ArticlePDF Available

H 2 S Protects Against Methionine–Induced Oxidative Stress in Brain Endothelial Cells

Authors:

Abstract and Figures

Homocysteine (Hcy) causes cerebrovascular dysfunction by inducing oxidative stress. However, to date, there are no strategies to prevent Hcy-induced oxidative damage. Hcy is an H2S precursor formed from methionine (Met) metabolism. We aimed to investigate whether H2S ameliorated Met-induced oxidative stress in mouse brain endothelial cells (bEnd3). The bEnd3 cells were exposed to Met treatment in the presence or absence of NaHS (donor of H2S). Met-induced cell toxicity increased the levels of free radicals in a concentration-dependent manner. Met increased NADPH-oxidase-4 (NOX-4) expression and mitigated thioredxion-1(Trx-1) expression. Pretreatment of bEnd3 with NaHS (0.05 mM) attenuated the production of free radicals in the presence of Met and protected the cells from oxidative damage. Furthermore, NaHS enhanced inhibitory effects of apocynin, N-acetyl-l-cysteine (NAC), reduced glutathione (GSH), catalase (CAT), superoxide dismutase (SOD), Nomega-nitro-l-arginine methyl ester (L-NAME) on ROS production and redox enzymes levels induced by Met. In conclusion, the administration of H2S protected the cells from oxidative stress induced by hyperhomocysteinemia (HHcy), which suggested that NaHS/H2S may have therapeutic potential against Met-induced oxidative stress.
Content may be subject to copyright.
ANTIOXIDANTS & REDOX SIGNALING
Volume 11, Number 1, 2009
© Mary Ann Liebert, Inc.
DOI: 10.1089/ars.2008.2073
Original Research Communication
H
2
S Protects Against Methionine–Induced Oxidative
Stress in Brain Endothelial Cells
Neetu Tyagi, Karni S. Moshal, Utpal Sen, Thomas P. Vacek, Munish Kumar,
William M. Hughes Jr., Soumi Kundu, and Suresh C. Tyagi
Abstract
Homocysteine (Hcy) causes cerebrovascular dysfunction by inducing oxidative stress. However, to date, there
are no strategies to prevent Hcy-induced oxidative damage. Hcy is an H
2
S precursor formed from methionine
(Met) metabolism. We aimed to investigate whether H
2
S ameliorated Met-induced oxidative stress in mouse
brain endothelial cells (bEnd3). The bEnd3 cells were exposed to Met treatment in the presence or absence of
NaHS (donor of H
2
S). Met-induced cell toxicity increased the levels of free radicals in a concentration-depen-
dent manner. Met increased NADPH-oxidase-4 (NOX-4) expression and mitigated thioredxion-1(Trx-1) ex-
pression. Pretreatment of bEnd3 with NaHS (0.05 mM) attenuated the production of free radicals in the pres-
ence of Met and protected the cells from oxidative damage. Furthermore, NaHS enhanced inhibitory effects of
apocynin, N-acetyl-l-cysteine (NAC), reduced glutathione (GSH), catalase (CAT), superoxide dismutase (SOD),
N
-nitro-l-arginine methyl ester (L-NAME) on ROS production and redox enzymes levels induced by Met. In
conclusion, the administration of H
2
S protected the cells from oxidative stress induced by hyperhomocys-
teinemia (HHcy), which suggested that NaHS/H
2
S may have therapeutic potential against Met-induced ox-
idative stress. Antioxid. Redox Signal. 11, 25–33.
25
Introduction
M
ETHIONINE
(M
ET
)
IS AN ESSENTIAL AMINO ACID
and is me-
tabolized to homocysteine (Hcy), a sulfhydryl-contain-
ing nonprotein amino acid (32). It has been suggested that hy-
perhomocysteinemia (HHcy) is an independent risk factor for
neurodegenerative diseases such as dementia, Alzheimer’s
disease (AD) and stroke (5, 16, 22, 28, 29, 33). Previous stud-
ies have shown that Hcy is critically involved in the patho-
genesis of neurodegenerative disorders (11, 12). These unfa-
vorable vascular effects of Hcy are believed to be due to one
or both of the following: generation of reactive oxygen species
(ROS) [including hydrogen peroxide (H
2
O
2
) and superoxide
anion (O
2
-) (10, 20, 47)], and a decrease in endothelial nitric
oxide (NO) bioavailability (27, 31, 37) that play a critical role
in endothelial cell damage and dysfunction.
Although hydrogen sulfide (H
2
S) has been recognized as
a toxic gas, recent H
2
S research has been focused on its pro-
tective role in cardiovascular disease conditions. Like nitric
oxide (NO) and carbon monoxide (CO) (40, 41), which are
considered two gaseous transmitters, H
2
S has been shown
to be the third gaseous transmitter (39); moreover, H
2
S plays
important roles in several diseases. NO, CO, and H
2
S share
distinct properties which qualify them as gasotransmitters:
a) they are small molecules of gas; b) they are freely perme-
able across membrane, do not act via specific membrane re-
ceptors; c) they are produced enzymatically (17). H
2
S has
been demonstrated to stimulate heme oxygenase expression
and CO production and has bidirectional effects on the in-
ducible NO synthase (23). However, much less is known
about the physiological role of H
2
S.
H
2
S is endogenously generated in various mammalian tis-
sues from Met–Hcy–Cys metabolism through the action of
cystathione--synthetase (CBS) and/or cystathionine -lyase
(CSE) enzymes (21). H
2
S is a toxic gas and may act as a func-
tional regulator in the nervous and cardiovascular systems
Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky.
(14). Although its neuromodulatory role has been demon-
strated, little is known about its protective role in oxidative
stress. Interestingly, recent studies have shown that H
2
S is
neuroprotective (4, 5, 18, 19). Physiological concentrations of
H
2
S in plasma have been reported to be between 45 Mand
300 M(48, 50). At physiological concentrations, H
2
S inhibits
smooth muscle cell proliferation via the mitogen-activated
protein kinase pathway and protects the following tis-
sues/cells against oxidative stress: neurons, cardiomyocytes,
pancreatic -cells, and vascular smooth muscle cells (19.3).
H
2
S induces apoptosis by activating ERK and pro-caspase-3
(44). It has also been established that H
2
S directly opens the
K
ATP
channel and causes reduction of vasorelaxation and
transient blood pressure (49). However, the role of H
2
S in
the regulation of oxidative stress in endothelial cells is still
unclear. In addition, H
2
S acts as an endogenous scavenger
for reactive oxygen species and reactive nitrogen species
(RNS) (8, 9, 42). Therefore, the purpose of the present study
was to determine a potential role of H
2
S in preventing Met-
induced oxidative damage in bEnd3 endothelial cells by
modulating the production of ROS/RNS.
Materials and Methods
Materials
DCFH-DA(2,7-dichlorodihydrofluoresceindiacetate),
DL-methionine (Met), Dulbecco’s modified Eagle’s medium
(DMEM), dimethyl sulfoxide (DMSO), N-acetyl-l-cysteine
(NAC), sodium hydrosulfide (NaHS), reduced glutathione
(GSH), catalase (CAT), apocynin, superoxide dismutase
(SOD), N
-nitro-l-arginine methyl ester (L-NAME), and DL-
propargylglycine (PAG) were purchased from Sigma Chem-
ical Company (St. Louis, MO). Specific antibodies against
NOX-4 and Trx-1 were purchased from Santa Cruz Biotech-
nology (Santa Cruz, CA). Fetal bovine serum (FBS), phos-
phate-buffered saline (PBS), penicillin, and streptomycin were
obtained from Gibco (Grand Island, NY). 3-[4,5-dimethyl-
thiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT), super-
oxide dismutase (SOD) assay kit were obtained from Cay-
man Chemical (Ann Arbor, MI).
Methods
Cell culture. bEnd3, an immortalized mouse brain endo-
thelial cell line originally generated in 1990 (26), now com-
mercially available at American Type Culture Collection,
Manassas (ATCC, VA), was grown according to the sup-
plier’s instructions in DMEM supplemented with 4.5 g/l glu-
cose, 3.7 g/l sodium bicarbonate, 4 mMglutamine, 10% FBS,
100 U/ml penicillin, and 100 g/ml streptomycin, pH 7.4.
Cells were maintained in a humid chamber at 37°C in an
atmosphere of 95% air and 5% CO
2
in 25 cm
2
tissue culture
flasks (38). Confluent 25 cm
2
flasks were trypsinized and
seeded at a density of 0.5–1.0 10
4
cells/cm
2
on to 6–12-
well cell culture plate, and allowed to grow to 70%–80%
confluence.
