ArticlePDF Available

Hydrogen sulfide ameliorates hyperhomocysteinemia-associated chronic renal failure

Authors:

Abstract

Elevated level of homocysteine (Hcy), known as hyperhomocysteinemia (HHcy), is associated with end-stage renal diseases. Hcy metabolizes in the body to produce hydrogen sulfide (H(2)S), and studies have demonstrated a protective role of H(2)S in end-stage organ failure. However, the role of H(2)S in HHcy-associated renal diseases is unclear. The present study was aimed to determine the role of H(2)S in HHcy-associated renal damage. Cystathionine-beta-synthase heterozygous (CBS+/-) and wild-type (WT, C57BL/6J) mice with two kidney (2-K) were used in this study and supplemented with or without NaHS (30 micromol/l, H(2)S donor) in the drinking water. To expedite the HHcy-associated glomerular damage, uninephrectomized (1-K) CBS(+/-) and 1-K WT mice were also used with or without NaHS supplementation. Plasma Hcy levels were elevated in CBS(+/-) 2-K and 1-K and WT 1-K mice along with increased proteinuria, whereas, plasma levels of H(2)S were attenuated in these groups compared with WT 2-K mice. Interestingly, H(2)S supplementation increased plasma H(2)S level and normalized the urinary protein secretion in the similar groups of animals as above. Increased activity of matrix metalloproteinase (MMP)-2 and -9 and apoptotic cells were observed in the renal cortical tissues of CBS(+/-) 2-K and 1-K and WT 1-K mice; however, H(2)S prevented apoptotic cell death and normalized increased MMP activities. Increased expression of desmin and downregulation of nephrin in the cortical tissue of CBS(+/-) 2-K and 1-K and WT 1-K mice were ameliorated with H(2)S supplementation. Additionally, in the kidney tissues of CBS(+/-) 2-K and 1-K and WT 1-K mice, increased superoxide (O(2)(*-)) production and reduced glutathione (GSH)-to-oxidized glutathione (GSSG) ratio were normalized with exogenous H(2)S supplementation. These results demonstrate that HHcy-associated renal damage is related to decreased endogenous H(2)S generation in the body. Additionally, here we demonstrate with evidence that H(2)S supplementation prevents HHcy-associated renal damage, in part, through its antioxidant properties.
Hydrogen sulfide ameliorates hyperhomocysteinemia-associated chronic
renal failure
Utpal Sen, Poulami Basu, Oluwasegun A. Abe, Srikanth Givvimani, Neetu Tyagi, Naira Metreveli,
Karan S. Shah, John C. Passmore, and Suresh C. Tyagi
Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, Kentucky
Submitted 11 March 2009; accepted in final form 19 May 2009
Sen U, Basu P, Abe OA, Givvimani S, Tyagi N, Metreveli N,
Shah KS, Passmore JC, Tyagi SC. Hydrogen sulfide ameliorates
hyperhomocysteinemia-associated chronic renal failure. Am J Physiol
Renal Physiol 297: F410 –F419, 2009. First published May 27, 2009;
doi:10.1152/ajprenal.00145.2009.—Elevated level of homocysteine
(Hcy), known as hyperhomocysteinemia (HHcy), is associated with
end-stage renal diseases. Hcy metabolizes in the body to produce
hydrogen sulfide (H
2
S), and studies have demonstrated a protective
role of H
2
S in end-stage organ failure. However, the role of H
2
Sin
HHcy-associated renal diseases is unclear. The present study was
aimed to determine the role of H
2
S in HHcy-associated renal damage.
Cystathionine--synthase heterozygous (CBS/) and wild-type
(WT, C57BL/6J) mice with two kidney (2-K) were used in this study
and supplemented with or without NaHS (30 mol/l, H
2
S donor) in
the drinking water. To expedite the HHcy-associated glomerular
damage, uninephrectomized (1-K) CBS(/) and 1-K WT mice were
also used with or without NaHS supplementation. Plasma Hcy levels
were elevated in CBS(/) 2-K and 1-K and WT 1-K mice along
with increased proteinuria, whereas, plasma levels of H
2
S were
attenuated in these groups compared with WT 2-K mice. Interestingly,
H
2
S supplementation increased plasma H
2
S level and normalized the
urinary protein secretion in the similar groups of animals as above.
Increased activity of matrix metalloproteinase (MMP)-2 and -9 and
apoptotic cells were observed in the renal cortical tissues of
CBS(/) 2-K and 1-K and WT 1-K mice; however, H
2
S prevented
apoptotic cell death and normalized increased MMP activities. In-
creased expression of desmin and downregulation of nephrin in the
cortical tissue of CBS(/) 2-K and 1-K and WT 1-K mice were
ameliorated with H
2
S supplementation. Additionally, in the kidney
tissues of CBS(/) 2-K and 1-K and WT 1-K mice, increased
superoxide (O
2
) production and reduced glutathione (GSH)-to-
oxidized glutathione (GSSG) ratio were normalized with exogenous
H
2
S supplementation. These results demonstrate that HHcy-associ-
ated renal damage is related to decreased endogenous H
2
S generation
in the body. Additionally, here we demonstrate with evidence that
H
2
S supplementation prevents HHcy-associated renal damage, in part,
through its antioxidant properties.
cystathionine-synthase; cystathionine-lyase; homocysteine; ma-
trix metalloproteinase
HYDROGEN SULFIDE (H
2
S) is known as a toxic gas with a very
strong repulsive odor. Despite its toxicity, many prokaryotic
and eukaryotic organisms thrive in sulfidic habitats (11). Re-
cently, the presence of tissue H
2
S from sulfidic organisms to
the animals who live in a sulfur-free environment has been
confirmed by several independent investigations (10, 49).
These findings unambiguously suggest that H
2
S is a constituent
of cellular milieu. However, to date, very little is known about
the physiological role of H
2
S in normal animal and during
pathological stages of renal diseases.
Endogenously, H
2
S is generated in the mammalian tissue
from a nonprotein amino acid, homocysteine (Hcy). This is a
sulfur-containing amino acid, which is an intermediate product
of methionine metabolism. In the body, two enzymes, cysta-
thionine -synthase (CBS) and cystathionine -lyase (CSE), of
the transulfuration pathway of methionine metabolism catalyze
conversion of H
2
S from Hcy. Studies from independent labo-
ratories reported that, at low level, H
2
S defends organs from
several pathophysiological conditions, such as oxidative stress,
ischemia-reperfusion, and hypertension (20, 52, 59). Recently,
Yan et al. (51) postulated that, being a reducing molecule, H
2
S
may modulate redox tone and redox cell signaling. These
authors, however, reported that at high levels H
2
S induces
reactive oxygen species (ROS) and reactive nitrogen species
(RNS) formation but at low levels decreases hydrogen perox-
ide (H
2
O
2
), peroxinitrite (ONOO
), and superoxide anion
(O
2
) generation induced by Hcy in a cell culture model (51).
