Vous êtes sur la page 1sur 6

Hydrogen Sulfide Is a Mediator of Cerebral

Ischemic Damage
Kun Qu, BSc; Christopher P.L.H. Chen, BMBCh, FRCP; Barry Halliwell, DPhil, DSc;
Philip K. Moore, PhD; Peter T.-H. Wong, PhD

Background and PurposeWe observed recently that elevated plasma cysteine levels are associated with poor clinical
outcome in acute stroke patients. In a rat stroke model, cysteine administration increased the infarct volume apparently
via its conversion to hydrogen sulfide (H2S). We therefore investigated the effects of H2S and the inhibition of its
formation on stroke.
MethodsCerebral ischemia was studied in a rat stroke model created by permanent occlusion of the middle cerebral
artery (MCAO). The resultant infarct volume was measured 24 hours after occlusion.
ResultsAdministration of sodium hydrosulfide (NaHS, an H2S donor) significantly increased the infarct volume after
MCAO. The NaHS-induced increase in infarct volume was abolished by the administration of dizolcilpine maleate (an
Downloaded from http://stroke.ahajournals.org/ by guest on September 7, 2017

N-methyl-D-aspartate receptor channel blocker). MCAO caused an increase in H2S level in the lesioned cortex as well
as an increase in the H2S synthesizing activity. Administration of 4 different inhibitors of H2S synthesis reduced
MCAO-induced infarct volume dose dependently. The potency of these inhibitors in effecting neuroprotection in vivo
appeared to parallel their potency as inhibitors of H2S synthesis in vitro. It also appeared that most of the H2S
synthesizing activity in the cortex results from the action of cystathionine -synthase.
ConclusionsThe present results strongly suggest that H2S plays a part in cerebral ischemic damage after stroke.
Inhibition of H2S synthesis should be investigated for its potential as a novel neuroprotective stroke therapy. (Stroke.
2006;37:889-893.)
Key Words: hydrogen sulfide N-methyl-D-aspartate receptor stroke

I n addition to being a risk factor of cardiovascular diseases,


elevated plasma homocysteine (Hcy) is strongly linked with
increased risk of acute stroke.1,2 Hcy may be converted by the
occur during brain ischemia and contribute to the pathophys-
iology of ischemic brain injury. Cys is not an agonist at the
N-methyl-D-aspartate (NMDA) receptor but is known to cause
action of cystathionine -synthase (CBS; EC 4.2.1.22) to cysta- neuronal cell death that can be prevented by NMDA antago-
thionine, which, in turn, is acted on by cystathionine -lyase nists.9,10 In this respect, it has been suggested that Cys exerts an
(CSE; EC 4.4.1.1) to form cysteine (Cys), thus suggesting a indirect action on the NMDA receptors via the excitatory amino
possible association of the actions of Cys and Hcy in stroke. acids or Cys derivatives such as S-nitrosocysteine or Cys
Plasma Cys has been reported to be as important a risk factor as sulfinate.9,11
Hcy in coronary heart disease.3 However, it has also been However, it is now known that Cys is also the precursor of
reported that plasma Cys remained unchanged in stroke patients the novel gaseous mediator hydrogen sulfide (H2S).12 It is
with hyperhomocysteinemia.4 interesting to note that the 2 enzymes involved in the
Cys has been demonstrated to cause neuronal death when conversion of Hcy to Cys are also the major enzymes
given orally to infant mice5 and is also an important mediator involved in the production of H2S from Cys, namely CBS and
of the pathology of brain injury in immature animals sub- CSE.13,14 Both enzymes use Cys as substrate to form H2S with
jected to hypoxic-ischemic brain injury.6 In rat hippocampal the byproduct serine, in the case of CBS, or pyruvate and
slices, Cys appears to be innocuous under normal conditions ammonia, in the case of CSE. The actions of H2S include, in
but causes toxicity to neurons deprived of glucose, oxygen, or the cardiovascular system, vasodilation and thus lowering of
both.7 Extracellular levels of Cys are also markedly elevated blood pressure, and in the brain, the enhancement of NMDA
after ischemic brain injury caused by carotid artery ligation in receptor-mediated responses such as the facilitation of the
Mongolian gerbils.8 Thus, elevation in extracellular Cys may induction of hippocampal long-term potentiation.15 The level

Received November 14, 2005; accepted November 30, 2005.