Cell viability assay. Cell viability was determined by an
MTT assay as originally described by Mosmann (27). In brief,
bEnd3 cells were plated at a density of 10
5
cells/well on
to 96-well tissue culture plates and incubated with Met
(0.114 2.3 mM), NaHS (0.05 –0.5 mM), and 500 MPAG (in-
hibitor of CSE) in serum-free DMEM/F12 at 37°C for 24 h.
Then, 10 l of MTT reagent (5 mg/ml) was added to each
well, and the plates were incubated for another 4 h. The
medium was removed and wells were rinsed twice with PBS.
To each well, 100 l of crystal dissolving solution was added
at room temperature to dissolve the formazan crystals for 5
min. The absorbance was measured at 570 nm with a spectra-
max3000 plate reader (Molecular devices, Sunnyvale, CA).
Measurements of homocysteine. Homocysteine was mea-
sured by using high pressure liquid chromatography
(HPLC) as described earlier (33). HPLC analyses were per-
formed using Class-VP 5.0 chromatograph (Shimadzu,
Tokyo, Japan) containing a LC-10ADvp pump, a SIL-
10ADvp auto-injector, a CTO-10Avp column oven, and a
SPD-10Avp detector. The temperature inside the column was
maintained at 37°C during analyses.
Sample preparation. Culture supernatants were collected
and centrifuged to remove cell debris. To determine the Hcy
level in the supernatants, 200 l of supernatant was diluted
with 100 l of water and then 300 l of 9 Murea (pH 9.0)
was added. Fifty microliters of n-amyl alcohol was added to
the solution as an antifoaming agent. Reduction of disulfides
and cleavage of the protein-bound sulfur-containing amino
acids was performed by the addition of 50 l of NaBH
4
so-
lution (10%, wt/vol) in 0.1 NNaOH. To perform the reac-
tion, samples were incubated in a water bath at 50°C for 30
min. Samples were cooled at room temperature, and the re-
action was stopped by the addition of 500 l of 20%
trichloroacetic acid. The proteins were separated by cen-
trifugation for 4 min at 12,000 g, and supernatants were fil-
tered using a 0.45 m Millipore filter (32).
Measurement of H
2
S production. bEnd3 (10
5
cells/well)
were grown briefly in a 10 cm
2
dish. Cells became confluent
following 24 h of treatment with Met at different concentra-
tions. H
2
S concentration in bEnd3 cells was measured as de-
scribed previously (7, 48, 50).
Intracellular fluorescence measurement of reactive oxygen
species. In order to measure the oxidized DCF levels in cells,
we used the probe, 2-7-dichlorodihydrofluorescein diacetate
(H
2
DCF-DA), as described previously (36). This membrane-
permeable probe enters the cells and produces a fluorescent
signal after intracellular oxidation by ROS. bEnd3 (10
5
cells/well) were grown briefly in 96-well plates. Cells were
grown to confluence and treated for 24 h in serum-free
DMEM/F12 media with or without the following: Met, NaHS,
other various agents at different concentrations. The cells were
washed with PBS, loaded with probe, DCFH-DA (10 M), and
incubated in dark for 2 h at 37°C in PBS. Thereafter, the cells
were washed three times with PBS to remove the excess probe.
Oxidized DCF was quantified by monitoring the DCF fluo-
rescence intensity with excitation at 485 nm and emission at
530 nm with a spectra-max3000 plate reader (Molecular de-
vices). Values were expressed in arbitrary units.
In situ labeling of ROS. bEnd3 cells were grown on 8-well
cover glass plates and serum starved before the treatments
with the following: 1.14 mMMet, 0.05 mMNaHS, or 1.14
mMMet
0.05 mMNaHS for 24 h. Ros formation was vi-
sualized as described previously (36, 37).
TYAGI ET AL.26
Measurement of intracellular superoxide levels. The intra-
cellular superoxide anion radicals were detected using su-
peroxide dismutase (SOD) assay, using a kit from Cayman
Chemicals. Briefly, bEnd3 were plated and treated with dif-
ferent agents in serum free DMEM/F12 media at 37°C for 24
h. Then cells were scraped with a rubber policeman and son-
icated in cold 20 mMHEPES buffer, pH 7.2, containing 1 mM
EGTA, 210 mMmannitol, and 70 mMsucrose, according to
the manufacturer’s recommendation. SOD activity was de-
termined by spectrophotometry as the ability to inhibit the
reduction of nitroblue tetrazolium (NBT) induced by xan-
thine-xanthine oxidase (1).
Immunoblot analysis. Cells were lysed in ice-cold-modi-
fied RIPA lysis buffer (Tris-HCl, 50 mM, pH 7.4; NP-40, 1%;
0.25% Na-deoxycholate, 150 mMNaCl; 1 mMEDTA; 1 mM
PMSF; 1 g/ml each of aprotinin, leupeptin, pepstatin; 1 mM
Na
3
VO
4
; 1 mMNaF). Protein content of the lysate was de-
termined using BCA protein assay (Pierce, Rockford, IL) kit.
Protein samples were mixed with 1:1 vol/vol ratio with 2X
sample loading buffer [800 l glycerol, 1 ml 0.5 mMTris-
HCl (pH 6.8), 1.6 ml 10% (wt/vol) SDS, 400 l 2-mercap-
toethanol, 400 l 0.05% (wt/vol) Bromophenol blue], boiled
at 95–100°C for 5 min. Samples were cooled to room tem-
perature and centrifuged to precipitate cell debris. Equal
amounts of protein (20 g) for each group were resolved by
10–15% SDS-PAGE. Protein was then electrophoretically
transferred to a nitrocellulose membrane (BioRad, Hercules,
CA). Transferred protein was blocked with 5% nonfat dry
milk in TBS-T (50 mMTris-HCl, 150 mMNaCl, 0.1% Tween-
20, pH 7.4) for 1 h at room temperature. The blot was then
incubated with appropriate primary antibody in blocking so-
lution according to the supplier’s specific instructions. Next,
the blot was washed with TBS-T three times for 10 min each.
The blots were incubated with appropriate horseradish per-
oxidase-conjugated secondary antibody for 2 h at room tem-
perature. Four more 10 min washes were performed, and
ECL Plus substrate (Amersham Biosciences, Pittsburgh, PA)
was applied to the blot for 5 min. The blot was developed
using X-ray film (RPI Corp, Inc., Mount Prospect, IL) with a
Kodak 2000A developer (Eastman Kodak, Rochester,NY).
Image analysis was performed using UMAX PowerLock II
(Taiwan, Republic of China).
Data analysis and statistics. Results were expressed as
means SEM from at least seven independent experiments.
Both paired and unpaired Student’s ttests were used, where
appropriate, for comparing the mean values between con-
trol and tested groups. The difference between mean values
of multiple groups was analyzed by one-way analysis of
variance (ANOVA), followed by a Scheffe’s post-hoc analy-
sis. Statistical significance was considered at p0.05. The
arbitrary densitometry units (AU) were represented as per-
centage relative to control.
Results
Effect of NaHS on Met-induced cytotoxicity
To determine the protected role of hydrogen sulfide on
Met-induced cytotoxicity, bEnd3 were cultured with Met
with or without NaHS (donor of H
2
S) at different concen-
trations for 24 h. The cell death was significantly increased
in Met (0.114–2.3 mM) and NaHS (0.25–0.5 mM)-treated cells
after 24 h compared to untreated cells. This effect was not
observed when cells were treated with lower concentrations
of Met or NaHS (Fig. 1A and B). The increase in cell death
by high Met (1.14 mM) was attenuated by the pre-treatment
with NaHS (0.05 and 0.1 mM), respectively (Fig. 1C). But this
effect was reduced significantly by PAG (inhibitor of CSE)
pretreatment (Fig. 1D).
H
2
S INHIBITS METHIONINE–INDUCED OXIDATIVE STRESS 27
FIG. 1. Effect of hydrogen sulfide
on Met–induced cytotoxicity. (A)
Viability of bEnd3 cells treated with
different concentrations of Met for
24 h; n7; *p0.05; **p0.01. (B)
Cell viability measured after treat-
ment with different concentrations
of NaHS for 24 h; n7; *p0.05;
**p0.01. (Cand D) Cells were in-
cubated with Met (1.14 mM) for 24
h in the presence or absence of
NaHS or PAG; in (C) n7 in each
group; *p0.05; **p0.05; ***p
0.05. In (D), n7 in each group,
*p0.05;
#
p0.05;
$
p0.01;
p
0.01.