These antioxidant properties were partly mediated by increas-
ing the antioxidant properties of cellular antioxidant molecules.
Nevertheless, the endogenous status of H
2
S and its role in
hyperhomocysteinemia (HHcy)-associated renal failure remain
unknown.
Previously, we have reported that reduction in renal function
leads to an increased plasma Hcy level (36, 38). The elevated
plasma Hcy, in turn, causes renal insufficiency, which leads to
a vicious cycle (22). Accumulated evidences from independent
laboratories, including our own, suggested that, at an elevated
level, Hcy is an independent and graded risk factor for car-
diorenovascular diseases (7, 27, 38). Reports are also available
that it may contribute to the pathogenesis of atherosclerosis (2,
26), including glomerulosclerosis, cellular apoptosis, podocyte
injury, and proteinuria (22, 56). These pathophysiological
effects of Hcy are mediated through generation of ROS,
including O
2
and H
2
O
2
, and reduction in endothelial nitric
oxide (NO) bioavailability (41, 60).
It is well known that ROS plays a major role in the extra-
cellular matrix (ECM) remodeling during various pathophysi-
ological conditions. One of the early events of the ECM
remodeling process is the activation of matrix metalloprotein-
ases (MMPs) (8, 39). Among MMPs, elastinases and collag-
enases are particularly important of matrix degradation in
fibrotic occlusive diseases including renal fibrosis (5, 21). We
have previously reported that MMP-2 and MMP-9 play an
important role in matrix accumulation associated with diabetic
nephropathy and HHcy (39). However, the role of H
2
S, if any,
in the regulation of these two MMPs in HHcy-associated renal
diseases was not defined. Also, the physiological status of
glomerular podocytes, injured podocyte marker, desmin, as
Address for reprint requests and other correspondence: Utpal Sen, Dept. of
Physiology & Biophysics, Univ. of Louisville School of Medicine, 500 S.
Preston St., Louisville, KY 40202 (e-mail: u0sen001@louisville.edu).
Am J Physiol Renal Physiol 297: F410–F419, 2009.
First published May 27, 2009; doi:10.1152/ajprenal.00145.2009.
0363-6127/09 $8.00 Copyright ©2009 the American Physiological Society http://www.ajprenal.orgF410
by 10.220.33.4 on November 27, 2017http://ajprenal.physiology.org/Downloaded from
well as normal slit diaphragm component nephrin during HHcy
is not clear. Therefore, the present study aimed to determine
whether HHcy was associated with decreased plasma H
2
S level
and increased oxidative stress during chronic renal failure by
modulating MMP-2 and -9 activities, glomerular cell apopto-
sis, and increased urinary protein secretion. Additionally, the
possible preventive role of H
2
S, if any, has been explored to
these deleterious effects in the failing kidney.
MATERIALS AND METHODS
Animals. Wild-type (WT, C57BL/6J) male mice aged 8 wk were
obtained from Jackson Laboratories (Bar Harbor, ME) and housed in
the animal care facility at University of Louisville. Mice were
acclimatized for 2 wk before the start of experiments. WT (C57BL/
6J) and heterozygous CBS(/), a model for hyperhomocysteine-
mic mice, were used for this study. Mice were divided into two
sets, the first set of mice had two kidneys (2-K), and the groups were
as follows: 1) WT, 2) CBS(/), 3)WTNaHS (30 mol/l, H
2
S
donor), and 4) CBS(/)NaHS (30 mol/l). To speed up the renal
damaging effects, the second set of mice was uninephrectomized
(1-K) with all above four groups. NaHS were supplied for 8 wk to the
appropriate groups. At the end of the experiments, mice were deeply
anesthetized, blood was collected, and the animals were killed to
harvest the tissues. All animal procedures were in accordance with the
National Institute of Health Guidelines for animal research and were
approved by the Institutional Animal Care and Use Committee of the
University Of Louisville School Of Medicine.
Rationale for NaHS (H
2
S donor) dose. The physiological concen-
tration of H
2
S ranges from 10 –100 mol/l (59). NaHS in the aqueous
phase produces exactly equal concentration of H
2
S gas in the solution.
Therefore, we used 30 mol/l NaHS in the drinking water to supple-
ment animals with 30 mol/l of H
2
S.
Antibodies and reagents. Rabbit polyclonal antibodies to desmin
and nephrin were purchased from Abcam (Cambridge, MA). Anti--
actin antibody, NaHS, and other analytical reagents were from Sigma-
Aldrich (St. Louis, MO). Horseradish peroxidase-linked anti-rabbit
IgG antibody was from Santa Cruz Biotechnology (Santa Cruz, CA).
PVDF membrane was from Bio-Rad (Hercules, CA).
Measurement of plasma Hcy. The high-performance liquid chro-
matography (HPLC) apparatus, chromatographic conditioning, and
sample preparation for plasma Hcy measurement were adopted from
a previously reported procedure (25) with modifications as described
earlier (39).
Measurement of plasma H
2
S. To measure the H
2
S concentration in
the plasma of each of the experimental groups, 100 l of aliquots were
mixed with 50 l of water in microcentrifuge tubes containing 300 l
of zinc acetate (1% wt/vol) to trap H
2
S. The reaction was stopped after
5 min by adding 200 lofN,N-dimethyl-p-phenylenediamine sulfate
(20 mM in 7.2 M HCl), immediately followed by addition of 200 l
of FeCl
3
(30 mM in 1.2 M HCl). The mixture was kept in the dark for
20 min. To precipitate protein from the plasma, 150 l of trichloro-
acetic acid (10% wt/vol) was added. The mixture was then centrifuged
at 10,000 gfor 10 min, and the absorbance of the resulting supernatant
was determined at 670 nm (53) in a 96-well plate using a spectro-
photometer (Spectramax M2; Molecular Devices, Sunnyvale, CA).
All samples were assayed in duplicate, and H
2
S concentration in the
plasma was calculated against a calibration curve of NaHS (3.125–
100 M).
Urinary protein measurement. Mice were housed in metabolic
cages to collect 24-h urine samples for quantitative determination of
protein content in the urine. Quantitative urinary protein concentration
was determined using the Bio-Rad protein assay reagent on the basis
of the Bradford dye-binding procedure (4). Protein concentrations
were calculated using the calibration curve prepared from a standard
solution (0 –2 mg/ml) of BSA (Sigma Chemical).
In vitro MMP-2, -9 activities assay. Gelatin zymography was
performed using 1.5% in gel gelatin as described elsewhere (28). In
brief, glomerular tissues were minced into small pieces in ice-cold
extraction buffer (1:3 wt/vol) containing (in mmol/l) 10 cacodylic
acid, 20 ZnCl, 1.5 NaN
3
, and 0.01% Triton X-100 (pH 5.0) and
incubated overnight at 4°C with gentle shaking. The homogenate was
then centrifuged for 10 min at 800 g, and supernatant was collected.