From the Departments of Pharmacology (K.Q., C.P.L.H.C., P.K.M., P.T.-H.W.) and Biochemistry (B.H.), Yong Loo Lin School of Medicine, National
University of Singapore, Kent Ridge, Singapore; and Department of Neurology (C.P.L.H.C.), National Neuroscience Institute, Singapore General Hospital
Campus, Singapore General Hospital.
Correspondence to Dr Peter T.-H. Wong, Department of Pharmacology, National University of Singapore, 18 Medical Dr, Singapore 117597. E-mail
phcwth@nus.edu.sg
2006 American Heart Association, Inc.
Stroke is available at http://www.strokeaha.org DOI: 10.1161/01.STR.0000204184.34946.41

889
890 Stroke March 2006

of endogenous H2S in the brain has been estimated at 50 the calculation.22 The calibration curve was linear from 0 to
to 160 mol/L,16,17 concentrations high enough to suggest 320 mol/L NaHS or 96 mol/L H2S.
physiological functions. Most recently, we observed that high
plasma Cys/cystine levels are associated with poor clinical Measurement of Endogenous Level of H2S
Rats were killed and the head was flash-frozen with liquid nitrogen.
outcome in acute stroke patients and that Cys neurotoxicity in
Cortical tissues were removed without thawing and then homoge-
cerebral ischemia in the rat is attenuated by inhibition of nized in ice-cold 50 mmol/L potassium phosphate buffer, pH 8.0
CBS.18 We therefore investigated the effects of H2S and the (12% wt/vol), with a Polytron homogenizer. The homogenate was
inhibition of its formation in a rat model of stroke. centrifuged (47 000g; 10 minutes; 4C) and the supernatant (75 L)
was mixed with 0.25 mL Zn acetate (1%) and 0.45 mL water for 10
Materials and Methods minutes at room temperature. TCA (10%; 0.25 mL) was then added,
centrifuged (14 000g; 10 minutes; 4C), and the clear supernatant
Animals and Drugs was collected and mixed with N,N-dimethyl-p-phenylenediamine
Male Wistar rats (200 to 250 g) obtained from the University Laboratory sulfate (20 mmol/L; 133 L) in 7.2 mol/L HCl and FeCl3
Animal Center, National University of Singapore were housed singly (30 mmol/L; 133 L) in 1.2 mol/L HCl. After 20 minutes, absor-
under natural light/dark (12 to 12 hours) cycle with food and water bance was measured as described above.
available ad libitum. All procedures were performed in accordance
with institutional guidelines, and all efforts were made to minimize
suffering and the number of rats used.
Reverse TranscriptasePolymerase Chain Reaction
Cys, DLpropargylglycine (PAG), aminooxyacetic acid (AOAA), Total mRNA from brain tissues were extracted using TriZol reagent
sodium hydrosulfide (NaHS), hydroxylamine (HA), -cyanoalanine (Invitrogen) as described.19 RNA (10 g) was reverse transcribed
Downloaded from http://stroke.ahajournals.org/ by guest on September 7, 2017