Role of NaHS in Hcy accumulation
Cell culture medium was collected after 24 h from control
(0.114 mMMet), high (1.14 mMMet), NaHS (0.05 mM), and
1.14 mMMet NaHS (0.05 mM) treated cells. The medium
was analyzed by HPLC. There was increased accumulation
of Hcy in the high Met (1.14 mMMet)-treated group com-
pared to that in control (Fig. 2A). Hcy levels were 3.6 0.5,
23.38 3.4, 5.1 0.6, and 11.3 1.2 Min control Met, high
Met, NaHS, and high Met NaHS treated cells, respectively.
These levels were similar and largely comparable with in vivo
studies (4). The increase in Hcy accumulation in the high Met
group was attenuated by NaHS treatment, which suggested
that H
2
S was a potent inhibitor of Hcy formation.
H
2
S levels
To determine the effect of endogenously generated H
2
S,
bEnd3 cells were treated with different concentrations (0.114–
2.3 mM) of Met for 24 h. H
2
S production rate was markedly
increased in a concentration-dependent manner (Fig. 2B).
NaHS attenuated Met-induced increases
in intracellular ROS
Because the cytotoxicity of Met is known to be mediated
mainly by oxidative stress, we investigated whether NaHS
affects ROS formation by high Met using DCFH-DA fluo-
rescence. Incubation of cells with different concentrations
(0.114–2.3 mM) of Met for 24 h resulted in significant in-
creases in ROS production in comparison with untreated
cells (Fig. 2C). When the cells were treated with Met (1.14
mM) for different time periods (6, 12, 24 h), the 24 h treat-
ment showed significant increases in ROS production (data
not shown). When cells were incubated with antioxidant
(NAC) or H
2
O
2
scavenger (CAT), there were marked de-
creases in ROS production induced by the treatment with
Met (1.14 mM, Fig. 2D). Furthermore, addition of NaHS (0.05
mM) significantly increased the inhibitory effects of NAC on
Met-induced ROS production, while the effect could not be
found in the CAT-treated group.
Interestingly, in-situ labeling (Fig. 3A–D) showed that the
levels of intracellular ROS were increased in cells treated
TYAGI ET AL.28
FIG. 2. Effect of hydrosulfide on homo-
cysteine accumulation. (A) Data from chro-
matography of homocysteine from cell cul-
ture medium. The effect of the addition of
Met (1.14 mM) and NaHS (0.05 mM) on Hcy
levels; *p0.05 compared to control (CT),
**p0.01compared to Met. (B) The mea-
surement of H
2
S in culture media after treat-
ment with different concentrations of Met
for 24 h. The cells were collected and ho-
mogenized to measure the H
2
S production
rate. N 7 for each group, *p0.05; **p
0.01. (C) ROS production, detected by 5-(6)-
chloromethyl-2,7-dichlorodihy-
droflurorescein diacetate (DCFH-DA) stain-
ing after incubation of cells with Met at
various concentrations for 24 h, *p0.05;
**p0.01. (D) Effect of hydrogen sulfide on
Met-induced reactive oxygen species (ROS)
production in bEnd3 cells, ROS production
was detected after treatment with Met (1.14
mM) in the presence or absence of NAC (50
M), CAT (5 /ml) with or without NaHS,
respectively; n7 for each group, *p
0.05; **p .01;
##
p0.01;

p0.01.
FIG. 3. In situ labeling of ROS. bEnd3 cells were grown on
glass 8-well chambers and exposed to different treatments for
24 h. ROS production was evaluated by staining the cells with
DCFH-DA. Images were acquired by laser confocal microscope
(FluoView 1000) at an excitation of 488 nm and emission of 525
nm representative micrograph. (A) Control; (B) 1.14 mMMet;
(C) 0.05 mMNaHS, (D) 1.14 mMMet 0.05 mMNaHS; mag-
nification 100. (For interpretation of the references to color in
this figure legend, the reader is referred to the web version of
this article at www.liebertonline.com/ars).
with Met (1.14 mM, Fig. 3B), indicated by the increases in
DCFH-DA fluorescence. However, when cells were treated
with both NaHS (0.05 mM) and Met (1.14 mM), DCFH-DA
fluorescence was decreased (Fig. 3D). This suggested that
Met-induced intracellular ROS accumulation was attenuated
by hydrogen sulfide. Cells treated with NaHS (0.05 mM)
alone showed weak DCFH-DA fluorescence (Fig. 3C), simi-
lar to that of untreated cells (Fig. 3A).
NaHS attenuated Met-induced peroxynitrite formation
The generation of ONOO
requires rapid interaction of
NO and O
2
.
. To determine the effects of the interaction of
Met and NaHS on ONOO
formation, cells were treated
with different concentrations (0.114–2.3 mM) of Met for 24 h
in the presence or absence of the NOS inhibitor, L-NAME
(100 M), NADPH oxidase inhibitor apocynin (100 M),
O
2
.
scavenger SOD (200 U/ml), with or without NaHS (0.05
mM). Figure 4A shows a concentration-dependent increase
in Met-induced ONOO
formation. However, when bEnd3
cells were co-treated with NaHS, Met (1.14 mM), apocynin,
or SOD, it ameliorated the inhibitory effect of apocynin, SOD,
and L-NAME, respectively (Fig. 4B).
NaHS attenuated Met-induced superoxide
anion production
To determine the protected role of hydrogen sulfide on
Met-induced superoxide anion, we examined the release of
superoxide anion by chemiluminescence assay. Figure 4C
H
2
S INHIBITS METHIONINE–INDUCED OXIDATIVE STRESS 29
FIG. 4. Effect of H
2
S on Met-induced
generation of peroxynitrite (ONOO
).
(A) The bEnd3 cells were treated with Met
1.14 mM, Met 1.14 mML-NAME (100
M) or Met (1.14 mM), L-NAME (100
M)NaHS (0.05 mM) for 24 h and ROS
were detected by using DCFH-DA probe;
**p0.05 compared to corresponding Met
treatment. p0.05 compared to Met
and L-NAME (100 M) treatment. (B) ROS
production was detected after bEnd3 cells
were incubated for 24 h with Met (1.14
mM) in the presence or absence of apoc-
ynin (100 M) or SOD (200 U/ml), with or
without NaHS (0.05 mM), respectively.
N7 in each group; *p0.05; **p0.01;
##
p0.01;

p0.05;
p0.05. (C) Met-
induced superoxide (O
2
.
) production and
effect of H
2
S in bEnd3: O
2
.production
was measured after bEnd3 cells were
treated with different concentrations of
Met for 24 h. (D) O
2
.
production was de-
termined in the treated cells after 24 h in-
cubation with NaHS at different concen-
trations. N7 for each group. *p 0.05
compared to control.
FIG. 5. Effect of hydrogen sulfide on Met-sup-
pressed thioredoxin (Trx) expression. (A) bEnd3
cells were cultured and treated with different con-
centrations (0.05–0.1 mM) of NaHS alone for 24h;
*p0.05 compared to control. (B) bEnd3 cells were
treated with Met (1.14 mM) in the presence or ab-
sence of the antioxidant NAC (50 M), GSH (1 mM),
with or without NaHS, respectively, for 24 h. Trx
protein was measured in cell lysates by Western
blot analysis, and membranes were stripped and
reprobed with -actin for equal loading. Bottom:
graphical presentation of Trx (fold change over con-
trol). N7 for each group. *p 0.05 vs. untreated
cells,
#
p0.05 or
##
p0.01 vs. cells treated with
Met in.
p0.05 vs. cells treated with Met NAC,

p0.05 vs. cells treated with Met GSH.
shows that 24 h of incubation with Met (1.14 mM) signifi-
cantly increased O
2
.
levels in a concentration-dependent
manner. The bEnd3 cells were incubated with different con-
centrations (0.05–0.25 mM) of NaHS for 24 h. The O
2
.
pro-
duction was significantly increased only in 0.25 mMNaHS-
treated cells, as compared to untreated cells (Fig. 4D).
Effect of NaHS on redox enzymes levels
To determine the protective role of hydrogen sulfide on
Met-induced imbalance between the redox enzymes, we ex-
amined the Trx-1 and NOX-4 protein levels by Western blot
analysis. Incubation of cells with 0.05–0.1 mMNaHS for 24
h resulted in a statistically significant increase in Trx-1 pro-
tein expression level (Fig. 5A) in comparison with untreated
cells. Met (1.14 mM) alone resulted in a significant decrease
in Trx-1 protein expression levels in comparison to the un-
treated control (Fig. 5B). Simultaneous incubation of Met
(1.14 mM) with either NAC (50 M) or GSH (1 mM) resulted
in a significant increase of the Met downregulated Trx-1 pro-
tein expression compared with Met alone. Furthermore,
when bEnd3 cells were treated with a combination [Met (1.14
mM); Met NAC; Met NAC NaHS; Met GSH; or
Met GSH NaHS], the treatment reduced the effect of
Met on Trx-1 protein expression as shown in Fig. 5B.