Protein concentration in the sample was measured using Bradford
method, and 100 g of the protein was electrophoretically resolved
for each sample in 8% SDS-PAGE containing 1.5% gelatin as MMP
substrate. Gels were washed in 2.5% Triton X-100 for 30 min to
remove SDS, rinsed in water, and incubated for at least 24 h in
activation buffer (50 mmol/l TrisHCl, 5 mmol/l CaCl
2
, and 0.02%
NaN
3
, pH 7.5) at 37°C in a water bath with gentle shaking. Gels were
then transferred to staining solution (acetic acid:methanol:water, 10:
50:40) containing 0.5% Coomassie blue for1hatroom temperature.
MMP activity in the gel was detected in a dark blue background with
white bands.
Cryosectioning. The kidneys were excised, and appropriate por-
tions of the kidney were cryopreserved in Peel-A-Way disposable
plastic tissue embedding molds (Polysciences, Warrington, PA) con-
taining tissue freezing media (Triangle Biomedical Sciences, Durham,
NC). These molds were kept frozen (70°C) until serial 5-m tissue
sections were made in Cryocut 1800 (Reichert-Jung). Cryosections
were placed on Superfrost/plus microscope slides and air dried.
TUNEL staining. Apoptotic cells were detected and quantified
using an in situ Apoptosis Detection Kit [TACS Terminal deoxynucleo-
tidyl Transferase (TdT) kit; R&D Systems, Minneapolis, MN] following
manufacturer instructions. Briefly, 5-m kidney tissue cryosections were
fixed with 3.7% formaldehyde solution and the permeabilized with
proteinase K. Endogenous peroxidase was quenched using hydrogen
peroxide (H
2
O
2
). Next, biotinylated nucleotides were incorporated into
3-OH ends of the DNA fragments by TdT. Positive controls were
generated by treating samples with TACS-Nuclease before TdT labeling.
The biotinylated nucleotides were detected using streptavidin-horseradish
peroxidase conjugate followed by the substrate, TACS Blue Label.
Apoptotic cells were detected by those that exhibit blue nuclear staining.
Western blots. The kidney tissue homogenates were prepared using
protein extraction buffer (0.01 M cacodylic acid pH 5.0, 0.15 M NaCl,
1M ZnCl
2
, 0.02 M CaCl
2
, 0.0015 M NaN
3
, and 0.01% vol/vol
Triton X-100). The extracted proteins were collected, and pH rose to
7.5 by adding 0.1 M Tris. Equal amounts of protein were analyzed on
10% SDS-PAGE, transferred to PVDF membrane, and probed with
appropriate antibodies following the earlier adopted method (39).
Detection of ROS. The method to detect ROS, specifically super-
oxide, was adopted from Dayal et al. (9). The oxidative fluorescent
dye, dihydroethidium (DHE; Invitrogen, Carlsbad, CA), was used in
frozen kidney sections, and the intensity of the fluorescent was
measured by laser scanning confocal microscopy (Fluo View 1000,
Olympus). Control sections were preincubated for 30 min with 250
U/ml polyethylene glycol-superoxide dismutase (PEG-SOD; Sigma-
Aldrich) before incubation with DHE. Fluorescent images were ana-
lyzed with ImagePro software (Media Cybernetics, Bethesda, MD).
Glutathione assay. Kidney tissue levels of reduced glutathione
(GSH) and oxidized glutathione (GSSG) were measured using a
commercially available kit (Cayman Chemicals, Ann Arbor, MI). The
GSH-to-GSSG ratio was calculated for each sample according to
manufacturer’s instructions.
Statistical analysis. Values are given as means SE from n
number of animals in each group as mentioned in each of the figure
legends. Differences between groups were tested with the use of
two-way ANOVA for repeated measures. Comparisons between
groups were made with the use of Student’s independent t-test.
Significance was accepted at P0.05.
F411H
2
S MODULATES RENAL INSUFFICIENCY
AJP-Renal Physiol VOL 297 AUGUST 2009 www.ajprenal.org
by 10.220.33.4 on November 27, 2017http://ajprenal.physiology.org/Downloaded from
RESULTS
Total plasma Hcy significantly increased in CBS(
/
)
mice. HPLC was performed to measure plasma levels of Hcy
from the experimental and control samples. Hcy from the
samples were identified according to the retention times and
cochromatography with standards. In CBS(/) 2-K mice, the
plasma Hcy level was found to be significantly higher com-
pared with WT 2-K mice (Fig. 1). This increase of Hcy level
in 8-wk-postsurgery mice was even higher and more dramatic
in CBS(/) 1-K mice compared with age-matched WT 1-K
mice. Although there was a tendency of higher plasma Hcy
level in WT 1-K mice compared with WT 2-K mice, the
difference was not significant. Additionally, there were no
further changes of plasma Hcy levels in the similar groups of
mice supplemented with NaHS (Fig. 1). These results clearly
suggest that, although 2-K CBS(/) mice develop high Hcy,
this effect was more acute in CBS(/) 1-K mice, and H
2
S
supplementation does not have a role in the plasma Hcy level.
NaHS supplementation increased total plasma H
2
S level.
Plasma total H
2
S levels were decreased 12% in WT 1-K
mice compared with WT 2-K mice, whereas there was virtually
no difference between WT 2-K mice supplemented with or
without NaHS (Fig. 2). However, a significant increase in
plasma H
2
S was observed in WT 1-K mice supplemented with
NaHS, which was comparable to WT 2-K mice. H
2
S level in
CBS(/) 2-K mice was found significantly lower than in WT
2-K littermates. This difference was even higher in CBS(/)
1-K mice. Interestingly, NaHS treatment in CBS(/) 2-K
mice showed a significant increase (32%, compared with its
2-K littermates without NaHS supplementation) in plasma H
2
S
level, which was comparable to WT 2-K mice. Similar results
were observed (29% increase of plasma H
2
S level, compared
with its 1-K littermates without NaHS supplementation) in
CBS(/) 1-K mice treated with NaHS (Fig. 2). These results
suggest that HHcy is associated with decreased plasma H
2
S
level and that exogenous supplementation of NaHS increases
plasma total H
2
S level.
Hydrogen sulfide prevented proteinuria in 1-K mice. Urinary
protein concentration was higher in CBS(/) 2-K mice
compared with WT 2-K mice (Fig. 3). In these animals,
however, NaHS did not affect total urinary protein excretion.
Compared with WT 2-K mice, WT 1-K mice showed signifi-
cant hyperproteinuria. This effect was even more acute in
CBS(/) 1-K mice. Interestingly, in response to NaHS, the
increased proteinuria in both the WT 1-K and CBS 1-K mice
was normalized. This result suggests that increased protein-
uria was attributable to HHcy-associated glomerular dam-
age, and this damage can be partially prevented by H
2
S
supplementation.