(-CNA), and dizolcilpine maleate (MK-801) were obtained from into cDNA using Avian Myeloblastosis Virus reverse transcriptase
Sigma. All drugs were dissolved in saline and administered by (Promega) and oligo dT12 as primers. The sequence of primers,
intraperitoneal injection. Control rats received saline only. annealing temperature/time, and PCR cycle are as follows: -actin
(internal standard; 870 bp; 60C/2 minutes; 29 cycles); sense:
Focal Cerebral Ischemic Stroke Model 5-ATCTGGCACCACACCTTCTACAATGAGCTGCG-3; anti-
Cerebral ischemia was induced by permanent unilateral occlusion of sense: 5-CGTCATACTCCTGCTTGCTGATCCACATCTGC-3;
the left middle cerebral artery (MCA) using a modified subtemporal CBS (559 bp; 58C/2 minutes; 32 cycles); sense: 5-ATGCTGCAG-
approach described previously.19 Briefly, the MCA was occluded AAAGGCTTCAT-3; antisense: 5-GTGGAAACCAGTCGGTGT-
proximal to the origin of the lenticulo-striate branches extending to CT-3; CSE (579 bp; 58C/2 minutes; 32 cycles); sense: 5-CGCA-
the point where it intersects the inferior cerebral vein by electrocau- CAAATTGTCCACAAAC-3; antisense: 5-GCTCTGTCCTTCT-
terizaton. Rectal temperature was maintained at 370.5C until CAGGCAC-3.
recovery from anesthesia. Control rats were sham operated by PCR amplification products were analyzed on a 1% agarose gel.
omitting only the occlusion. Bands were visualized with an ultraviolet transilluminator and band
intensities were quantified by a SynGene Multi Genius Bio Imaging
Measurement of Infarct Volume System.
Rats were decapitated 24 hours after MCA occlusion (MCAO).
Coronal sections (2 mm) of the cerebrum were then stained with Statistical Analysis
0.1% 2,3,5-triphenyltetrazolium chloride (TTC) solution at 37C for All comparisons were performed by 1-way ANOVA followed by
30 minutes and fixed using 4% formaldehyde in PBS. The size of the post hoc analysis with Bonferroni correction using SPSS for Win-
unstained infarcted area per section was analyzed (Olympus Micro
dows (v13). Data are expressed as meanSEM. The critical P level
Image Lite 4.0 system), and the true infarct volume was obtained by
set for significance is 0.05.
correcting for edema.20

H2S Synthesizing Activity in Cortical Homogenate


H2S production in cortical homogenates was studied as described by
Abe and Kimura12 with modification. Rat cerebral cortex was
homogenized in ice-cold 50 mmol/L potassium phosphate buffer, pH
8.0 (12% wt/vol), with a Polytron homogenizer. Homogenate (0.9
mL), preincubated at 37C with or without an inhibitor for 5 minutes
in a 20-mL glass vial, was then cooled on ice for 10 minutes before
L-Cys (10 mmol/L final concentration) and pyridoxal 5-phosphate
(2 mmol/L) were added. The final volume was 1 mL. A 2-mL tube
containing a piece of filter paper (0.51.5 cm) soaked with zinc
acetate (1%; 0.3 mL) was put inside the vial. The vial was then
flushed with a slow stream of nitrogen gas for 20 seconds and then
capped with an airtight serum cap. The vials were then transferred to
a 37C shaking water bath. After 90 minutes, trichloroacetic acid
(TCA; 50%; 0.5 mL) was injected into the reaction mixture through
the serum cap. Another 60 minutes was allowed for the trapping of
evolved H2S by the Zn acetate solution as Zn sulfide. Then the serum
cap was removed and N, N-dimethyl-p-phenylenediamine sulfate Figure 1. Dose-dependent effect of NaHS on infarct volume 24
(20 mmol/L; 50 L) in 7.2 mol/L HCl and FeCl3 (30 mmol/L; 50 hours after MCAO. NaHS (0.18 mmol/kg or otherwise stated in
L) in 1.2 mol/L HCl were added to the inner tube. After 20 minutes, parentheses) was injected intraperitoneally 10 minutes before
absorbance at 670 nm was measured with a microplate reader MCAO. MK-801 (3 mol/kg), injected intraperitoneally 10 min-
(Sunrise; TECAN). utes before NaHS administration, completely abolished the
The calibration curve of absorbance versus H2S concentration was effect of NaHS. The mean infarct volume was 17910 mm3
obtained by using NaHS solution of varying concentrations. When (100%) for control rats receiving saline only. n4. One-way
NaHS is dissolved in water, HS is released and forms H2S with H. ANOVA: F(3,12)7.389; P0.01. *P0.01 against control group
H2S concentration was taken as 30% of the NaHS concentration in by post hoc analysis with Bonferroni correction.
Qu et al Hydrogen Sulfide and Stroke 891