There was a concentration-dependent decrease in NOX-4
protein expression (Fig. 6A) after cells were incubated with
NaHS (0.05 or 0.1 mM) for 24 h. Met (1.14 mM) alone sig-
nificantly induced NOX-4 protein expression (Fig. 6A). Met-
induced NOX-4 protein expression was markedly decreased
in cells when they were pretreated with either NAC (50 M)
or GSH (1 mM). Furthermore, addition of NaHS (0.05 mM)
significantly increased the inhibitory effects of NAC and
GSH on Met-induced NOX-4 protein production in compar-
ison with that in the absence of NaHS (Fig. 6).
Discussion
Our present study attempted to examine a novel link be-
tween the protective role of H
2
S towards oxidative stress
caused by HHcy in brain endothelial cells. Methionine, an
essential amino acid, is converted to Hcy that then promotes
neurodegenerative diseases through endothelial dysfunction
(24, 25, 34). It is important to note that there are other re-
spondents to increases in ROS, such as HO-1. Homocysteine
generates ROS via the auto-oxidation of the thiol group (36,
44) or by decreasing the endothelial heme oxygenase-1 (HO-
1) activity (30). The transcriptional upregulation of the HO-
1 gene downregulated intracellular ROS (29). H
2
S is an en-
dogenous metabolic product of Met by the trans-sulfuration
pathway that is dependent on two important enzymes: CBS
and CSE (39). It has been demonstrated that H
2
S reacts with
at least four different ROS, superoxide radical anion, hy-
drogen peroxide, peroxynitrite, and hypochorite (8, 9, 41).
All these compounds are highly reactive and their interac-
tion with H
2
S resulted in the protection of proteins and lipids
from ROS/RNS-mediated damage (41, 42). Therefore, we hy-
pothesize that H
2
S may protect the cerebro-vasculature
against Met-induced endothelial damage.
Our present study results show that high Met (1.14 mM)
significantly decreased bEnd3 cells viability. Expectedly, the
addition of NaHS (0.05 or 0.1 mM) significantly increased
the cell viability as compared to the cells treated with high
Met. Although H
2
S (0.1 mM) reduced cell viability, this
was merely the effect of chemical cytotoxicity rather than
physiological effect. But this effect was reduced significantly
by PAG pretreatment (Fig. 1D), a result consistent with pre-
TYAGI ET AL.30
FIG. 6. Effect of hydrogen sulfide on Met-In-
duced NOX-4 expression. (A) Serum-starved
bEnd3 cells were treated for 24 h with different
concentration (0.05–0.1 mM) of NaHS alone for 24
h; *p 0.05 compared to control. (B) Serum-
starved bEnd3 cells were either left untreated or
were treated with Met (1.14 mM) in the presence
or absence of the antioxidant NAC (50 M), GSH
(1 mM) with or without NaHS, respectively, for
24 h. After treatment, cell lysates were analyzed
with 10–15% SDS-PAGE and subsequently by
Western blot analysis; membranes were stripped
and reprobed with -actin for equal loading. Bot-
tom: graphical presentation of NOX-4 (fold change
over control). N7 for each group, *p 0.05 vs.
untreated cells.
p0.05;

p0.05 vs. cells
treated with Met group.
#
p0.05 vs. cells treated
with Met NAC,
##
p0.01 vs. cells treated with
Met GSH.
FIG. 7. Schematic presentation of proposed mechanism
for the protective role of H
2
S towards Met-induced oxida-
tive stress in bEnd3 cells.
vious studies (32, 36, 43). PAG is an inhibitor of CSE, an en-
zyme responsible for endogenous H
2
S formation. We found
that, in the presence of PAG, cell viability was decreased in
the setting of high Met.
The rapid interaction of superoxide with nitric oxide gen-
erated peroxynitrite, a potent mediator of oxidant-induced
cellular injury (15, 34). In the present study, we demonstrated
that ROS production was increased in a concentration-de-
pendent manner after bEnd3 cells were treated with differ-
ent doses of Met for 24 h. Met-induced ROS production was
effectively blocked by NAC and H
2
O
2
scavenger, CAT, and
concentration-dependently inhibited by the NOS inhibitor,
L-NAME. This provides evidence that Met can induce not
only H
2
O
2,
but also ONOO
generation in mouse brain en-
dothelial cells (Fig. 2). This finding is supported by the ob-
served inhibitory effect of SOD and the effect of NADPH ox-
idase inhibitor, apocynin, on ROS formation (Fig. 3).
According to previous studies, H
2
S worked as a scavenger
of oxygen-derived free radicals (9), which could contribute
to the protective role of NaHS against the toxicity of H
2
O
2
in vitro and in vivo model (45). We observed that Met-induced
O
2
production was markedly reduced by the O
2
scav-
engers (SOD) as well as by NaHS (Fig. 4).
In addition, H
2
S enhanced NO production via ERK1/2 ac-
tivation, which suggested that H
2
S may cooperate with NO
in modulating their biological effects (13). Our results
showed that high levels of H
2
S can induce ROS and RNS for-
mation, but low levels of H
2
S can decrease H
2
O
2
, ONOO
,
and O
2
generation induced by Met in bEnd3 cells. Fur-
thermore, our results indicated that low concentrations of
H
2
S combined with certain agents, such as NAC, apocynin,
SOD, or L-NAME, may synergistically increase their antiox-
idant effects, scavenge ROS, and protect vascular endothe-
lium from Met-induced oxidant stress and cytotoxicity (Fig.
4). This is in contrast to functioning directly as an antioxi-
dant. This protective role of H
2
S may be similar to the pro-
tection of neurons from oxidative stress (18). The failure of
NaHS combined with CAT to reduce Met-induced ROS for-
mation might be explained by the proposed low efficiency
of CAT compared with NAC in removing H
2
O
2
at low con-
centrations (46). H
2
S protects the cell damage by decreasing
the ROS production by increasing SOD. The mechanism of
Met-induced oxidative stress is not yet well known. Some
studies suggested that Met-induced oxidative stress is pos-
sible through an NADPH oxidase-mediated pathway (36).
Our results showed that Met (1.14 mM) significantly in-
creased NOX-4 expression and, consequently, decreased Trx
expression. bEnd3 cells treated with a combination of high
Met, antioxidant, and NaHS may synergistically increase Trx
expression and decrease NOX-4 expression, in comparison
with the group treated with Met alone.
In this study, we employed different approaches to de-
termine the protective role of H
2
S on Met–induced oxidative
damage. We presented evidence that H
2
S is an important
modulator of cellular cytotoxicity via redox cell pathways in
the pathogenic conditions associated with HHcy. In conclu-
sion, the data presented here provides a new mechanism by
which H
2
S reduces oxidative damage induced by Met. Thus,
H
2
S protected bEnd3 cells from oxidative damage. When
considering previous studies along with our results with re-
spect to antioxidant activity of H
2
S and its effect on Met, we
suggest that administration of H
2
S might be an interesting
potentially preventive strategy for reducing cerebrovascular
complications in hyperhomocysteinemia (Fig. 7). However,
the molecular mechanisms of a putative protective role of
H
2
S in neurogenerative pathogenesis should be further in-
vestigated.
Limitations
In “mild” human hyperhomocysteinemia (which is asso-
ciated with an increased neurodegenerative diseases),
plasma Hcy levels range from 15 to 30 mol/l. However,
only a fraction of total plasma Hcy is in the reduced form in
vivo. The concentrations used in the present study represent
a 100-fold dose. Three ranges of hyperhomocysteinemia
are defined as follows: moderate (16–30 M), intermediate
(31–100 M), and severe (100 M) (2, 6). Extracellular thi-
ols are oxidized. Only a fraction of total plasma Hcy is in the
reduced form in vivo and in vitro. NaHS, a donor of H
2
S, was
used at physiologically relevant concentrations (41).
Abbreviations
bEnd3, mouse brain microvascular endothelial cells; CAT,
catalase; CBS, cystathione--synthetase; CSE, cystathionine
-lyase; GSH, reduced glutathione; Hcy, homocysteine;
HHcy, hyperhomocysteinemia; H
2
S, hydrogen sulfide; L-
NAME, N
-nitro-l-arginine methyl ester; Met, methionine;
NAC, N-acetyl-l-cysteine; NaHS, sodium hydrogen sulfide;
NOX4, NADPH-oxidase-4; PAG, DL-propargylglycine. ROS,
reactive oxygen species; SOD, superoxide dismutase; Trx-1
,thioredxion-1.
Acknowledgments
Supported in part by the National Institutes of Health
grants HL-75185, HL-71010, and NS-51568.