MMP activities attenuated by H
2
S. MMPs are involved in
the remodeling process in the normal and diseased vascular
beds. Hence, we examined the MMP-2 and -9 activities in the
kidney because these two MMPs are critically involved in the
glomerular matrix remodeling process. As shown in the Fig. 4,
Fig. 1. Effect of uninephrectomy (1-K) and H
2
S supplementation on plasma
homocysteine (Hcy) level. Total homocysteine was extracted from plasma and
analyzed by high-performance liquid chromatography as described in MATE-
RIALS AND METHODS using our previously adopted procedure. Data represent
the means SE, n4; NaHS, 30 mol/l. *Significant difference (P0.05)
compared with respective wild-type (WT) two kidney mice (2-K). **Signifi-
cant difference (P0.05) compared with respective WT 1-K. CBS, cystathi-
onine--synthase.
Fig. 2. Effect of NaHS supplementation on plasma H
2
S level. Reduced H
2
S
production was observed in CBS(/) 2-K, as well as CBS(/) 1-K and
WT 1-K mice. Interestingly, NaHS supplementation increased total plasma
H
2
S level in these mice. *Significant differences (P0.05) compared with
WT 2-K mice. **P0.05 vs. CBS(/) 2-K mice; ⌽⌽P0.05 vs.
CBS(/) 1-K. Data represent the means SE, n6 animals in each group.
Fig. 3. Role of H
2
S in homocysteine-associated proteinuria. Urinary protein
was measured using Bradford method. *Significant difference (P0.05)
compared with WT 2-K. **P0.05 vs. WT 1-K mice; P0.05 vs.
CBS(/) 2-K; ⌽⌽P0.05 vs. CBS(/) 1-K. Data represent the means
SE, n8 animals per group.
F412 H
2
S MODULATES RENAL INSUFFICIENCY
AJP-Renal Physiol VOL 297 AUGUST 2009 www.ajprenal.org
by 10.220.33.4 on November 27, 2017http://ajprenal.physiology.org/Downloaded from
WT 2-K mice showed basal level of the proform and active
form of MMP-9 activity as well as active form of MMP-2
activity in the cortical tissue extract. The activities of these
MMPs were elevated in WT 1-K mice. Contrary to WT 2-K
mice, CBS(/) 2-K mice showed a very high level of these
MMP activities, which were further elevated in CBS(/)
1-K mice (Fig. 4). Interestingly, H
2
S supplementation almost
normalized these MMP activities in WT 1-K and both in
CBS(/) 2-K and 1-K mice. These results suggest that both
the MMP-2 and -9 play a major role in high Hcy-associated
renopathy. Importantly, modulation of these two MMPs by
H
2
S suggests a possible trigger of both these of MMPs during
HHcy in the renal cortex.
H
2
S prevented HHcy-associated glomerular cell death. We
determined the glomerular cell apoptosis by TUNEL staining.
Representative images of TUNEL staining showed that, in the
glomerulus of WT 2-K kidney, there were very few apoptotic
podocytic cells (Fig. 5). Similarly, WT 2-K mice treated with
NaHS showed very few dead cells. WT 1-K mice showed a
relatively high number of apoptotic cells (Fig. 5). These in-
creases in apoptotic cells were absent when WT 1-K mice were
supplemented with NaHS.
In the CBS(/) 2-K mice, however, although a very
minimal number of apoptotic cells was identified, the number
of apoptotic cells was higher than in WT 2-K littermates (Fig.
5). This number was dramatically increased in CBS(/) 1-K
mice, which was almost completely prevented in the mice
treated with NaHS. Likewise, in the kidney of CBS(/) 2-K
mice treated with NaHS, a minimal number of apoptotic cells
was observed.
Expression of desmin and nephrin during HHcy. A basal
level of desmin expression was observed in CBS(/) 2-K
mice. This expression was almost comparable with WT 2-K
mice; however, the expression was increased significantly in
both WT 1-K and CBS(/) 1-K mice although the expres-
sion was higher in CBS(/) 1-K mice (Fig. 6). WT 1-K and
CBS(/)1-K mice supplemented with NaHS prevented the
increased expression of desmin, and the levels were compara-
ble to their respective 2-K littermates. Contrary to the desmin
expression, the expression of nephrin in the kidney was found
to be opposite in the respective experimental groups. NaHS
treatment, however, prevented the changes of nephrin expres-
sions in WT 1-K and both the CBS(/) 2-K and CBS(/
)1-K mice (Fig. 6).
Increased ROS production was associated with HHcy. Pro-
duction of ROS in the glomerulus was measured using oxida-
tive fluorescent dye, DHE (Fig. 7). The DHE fluorescence
indicates ROS production. When CBS(/) mice were sup-
plemented with exogenous H
2
S, a significant decrease in DHE
fluorescence was observed. The fluorescence intensity in
CBS(/) 2-K mice was higher compared with WT 2-K
age-matched littermates. Interestingly, in CBS(/) 1-K
mice, a robust increase of DHE fluorescence was observed.
PEG, which is covalently linked to SOD (PEG-SOD) and a
potent superoxide (O
2
) anion scavenger, dramatically dimin-
ished DHE fluorescence, suggesting that the increased DHE
fluorescence observed in CBS(/)1-K was attributable to
increased production of O
2
.
H
2
S increased cortical tissue GSH-to-GSSG ratio. GSH is a
potent intracellular antioxidant, and GSH-to-GSSG ratio cor-
responds to the capacity of a cell to attenuate oxidative stress
(13). To assess the oxidative redox state in our experimental
groups, we measured the cortical-tissue reduced GSH and
GSH-to-GSSG ratio, and the calculated data are shown in
Fig. 8. There was a significant decrease of GSH level in WT
1-K and both in CBS(/) 2-K and CBS(/)1-K mice
compared with WT 2-K control groups. Also, a significant
decrease was observed in GSH/GSSG ratio in these groups.
Interestingly, H
2
S supplementation normalized the GSH and
GSH/GSSG ratio, suggesting that H
2
S maintained the intracel-
lular antioxidant capacity.
Fig. 4. H
2
S regulates matrix metalloproteinase (MMP)-2
and -9 activities during hyperhomocysteinemia. In gel, gelatin
zymography was performed to measure MMP-2 and -9 activi-
ties in the kidney cortex tissue-extracted protein. Both proforms
and active forms of MMP-9 and active form of MMP-2 in WT
1-K, CBS(/) 2-K, and 1-K mice were found to be signifi-
cantly higher than WT 2-K littermates. These MMP activities
were attenuated by H
2
S supplementation (NaHS, 30 mol/l) for
the period of 8 wk postsurgery; data represent means SE, n
5 for WT mice per group, and n6 for CBS(/) mice per
group; *P0.01 vs. WT 2-K.
