflammatory properties [36]. The action of H2S influence on vascular inflammatory process is of particular interest because H2S has an antiinflammatory influence on macrophages [34] and it has an proinflammatory effect on vascular smooth muscle cells [35]. Various groups have demonstrated the effect of H2S on organ injury and postischemic reperfusion disorders [1]. Moreover, anti-aggregatory, anti-coagulatory and anti-thrombotic properties of H2S were observed [2,24,37]. Hydrogen sulfide may decrease arterial pressure (about 125 lM H2S) [38], hyperpolarize blood vessels in vivo by KATP channels activation (about 200 lM H2S) [32]. The relaxing properties of hydrogen sulfide was shown in endothelial cells and in smooth muscle cells of gastrointestinal system and airways. The results of Mazza et al. [39] indicate that Akt/eNOS signaling and S-sulfhydration of cardiac proteins are involved in H2S-dependent cardiac relaxation in frog and rat. The experiments of Castro-Piedras and Perez-Zaghbi [40] have shown that hydrogen sulfide inhibits Ca2+ release through inositol-1,4,5-trisphosphate receptors and relaxes airway smooth muscle. Other results have reported that hydrogen sulfide plays an important function in the peripheral nervous system [41]. Qu et al. [42] have observed that H2S concentration in the cerebral cortex is increased in stroke. The authors suggest that high concentration of cysteine, probably is associated with high concentration of hydrogen sulfide and inversely correlates with clinical outcome of patients which suffer from ischemic stroke [43]. The role of H2S in oxidative stress is still uncertain. Recent studies have demonstrated that hydrogen sulfide reacts with reactive oxygen and nitrogen species, i.e. hydrogen peroxide (H2O2), super oxide anion radical (O 2 ), peroxynitrite (ONOO ) and hypochlorite [44–48].

143

4. Regulation of hemostasis by hydrogen sulfide

192

The hemostatic system consists of a complex array of processes that maintains blood flow under physiological conditions. Hemostasis involves an explosive reaction, designed to curtail blood loss, restore vascular integrity and ultimately preserve life. The rapid transformation of blood from its fluid state into a localized thrombus at the site of tissue damage is controlled by an intricate interplay of four key components – the vascular endothelium, blood platelets, the coagulation pathway and fibrinolysis. Disruption of this tightly regulated hemostatic process can result in pathological thrombosis or hemorrhage, both a frequent complication of surgery, illness and trauma. Blood platelets circulate in close contact

193

Please cite this article in press as: B. Olas, Hydrogen sulfide in hemostasis: Friend or foe?, Chemico-Biological Interactions (2014), http://dx.doi.org/ 10.1016/j.cbi.2014.04.006

144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191

194 195 196 197 198 199 200 201 202 203

CBI 7027

No. of Pages 8, Model 5G

19 April 2014 3

B. Olas / Chemico-Biological Interactions xxx (2014) xxx–xxx

Pyruvate + thiosulfate +

L-cysteine + α-ketobutyrate + NH4+

H2 S

Mercaptopuryvate sulphurtransferase

Cystathionine-γ-lyase

3-mercaptopyruvate

Cystathionine + Cysteine aminotransferase

L-glutamate

H2 S

Cystathionine-β -synthase

Homocysteine

α-ketobutyrate L-cysteine

Cysteine dioxygenase

L-cysteinylosulfinate

Cystine Cystathionine - β -synthase

α-ketobutyrate Hipotaurine

L-glutamate

Pyruvate + thiocysteine + NH4+

β-sulfinatopyruvate Taurine Pyruvate + sulfite

L-cysteine +

H2 S

Sulfite oxidase

Sulfate Fig. 1. Pathways of synthesis of endogenous hydrogen sulfide. The substrate for the production of endogenous H2S is L-cysteine, which may be metabolized by three enzymes: cystathionine-b-synthase, cystathionine-c-lyase and mercaptopyruvate sulfurtransferase [[17–20,96,97], modified].

204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225

with the endothelial wall of blood vessels, facilitating rapid recognition of a disrupted or injured endothelial cell surface. Later, the formation of a platelet and fibrin hemostatic plug is observed. The process of platelet plug formation is called primary hemostasis, as opposed to the secondary events of the procoagulant system. Primary hemostasis consists of several platelet events; adhesion, secretion and aggregation [49]. Platelets are multiresponding cells, both with respect to the different number of agonists and compounds. They can be activated by several physiologically important compounds including collagen, ADP and thrombin. Platelet aggregation under physiological conditions is an important process to stop bleeding, but it is considered that excessive platelet aggregation causes thrombosis and atherosclerosis. Recent studies have shown that H2S acts antithrombotic, thereby inhibiting different steps of platelet activation (i.e. platelet adhesion, secretion and aggregation) and thrombus formation [2,37,50–52] (Fig. 3). The total inhibition of platelet aggregation was observed at 10 mM NaHS concentration, independently from the agonists in vitro. Studies conducted by Zagli et al. [37] showed that NaHS prevented in a concentration-dependent manner platelet aggregation induced by different agonists: ADP, U46619, collagen, epinephrine, thrombin and arachidonic acid. The highest

concentration by 2 lM ADP, thus excluding the possibility that NaHS acts via endogenous NO generation. Inhibition of adenylyl cyclase by SQ 22,536 (100 lM) and of guanylyl cyclase by ODQ (100 lM) did not affect NaHS-induced inhibition of ADPstimulated aggregation (2 lM). Significant inhibition of platelet aggregation induced by a moderate ADP concentration (0.8 lM), was also obtained with NaHS concentrations as low as 30 lM. Results of Nishikawa et al. [50] have demonstrated that H2S suppresses rabbit platelet aggregation (induced by collagen and ADP) by interfering with both upstream and downstream signals of cytosolic Ca2+ mobilization in a cAMP-dependent manner. The effective concentration range of NaHS was 0.1–0.3 mM in platelet-rich plasma and 1–3 mM in washed platelets. However, the potency of H2S to inhibit platelet aggregation depends on the strength of the stimulus, and H2S concentrations close to those found in human plasma exhibited antiaggregating effects. Thus, it could be theorized that physiological H2S concentrations in plasma may be sufficient to prevent platelet aggregation induced by moderate stimulation. Moreover, experiments of Grambow et al. [2] suggest that the anti-aggregatory effect of hydrogen sulfide might be due to S-sulfhydration of blood platelet proteins with concomitantly reduced granule exocytosis and shape change upon

Please cite this article in press as: B. Olas, Hydrogen sulfide in hemostasis: Friend or foe?, Chemico-Biological Interactions (2014), http://dx.doi.org/ 10.1016/j.cbi.2014.04.006

226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247

CBI 7027

No. of Pages 8, Model 5G

19 April 2014 4

B. Olas / Chemico-Biological Interactions xxx (2014) xxx–xxx

Fig. 2. Hydrogen sulfide-mediated pathways. H2S may stimulate the expression of heme oxygenase 1 (HO-1) through a signaling pathway depending on extracellular signal-regulated kinase (ERK). H2S may also stimulate phosphorylation mitogen activated kinase (p-38 MAPK). The HO-1 reduces production of NO by inhibiting the expression of inducible nitric oxide synthase (iNOS).