Figure 2. H2S levels in cortical tissues 24 hours after MCAO


with or without Cys loading. Cys (10 mmol/kg) was injected in- Figure 4. Cortical expression of CBS and CSE mRNA in sham-
traperitoneally 50 minutes before MCAO. n4. One-way operated and MCAO rats. RT-PCR was performed as described
ANOVA: F(2,9)52.917; P0.001. *P0.001 against the sham- in the Methods. No significant difference in expression of either
operated control group and **P0.02 against MCAO group by enzyme was detected between sham-operated and MCAO rats
post hoc analysis with Bonferroni correction. (independent sample t test). n3. Inset, Representative bands
for CBS, CSE, and -actin RT-PCR products obtained by gel
Downloaded from http://stroke.ahajournals.org/ by guest on September 7, 2017

electrophoresis.
Results
MCAO causes widespread tissue infarction predominantly in To gain further insight into the role played by H2S in this
the cortex of the occluded side of the brain. Administration of stroke model, the inhibitory effects of 2 CBS inhibitors
NaHS at 0.09 mmol/kg (IP) before MCAO had no significant (AOAA and HA) and 2 CSE inhibitors (-CNA and PAG) on
effect on the infarct volume, but at 0.18 mmol/kg, the infarct the H2S synthesizing activity in cortical homogenate was
volume was increased to 150% of control. NaHS was studied. All 4 inhibitors inhibited H2S production in vitro in
administered at a sublethal dose,17 and no increase in mortality a dose-dependent manner (Figure 5). AOAA exhibited the
was observed in this group of rats. Coadministration of the greatest potency with an IC50 value of 12.6 mol/L, reaching
NMDA receptor channel blocker MK-801 (3 mol/kg) com- 98% inhibition at a concentration of 0.5 mmol/L (data not
pletely abolished the proinfarct effect of NaHS (Figure 1). shown in Figure 5). The IC50 values for the other inhibitors
The endogenous level of H2S in the cerebral cortex almost are 0.5 mmol/L (HA), 2.5 mmol/L (-CNA), and 7.1 mmol/L
doubled in the lesioned cortex after MCAO when compared (PAG). In addition, the CSE inhibitors achieved only 70%
with sham-operated controls. As expected, H2S levels in- (-CNA) and 55% (PAG) inhibition at the highest concen-
creased further after previous Cys loading (Figure 2). In tration used (10 mmol/L).
addition, the H2S synthesizing activity, at maximal Cys Administration of each of the 4 inhibitors to rats before
concentration, in cortical homogenates also increased after MCAO revealed that all 4 compounds were able to reduce
MCAO (Figure 3). However, when the cortical expression of infarct volume in a dose-dependent manner (Figure 6).
CBS and CSE were investigated using RT-PCR, no signifi- Notably, their effectiveness in this respect paralleled their
cant difference in the expression of either enzyme was ability to inhibit H2S synthesis in vitro, thus AOAA was the
observed between sham-operated and MCAO rats (Figure 4). most effective at a dose of 0.05 mmol/kg followed by HA,
It is noted that the expression of CSE was observed to be
higher than that of CBS in both groups of animals.

Figure 5. Inhibition of H2S synthesizing activity in cortical ho-


mogenate by AOAA (), HA (), -CNA () and PAG (). The
H2S assay was performed as described in the Methods. Each
Figure 3. H2S synthesizing activity in cortical homogenate 24 point represents the meanSEM of 3 independent experiments
hours after MCAO. H2S assay was performed as described in determined in duplicate. The calculated IC50 values were
the Methods. n4. *P0.05 against sham-operated control rats 12.6 mol/L (AOAA), 0.5 mmol/L (HA), 2.5 mmol/L (-CNA),
by independent sample t test; t2.970; df6. and 7.1 mmol/L (PAG).
892 Stroke March 2006

MCAO lesioned cortex (Figures 2 and 3), these various


observations strongly suggest that H2S plays an important
role in tissue damage in the ischemic brain, possibly through
enhancement of NMDA receptor-mediated calcium overload.
Another possibility is that H2S influences the ischemic
infarction by altering cerebral blood flow. However, because
H2S causes vasodilation and vasodilators are generally cere-
broprotective, leading to reduced infarct size;23 this possible
mechanism is much less likely.
The ability of cortical tissue to increase production of H2S
very quickly after MCAO or Cys loading suggests that the
enzyme responsible for this conversion is not saturated by its
substrate in vivo. Abe and Kimura12 noted that CBS inhibitors
including AOAA completely inhibited the production of H2S
Figure 6. Dose-dependent effect of AOAA, HA, -CNA, and in rat whole brain homogenates, whereas CSE inhibitors,
PAG on infarct volume 24 hours after MCAO. Inhibitors were including PAG, were ineffective at a concentration of
injected intraperitoneally at various doses 50 minutes before 2 mmol/L. Abe and Kimura12 concluded that CBS is the
MCAO. The mean infarct volume was 19712 mm3 (100%) for
predominant enzyme responsible for H2S production in the
Downloaded from http://stroke.ahajournals.org/ by guest on September 7, 2017