F413H
2
S MODULATES RENAL INSUFFICIENCY
AJP-Renal Physiol VOL 297 AUGUST 2009 www.ajprenal.org
by 10.220.33.4 on November 27, 2017http://ajprenal.physiology.org/Downloaded from
DISCUSSION
The present study clearly demonstrates that plasma Hcy was
increased in CBS(/) mice, and this increase was further
elevated when renal function was reduced by the removal of
one kidney. The plasma Hcy was also correlated with the
extent of renal damage as detected by proteinuria. Plasma H
2
S
levels were found to be the opposite to that of the Hcy levels,
and this decrease in H
2
S level was elevated back toward
normal with exogenous supplementation of NaHS, a donor of
H
2
S. The increased MMP-2 and -9 activities and the occur-
Fig. 5. Effect of H
2
S on glomerular cell death. Histological kidney sections were analyzed for in situ apoptosis as described in MATERIALS AND METHODS. Numbers
of dead cells were counted under the microscope from 15 randomized fields in each group, quantitated, and plotted as bar diagram as shown; n7 in each group;
200 magnification.
2K WT
2K WT
Fig. 6. H
2
S normalized tissue expression of desmin and
nephrin. Protein was extracted from the kidney tissue and
was analyzed by Western blot. Equal amount of protein
was loaded in each well, and the expression of each of
the proteins was normalized with -actin. *Significant
difference (P0.05) compared with WT 2-K. **P
0.05 vs. WT 1-K mice; P0.05 vs. CBS(/) 2-K;
⌽⌽P0.05 vs. CBS(/) 1-K. Data represent the
means SE, n6 per group.
F414 H
2
S MODULATES RENAL INSUFFICIENCY
AJP-Renal Physiol VOL 297 AUGUST 2009 www.ajprenal.org
by 10.220.33.4 on November 27, 2017http://ajprenal.physiology.org/Downloaded from
rence of glomerular cell death were associated with the in-
creased oxidative stress, which has been shown to be associ-
ated with HHcy. In addition to that, the increased proteinuria
was corroborated with the renal damage, as evidenced by
increased expression of podocyte injury marker, desmin, and
decreased expression of glomerular slit diaphragm protein,
nephrin. Interestingly, the extent of HHcy-associated renal
damage was ameliorated by H
2
S supplementation. Although it
is not completely understood, it appears that the defense
mechanism of H
2
S was exhibited, in part, through reducing
oxidative stress and maintaining intracellular GSH-to-GSSG
ratio. This mechanism, in addition to normalizing the expres-
sion of desmin and nephrin, prevented glomerular cell apopto-
sis and restored basal MMP activities. These resulted in rever-
sals of proteinuria in our hyperhomocysteinemic experimental
animal model treated with H
2
S.
HHcy is a risk factor for chronic kidney disease and is
associated with end-stage renal disease. In disease condition,
such as in diabetic nephropathy, Hcy disposal and clearance
are impaired and therefore accumulate in the body, resulting in
increase of plasma and tissue levels of Hcy (43). This, in turn,
causes renal microvascular impairment and vasoconstriction
(37), which lead to renal volume retention and further accu-
mulation of Hcy (38). This is a vicious cycle and is often
associated with chronic renal failure. The purpose of unine-
phrectomy (1-K), in the present study, was to impair the renal
function to subnormal level, which further accumulates Hcy
through increased renal volume retention. The results from our
study suggest that 1-K mice have more Hcy in the plasma than
their respective 2-K control groups. Therefore, this model was
created and used to investigate Hcy-associated renal damage
and remodeling process in the kidney.
In the body, Hcy is metabolized by two enzymes, CBS and
CSE, and produces a gaseous substance, H
2
S (16, 42). CBS is
a predominant H
2
S-generating enzyme in the brain and ner-
vous tissues (1, 12), and CSE is mainly expressed in the liver,
kidney, and vascular smooth muscle cells (3, 61). Although
Hcy has been shown to promote CSE activity at its lower
Fig. 7. H
2
S mitigated reactive oxygen species (ROS) production. Production of ROS in the frozen kidney sections was measured using oxidative fluorescent dye,
dihydroethidium (DHE), as described in MATERIALS AND METHODS. DHE fluorescence was detected at a higher level in the glomerulus of CBS(/) 1-K mice.
Interestingly, CBS(/) 1-K mice supplemented with exogenous H
2
S showed a significant decrease in DHE fluorescence. In addition, preincubation of
CBS(/) 1-K kidney sections with polyethylene glycol-superoxide dismutase (PEG-SOD) dramatically diminished DHE fluorescence, suggesting that the
major portions of ROS are superoxide (O
2
). *Significant difference (P0.05) vs. CBS(/) 1-K (means SE, n5– 6 animals in each group).
Magnification, 200.
F415H
2
S MODULATES RENAL INSUFFICIENCY
AJP-Renal Physiol VOL 297 AUGUST 2009 www.ajprenal.org
by 10.220.33.4 on November 27, 2017http://ajprenal.physiology.org/Downloaded from
concentrations, inhibition of CSE activity was reported at its
higher concentration in the rat liver (54). At pathological
conditions, elevated levels of Hcy alter the transulfuration
pathway by inhibiting CSE enzyme activity (6), thereby reduc-
ing endogenous production of H
2
S in the body. In the present
study, we investigated the effect of an increased level of
plasma Hcy, not the glomerular tissue level of Hcy, on the
glomerulus and its consequences in the renal remodeling pro-
cess. CBS(/) mice show higher levels of plasma Hcy and a
very reliable model to test HHcy-related disease process.
Additionally, it is also reported that HHcy inhibits CSE (6);
therefore, the use of CBS(/) mice is advantageous over
CSE(/). Hence, we used CBS(/) mice for the present
study to investigate the HHcy-related renal-damaging effect.
The growing body of evidence suggests that, despite its past
reputation as a noxious gas, H
2
S is rapidly emerging as a third
gaseous transmitter, in addition to nitric oxide and carbon
monoxide (16, 17, 46); the physiological function of endoge-
nous H
2
S, however, is not clear. It is reported that H
2
Sis
involved in the regulation of vascular tone (24) and hyperten-
sion (52) and protects neuronal cells from oxidative stress by
increasing the intracellular concentration of antioxidant and
GSH (19, 48). Recently, Tripatara et al. (44) demonstrated that
generation of endogenous H
2
S limits renal ischemia/reperfu-
sion injury and dysfunction. In the present study, to test any
protective effects of H
2
S in HHcy-associated renal damage, we
used CBS(/) mutant mice. These mice have an 50%
reduction in CBS mRNA and enzyme activity in the liver and
have twice the normal plasma Hcy levels than WT littermates
(35). Thus the CBS(/) mice develop mild HHcy and are a
very good model to study HHcy-related disease processes,
including cardiovascular-renal diseases. Reports are available
that CBS inhibitors hydroxylamine and amino-oxyacetate sup-
press the production of H
2
S and that a CBS activator, S-
adenosyl-L-methionine, enhances H
2
S production in the brain
tissue (1). A similar trend was observed by Xia et al. (50)
where inhibition of CBS reduced endogenous generation of
H
2
S in the kidney tissues. These reports indicated the regula-
tory role of CBS enzyme in the production of endogenous H
2
S.