248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281

activation. Other results of Grambow et al. [2] showed that hydrogen sulfide also reduces intracellular calcium levels in blood platelets, leading to both reduced exocytosis and shape changes. Next experiments demonstrated the inhibitory action of hydrogen sulfide on blood platelet adhesion [51]. Moreover, H2S modifies the adhesive properties of collagen and fibrinogen [52]. The authors assume that the interaction of modified adhesive proteins may cause impaired adhesion [52]. Flow cytometry of TRAP (peptide which is ‘‘tethered’’ ligand domain of thrombin receptor; peptide with the sequence Ser-Phe-Leu-Leu-Arg-Asn; SFLLRN) -activated blood platelets revealed that hydrogen sulfide exposure decreased adhesion molecule expression (i.e. P-selectin expression) in a dose dependent manner [2]. Studies of Morel et al. [51] indicated that H2S seems to display multiple effects of blood platelet activation since it inhibits action of both, the thrombin-mediated proteolytic cleavage of PAR(s) and non-proteolytic agonist – TRAP, but reduction of platelet responses causes by thrombin is stronger than by TRAP. These results suggest that NaHS–H2S donor may inhibit the activation cascade downstream of G-protein activation. Moreover, the tested concentrations (0.00001–0.1 mM) of NaHS did not cause the lyses of blood platelets determined as a leakage of lactic dehydrogenase into the extracellular medium [51]. During agonist induced blood platelet activation, a signal is transmitted into the platelet that produces different biochemical events, including reactive oxygen/nitrogen species (ROS/RNS) formation. ROS/RNS may behave as second messengers and may regulate blood platelet activation [41,53]. H2S may have pro- and antioxidative effects, which depend on its concentration. Inactive nitrozothiols are the products of a chemical reaction between H2S with nitric oxide. H2S decreases NO production during inflammation and limits his bioavailability of NO in physiological levels. Low concentrations of NaHS (30–50 lM) potentiate the protective antioxidative role of N-acetylcysteine, glutathione, superoxide dismutase, catalase or vitamin C.

Morel and co-workers [51], using NaHS as H2S donor, demonstrated in vitro that NaHS inhibits O 2 generation in blood platelets. The authors suppose that NaHS reacts with O 2 in resting blood platelets and thrombin- or TRAP-activated platelets. They showed that the strongest inhibitory action of NaHS on O 2 generation takes place in blood platelets activated by thrombin. Antioxidative properties of NaHS may be involved in the antiplatelet action of this compound. Platelet stimulation by thrombin causes the activation of phosphoinositides 3-kinase (PI 3-K), which phosphorylates the D3 position of the phosphatidylinositol ring to produce phosphatidylinositol (3,4) – bisphosphate (PI3, 4 P2) and phosphatidylinositol (3,4,5) – triphosphate (PI3, 4, 5 P3) [54,55]. The generation of PI3, 4 P2 would be required for the prolonged activation of integrin aIIbb3 and stabilization of platelet aggregates [54,55]. PI 3-kinase is also involved in receptor-mediated ROS production in blood platelets (activated by thrombin) and supports the importance of ROS signaling in platelet functions [56]. Hu et al. [57] demonstrated that cardioprotection induced by hydrogen sulfide involves PI 3-kinase pathway, therefore Morel et al. [51] postulate that PI 3-kinase may be involved in the mechanism(s) of H2S action in blood platelets. The effects of hydrogen sulfide on the complex coagulation system and fibrinolysis are manifold due to its pleiotropic character. We have observed that NaHS (at concentration 0.00001–0.1 mM) prolonged clotting time, decreased the maximal velocity of fibrin polymerization and stimulated the fibrinolysis in human plasma. The obtained in vitro results indicate the anticoagulant activities of H2S [58]. Therefore, we suppose that hydrogen sulfide may be a promising compound to prevent thrombosis in pathological processes where plasma procoagulant activity is observed. We suppose that modifications of various proteins of hemostatic system (including fibrinogen, plasminogen and thrombin) induced by H2S may be associated with changes of coagulation process and fibrinolysis. Experiments of Li et al. [59] showed that H2S promotes sulfhydrylation of protein cysteine. Results of other authors demonstrated that compound with thiol group(s) enhances plasma factor XIII-mediated fibrinogen cross linking [60,61]. It is possible that H2S is involved in this process.

282

5. Effects of hydrogen sulfide on the cardiovascular system

320

Hemostasis is a complex process that regulates in vivo the flowing properties of blood. However, changes in hemostatic process may play a very important role in the modulation of cardiovascular system and in the pathogenesis of cardiovascular diseases. Experiments performed on many various animals, and tissues or cells defined the functional role of H2S in the cardiovascular system [13]. It has known that H2S induces vasorelaxation in arterial hypertension. Researches on some animal models have shown that plasma H2S concentration, aortic CSE expression and enzymatic activity are lower in spontaneously hypertensive rats (SHR) in comparison with Wistar–Kyoto rats. In other words, chronic administration of NaHS lowers blood pressure in SHR but not in normotensive rats [62]. The CSE inhibitor D,L-propargylglicyna (PAG) decreases plasma H2S concentration and increases blood pressure in normotensive rats but not in SHR. This indicates that vascular H2S is involved in the regulation of vascular tone under baseline conditions thus H2S-generating system is suppressed in the hypertensive strain [63]. Other experiments have shown that low doses of NaHS increase the mean arterial pressure in anesthetized rats [64]. On the other hand, a transient dose-dependent decrease in mean arterial pressure in anesthetized rats is induced by intravenous bolus injection of hydrogen sulfide [11]. Moreover, NaHS relaxed rat thoracic aorta and portal vein preconstricted with norepinephrine, in vivo. H2S