the control rats receiving saline only. n5 to 6. One-way


ANOVA for AOAA: F(4,20)7.389, P0.01; HA: F(3,16)27.532, brain. It has also been reported that CBS is localized in most
P0.001; -CNA: F(3,18)5.753, P0.01; PAG: F(3,16)41.150, areas of the adult mouse brain but predominantly in the cell
P0.001. *P0.05 and **P0.01 against control group by post bodies and neuronal processes of Purkinje cells and Ammons
hoc analysis with Bonferroni correction.
horn neurons.24
Consistently, our present results (Figure 5) also suggest a
which was effective at 0.5 to 1.0 mmol/kg. It is interesting to
predominantly CBS-catalyzed production of H2S in the cere-
note that AOAA was not effective at higher doses (ie, up to
bral cortex. AOAA inhibited H2S production effectively with
0.5 mmol/kg). In contrast, -CNA and PAG were effective at
an IC50 value of 12.6 mol/L and caused almost complete
much higher doses of 1 or 2 mmol/kg. However, relative to
inhibition of H2S production at 0.5 mmol/L. In contrast, PAG,
-CNA, PAG appeared to be more effective in reducing
a potent CSE inhibitor, inhibited cortical H2S production with
infarct volume than its potency in inhibiting H2S production an IC50 value of 7.1 mmol/L, suggesting that PAG may be
suggested (Figure 5). acting as a low-affinity inhibitor of CBS in this instance
rather than as an inhibitor of CSE. It has been reported that
Discussion CSE is not expressed at detectable levels or expressed at a
Cys has been shown previously to increase infarct volume barely detectable level in the rat and mouse brain by Northern
after MCAO in a dose-dependent manner.18 This effect of or Western blot analysis.12 However, it has to be noted that
Cys was abolished by AOAA, a CBS inhibitor, suggesting the expression of CSE mRNA (by RT-PCR) is apparently
that Cys exerts its effect after conversion to H2S via the action higher than that of CBS (Figure 4), which is in stark contrast
of CBS in the brain. We show here that NaHS, an H2S donor, to the data obtained in the in vitro assay (Figure 6). It is
is also able to enhance the destructive effects of cerebral possible that CSE is expressed at the mRNA level but not at
ischemia, leading to a marked increase in the extent of tissue the protein level. More conclusive studies can be made only
damage. At a dose of 0.18 mmol/kg of NaHS, the effective when antibodies to both CBS and CSE become available.
dose of H2S is 0.06 mmol/kg based on a yield of 30%;21 All 4 inhibitors used reduced the MCAO-induced infarct
this is equivalent to only 0.6% of the effective dose of Cys at volume in a dose-dependent manner. The rank order of
10 mmol/kg. This is therefore consistent with the possibility potency was AOAAHAPAG-CNA (Figure 6).
that Cys increased the cerebral infarct by production of H2S. Significantly, the observed potencies of the compounds as
Moreover, the effects of both Cys (K. Qu and P.T.H. Wong, H2S synthesis inhibitors in vitro paralleled their effectiveness
unpublished data, 2005) and NaHS (Figure 1) were abolished in reducing MCAO infarct size in vivo. AOAA, as the most
by MK-801 pretreatment, confirming that H2S acts most potent inhibitor, significantly reduced infarct volume at a
likely by an effect via NMDA receptors. dose of 0.05 mmol/kg. Interestingly, at higher doses, AOAA
It has been reported previously that physiological concen- no longer exhibited any protective effects, probably indicat-
trations of H2S enhances NMDA receptor function through ing overinhibition of H2S formation, leading to detrimental
activation of adenylyl cyclase. Increased production of effects, supporting an important neuromodulator role for H2S
cAMP, observed in primary cultures of both neuronal and in the brain. It was further noted that at doses 0.5 mmol/kg,
glial cells, may lead to phosphorylation of the NMDA rats showed an unacceptably high mortality rate (data not
receptor subunits at specific sites by protein kinase A, shown).
resulting in the activation of NMDA receptor-mediated exci-
tatory postsynaptic current.22 Thus, in cerebral ischemia, H2S Summary
may enhance the NMDA receptor mediated excitotoxicity of The present results clearly demonstrate that H2S, produced
glutamate. Together with the observed increase in the endog- from Cys in the cerebral cortex most probably by CBS, is an
enous level of both H2S and H2S synthesizing activity in the important mediator of ischemic damage. H2S acts via the
Qu et al Hydrogen Sulfide and Stroke 893