Our present result (Fig. 1) suggested that HHcy was associated
with CBS deficiency, where CBS(/) mutant mice exhibited
a higher level of plasma Hcy. This increase of plasma Hcy
level was even higher in CBS(/) mice after uninephrec-
tomy (1-K). Thus the results clearly indicated a strong rela-
tionship between renal insufficiency and plasma Hcy level.
Because Hcy is one of the precursors of endogenous H
2
S
generation, it was expected that increase of plasma Hcy level
would eventually be elevated in the plasma H
2
S level. Contrary
to this mechanism, the data presented in Fig. 2 showed that the
plasma H
2
S level was, in fact, inversely regulated by plasma
Hcy level, where increased Hcy repressed H
2
S level in the
plasma. This may be due to a negative feedback mechanism,
where increased plasma Hcy inhibited its metabolizing en-
zyme, CSE and/or CBS, resulting in low production of H
2
S.
We previously reported that stress, such as volume overload,
decreased CSE expression and subsequent H
2
S generation in
the cardiac tissue (40). Whether this mechanism is still appli-
cable to HHcy-associated oxidative stress and regulates the
plasma H
2
S level needs to be verified. Nevertheless, the results
from our present study indicated the possibility of such mech-
anisms that may be involved in part, if not the only mechanism,
which triggered endogenous generation of H
2
S and subsequent
plasma level of this gaseous substance.
Along the same line, Wei et al. (48) showed that severe
oxidative stress was present in a model of hypoxic pulmonary
hypertension and was accompanied by a decrease in the en-
dogenous production of H
2
S in the lung tissue. H
2
S, however,
acted as an antioxidant during this oxidative stress, which was
a result of the attenuated GSSG content. More recently, per-
fusion of H
2
S in ischemia-reperfusion-injured lung has been
shown to reduce malondialdehyde production, potentiated
SOD, and catalase (CAT) activities and restrain superoxide
(O
2
) production in the lung, resulting in an attenuated oxi-
dative lung injury (15). This emerging evidence suggests the
potential antioxidant properties of H
2
S in normal and patho-
physiological conditions. To determine the antioxidant role of
H
2
S, we measured the reduced GSH content and reduced
GSH-to-GSSG ratio in our study. Our results suggested that
H
2
S supplementation normalized the reduced GSH and re-
duced-GSH-to-GSSG ratio associated with HHcy. In our ex-
periments, although we have found that H
2
S supplementation
increased GSH/GSSG ratio in the kidney tissues, the exact
mechanisms, however, were not elucidated. Recently, Liu et al.
(23) reported that H
2
S protected intestinal ischemia-reperfu-
sion injury by increasing serum and intestinal level of SOD and
GSH peroxidase (23). Our laboratory has previously shown
that, in vitro, H
2
S enhanced the inhibitory effects of CAT and
SOD in methionine-loaded oxidative stress in mouse brain
endothelial cells (45). This mechanism has clearly indicated
Fig. 8. Decreased glutathione (GSH)/oxidized glutathione (GSSG) ratio was
normalized by H
2
S. Cortical tissue reduced GSH, and GSH-to-GSSG ratio was
measured and calculated as described in MATERIALS AND METHODS.H
2
S therapy
significantly improved GSH level in WT 1-K and in CBS(/)2-K and
CBS(/)1-K mice. Additionally, H
2
S preserved the GSH/GSSG ratio com-
pared with respective nontreated groups. Data represent means SE, n7
per group. *P0.05 vs. 2-K WT.
F416 H
2
S MODULATES RENAL INSUFFICIENCY
AJP-Renal Physiol VOL 297 AUGUST 2009 www.ajprenal.org
by 10.220.33.4 on November 27, 2017http://ajprenal.physiology.org/Downloaded from
the antioxidant property of H
2
S and was partly mediated by
increasing intracellular CAT and SOD. The present study did
not aim to elucidate this mechanism; however, whether or not
the same mechanism applies to HHcy-associated renal remod-
eling needs to be explored comprehensively. Interestingly, in
the present study, increased superoxide (O
2
) production was
attenuated with H
2
S supplementation in the mice that exhibited
high Hcy. These results suggest that H
2
S protects glomerular
tissue, at least in part, through its antioxidant properties.
H
2
S had been reported to be a general protective mechanism
for degenerative organ damages. Herein, we have focused our
study to ameliorate HHcy-associated kidney damages, if any,
through exogenous H
2
S supplementation. It is a need of future
investigation to elucidate whether H
2
S could be such a protec-
tive factor in the hypertensive kidney damages. For example,
does H
2
S protect DOCA salt-induced kidney damage? It is
reported that DOCA salt-induced hypertension decreased SOD
in the rat aorta and that antioxidant therapy increased SOD
activity (31). Given the fact that both DOCA salt and Hcy
induce oxidative stress through inhibition of SOD, and H
2
Sis
an enhancer of intracellular SOD, it is possible that H
2
S may
also play a role to increase SOD activity in the kidney of
DOCA salt-induced hypertensive rat. Although this is a very
interesting area to study antioxidant properties of H
2
S, the
scope of the present investigation, however, was limited to
investigate the protective role of H
2
S in HHcy-associated renal
damages.
Clinical studies have implicated proteinuria as a key prog-
nostic factor for renal complications in hypertension; however,
the pathogenesis causing proteinuria is poorly understood (30).
It is reported that dysfunction of podocytes, the final filtration
barrier in the glomerulus, plays a pivotal role in proteinuria
(29, 32, 47). Yi et al. (55) have shown that urinary albumin
excretion increased at the second week of methionine, the
precursor of Hcy, treatment. These reports established a strong
correlation between high Hcy and kidney disease. In our
present study, we have found that proteinuria was strongly
correlated with high Hcy and attenuated H
2
S in the plasma.
Increased Hcy levels were associated with apoptosis of podo-
cytes, in part attributable to reduction of H
2
S and increased
oxidative stress, and therefore damaged the final filtration
barrier of the kidney. These changes in the glomerulus allowed
excessive protein excretion in the urine. On the contrary to this
mechanism, H
2
S supplementation prevented podocyte apopto-
sis, and this provided protection to the kidney against HHcy-
associated renal damage and proteinuria. It is important to men-
tion that, although the level of protein excretion was high in CBS
(/) 2-K mice compared with WT 2-K mice, the difference was
not significant even though the reduction of plasma H
2
S levels
was significant in CBS(/) 2-K mice compared with WT 2-K
mice. Herein, it is possible that other antioxidants in the tissue,
such as CAT, SOD, lipid peroxidase, etc., may have played a
similar role as of H
2
S, to repress the oxidative stress, and therefore
ameliorated proteinuria in CBS(/) 2-K mice. Thus the ob-
served proteinuria in CBS(/) mice was not parallel with the
reduction of H
2
S levels. Further understanding of the regulatory
mechanisms of proteinuria by H
2
S in HHcy-associated renal
failure may provide a new insight into the molecular mechanisms
of this disease process.