321

Please cite this article in press as: B. Olas, Hydrogen sulfide in hemostasis: Friend or foe?, Chemico-Biological Interactions (2014), http://dx.doi.org/ 10.1016/j.cbi.2014.04.006

283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319

322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344

CBI 7027

No. of Pages 8, Model 5G

19 April 2014 5

B. Olas / Chemico-Biological Interactions xxx (2014) xxx–xxx

H2 S

S-sulfhydration

Blood platelet proteins

inhibition

O2 generation changes adhesive properties of collagen

Blood platelet activation

Collagen

Cellular injury Tissue factor

Contact system Blood platelet adhesion Blood platelet secretion

inhibition inhibition

H2 S

inhibition

Blood platelet aggregation Fibrinogen

Fibrin

Thrombus

changes biological properties of fibrinogen

H2 S

Fig. 3. The role of H2S in hemostatic system. H2S may display multiple effect on hemostasis. H2S may inhibit different steps of blood platelet activation: platelet adhesion, secretion and aggregation. H2S also modifies the adhesive properties of collagen and fibrinogen. Moreover, H2S inhibits O 2 production in blood platelets and induces Ssulhydration of platelet proteins.

345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372

relaxed also rat mesenteric arteries which are peripheral resistance vessel. This effect exerted hydrogen sulfide at concentrations lower than required to relax the aorta. Results of Zhu et al. [65] showed that H2S plays an important role in myocardial ischemia in experimental rats. Animals were randomly divided into three groups and received either vehicle, 14 lmol/kg of NaHS or 50 mg/kg PAG everyday for 1 week before surgery, and the treatment was continued for a further 2 days after myocardial ischemia when the animals were killed. The authors observed that endogenous H2S was cardioprotective in the rat model of myocardial ischemia. PAG reduced endogenous H2S generation after myocardial ischemia by inhibiting cystathionine-gamma-lyase. The mechanisms of action explicated by hydrogen sulfide are multiple and range from vasodilatation and activation of KATPchannels to the differential regulation of pro- and anti-inflammatory genes [1,24]. Other experiments suggest that the interaction between NO and H2S is involved in regulation of not only cellular functions, inflammatory/immune function, but also cardiovascular responses [1,6,16,28]. There is a synergy of NO and H2S on vascular smooth muscle relaxation and the cysteine residues of hydrogen sulfide producing enzyme CSE are potential targets for NO-induced S-nitrosylation, thereby enhancing the activity of CSE [24,66]. Beside the action of NO on CSE protein, nitric oxide may stimulate CSE by increasing the activity of cGMP-dependent kinases [11,24]. On the other hand, hydrogen sulfide upregulates the NO generation from endothelial nitric oxide synthase (eNOS) through Akt kinasedependent Ser 1177 phosphorylation [67]. Moreover, hydrogen

sulfide augments NO production by potentiating the IL-1b-induced NFjB activation in rat vascular smooth muscle cells [35]. Results of Minamishima et al. [68] have demonstrated that hydrogen sulfide increases the phosphorylation of eNOS in the left ventricle of mice undergoing cardiac arrest and resuscitation. Issa et al. [69] demonstrated that NaHS when given before reperfusion protects against the effects of haemorrhage-induced ischemia–reperfusion by acting primarily through a decrease in both proinflammatory cytokines and inducible nitric oxide synthase pathway. Experiments of Huang et al. [70] indicate that endogenous H2S in the early reperfusion phase is the key mechanism of cardioprotection induced by ischemic postconditioning. Studies of Sun et al. [71] have shown that pretreating rat neonatal cardiomyocytes with NaHS reduced the level of ROS during the hypoxia/reoxygenation condition. They found that H2S inhibited mitochondrial complex IV activity and increased the activities of superoxide dismutases, including Mn-SOD and CuZn-SOD. Experiments of Kram et al. [24] have shown that H2S has antithrombotic action, i.e. prolonging the time until both initial occlusion of blood flow. The anti-thrombotic efficacy of H2S involves the NOS pathway. The recent studies have demonstrated that hydrogen sulfide may have some direct effects on the vascular wall [62,63,72]. Hydrogen sulfide causes apoptosis of human aortic smooth muscle cells [73,74] and reduces the growth of artherosclerotic lesions. A novel H2S releasing derivative of the non-steroidal anti-inflammatory drug – ‘‘S-diclofenac’’ has pro-apoptotic effects in human aortic smooth muscle cells over-expressing the H2S synthesizing

Please cite this article in press as: B. Olas, Hydrogen sulfide in hemostasis: Friend or foe?, Chemico-Biological Interactions (2014), http://dx.doi.org/ 10.1016/j.cbi.2014.04.006