NMDA receptor, which has become a prime target for stroke 12. Abe K, Kimura H. The possible role of hydrogen sulphide as an endog-
research over the past decade. Indeed, some NMDA antago- enous neuromodulator. J Neurosci. 1996;16:1066 1071.
13. Stipanuk MH, Beck PW. Characterization of the enzymic capacity for
nist and glycine antagonists have shown promise in clinical cysteine desulfhydration in liver and kidney of the rat. Biochem J. 1982;
trials.25,26 Current evidence suggests that H2S promotes ische- 206:267277.
mic damage by a direct degenerative effect on cerebral 14. Griffith OW. Mammalian sulfur amino acid metabolism: an overview.
neurons, although effect on cerebral blood flow may not be, Methods Enzymol. 1987;143:366 376.
15. Moore PK, Bhatia M, Moochhala S. Hydrogen sulfide: from the smell of
as yet, excluded. Whatever the mechanism of action, these
the past to the mediator of the future? Trends Pharmacol Sci. 2003;24:
results suggest, for the first time, that inhibition of H2S 609 611.
production using a CBS inhibitor may represent a novel 16. Savage JC, Gould DH. Determination of sulfide in brain tissue and rumen
therapeutic approach to the treatment of stroke. fluid by ion-interaction reversed-phase high-performance liquid chroma-
tography. J Chromatogr. 1990;526:540 545.
17. Goodwin LR, Francom D, Dieken FP, Taylor JD, Warenycia MW,
Acknowledgments Reiffenstein RJ, Dowling G. Determination of sulfide in brain tissue by
This work was supported by a Biomedical Research Council of gas dialysis/ion chromatography: post-mortem studies and two case
Singapore grant awarded to P.T.H.W. (PI), C.P.L.H.C., and B.H. reports. J Anal Toxicol. 1989;13:105109.
18. Wong PTH, Qu K, Chimon GN, Seah ABH, Chang HM, Wong MC, Ng YK,
References Rumpel H, Halliwell B, Chen CPLH. High plasma cyst(e)ine level may
1. Boysen G, Brander T, Christensen H, Gideon R, Truelsen T. Homo- indicate poor clinical outcome in acute stroke patients: possible involvement
cysteine and risk of recurrent stroke. Stroke. 2003;34:1258 1261. of hydrogen sulfide. J Neuropathol Exp Neurol. 2006;65:in press.
2. Del Ser T, Barba R, Herranz AS, Seijas V, Lopez-Manglano C, Domingo J, 19. Lu Q, Zhu YZ, Wong PTH. Angiotensin receptor gene expression in can-
Downloaded from http://stroke.ahajournals.org/ by guest on September 7, 2017