Proteinuria is a hallmark of renal complication and a major
deteriorating factor for the progression to end-stage renal
diseases (34). The outer aspect of glomerular basement mem-
brane is lined up with very specialized visceral epithelial cells,
named podocytes, and these podocytes serve as the final
defense against urinary protein loss in the normal glomerulus
(30). Any damage to the podocytes and their slit diaphragm is
intimately associated with proteinuria (32). Biochemical as-
sessment of normal slit diaphragm component, such as nephrin
(18), and injured podocyte marker desmin (14) are now there-
fore considered as two major sensitive markers of podocyte
injury and subsequently glomerulopathy in renal diseases.
Therefore, we measured desmin and nephrin in the kidney
tissue extract to asses whether the kidney injury is associated
with a high level of plasma Hcy and whether H
2
S supplemen-
tation can ameliorate this change. In our present study, we have
found a conspicuous increase in the expression of desmin,
whereas expression of nephrin was decreased in the mice
showing high Hcy levels and decreased plasma H
2
S level. H
2
S
supplementation reversed the effect on these two protein ex-
pressions associated with high Hcy, suggesting the regulatory
role of H
2
S on these two proteins during HHcy.
High Hcy has been reported to produce a sustained and
abnormal elevation of glomerular arterial wall stress through
generation of ROS (57, 58). This stress initiates a complex and
progressive glomerular remodeling, including activation of
MMPs, collagen degradation, glomerular hypertrophy, and
dysfunction (22, 37). One of the major causes of glomerular
sclerosis and renal dysfunction is the increase of glomerular
ECM. Glomerular ECM, which is composed of mesangial
matrix and basement membrane, plays an important role in
physical, mechanical, and structural functions of the glomeru-
lus. MMPs degrade both the collagenous and noncollagenous
components of the ECM and are thereby actively involved in
matrix turnover. In pathological conditions, collagenases initi-
ate the degradation process of ECM and denature collagen into
nonhelical gelatin derivatives. Gelatinases, which are a mem-
ber of the family of MMPs, digest these products into smaller
peptides. Of particular interest are gelatinases MMP-2 and
MMP-9, which have potential capability to disrupt the kidney
architecture by virtue of their specificity for various compo-
nents of basement membrane (33). Therefore, the measurement
of MMP-2 and -9 activities allows for an estimation of the
remodeling process during HHcy-associated glomerulopathies.
Data from our present report suggested that high Hcy induced
elevation of superoxide (O
2
) production in the CBS(/)
kidney, and this production of O
2
was even greater in 1-K
mice. The level of MMP-2 and -9 activities followed the
increasing trend of O
2
production in both 2-K and 1-K
CBS(/) mice. H
2
S supplementation normalized both MMP
activity level and the increased level of O
2
production in
these mice. This result suggests that the activities of MMP-2
and -9 are associated with increased O
2
production and that
H
2
S scavenges O
2
production, thereby regulating MMP
activities in our experimental condition. This regulatory mech-
anism of O
2
by H
2
S prevented renal damage and subsequent
renal failure associated with HHcy.
In summary, we have shown that elevation of plasma Hcy
level causes a decrease in the level of plasma H
2
S and is
associated with renal impairment. This increase of plasma Hcy
level induced glomerular oxidative stress, resulting in aug-
mented MMP activities and induction in glomerular cell apop-
tosis. Additionally, HHcy altered expression of desmin and the
F417H
2
S MODULATES RENAL INSUFFICIENCY
AJP-Renal Physiol VOL 297 AUGUST 2009 www.ajprenal.org
by 10.220.33.4 on November 27, 2017http://ajprenal.physiology.org/Downloaded from
final filtration barrier regulatory protein, nephrin. These
changes in cellular and protein level indicated damage in the
kidney, which was exhibited by proteinuria, a marker of renal
failure. H
2
S supplementation, however, showed the reversal of
these deleterious changes associated with HHcy and is there-
fore protective to the kidney.
ACKNOWLEDGMENTS
This study was supported, in part, by NIH grants HL-71010, HL-88012,
HL-74185, and NS-51568.
REFERENCES
1. Abe K, Kimura H. The possible role of hydrogen sulfide as an endoge-
nous neuromodulator. J Neurosci 16: 1066 –1071, 1996.
2. Austin RC, Lentz SR, Werstuck GH. Role of hyperhomocysteinemia in
endothelial dysfunction and atherothrombotic disease. Cell Death Differ
11, Suppl 1: S56 –S64, 2004.
3. Barber T, Triguero A, Martinez-Lopez I, Torres L, Garcia C,
Miralles VJ, Vina JR. Elevated expression of liver gamma-cystathionase
is required for the maintenance of lactation in rats. J Nutr 129: 928 –933,
1999.
4. Bradford MM. A rapid and sensitive method for the quantitation of
microgram quantities of protein utilizing the principle of protein-dye
binding. Anal Biochem 72: 248 –254, 1976.
5. Chang HR, Yang SF, Li ML, Lin CC, Hsieh YS, Lian JD. Relation-
ships between circulating matrix metalloproteinase-2 and -9 and renal
function in patients with chronic kidney disease. Clin Chim Acta 366:
243–248, 2006.
6. Chang L, Geng B, Yu F, Zhao J, Jiang H, Du J, Tang C. Hydrogen
sulfide inhibits myocardial injury induced by homocysteine in rats. Amino
Acids 34: 573–585, 2008.
7. Clarke R, Daly L, Robinson K, Naughten E, Cahalane S, Fowler B,
Graham I. Hyperhomocysteinemia: an independent risk factor for vascu-
lar disease. N Engl J Med 324: 1149 –1155, 1991.
8. Cruz CI, Ruiz-Torres P, del Moral RG, Rodriguez-Puyol M, Rodri-
guez-Puyol D. Age-related progressive renal fibrosis in rats and its
prevention with ACE inhibitors and taurine. Am J Physiol Renal Physiol
278: F122–F129, 2000.
9. Dayal S, Wilson KM, Leo L, Arning E, Bottiglieri T, Lentz SR.
Enhanced susceptibility to arterial thrombosis in a murine model of
hyperhomocysteinemia. Blood 108: 2237–2243, 2006.
10. Doeller JE, Grieshaber MK, Kraus DW. Chemolithoheterotrophy in a
metazoan tissue: thiosulfate production matches ATP demand in ciliated
mussel gills. J Exp Biol 204: 3755–3764, 2001.
11. Doeller JE, Isbell TS, Benavides G, Koenitzer J, Patel H, Patel RP,
Lancaster JR Jr, Darley-Usmar VM, Kraus DW. Polarographic mea-
surement of hydrogen sulfide production and consumption by mammalian
tissues. Anal Biochem 341: 40 –51, 2005.
12. Eto K, Ogasawara M, Umemura K, Nagai Y, Kimura H. Hydrogen
sulfide is produced in response to neuronal excitation. J Neurosci 22:
3386 –3391, 2002.