373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400

CBI 7027

No. of Pages 8, Model 5G

19 April 2014 6 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451

B. Olas / Chemico-Biological Interactions xxx (2014) xxx–xxx

enzyme CSE. Exogenous H2S induces apoptosis in the concentration-dependent manner through activation of MAPK pathway in human aortic smooth muscle cells. Surprisingly, this proapoptotic properties of hydrogen sulfide may be important for the prevention of cell proliferation in some diseases, such as: vascular graft occlusion, neointimal hyperplasia and atherosclerosis [13,75]. However, it was confirmed that hydrogen sulfide has antiatheroscerotic effects in different ways (Fig. 4). Decreased endogenous H2S production predisposes the animals to vascular remodeling and early development of atherosclerosis. Results of Mani et al. [76] suggest that the CSE/H2S pathway is an important therapeutic target for protection against atherosclerosis. Some facts support the hypothesis that H2S deficiency might contribute to atherogenesis and if it’s feasible, one should focus on patients who suffer from hyperhomocysteinemia. Despite elevated plasma Hcy, the risk of acute cardiovascular effects is not increased in humans with reduced activity of methylenetetrahydrofolate reductase (MTHFR) [77]. As proven previously, MTHFR deficiency should increase the level of H2S which may partly attenuate the proatherogenic effect of Hcy. The risk of cardiovascular events is increased in patients with homocystinuria, which were treated with vitamin B6, but is not as high as in patients with mild hyperhomocysteinemia, even though plasma Hcy is much greater in the former group [78]. To sum up, progression of atherosclerosis is slower in patients with Down syndrome, a state of H2S overproduction [79]. In in vivo experiments it was shown the pro-angiogenic effects of H2S in endothelial cells and this action was inhibited by the inhibitor of phosphoinositide 3-kinase. Several studies indicated that H2S is involved in the pathogenesis of hyperhomocysteinemia. Yan et al. [80] demonstrated that H2S plays a protective role on hyperhomocysteinemia-induced cell injury by its anti-oxidative stress effect. It was demonstrated that low concentrations of H2S (30 and 50 lM) increased homocysteine (100 lM)-incubated cell survival by 7% and 61%, respectively. It was also shown that H2S (30 lM) attenuated Hcy (100 lM)induced overproduction of O 2 by 55.8%. Chang et al. [81] observed that injection of H2S reduced the concentration of Hcy in plasma, and decreased lipid peroxidation. These results suggest that there is a protective role of H2S in hyperhomocysteinemia induced by anti-oxidative stress, which may interfere with the promoting effect of hyperhomocysteinemia on cardiovascular disease. Some studies have demonstrated the role of hydrogen sulfide as therapeutic agent in various diseases, including cardiovascular diseases. An injectable Na2S (IK-1001), which is an H2S donor, has been developed for clinical use. It is currently undergoing phase I and II trials for therapy in ischemia reperfusion injury and renal injury [82]. S-allylcysteine, which may derive with garlic reduced blood platelet aggregation, and this action may be mediated through H2S [83,84]. Table 1 has shown hydrogen sulfide as therapeutic agent.

H2 S oxidative stress

adhesion

vascular remodeling

inflammation

Ox-LDL Fig. 4. Anti-atherosclerotic actions of hydrogen sulfide. H2S has a number of antiatherosclerotic effects, like reducing inflammation, attenuating oxidative stress, inhibiting blood platelet adhesion and preventing proliferation of vascular smooth muscle cells [[98], modified].

Table 1 Hydrogen sulfide as therapeutic agent [83,84,91–95]. Therapeutic agent

Functions

Na2S (IK-100)

Phase I and II trials for therapy in ischemia reperfusion injury and renal injury Anti-inflammatory role

Morpholin-4-ium-4methoxyphenyl(morpholino) phosphinodithioate (GYY4137) 5-Amino-2-hydroxybenzoic acid 4(5-thioxo-5H-[1,2]dithiol-3yl)phenyl ester (ATB-429; mesalazine) 2-(6-Methoxynapthalen-2-yl)propionic acid 4thiocarbomylphenyl ester (ATB3346; naproxen derivative) 2-Acetyloxybenzoic acid 4-(3-thioxo3H-1,2-dithiol-5-yl) ester (ACS-14; aspirin derivative) 4-(3-Thioxo-3H-1,2-dithiol-4-yl)benzoic acid (ACS48; dopamine derivative) [2-Methoxy-4-3-thioxo-3H-1,2dithiol-5-yl)-phenoxy] acetic acid (ACS50; dopamine derivative) 1,3-Dithiolate-2-thioxo-4-caboxylic acid (ACS5; dopamine derivative) 3-(Prop-2-en-1-yldisufanyl) propanoic acid (ACS81; dopamine derivative) Garlic derived compounds (at dietary doses) – S-allylcysteine

Crohn’s disease and ulcerative colitis treatment

Inhibits activity of cyclooxygenase-2 and protects gastrointestinal mucosa

Plays anti-inflammatory and analgestic role Parkinson’s disease

Parkinson’s disease

Parkinson’s disease Parkinson’s disease

Cardioprotective effects by inhibition of blood platelet aggregation

6. Conclusions

452

Endogenous metabolism and physiological functions position H2S in the novel family of endogenous gaseous transmitters – called ‘gasotransmitters’, besides NO and CO. So H2S is another inorganic gasotransmitter in the cardiovascular and nervous system [15]. Because there are some controversies in the field surrounding the physiologically significant H2S concentrations and its biological effects is in the midst of the lively expansion [85,86]. H2S has been demonstrated to stimulate or inhibit certain intracellular transduction pathways, to stimulate [87] or inhibit [72,88] cell proliferation, to active [73,74] or block [29] apoptosis, to be pro- [32] or anti-inflammatory [89], active or block hemostasis [17]. Some of this effects are still controversial [15]. Only few studies have shown alterations in H2S level in human diseases (e.g. Down syndrome, septic shock or ischemic stroke) and what is important, in most cases it was done indirectly by measuring H2S-related compounds – thiosulfate or sulfhemoglobin [90] rather than H2S itself. Due to its vasorelaxative and also vasoprotective properties, H2S might be useful in treating arterial hypertension by decreasing peripheral resistance [86]. Novel exciting and important aspects of H2S research continuously emerge. For example, considering the role of oxidative stress in many diseases (such as atherosclerosis, Alzheimer’s disease, etc.) one might wonder if excessive ROS production may cause H2S deficiency [45]. Its effect on hemostasis: blood platelet activation or hemostatic proteins is still unknown? Recognition of H2S role (the friend or the foe) in hemostasis may bring benefits in prophylaxis and treatment many diseases, such as: cardiovascular diseases, which are the highest risk of death. Researchers speculate the positive role of hydrogen sulfide (as the friend) in the cardiovascular system.

453

Conflict of Interest

482

The authors declare that there are no conflicts of interest.

Please cite this article in press as: B. Olas, Hydrogen sulfide in hemostasis: Friend or foe?, Chemico-Biological Interactions (2014), http://dx.doi.org/ 10.1016/j.cbi.2014.04.006

454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481

483

CBI 7027

No. of Pages 8, Model 5G

19 April 2014 B. Olas / Chemico-Biological Interactions xxx (2014) xxx–xxx 484

Transparency Document

485 487 486

The Transparency document associated with this article can be found in the online version.

488

Acknowledgements

489 491

Special thanks goes to A. Morel, J. Malinowska and J. Tadeusiewicz (Department of General Biochemistry, University of Lodz) for help in preparing of the manuscript.