References
1. Beauchamp C and Fridovich I. Superoxide dismutase: Im-
proved assays and an assay applicable to acrylamide gels.
Anal Biochem 44: 276–287, 1971.
2. Bonaventura D, Tirapelli CR, Haddad R, Höehr NF, Eberlin
MN, and de Oliveira AM. Chronic methionine load-induced
hyperhomocysteinemia enhances rat carotid responsiveness
for angiotensin II. Pharmacology 70: 91–99, 2004.
3. Elrod JW, Calvert JW, Morrison J, Doeller JE, Kraus DW, Tao
L, Jiao X, Scalia R, Kiss L, Szabo C, Kimura H, Chow CW,
and Lefer DJ. Hydrogen sulfide attenuates myocardial isch-
emia-reperfusion injury by preservation of mitochondrial
function. Proc Natl Acad Sci USA. 104: 15560–15565, 2007.
4. Dayal S, Wilson KM, Leo L, Arning E, Bottiglieri T, and
Lentz SR. Enhanced susceptibility to arterial thrombosis in
a murine model of hyperhomocysteinemia. Blood 108:
2237–2243, 2006.
5. Faraci FM and Lentz SR. Hyperhomocysteinemia oxidative
stress and cerebral vascular dysfunction. Stroke 35: 345–347,
2004.
6. Garlick PJ. Toxicity of methionine in humans. J Nutr. 136:
1722S–1725S, 2006.
7. Geng B, Cui Y, Zhao J, Fang Yu, Zhu Y, Xu G, Zhang Z,
Tang C, and Du J. Hydrogen sulfide downregulates the aor-
tic L-arginine/nitric oxide pathway in rats. Am J Physiol
Regul Integr Comp Physiol 293: R1608-R1618, 2006.
8. Geng B, Yang J, Qi Y, Zhao J, Pang Y, Du J, and Tang C. H
2
S
H
2
S INHIBITS METHIONINE–INDUCED OXIDATIVE STRESS 31
generated by heart in rat and its effects on cardiac function.
Biochem. Biophys. Res. Commun 313: 362–368, 2004.
9. Geng B, Chang L, Pan C, Qi Y, Zhao J, Pang Y, Du Y, and
Tang C. Endogenous hydrogen sulfide regulation of myo-
cardial injury induced by isoproterenol. Biochem Biophys Res
Commun 318: 756–763, 2004.
10. Heinecke JW, Rosen H, Suzuki LA, and Chait A. The role of
sulfur-containing amino acids in superoxide production and
modification of low density lipoprotein by arterial smooth
muscle cells. J Biol Chem 262: 10098–10103, 1987.
11. Ho PI, Collins SC, Dhitavat S, Ortiz D, Ashline D, Rogers E,
and Shea TB. Homocysteine potentiates beta-amyloid neu-
rotoxicity: Role of oxidative stress. J. Neurochem 78: 249–253,
2001.
12. James SJ, CutlerP, Melnyk S, Jernigan S, Janak l, Gaylor DW,
and Neubrander JA. Metabolic biomarkers of increased ox-
idative stress and impaired methylation capacity in children
with autism. Am J Clin Nutr 80: 1611–1617, 2004.
13. Jeong SO, Pae HO, Oh GS, Jeong GS, Lee BS, Lee S, Kim YD,
Rhew HY, Lee KM, and Chung HT. Hydrogen sulfide po-
tentiates interleukin-1-induced nitric oxide production via
enhancement of extracellular signal-regulated kinase acti-
vation in rat vascular smooth muscle cells. Biochem Biophys
Res Commun 345: 938–944, 2006.
14. Kamoun P. Endogenous production of hydrogen sulfide in
mammals. Amino acids 26: 243–254, 2004.
15. Kanani PM, Sinkey CA, Browning RL, Allaman M, Knapp
HR, and Haynes WG. Role of oxidant stress in endothelial
dysfunction produced by experimental hyperhomocyst(e)-
inemia in humans. Circulation 100: 1161–1168, 1999.
16. Kang SS, Wong PWK, and Malinow MR. Hyperhomo-
cyst(e)inemia as a risk factor for occlusive vascular disease.
Ann Rev Nutr 12: 279–298, 1992.
17. Kasparek MS, Linden DR, Kreis ME, and Sarr MG. Gaso-
transmitters in the gastrointestinal tract. Surgery 143: 455–
459. 2008.
18. Kimura Y and Kimura H. Hydrogen sulfide protects neu-
rons from oxidative stress. FASEB J 18: 1165–1167, 2004.
19. Kimura Y, Dargusch R, Schubert D, and Kimura H. Hydro-
gen sulfide protects HT22 neuronal cells from oxidative
stress. Antioxid Redox Signal 8: 661–670, 2006.
20. Kotamraju S, Konorev EA, Joseph J, and Kalyanaraman B.
Doxorubicin-induced apoptosis in endothelial cells and car-
diomyocytes is ameliorated by nitrone spin traps and ebse-
len. Role of reactive oxygen and nitrogen species. J Biol Chem
275: 33585-92, 2000.
21. Levonen AL, Lapatto R, Saksela M, and Raivio KO. Human
cystathionine gamma-lyase: Developmental and in vitro ex-
pression of two isoforms. Biochem J 347: 1–5, 2000.
22. Loscalzo J. The oxidant stress of hyperhomocyst(e)inemia. J
Clin Invest 98: 5–7, 2000.
23. úowicka E and Be towski J. Hydrogen sulfide (H2S)—the
third gas of interest for pharmacologists. Pharmacol Rep 59:
24, 2007.
24. Mattson MP and Duan W. Apoptotic biochemical cascades
in synaptic compartments: roles in adaptive plasticity and
neurodegenerative disorders. J. Neurosci Res 58:152–166,
1999.
25. Mattson MP, Pedersen WA, Duan W, Culmsee C, and Ca-
mandola S. Cellular and molecular mechanisms underlying
perturbed energy metabolism and neuronal degeneration in
Alzheimer’s and Parkinson’s diseases. Ann NY Acad Sci
893:154–175, 1999.
26. Montesano R, Pepper MS, Möhle–Steinle U, Risau W, Wag-
ner EF, and Orci L. Increased proteolytic activity is respon-
sible for the aberrant morphogenetic behavior of endothe-
lial cells expressing the middle T oncogene. Cell 10: 62: 435–
45, 1990.
27. Mosmann T. Rapid colorimetric assay for cellular growth
and survival: application to proliferation and cytotoxicity as-
says. J Immunol Methods 65: 55–63, 1983.
28. Muijsers RB, van Den Worm E, Folkerts G, Beukelman CJ,
Koster AS, Postma DS, and Nijkamp FP. Apocynin inhibits
peroxynitrite formation by murine macrophages. Br J Phar-
macol 130: 932–936, 2000.
29. Obeid R and Herrmann W. Mechanisms of homocysteine
neurotoxicity in neurodegenerative diseases with special ref-
erence to dementia. FEBS Lett 580: 2994–3005, 2006.
30. Rodrigo R, Passalacqua W, Araya J, Orellana M, and Rivera
G . Homocysteine and essential hypertension. J Clin Phar-
macol 43: 1299–1306, 2003.
31. Ryter SW and Choi AM. Heme oxygenase-1: Molecular
mechanisms of gene expression in oxygen-related stress. An-
tioxid Redox Signal 4: 625–632, 2002.
32. Sawle P, Foresti R, Green CJ, and Motterlini R. Homocys-
teine attenuates endothelial haem oxygenase-1 induction by
nitric oxide (NO) and hypoxia. FEBS Lett 23: 508: 403–406.
2001.
33. Selhub J. Homocysteine metabolism. Annu Rev Med 19:
217–246, 1999.
34. Sen U, Tyagi N, Kumar M, Moshal KS, Rodriguez WE,
and Tyagi SC. Cystathionine-beta-synthase gene transfer
and 3-deazaadenosine ameliorate inflammatory response in
endothelial cells. Am J Physiol Cell Physiol 293: C1779–87,
2007.
35. Stamler JS, Osborne JA, Jaraki O, Rabbani LE, Mullins M,
Singel D, and Loscalzo J. Adverse vascular effects of homo-
cysteine are modulated by endothelium-derived relaxing
factor and related oxides of nitrogen. J Clin Invest 91:
308–318, 1999.
36. Stampfer MJ, Malinow MR, Willett WC, Newcomer LM, Up-
son B, Ullmann D, Tishler PV, and Hennekens CH. A
prospective study of plasma homocyst(e)ine and risk of
myocardial infarction in US physicians. J Am Med Assoc 268:
877–881, 1992.