Pondal M. Hyperhomocyst(e)inemia is a risk factor of secondary vascular desartan mediated neuroprotection. NeuroReport. 2004;15:26432646.
events in stroke patients. Cerebrovasc Dis. 2001;12:9198. 20. Lin TN, He YY, Wu G, Khan M, Hsu CY. Effect of brain edema on
3. Ozkan Y, Ozkan E, Simsek B. Plasma total homocysteine and cysteine infarct volume in a focal cerebral ischemia model in rats. Stroke. 1993;
levels as cardiovascular risk factors in coronary heart disease. Int 24:117121.
J Cardiol. 2002;82:269 277. 21. Dombkowski RA, Russell MJ, Olson KR. Hydrogen sulfide as an endog-
4. Andersson A, Hutlberg B, Lindgren A. Redox status of plasma homo- enous regulator of vascular smooth muscle tone in trout. Am J Physiol
cysteine and other plasma thiols in stroke patients. Atherosclerosis. 2000; Regul Integr Comp Physiol. 2004;286:R678 R685.
151:535539. 22. Kimura H. Hydrogen sulfide induces cyclic AMP and modulates the
5. Olney JW, Ho OL, Rhee V. Cytotoxic effects of acidic and sulphur- NMDA receptor. Biochem Biophys Res Comm. 2000;267:129 133.
containing amino acids on the infant mouse central nervous system. Exp 23. Fournier A, Achard JM, Boutitie F, Mazouz H, Mansour J, Oprisiu R,
Brain Res. 1971;14:6176.
Fernandez L, Messerli F. Is the angiotensin II Type 2 receptor cerebro-
6. Barks JDE, Silverstein FS. Excitatory amino acids contribute to the
protective? Curr Hypertens Rep. 2004;6:182189.
pathogenesis of perinatal hypoxic-ischaemic brain injury. Brain Pathol.
24. Robert K, Vialard F, Theiry E, Toyama K, Sinet PM, Janel N, London J.
1992;2:235243.
Expression of the cystathionine synthase (CBS) gene during mouse
7. Schurr A, West CA, Heine MF, Rigor BM. The neurotoxicity of sulphur-
containing amino acids in energy-deprived rat hippocampal slices. Brain development and immunolocalization in adult brain. J Histochem
Res. 1993;601:317320. Cytochem. 2003;51:363371.
8. Slivka A, Cohen B. Brain ischaemia markedly elevates levels of the 25. Lees KR, Asplund K, Carolei A, Davis SM, Diener HC, Kaste M,
neurotoxic amino acid cysteine. Brain Res. 1993;608:3337. Orgogozo JM, Whitehead J. Glycine antagonist (gavestinel) in neuropro-
9. Olney JW, Zorumski C, Price MT, Labruyere J. L-Cysteine, a bicarbonate- tection (GAIN International) in patients with acute stroke: a randomised
sensitive endogenous excitotoxin. Science. 1990;248:596599. controlled trial. GAIN International Investigators. Lancet. 2000;355:
10. Mathisen GA, Fonnum F, Paulsen RE. Contributing mechanisms for 1949 1954.
cysteine excitotoxicity in cultured cerebellar granule cells. Neurochem 26. Lees KR, Lavelle JF, Cunha L, Diener HC, Sanders EA, Tack P, Wester P;
Res. 1996;21:293298. GAIN Phase II European Study Group. Glycine antagonist (GV150526) in
11. Janaky R, Varga V, Hermann A, Saransaari P, Oja SS. Mechanisms of acute stroke: a multicentre, double-blind placebo-controlled phase II trial.
L-cysteine Neurotoxicity. Neurochem Res. 2000;25:13971405. Cerebrovasc. Dis. 2001;11:2029.
Hydrogen Sulfide Is a Mediator of Cerebral Ischemic Damage
Kun Qu, Christopher P.L.H. Chen, Barry Halliwell, Philip K. Moore and Peter T.-H. Wong

Stroke. 2006;37:889-893; originally published online January 26, 2006;


Downloaded from http://stroke.ahajournals.org/ by guest on September 7, 2017

doi: 10.1161/01.STR.0000204184.34946.41
Stroke is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231
Copyright 2006 American Heart Association, Inc. All rights reserved.
Print ISSN: 0039-2499. Online ISSN: 1524-4628

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://stroke.ahajournals.org/content/37/3/889

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Stroke can be obtained via RightsLink, a service of the Copyright Clearance Center, not the Editorial Office.
Once the online version of the published article for which permission is being requested is located, click
Request Permissions in the middle column of the Web page under Services. Further information about this
process is available in the Permissions and Rights Question and Answer document.

Reprints: Information about reprints can be found online at:


http://www.lww.com/reprints

Subscriptions: Information about subscribing to Stroke is online at:


http://stroke.ahajournals.org//subscriptions/

Vous aimerez peut-être aussi