492

References

493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560

[1] C. Szabo, Hydrogen sulfide and its therapeutic potential, Nat. Rev. Drug Discov. 6 (2007) 917–935. [2] E. Grambow, F. Mueller-Graf, E. Delyagina, M. Frank, A. Kuhla, B. Vollmar, Effect of the hydrogen sulfide donor GYY4137 on platelet activation and microvascular thrombus formation in mice. Platelets, 2013 (in press). [3] A.J. Ellis, N.B. Milestone, The ionization constants of hydrogen sulphide from 20 to 90 °C, Geochim. Cosmochim. Acta 31 (1967) 615–620. [4] M.N. Huges, M.N. Centelles, K.P. Moore, Making and working with hydrogen sulfide: the chemistry and generation of hydrogen sulfide in vitro and its measurement in vivo: a review, Free Radic. Biol. Med. 47 (2009) 1346–1353. [5] M. Ishigami, K. Hiraki, K. Umemura, Y. Ogasawara, K. Ishii, H. Kimura, A source of hydrogen sulfide and a mechanism of its release in the brain, Antioxid. Redox Signal. 11 (2009) 205–214. [6] R. Wang, The gasotransmitter role of hydrogen sulfide, Antioxid. Redox Signal. 5 (2003) 493–501. [7] L. Moore, P.K. Li, Putative biological roles of hydrogen sulfide in health and disease: a breath of not so fresh air?, Trends Pharmacol Sci. 29 (2008) 84–90. [8] K. Abe, H. Kimura, The possible role of hydrogen sulfide as an endogenous neuromodulator, J. Neurosci. 16 (1996) 1066–1071. [9] M. Whiteman, P.K. Moore, Hydrogen sulfide and the vasculature: a novel vasculoprotective entity and regulator of nitric oxide, J. Cell. Mol. Med. 13 (2009) 488–507. [10] P.J. Hogg, Contribution of allosteric disulfide bonds to regulation of hemostasis, J. Thromb. Haemost. 7 (Suppl. 1) (2009) 13–16. [11] W. Zhao, J. Zhang, Y. Lu, R. Wang, The vasorelaxant effect of H(2)S as a novel endogenous gaseous K(ATP) channel opener, EMBO J. 20 (2001) 6008–6016. [12] M. Whiteman, N. Cheung, Y. Zhu, S.H. Chu, J.L. Siau, B.S. Wong, J.S. Armstrong, P.K. Moore, Hydrogen sulphide: a novel inhibitor of hypochlorous acidmediated oxidative damage in the brain?, BBRC 326 (2005) 794–798 [13] D.J. Elsey, R.C. Fowkes, G.F. Baxter, Regulation of cardiovascular cell function by hydrogen sulfide (H(2)S), Cell Biochem. Funct. 28 (2010) 95–106. [14] M. Ishigami, K. Hiraki, K. Umemura, Y. Ogasawara, K. Ishii, H. Kimura, A source of hydrogen sulfide and a mechanism of its release in the brain, Antioxid. Redox Signal. 11 (2009) 205–214. [15] E. Lowicka, J. Beltowski, Hydrogen sulfide (H2S) – the third gas of interest for pharmacologists, Pharmacol. Rep. 59 (2007) 4–24. [16] R. Wang, Two’s company, three’s a crowd: can H2S be the third endogenous gaseous transmitter?, FASEB J 16 (2002) 1792–1798. [17] A. di Masi, P. Ascenzi, H2S: a ‘‘double face’’ molecule in health and disease, Biofactors 39 (2013) 186–196. [18] N. Shibuya, S. Koike, M. Tanaka, M. Ishigami-Yuasa, Y. Kimura, Y. Ogasawara, K. Fukui, N. Nagahara, H. Kimura, A novel pathway for the production of hydrogen sulfide from D-cysteine in mammalian cells, Nat. Commun. 4 (2013) 1366. [19] X. Chen, K.H. Jhee, W.D. Kruger, Production of the neuromodulator H2S by cystathionine beta-synthase via the condensation of cysteine and homocysteine, J. Biol. Chem. 279 (2004) 52082–52086. [20] K. Tanizawa, Production of H2S by 3-mercaptopyruvate sulphurtransferase, J. Biochem. 149 (4) (2011) 357–359. [21] N. Shibuya, M. Tanaka, M. Yoshida, Y. Ogasawara, T. Togawa, K. Ishii, H. Kimura, 3-Mercaptopyruvate sulfurtransferase produces hydrogen sulfide and bound sulfane sulfur in the brain, Antioxid. Redox Signal. 11 (2009) 703–714. [22] N. Shibuya, Y. Mikami, Y. Kimura, N. Nagahara, H. Kimura, Vascular endothelium expresses 3-mercaptopyruvate sulfurtransferase and produces hydrogen sulfide, J. Biochem. 146 (2009) 623–626. [23] Y. Ogasawara, S. Isoda, S. Tanable, Tissue and subcellular distribution of bound and acid-labile sulfur; and the enzymatic capacity for sulfide production in the rat, Biol. Pharm. Bull. 17 (1994) 1535–1542. [24] L. Kram, E. Grambow, F. Mueller-Graf, H. Sorg, B. Vollmar, The anti-thrombotic effect of hydrogen sulfide is partly mediated by an upregulation of nitric oxide synthases, Thromb. Res. 132 (2013) e112–e117. [25] E.G. Mueller, Trafficking in persulfides: delivering sulfur in biosynthetic pathways, Nat. Chem. Biol. 2 (2006) 185–195. [26] H.A. Beinert, A tribute to sulfur, Eur. J. Biochem. 267 (2000) 5657–5664. [27] J.I. Toohey, Sulfur signaling: is the agent sulfide or sulfane?, Anal Biochem. 413 (2011) 1–7. [28] L. Li, P.K. Moore, An overview of the biological significance of endogenous gases: new role for old molecules, Biochem. Soc. Trans. 35 (2007) 1138–1141.