37. Su JH, Anderson AJ, Cummings BJ, and Cotman CW. Im-
munohistochemical evidence for apoptosis in Alzheimer’s
disease. Neuroreport 5: 2529–2533, 1994.
38. Tawakol A, Omland T, Gerhard M, Wu JT, and Creager MA.
Hyperhomocyst(e)inemia is associated with impaired endo-
thelium-dependent vasodilation in humans. Circulation
95:1119–1121, 1997.
39. Tyagi N, Ovechkin AV, Lominadze D, Moshal KS, and Tyagi
SC. Mitochondrial mechanism of microvascular endothelial
cells apoptosis in hyperhomocysteinemia. J Cell Biochem 98:
1150–1162, 2006.
40. Tyagi N, Sedoris KC, Steed M, Ovechkin AV, Moshal KS,
and Tyagi SC. Mechanisms of homocysteine-induced ox-
idative stress. Am J Physiol Heart Circ Physiol 289: H2649–
H2656, 2005.
41. Tyagi N, Moshal KS, Tyagi SC, and Lominadze D. Gamma
aminobutyric acid A receptor mitigates homocysteine-in-
duced endothelial cell permeability. Endothelium 14: 315–323,
2007.
42. Wang R. Two’s company, three’s a crowd: Can H
2
S be the
third endogenous gaseous transmitter? FASEB J 16: 1792–
1798, 2002.
43. Wang R, Wang ZZ, and Wu L. Carbon monoxide-induced
vasorelaxation and the underlying mechanisms. Br J Phar-
macol 121: 927–934, 1997.
TYAGI ET AL.32
44. Wang R, Wu L, and Wang ZZ. The direct effect of carbon
monoxide on K
Ca
channels in vascular smooth muscle cells.
PflÅgers Arch 434: 285–291, 1997.
45. Whiteman M, Cheung NS, Zhu YZ, Chu SH, Siau JL, Wong
BS, Armstrong JS, and Moore PK. Hydrogen sulphide: A
novel inhibitor of hypochlorous acid-mediated oxidative
damage in the brain? Biochem Biophys Res Commun 28: 794–
798, 2005.
46. Whiteman M, Armstrong JS, Chu SH, Jia–Ling S, Wong BS,
Cheung NS, Halliwell B, and Moore PK. The novel neuro-
modulator hydrogen sulfide: an endogenous peroxynitrite
‘scavenger’? J. Neurochem 90: 765–768, 2005.
47. Yan SK, Chang T, Wang H, Wu L, Wang R, and Meng QH.
Effects of hydrogen sulfide on homocysteine-induced ox-
idative stress in vascular smooth muscle cells. Biochem Bio-
phys Res Commun 15: 485–491, 2006.
48. Yang G, Cao K, Wu L, and Wang R. Cystathionine gamma-
lyase overexpression inhibits cell proliferation via a H2S-
dependent modulation of ERK1/2 phosphorylation and
p21Cip/WAK-1. J Biol Chem 279: 49199–49205, 2004.
49. Yonezawa D, Sekiguchi F, Miyamoto M, Taniguchi E, Honjo
M, Masuko T, Nishikawa H, and Kawabata A. A protective
role of hydrogen sulfide against oxidative stress in rat gas-
tric mucosal epithelium. Toxicology 20: 11–18, 2007.
50. Yu BP. Cellular defenses against damage from reactive oxy-
gen species. Physiol. Rev 74: 139–162, 1994.
51. Zhang Q, Du J, Zhou W, Yan H, Tang C, and Zhang C. Im-
pact of hydrogen sulfide on carbon monoxide/heme oxy-
genase pathway in the pathogenesis of hypoxic pulmonary
hypertension. Biochem.Biophys.Res.Commun 317: 30–37, 2004.
52. Zhao W, Zhang J, Lu Y, and Wang R. The vasorelaxant ef-
fect of H
2
S as a novel endogenous gaseous K
ATP
channel
opener. EMBO J 20: 6008–6016, 2001.
53. Zhong C, Du J, Bu D, Yan H, Tang D, and Tang C. The reg-
ulatory effect of hydrogen sulfide on hypoxic pulmonary hy-
pertension in rats. Biochem Biophys Res Commun 302: 810–816,
2003.
54. Zhu YZ, Wang ZJ, Ho P, Loke YY, Zhu YC, Huang SH, Tan
CS, Whiteman M, Lu J, and Moore PK. Hydrogen sulfide
and its possible roles in myocardial ischemia in experimen-
tal rats. J Appl Physiol 102: 261–268. 2006.
Address reprint requests to:
Suresh C. Tyagi, PhD
Department of Physiology and Biophysics
School of Medicine
University of Louisville
Louisville, KY 40202
E-mail: suresh.tyagi@louisville.edu
Date of first submission to ARS Central, March 8, 2008; date
of final revised submission, July 16, 2008; date of acceptance,
July 16, 2008.
H
2
S INHIBITS METHIONINE–INDUCED OXIDATIVE STRESS 33
... Studies have shown that H 2 S increases levels of intracellular reduced glutathione (GSH), which is a major antioxidant in the brain [33,34] and spinal cord [150]. Past studies have shown that the H 2 S donor promotes glutamate uptake in astrocytes by enhancing glial glutamate transporter GLT-1 delivery, enhancing cystine transport, and as a result, GSH synthesis [32]. ...
... Trx-1 reduces hydrogen peroxide with peroxiredoxine (Prx), and oxidized Trx-1 is reduced with thioredoxine reductase. H 2 S has been shown to increase gene transcription and Trx-1 levels [34,69]. ...
Article
Full-text available
Injuries of the central (CNS) and peripheral nervous system (PNS) are a serious problem of the modern healthcare system. The situation is complicated by the lack of clinically effective neuroprotective drugs that can protect damaged neurons and glial cells from death. In addition, people who have undergone neurotrauma often develop mental disorders and neurodegenerative diseases that worsen the quality of life up to severe disability and death. Hydrogen sulfide (H2S) is a gaseous signaling molecule that performs various cellular functions in normal and pathological conditions. However, the role of H2S in neurotrauma and mental disorders remains unexplored and sometimes controversial. In this large-scale review study, we examined the various biological effects of H2S associated with survival and cell death in trauma to the brain, spinal cord, and PNS, and the signaling mechanisms underlying the pathogenesis of mental illnesses, such as cognitive impairment, encephalopathy, depression and anxiety disorders, epilepsy and chronic pain. We also studied the role of H2S in the pathogenesis of neurodegenerative diseases: Alzheimer's disease (AD) and Parkinson's disease (PD). In addition, we reviewed the current state of the art study of H2S donors as neuroprotectors and the possibility of their therapeutic uses in medicine. Our study showed that H2S has great neuroprotective potential. H2S reduces oxidative stress, lipid peroxidation, and neuroinflammation; inhibits processes associated with apoptosis, autophagy, ferroptosis and pyroptosis; prevents the destruction of the blood-brain barrier; increases the expression of neurotrophic factors; and models the activity of Ca2+ channels in neurotrauma. In addition, H2S activates neuroprotective signaling pathways in psychiatric and neurodegenerative diseases. However, high levels of H2S can cause cytotoxic effects. Thus, the development of H2S-associated neuroprotectors seems to be especially relevant. However, so far, all H2S modulators are at the stage of preclinical trials. Nevertheless, many of them show a high neuroprotective effect in various animal models of neurotrauma and related disorders. Despite the fact that our review is very extensive and detailed, it is well structured right down to the conclusions, which will allow researchers to quickly find the proper information they are interested in.
... Similarly, another study reported that 100 µM NaHS induces glutamate uptake by assisting glial glutamate transporter-1 (GLT-1) and enhances cysteine transport and GSH synthesis [224]. In support of this effect, multiple studies demonstrated that H 2 S induces cellular GSH in the brain [225], spinal cord [226], heart [227], lung [228], kidney [229], liver [228], and gastrointestinal tract [230,231]. Moreover, recent reports suggested that H 2 S could attenuate cellular oxidative stress by improving the activities of catalase [227,[232][233][234] and glutathione peroxidase [235][236][237]. ...
Article
Full-text available
A high level of homocysteine (Hcy) is associated with oxidative/ER stress, apoptosis, and impairment of angiogenesis, whereas hydrogen sulfide (H2S) has been found to reverse this condition. Recent studies have shown that cancer cells need to produce a high level of endogenous H2S to maintain cell proliferation, growth, viability, and migration. However, any novel mechanism that targets this balance of Hcy and H2S production has yet to be discovered or exploited. Cells require homocysteine metabolism via the methionine cycle for nucleotide synthesis, methylation, and reductive metabolism, and this pathway supports the high proliferative rate of cancer cells. Although the methionine cycle favors cancer cells for their survival and growth, this metabolism produces a massive amount of toxic Hcy that somehow cancer cells handle very well. Recently, research showed specific pathways important for balancing the antioxidative defense through H2S production in cancer cells. This review discusses the relationship between Hcy metabolism and the antiapoptotic, antioxidative, anti-inflammatory, and angiogenic effects of H2S in different cancer types. It also summarizes the historical understanding of targeting antioxidative defense systems, angiogenesis, and other protective mechanisms of cancer cells and the role of H2S production in the genesis, progression, and metastasis of cancer. This review defines a nexus of diet and precision medicine in targeting the delicate antioxidative system of cancer and explores possible future therapeutics that could exploit the Hcy and H2S balance.