490

Q3

7

[29] R.C. Zanardo, V. Brancaleone, E. Distrutti, S. Fiorucci, G. Cirino, J.L. Wallace, Hydrogen sulfide is an endogenous modulator of leukocyte-mediated inflammation, FASEB J. 20 (2006) 2118–2120. [30] A. Sivarajah, M. Collino, M. Yasin, E. Benetti, M. Gallicchio, E. Mazzon, S. Cuzzocrea, R. Fantozzi, C. Thiemermann, Anti-apoptotic and antiinflammatory effects of hydrogen sulfide in a rat model of reginal myosardial I/R, Shock 31 (2009) 267–274. [31] F.S. Gaskin, K. Kamada, M. Yusof, W. Durante, G. Gross, R.J. Korthuis, AICAR preconditioning prevents postischemic leukocyte rolling and adhesion: role of K(ATP) channels and heme oxygenase, Microcirculation 16 (2009) 167–176. [32] M. Bhatia, J. Sidhapuriwala, S.M. Moochhala, P.K. Moore, Hydrogen sulphide is a mediator of carrageenan-induced hindpaw oedema in the rat, Br. J. Pharmacol. 145 (2005) 141–144. [33] J. Du, Y. Hui, Y. Cheung, G. Ben, H. Jiang, X. Chen, C. Tang, The possible role of hydrogen sulfide as a smooth muscle cell proliferation inhibitor in rat cultured cells, Heart Vessels 19 (2004) 75–80. [34] G.S. Oh, H.O. Pae, B.S. Lee, B.N. Kim, J.M. Kim, H.R. Kim, S.B. Jeon, W.K. Jeon, H.J. Chae, H.T. Chung, Hydrogen sulfide inhibits nitric oxide production and nuclear factor-kappaB via heme oxygenase-1 expression in RAW264.7 macrophages stimulated with lipopolysaccharide, Free Radic. Biol. Med. 41 (2006) 106–119. [35] S.O. Jeong, H.O. Pae, G.S. Oh, G.S. Jeong, B.S. Lee, S. Lee, D.Y. Kim, H.Y. Rhew, K.M. Lee, H.T. Chung, Hydrogen sulfide potentiates interleukin-1beta-induced nitric oxide production via enhancement of extracellular signal-regulated kinase activation in rat vascular smooth muscle cells, Biochem. Biophys. Res. Commun. 345 (2006) 938–944. [36] S. Fiorucci, E. Antonelli, E. Distrutti, G. Rizzo, A. Mancarelli, S. Orlandi, R. Zanardo, B. Renga, M. Di Sante, A. Morelli, G. Cirino, J.L. Wallace, Inhibition of hydrogen sulfide generation contributes to gastric injury caused by antiinflammatory nonsteroidal drugs, Gastroenterology 129 (2005) 1210–1224. [37] G. Zagli, R. Patacchini, M. Trevisani, R. Abbate, S. Cinotti, G.F. Gensini, G. Masotti, P. Geppetti, Hydrogen sulfide inhibits human platelet aggregation, Eur. J. Pharmacol. 559 (2007) 65–68. [38] R. Hosoki, N. Matsuki, H. Kimura, The possible role of hydrogen sulfide as an endogenous smooth muscle relaxant in synergy with nitric oxide, Biochem. Biophys. Res. Commun. 237 (1997) 527–531. [39] R. Mazza, T. Pasqua, M.C. Cerra, T. Angelone, A. Gattuso, Akt/eNOS signaling and PLN S-sulfhydration are involved in H2S-dependent cardiac effects in frog and rat, Am. J. Physiol. Regul. Integr. Comp. Physiol. 305 (2013) R443–R451. [40] I. Castro-Piedras, J.F. Perez-Zoghbi, Hydrogen sulphide inhibits Ca2+ release through InsP3 receptors and relaxes airway smooth muscle, J. Physiol. (2013) (in press). [41] R. Patacchini, P. Santicioli, S. Giuliani, C.A. Maggi, Hydrogen sulfide (H2S) stimulates capsaicin-sensitive primary afferent neurons in the rat urinary bladder, Br. J. Pharmacol. 142 (2004) 31–34. [42] K. Qu, C.P. Chen, B. Halliwell, P.K. Moore, P.T. Wong, Hydrogen sulfide is a mediator of cerebral ischemic damage, Stroke 37 (2006) 889–893. [43] P.T. Wong, K. Qu, G.N. Chimon, A.B. Seah, H.M. Chang, M.C. Wong, Y.K. Ng, H. Rumpel, B. Halliwell, C.P. Chen, High plasma cyst(e)ine level may indicate poor clinical outcome in patients with acute stroke: possible involvement of hydrogen sulfide, J. Neuropathol. Exp. Neurol. 65 (2006) 109–115. [44] P. Kamoun, Mental retardation in Down syndrome: a hydrogen sulfide hypothesis, Med. Hypotheses 57 (2001) 389–392. [45] M. Whiteman, J.S. Armstrong, S.H. Chu, S. Jia-Ling, B.S. Wong, N.S. Cheung, B. Halliwell, P.K. Moore, The novel neuromodulator hydrogen sulfide: an endogenous peroxynitrite ‘scavenger’?, J Neurochem. 90 (2004) 765–768. [46] M. Whiteman, N.S. Cheung, Y.Z. Zhu, S.H. Chu, J.L. Siau, B.S. Wong, J.S. Armstrong, P.K. Moore, Hydrogen sulphide: a novel inhibitor of hypochlorous acid-mediated oxidative damage in the brain?, Biochem Biophys. Res. Commun. 326 (2005) 794–798. [47] M. Collin, C. Thiemermann, Hydrogen sulfide and sulfite: novel mediators in the pathophysiology of shock and inflammation, Shock 24 (2005) 595–596. [48] H. Mitsuhashi, H. Ikeuchi, Y. Nojima, Is sulfite an antiatherogenic compound in wine?, Clin Chem. 47 (2001) 1872–1873. [49] S.K. Austin, Haemostasis, Medicine 37 (2009) 133–136. [50] H. Nishikawa, H. Hayashi, S. Kubo, M. Tsubota-Matsunami, F. Sekiguchi, A. Kawabata, Inhibition by hydrogen sulfide of rabbit platelet aggregation and calcium mobilization, Biol. Pharm. Bull. 36 (2013) 1278–1282. [51] A. Morel, J. Malinowska, B. Olas, Antioxidative properties of hydrogen sulfide may involve in its antiadhesive action on blood platelets, Clin. Biochem. 45 (18) (2012) 1678–1682. [52] A. Morel, J. Malinowska, B. Olas, Hydrogen sulfide changes adhesive properties of fibrinogen and collagen in vitro, Platelets 25 (2014) 147–149. [53] B. Chadefaux, I. Ceballos, M. Hamet, M. Coude, M. Poissonnier, P. Kamoun, D. Allard, Is absence of atheroma in Down syndrome due to decreased homocysteine levels?, Lancet 2 (1988) 741 [54] S.E. Rittenhouse, Phosphoinositide 3-kinase activation and platelet function, Blood 12 (1996) 4401–4414. [55] J. Zhang, S.J. Shattil, M.C. Cunningham, S.E. Rittenhouse, Phosphoinositide 3kinase c and p85/phosphoinositide 3-kinase in platelets, J. Biol. Chem. 271 (1996) 6265–6272. [56] B. Wachowicz, B. Olas, H.M. Zbikowska, A. Buczynski, Generation of reactive oxygen species in blood platelets, Platelets 13 (2002) 175–182. [57] Y. Hu, X. Chen, T.T. Pan, K.L. Neo, S.W. Lee, E.S. Khin, P.K. Moore, J.S. Bian, Cardioprotection induced by hydrogen sulfide preconditioning involves activation of ERK and PI3K/Akt pathways, Pflugers Arch. 455 (2008) 607–616.

Please cite this article in press as: B. Olas, Hydrogen sulfide in hemostasis: Friend or foe?, Chemico-Biological Interactions (2014), http://dx.doi.org/ 10.1016/j.cbi.2014.04.006

Q4

561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646

CBI 7027

No. of Pages 8, Model 5G

19 April 2014 8 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708

B. Olas / Chemico-Biological Interactions xxx (2014) xxx–xxx

[58] B. Olas, B. Kontek, Anticoagulant properties of exogenous hydrogen sulfide, Toxicol. In Vitro (sub.). [59] L. Li, P. Rose, P.K. Moore, Hydrogen sulphide and cell signaling, Annu. Rev. Pharmacol. Toxicol. 109 (2011) 169–187. [60] R. Marchi, Z. Carvajal, J.W. Weisel, Comparison of the effect of different homocysteine concentrations on clot formation using human plasma and purified fibrinogen, Thromb. Haemost. 99 (2008) 451–452. [61] I.L. Quintana, M.V. Oberholzer, L. Kordich, A.M. Lauricella, Impaired fibrin gel permeability by high homocysteine levels, Thromb. Res. 127 (2011) 35–38. [62] H. Yan, J. Du, C. Tang, The possible role of hydrogen sulfide on the pathogenesis of spontaneous hypertension in rats, Biochem. Biophys. Res. Commun. 313 (2004) 22–27. [63] G. Zhong, F. Chen, Y. Cheng, C. Tang, J. Du, The role of hydrogen sulfide generation in the pathogenesis of hypertension in rats induced by inhibition of nitric oxide synthase, J. Hypertens. 21 (2003) 1879–1885. [64] M.Y. Ali, C.Y. Ping, Y.Y. Mok, L. Ling, M. Whiteman, M. Bhatia, P.K. Moore, Regulation of vascular nitric oxide in vitro and in vivo; a new role for endogenous hydrogen sulphide?, Br J. Pharmacol. 149 (2006) 625–634. [65] Y.Z. Zhu, Z.J. Wang, P. Ho, Y.Y. Loke, Y.C. Zhu, S.H. Huang, C.S. Tan, M. Whiteman, J. Lu, P.K. Moore, Hydrogen sulfide and its possible roles in myocardial ischemia in experimental rats, J. Appl. Physiol. 102 (2007) 261– 268. [66] J.R. Koenitzer, T.S. Isbell, H.D. Patel, G.A. Benavides, D.A. Dickinson, R.P. Patel, V.M. Darley-Usmar, J.R. Lancaster, J.E. Doeller, D.W. Kraus, Hydrogen sulfide mediates vasoactivity in an O2-dependent manner, Am. J. Physiol. Heart Circ. Physiol. 292 (2007) H1953–H1960. [67] B.L. Predmore, D. Julian, A.J. Cardounel, Hydrogen sulfide increases nitric oxide production from endothelial cells by an akt-dependent mechanism, Front. Physiol. 2 (2011) 104. [68] S. Minamishima, M. Bougaki, P.Y. Sips, J.D. Yu, Y.A. Minamishima, J.W. Elrod, D.J. Lefer, K.D. Bloch, F. Ichinose, Hydrogen sulfide improves survival after cardiac arrest and cardiopulmonary resuscitation via a nitric oxide synthase 3dependent mechanism in mice, Circulation 120 (2009) 888–896. [69] K. Issa, A. Kimmoun, S. Collin, F. Ganster, S. Fremont-Orlowski, P. Asfar, P.M. Mertes, B. Levy, Compared effects of inhibition and exogenous administration of hydrogen sulphide in ischaemia-reperfusion injury, Crit. Care 17 (2013) 1–12. [70] Y.E. Huang, Z.H. Tang, W. Xie, X.T. Shen, M.H. Liu, X.P. Peng, Z.Z. Zhao, D.B. Nie, L.S. Liu, Z.S. Jiang, Endogenous hydrogen sulfide mediates the cardioprotection induced by ischemic postconditioning in the early reperfusion phase, Exp. Ther. Med. 4 (2012) 1117–1123. [71] W.H. Sun, F. Liu, Y. Chen, Y.C. Zhu, Hydrogen sulfide decreases the levels of ROS by inhibiting mitochondrial complex IV and increasing SOD activities in cardiomyocytes under ischemia/reperfusion, Biochem. Biophys. Res. Commun. 421 (2012) 164–169. [72] J. Du, Y. Hui, Y. Cheung, G. Bin, H. Jiang, X. Chen, C. Tang, The possible role of hydrogen sulfide as a smooth muscle cell proliferation inhibitor in rat cultured cells, Heart Vessels 19 (2004) 75–80. [73] G. Yang, X. Sun, R. Wang, Hydrogen sulfide-induced apoptosis of human aorta smooth muscle cells via the activation of mitogen-activated protein kinases and caspase-3, FASEB J. 18 (2004) 1782–1784. [74] G. Yang, L. Wu, R. Wang, Pro-apoptotic effect of endogenous H2S on human aorta smooth muscle cells, FASEB J. 20 (2006) 553–555. [75] E. Zavaczki, V. Jeney, A. Agarwal, A. Zarjou, M. Oros, M. Katko, Z. Varga, G. Balla, J. Balla, Hydrogen sulfide inhibits the calcification and osteoblastic differentiation of vascular smooth muscle cells, Kidney Int. 80 (2011) 731–739. [76] S. Mani, H. Li, A. Untereiner, L. Wu, G. Yang, R.C. Austrin, J.G. Dickhout, S. Lhotak, Q.H. Meng, R. Wang, Decreased endogenous production of hydrogen sulfide accelerates atherosclerosis, Circulation 127 (2013) 2523–2534. [77] J. Ma, M.J. Stampfer, C.H. Hennekens, P. Frosst, J. Selhub, J. Horsford, M.R. Malinow, W.C. Willett, R. Rozen, Methylenetetrahydrofolate reductase polymorphism, plasma folate, homocysteine, and risk of myocardial infarction in US physicians, Circulation 94 (1996) 2410–2416.