Article
Full-text available
Cryopreservation of sperm can cause oxidative stress and damage, leading to decreased different functional parameters and fertilization potential. In this study, we evaluated two types of H2S donors: NaHS, a fast-releasing donor, and GYY4137, a slow-releasing donor during cryopreservation of goat sperm. Initially, we determined that 1.5 and 3 μM NaHS, and 15 and 30 μM GYY4137 are optimal concentrations that improved different sperm functional parameters including motility, viability, membrane integrity, lipid peroxidation, and ROS production during incubation at 38.5 °C for 90 min. We subsequently evaluated the impact of the optimal concentration of NaHS and GYY4137 supplementation on various functional parameters following thawing during cryopreservation. Our data revealed that supplementation of extender improved different parameters including post-thaw sperm motility, viability, membrane integrity, and reduced DNA damage compared to the frozen-thawed control group. The supplementation also restored the redox state, decreased lipid peroxidation, and improved mitochondrial membrane potential in the thawed sperm. Finally, we found that supplementation of the extender with NaHS and GYY4137 enhanced IVF outcomes in terms of blastocyst rate and quality of blastocysts. Our results suggest that both donors can be applied for cryopreservation as antioxidants to improve sperm quality and IVF outcomes of frozen-thawed goat sperm.
Article
Neurodegeneration is a complex progressive pathological process leading to the neuronal death, which is induced by various external and internal factors. Neurodegenerative diseases, injuries of the central and peripheral nervous system, mental disorders, and a number of other pathological conditions, accompanied by functional and structural degradation of neurons and their death, is a serious problem in the global healthcare system, as due to these diseases millions of people around the world become disabled or die every year. The situation is complicated by the lack of selective, clinically effective neuroprotective drugs. It has been shown that nitric oxide (NO) and hydrogen sulfide (H2S) are actively involved in neurodegeneration and cell death of neurons and glia, but their role is not completely clear. This review considers NO- and H2S-dependent signaling mechanisms underlying the pathogenesis of neurodegenerative processes. The prospects for further studies of the role of NO and H2S in the nervous tissue under conditions of pathological conditions associated with neurodegeneration are considered.
Article
Full-text available
Background Diabetic retinopathy is a progressive disease, and one of the key metabolic abnormalities in the pathogenesis of diabetic retinopathy, mitochondrial damage, is also influenced by the duration of hyperglycemia. Mitochondrial quality control involves a coordination of mitochondrial dynamics, biogenesis and removal of the damaged mitochondria. In diabetes, these processes are impaired, and the damaged mitochondria continue to produce free radicals. Diabetic patients also have high homocysteine and reduced levels of hydrogen sulfide, and hyperhomocysteinemia is shown to exacerbate diabetes-induced mitochondrial damage and worsen their dynamics. This study aims to investigate the temporal relationship between hyperhomocysteinemia and retinal mitochondrial quality control in diabetic retinopathy. Methods Human retinal endothelial cells incubated in 20 mM d -glucose for 24 to 96 h, in the absence or presence of 100 µM homocysteine, with/without a hydrogen sulfide donor GYY4137, were analyzed for mitochondrial ROS (MitoSox fluorescence), DNA damage (transcripts of mtDNA-encoded ND6 and CytB ), copy numbers, oxygen consumption rate (Seahorse XF analyzer) and mitophagy (mitophagosomes immunofluorescence labeling and flow cytometry). Results were confirmed in the retina from mice genetically manipulated for hyperhomocysteinemia (cystathionine β-synthase deficient mice, Cbs +/− ), streptozotocin-induced diabetic for 8 to 24 weeks. At 24 weeks of diabetes, vascular health was evaluated by counting acellular capillaries in the trypsin digested retinal vasculature and by fluorescein angiography. Results Homocysteine, in high glucose medium, exacerbated mitochondrial ROS production, mtDNA damage and impaired mitochondrial respiration within 24 h, and slowed down/worsened mitochondrial biogenesis and mitophagy, as compared to 48 to 96 h in high glucose alone. GYY4137 supplementation ameliorated homocysteine + high glucose-induced mitochondrial damage and impairment in biogenesis and mitophagy. Similar results were obtained from Cbs +/− mice-mitochondrial ROS, mtDNA damage and decline in biogenesis and mitophagy were observed within eight weeks of diabetes vs. 16 to 24 weeks of diabetes in Cbs +/+ mice, and at 24 weeks of diabetes, Cbs +/− mice had significantly higher acellular capillaries and vascular leakage. Conclusions Hyperhomocysteinemia, in a hyperglycemic environment, overwhelms the mitochondria, accelerating and exacerbating their dysfunction, and also delays/worsens their removal, augmenting the development of diabetic retinopathy. Thus, our results strengthen the importance of maintaining homocysteine-hydrogen sulfide balance during the early stages of diabetes for a patient to prevent/retard vision loss.
Article
Full-text available
Aromatic l-amino acid decarboxylase deficiency (AADC-DY) is caused by one or more mutations in the DDC gene, resulting in the deficit in catecholamines and serotonin neurotransmitters. The disease has limited therapeutic options with relatively poor clinical outcomes. Accumulated evidence suggests the involvement of neurodegenerative mechanisms in the etiology of AADC-DY. In the absence of neurotransmitters’ neuroprotective effects, the accumulation and the chronic presence of several neurotoxic metabolites including 4-dihydroxy-L-phenylalanine, 3-methyldopa, and homocysteine, in the brain of subjects with AADC-DY, promote oxidative stress and reduce the cellular antioxidant and methylation capacities, leading to glial activation and mitochondrial dysfunction, culminating to neuronal injury and death. These pathophysiological processes have the potential to hinder the clinical efficacy of treatments aimed at increasing neurotransmitters’ synthesis and or function. This review describes in detail the mechanisms involved in AADC-DY neurodegenerative etiology, highlighting the close similarities with those involved in other neurodegenerative diseases. We then offer novel strategies for the treatment of the disease with the objective to either reduce the level of the metabolites or counteract their prooxidant and neurotoxic effects. These treatment modalities used singly or in combination, early in the course of the disease, will minimize neuronal injury, preserving the functional integrity of neurons, hence improving the clinical outcomes of both conventional and unconventional interventions in AADC-DY. These modalities may not be limited to AADC-DY but also to other metabolic disorders where a specific mutation leads to the accumulation of prooxidant and neurotoxic metabolites.
Article
The poultry industry is one of the pillars of food security in the world, as it is relied upon to provide meat and eggs to meet the increasing food demands. Therefore, this study was designed to investigate the effect of L-carnitine and methionine supplementation to the standard diets of broiler chickens in productive performance of broiler (Ross 308). One Hundred- fifty broiler chicks unsexed (Ross 308) with an initial weight with 43 g, were obtained from Al-Habbaniya hatchery (commercial hatchery). All the animals were within an average weight of 40 g (one-day old chicks). The experimental groups were as follows: the animals in T1 group received basal diet without any addition, the animals in T1 group received basal diet supplemented with lead acetate 400 mg/kg feed , the animals in T3 group received diet supplemented with carnitine 300 mg + lead acetate 400 mg, the animals in T4 group received basal diet supplemented with methionine 100 mg + lead acetate 400 mg, the animals in T5 group received basal diet supplemented with methionine 100 mg + carnitine 300 mg + lead acetate 400 mg. Body weight gain and feed consumption were weekly recorded. Feed conversion ratio was also calculated. Results showed that Birds in (T5) fed diets with (carnitine + methionine) observed highest live body weights comparison with T3 (carnitine + lead acetate) and T4 (adding methionine+ lead acetate). Data of results showed no significant differences were recorded in body weight gain. Also, Results obtained increase with feed consumption for treatment T5, while birds in T1 and T4 recorded lowest means in feed consumed. However, birds in T4 and T5 observed best feed conversion ratio as compare with T1, T2 and T3. Therefore, it can conclude that addition carnitine and methionine enhanced broiler productive performance.