[78] D.W. Jacobsen, Hyperhomocysteinemia and oxidative stress: time for a reality check?, Arterioscler Thromb. Vasc. Biol. 20 (2000) 1182–1184. [79] P. Kamoun, M.C. Belardinelli, A. Chabli, K. Lallouchi, B. Chadefaux-Vekemans, Endogenous hydrogen sulfide overproduction in Down syndrome, Am. J. Med. Genet. A 116A (2003) 310–311. [80] S.K. Yan, T. Chang, H. Wang, L. Wu, R. Wang, Q.H. Meng, Effects of hydrogen sulfide on homocysteine-induced oxidative stress in vascular smooth muscle cell, Biochem. Biophys. Res. Commun. 351 (2006) 485–491. [81] L. Chang, B. Geng, F. Yu, J. Zhao, H. Jiang, J. Du, C. Tang, Hydrogen sulfide inhibits myocardial injury induced by homocysteine in rats, Amino Acids 34 (2008) 573–585. [82] B.L. Predmore, D.J. Lefer, Development of hydrogen sulphide-based therapeutics for cardiovascular diseases, J. Cardiovasc. Transl. Res. 3 (2010) 487–498. [83] S.C. Chuah, P.K. Moore, Y.Z. Zhu, S-allylcysteine mediates cardioprotection on an acute myocardial infarction rat model via a hydrogen sulphide-mediated pathway, Am. J. Physiol. Heart Circ. Physiol. 293 (2007) H2693–H2701. [84] G. Scharbert, M.L. Kalb, M. Duris, C. Marschalak, S.A. Kozek-Langenecker, Garlic at dietary doses not impair platelet function, Anesth. Analg. 105 (2007) 1214–1218. [85] O. Kabil, R. Banerjee, Redox biochemistry of hydrogen sulfide, J. Biol. Chem. 285 (2010) 21903–21907. [86] C.A. Wagner, Hydrogen sulfide: a new gaseous signal molecule and blood pressure regulator, J. Nephrol. 22 (2009) 173–176. [87] B. Deplancke, H.R. Gaskins, Hydrogen sulfide induces serum-independent cell cycle entry in nontransformed rat intestinal epithelial cells, FASEB J. 17 (2003) 1310–1312. [88] G. Yang, K. Cao, L. Wu, R. Wang, Cystathionine gamma-lyase overexpression inhibits cell proliferation via a H2S-dependent modulation of ERK1/2 phosphorylation and p21Cip/WAK-1, J. Biol. Chem. 279 (2004) 49199–49205. [89] L. Rinaldi, G. Gobbi, M. Pambianco, C. Micheloni, P. Mirandola, M. Vitale, Hydrogen sulfide prevents apoptosis of human PMN via inhibition of p38 and caspase 3, Lab. Invest. 86 (2006) 391–397. [90] J. Lyons, A. Rauh-Pfeiffer, Y. Ming-Yu, X.M. Lu, D. Zurakowski, M. Curley, S. Collier, C. Duggan, S. Nurko, J. Thompson, A. Ajami, S. Borgonha, V.R. Young, L. Castillo, Cysteine metabolism and whole blood glutathione synthesis in septic pediatric patients, Crit. Care Med. 29 (2001) 870–877. [91] E. Disrutti, L. Sediari, A. Mencarelli, B. Renga, S. Orlandi, G. Russo, G. Caliendo, V. Santagada, G. Crino, J.L. Wallece, S. Fiorucci, 5-Amino-2-hydroxybenzoic acid 4-(5-thioxo-5H-[1,2]dithiol-3yl)-phenyl ester (ATB-429), a hydrogen sulfide-releasing derivative of mesalamine, exerts antinociceptive effects in a model of postinflammatory hypersensitivity, JPET 319 (2006) 447–458. [92] S. Fiorucci, S. Orlandi, A. Mencarelli, G. Caliendo, V. Santagada, E. Distrutti, L. Santucci, G. Cirino, J.L. Wallace, Enhanced activity of a hydrogen sulphidereleasing derivative of mesalamine (ATB-429) in a mouse model of colitis, Br. J. Pharmacol. 150 (2007) 996–1002. [93] L. Li, M. Salto-Tellez, C. Tan, M. Whiteman, P.K. Moore, GYY4137, a novel hydrogen sulfide-releasing molecule, protects against endotoxic shock in the rat, Free Radic. Biol. Med. 47 (2009) 103–113. [94] A. Sparatore, E. Perrino, V. Tazzari, D. Giustarini, R. Rossi, G. Rossoni, K. Erdman, H. Schröder, P. Soldato, Pharmacological profile of a novel H2Sreleasing aspiryn, Free Radic. Biol. Med. 46 (2009) 586–592. [95] J.L. Wallace, G. Caliendo, V. Santagada, G. Cirino, Markedly reduced toxicity of a hydrogen sulphide-releasing derivative of naproxen (ATB-346), Br. J. Pharmacol. 159 (2010) 1236–1246. [96] M. Iciek, A. Bilska, L. Ksiazek, Z. Srebro, L. Wlodek, Allyl disulfide as donor and cyanide as acceptor of sulfane sulfur in the mouse tissues, Pharmacol. Rep. 57 (2005) 212–218. [97] M.H. Stipanuk, Sulfur amino acid metabolism: pathways for production and removal of homocysteine and cysteine, Annu. Rev. Nutr. 24 (2004) 539–577. [98] W. Qiao, T. Chaoshu, J. Hongfang, D. Junbao, Endogenous hydrogen sulfide is involved in the pathogenesis of atherosclerosis, Biochem. Biophys. Res. Commun. 396 (2010) 182–186.

709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771

Please cite this article in press as: B. Olas, Hydrogen sulfide in hemostasis: Friend or foe?, Chemico-Biological Interactions (2014), http://dx.doi.org/ 10.1016/j.cbi.2014.04.006

Hydrogen sulfide in hemostasis: friend or foe?

Hydrogen sulfide (H2S) is a well known toxic gas that is synthesized from the amino acids: cysteine (Cys) and homocysteine (Hcy) by three enzymes: cys...
837KB Sizes 3 Downloads 6 Views