Article
Hydrogen sulfide (H2S) plays a cytoprotective role during mitophagy by detoxifying superfluous reactive oxygen species (ROS), and its concentration fluctuates in this process. However, no work has been reported to reveal the variation in H2S levels during autophagic fusion of lysosomes and mitochondria. Herein, we present a lysosome-targeted fluorogenic probe, named NA-HS, for real-time monitoring of H2S fluctuation for the first time. The newly synthesized probe exhibits good selectivity and high sensitivity (detection limit of 23.6 nM). Fluorescence imaging results demonstrated that NA-HS could image exogenous and endogenous H2S in living cells. Interestingly, the colocalization results revealed that the level of H2S was upregulated after autophagy began because of the cytoprotective effect, and was finally gradually reduced during subsequent autophagic fusion. This work not only affords a powerful fluorescence tool to monitor the variations in H2S levels during mitophagy, but also offers new insights into targeting small molecules for elaborating the complex cellular signal pathways.
Article
Full-text available
Objective The aim of the present study was to explore influencing factors of cognitive impairments and their interrelationships in drug-naïve, first-episode schizophrenia (SCZ). Methods Patients with drug naïve, first episode SCZ and healthy controls (HCs) were enrolled. Cognitive function was assessed by the MATRICS Consensus Cognitive Battery (MCCB). Serum levels of oxidative stress indices, including folate, superoxide dismutase (SOD), uric acid (UA) and homocysteine (Hcy), were determined after an overnight fast. Hippocampal subfield volumes were measured using FreeSurfer. Mediation models were conducted using the SPSS PROCESS v3.4 macro. A false discovery rate (FDR) correction was applied for multiple comparisons. Results Sixty-seven patients with SCZ and 65 HCs were enrolled in our study. The patient group had significantly lower serum levels of folate and SOD and higher serum levels of HCY compared with the HCs (all p < 0.05). The patient group had a significantly smaller volume of the whole hippocampus than the HC group (p < 0.05). We also found significant volume differences between the two groups in the following subfields: CA1, molecular layer, GC-ML-DG and fimbria (all p < 0.05, uncorrected). The partial correlation analysis controlling for age and sex showed that the fimbria volume in the patient group was significantly positively associated with NAB scores (r = 0.382, pFDR = 0.024); serum levels of SOD in the patient group showed a significantly positive correlation with fimbria volume (r = 0.360, pFDR = 0.036). Mediation analyses controlling for age and sex showed that the serum levels of SOD in patients with SCZ had significant indirect effects on the NAB scores which were mediated by the fimbria volume [indirect effect = 0.0565, 95% CI from the bootstrap test excluding zero (0.0066 to 0.0891)]. Conclusion Oxidative stress, a reduction in hippocampal subfield volumes and cognitive impairments occur in early SCZ. Oxidative stress impairs cognitive function by affecting hippocampal subfield volumes.
Article
Full-text available
Cystathionine gamma-lyase (CGL) is the last enzyme of the trans-sulphuration pathway, which converts methionine into cysteine, To study the possible differences in enzymic activity of the two human cystathionine gamma-lyase isoforms characterized earlier, these were separately expressed in human kidney embryonic 293T cells. Furthermore, developmental changes in the expression of the two mRNA forms as well as the enzymic activity in human liver were studied, as it has been postulated that a change in the relative expression of CGL isoforms causes the postnatal increase in CGL activity. Transfection with the longer isoform increased the CGL activity 1.5-fold, while the activity of the cells transfected with the shorter form did not differ from the basal activity. In human liver samples, CGL activity was only detected in adult tissue (68 +/- 9 nmol of cysteine/h per mg of protein), whereas activity in fetal, premature and full-term neonatal liver tissue was undetectable. In contrast, strong mRNA expression of both mRNA isoforms was detected from the 19th gestational week onwards and the longer form of CGL appeared to be predominant. The expression of the two mRNA forms varied in parallel. In conclusion, we have shown that only cells overexpressing the longer form of CGL have increased activity, and CGL appears to be regulated at the post-transcriptional level during development.
Article
Apoptosis is a form of cell death historically defined by morphological and biochemical changes that occur in the cell body and nucleus. However, in contrast to nonneuronal cells in which apoptosis has been most intensively studied, neurons exhibit elaborate morphologies with synaptic connections often located at sites a great distance from the cell body. Signaling events occurring in synaptic terminals are believed to play important roles in either promoting (e.g., activation of glutamate receptors in postsynaptic spines) or preventing (e.g., activation of neurotrophic factors in presynaptic terminals) neuronal cell death in various physiological and pathological settings. We have found that apoptotic biochemical cascades can be activated locally in synaptic terminals and neurites and have shown that such cascades can result in local functional and morphological alterations and can also propagate to the cell body resulting in neuronal death. Prostate apoptosis response-4 production, caspase activation, loss of plasma membrane phospholipid asymmetry, mitochondrial dysfunction, and production of factors capable of inducing nuclear chromatin condensation and fragmentation can all occur locally in synaptic terminals in response to various stimuli. Activation of receptors for neurotrophic factors (e.g., basic fibroblast growth factor, secreted form of amyloid precursor protein α, and activity-dependent neurotrophic factor) and cytokines (e.g., tumor necrosis factor-α) in synaptic terminals can exert synaptoprotective actions that either can be transduced locally or may require signals to the nucleus and back. In addition to their roles in synaptic degeneration and neuron death, apoptotic cascades may play roles in synaptic plasticity. For example, we found that caspase activation can lead to proteolysis of certain glutamate receptor subunits and that this action of capases is correlated with reduced calcium responses to glutamate. We propose that apoptotic cascades function in a continuum in which low levels of activation play roles in adaptive responses to “stressors,” whereas higher levels of activation mediate synaptic degeneration and cell death. J. Neurosci. Res. 58:152–166, 1999. © 1999 Wiley-Liss, Inc.
Conference Paper
The literature has been searched to identify evidence relating to the possible toxicity of the amino acid methionine in human subjects. Nutritional and metabolic studies have employed amounts of methionine, including the D and DL isomers, both below and above the requirement and have not reported adverse effects in adults and children. Although methionine is known to exacerbate psychopathological symptoms in schizophrenic patients, there is no evidence of similar effects in healthy subjects. The role of methionine as a precursor of homocysteine is the most notable cause for concern. A "loading dose" of methionine (0.1 g/kg) has been given, and the resultant acute increase in plasma homocysteine has been used as an index of the susceptibility to cardiovascular disease. Although this procedure results in vascular dysfunction, this is acute and unlikely to result in permanent damage. However, a 10-fold larger dose, given mistakenly, resulted in death. Longer-term studies in adults have indicated no adverse consequences of moderate fluctuations in dietary methionine intake, but intakes higher than 5 times normal resulted in elevated homocysteine levels. These effects of methionine on homocysteine and vascular function are moderated by supplements of vitamins B-6, B-12, C, and folic acid. In infants, methionine intakes of 2-5 times normal resulted in impaired growth and extremely high plasma methionine levels, but no adverse long-term consequences were observed.
Article
RECENTLY, in vitro studies conducted in our laboratory and others have suggested that apoptosis may have a role in the neuronal cell death associated with Alzheimer's disease (AD). To evaluate this hypothesis, the hippocampi and entorhinal cortices of AD, aged control, and surgical biopsy tissue were examined using the ApopTag system for the detection of DNA fragmentation and DNA stains to reveal nuclear morphology. Numerous neuronal nuclei displaying distinct morphological characteristics of apoptosis were present within tangle-bearing neurons as well as non-tangle-bearing neurons in AD brain, whereas few or no such nuclei were detected in control brain. Our in vivo results support the hypothesis that apoptosis may be one mechanism leading neuronal cell death in AD.
Article
The cause of neuronal degeneration in Alzheimer's disease (AD) has not been completely clarified, but has been variously attributed to increases in cytosolic calcium and increased generation of reactive oxygen species (ROS). The β-amyloid fragment (Aβ) of the amyloid precursor protein induces calcium influx, ROS and apoptosis. Homocysteine (HC), a neurotoxic amino acid that accumulates in neurological disorders including AD, also induces calcium influx and oxidative stress, which has been shown to enhance neuronal excitotoxicity, leading to apoptosis. We examined the possibility that HC may augment Aβ neurotoxicity. HC potentiated the Aβ-induced increase in cytosolic calcium and apoptosis in differentiated SH-SY-5Y human neuroblastoma cells. The antioxidant vitamin E and the glutathione precursor N-acetyl-l-cysteine blocked apoptosis following cotreatment with HC and Aβ, indicating that apoptosis is associated with oxidative stress. These findings underscore that moderate accumulation of excitotoxins at concentrations that alone do not appear to initiate adverse events may enhance the effects of other factors known to cause neurodegeneration such as Aβ.