13. Exner R, Wessner B, Manhart N, Roth E. Therapeutic potential of
glutathione. Wien Klin Wochenschr 112: 610 616, 2000.
14. Floege J, Kriz W, Schulze M, Susani M, Kerjaschki D, Mooney A,
Couser WG, Koch KM. Basic fibroblast growth factor augments podo-
cyte injury and induces glomerulosclerosis in rats with experimental
membranous nephropathy. J Clin Invest 96: 2809 –2819, 1995.
15. Fu Z, Liu X, Geng B, Fang L, Tang C. Hydrogen sulfide protects rat
lung from ischemia-reperfusion injury. Life Sci 82: 1196 –1202, 2008.
16. Geng B, Yang J, Qi Y, Zhao J, Pang Y, Du J, Tang C. H2S generated
by heart in rat and its effects on cardiac function. Biochem Biophys Res
Commun 313: 362–368, 2004.
17. Hosoki R, Matsuki N, Kimura H. The possible role of hydrogen sulfide
as an endogenous smooth muscle relaxant in synergy with nitric oxide.
Biochem Biophys Res Commun 237: 527–531, 1997.
18. Kawachi H, Koike H, Kurihara H, Yaoita E, Orikasa M, Shia MA,
Sakai T, Yamamoto T, Salant DJ, Shimizu F. Cloning of rat nephrin:
expression in developing glomeruli and in proteinuric states. Kidney Int
57: 1949 –1961, 2000.
19. Kimura Y, Dargusch R, Schubert D, Kimura H. Hydrogen sulfide
protects HT22 neuronal cells from oxidative stress. Antioxid Redox Signal
8: 661– 670, 2006.
20. Kimura Y, Kimura H. Hydrogen sulfide protects neurons from oxidative
stress. FASEB J 18: 1165–1167, 2004.
21. Lelongt B, Legallicier B, Piedagnel R, Ronco PM. Do matrix metallo-
proteinases MMP-2 and MMP-9 (gelatinases) play a role in renal devel-
opment, physiology and glomerular diseases? Curr Opin Nephrol Hyper-
tens 10: 7–12, 2001.
22. Li N, Chen YF, Zou AP. Implications of hyperhomocysteinemia in
glomerular sclerosis in hypertension. Hypertension 39: 443– 448, 2002.
23. Liu H, Bai XB, Shi S, Cao YX. Hydrogen sulfide protects from intestinal
ischaemia-reperfusion injury in rats. J Pharm Pharmacol 61: 207–212,
2009.
24. Lowicka E, Beltowski J. Hydrogen sulfide (H
2
S)—the third gas of
interest for pharmacologists. Pharmacol Rep 59: 4 –24, 2007.
25. Malinow MR, Kang SS, Taylor LM, Wong PW, Coull B, Inahara T,
Mukerjee D, Sexton G, Upson B. Prevalence of hyperhomocyst(e)inemia
in patients with peripheral arterial occlusive disease. Circulation 79:
1180 –1188, 1989.
26. McCully KS. Chemical pathology of homocysteine. I. Atherogenesis. Ann
Clin Lab Sci 23: 477– 493, 1993.
27. McCully KS. Homocysteine and vascular disease. Nat Med 2: 386 –389,
1996.
28. Moshal KS, Rodriguez WE, Sen U, Tyagi SC. Targeted deletion of
MMP-9 attenuates myocardial contractile dysfunction in heart failure.
Physiol Res 57: 379 –384, 2008.
29. Mundel P, Shankland SJ. Podocyte biology and response to injury. JAm
Soc Nephrol 13: 3005–3015, 2002.
30. Nagase M, Shibata S, Yoshida S, Nagase T, Gotoda T, Fujita T.
Podocyte injury underlies the glomerulopathy of Dahl salt-hypertensive
rats and is reversed by aldosterone blocker. Hypertension 47: 1084–1093,
2006.
31. Ndisang JF, Lane N, Jadhav A. Crosstalk between the heme oxygenase
system, aldosterone, and phospholipase C in hypertension. J Hypertens 26:
1188 –1199, 2008.
32. Pavenstadt H, Kriz W, Kretzler M. Cell biology of the glomerular
podocyte. Physiol Rev 83: 253–307, 2003.
33. Rao VH, Lees GE, Kashtan CE, Nemori R, Singh RK, Meehan DT,
Rodgers K, Berridge BR, Bhattacharya G, Cosgrove D. Increased
expression of MMP-2, MMP-9 (type IV collagenases/gelatinases), and
MT1-MMP in canine X-linked Alport syndrome (XLAS). Kidney Int 63:
1736 –1748, 2003.
34. Remuzzi G, Bertani T. Pathophysiology of progressive nephropathies.
N Engl J Med 339: 1448 –1456, 1998.
35. Robert K, Chasse JF, Santiard-Baron D, Vayssettes C, Chabli A,
Aupetit J, Maeda N, Kamoun P, London J, Janel N. Altered gene
expression in liver from a murine model of hyperhomocysteinemia. J Biol
Chem 278: 31504 –31511, 2003.
36. Rodriguez WE, Sen U, Tyagi N, Kumar M, Carneal G, Aggrawal D,
Newsome J, Tyagi SC. PPAR gamma agonist normalizes glomerular
filtration rate, tissue levels of homocysteine, and attenuates endothelial-
myocyte uncoupling in alloxan induced diabetic mice. Int J Biol Sci 4:
236 –244, 2008.
37. Rodriguez WE, Tyagi N, Joshua IG, Passmore JC, Fleming JT,
Falcone JC, Tyagi SC. Pioglitazone mitigates renal glomerular vascular
changes in high-fat, high-calorie-induced type 2 diabetes mellitus. Am J
Physiol Renal Physiol 291: F694 –F701, 2006.
38. Sen U, Rodriguez WE, Tyagi N, Kumar M, Kundu S, Tyagi SC.
Ciglitazone, a PPARagonist, ameliorates diabetic nephropathy in part
through homocysteine clearance. Am J Physiol Endocrinol Metab 295:
E1205–E1212, 2008.
39. Sen U, Tyagi N, Kumar M, Moshal KS, Rodriguez WE, Tyagi SC.
Cystathionine--synthase gene transfer and 3-deazaadenosine ameliorate
inflammatory response in endothelial cells. Am J Physiol Cell Physiol 293:
C1779 –C1787, 2007.
40. Sen U, Vacek TP, Hughes WM, Kumar M, Moshal KS, Tyagi N,
Metreveli N, Hayden MR, Tyagi SC. Cardioprotective role of sodium
thiosulfate on chronic heart failure by modulating endogenous H2S gen-
eration. Pharmacology 82: 201–213, 2008.
41. Stamler JS, Osborne JA, Jaraki O, Rabbani LE, Mullins M, Singel D,
Loscalzo J. Adverse vascular effects of homocysteine are modulated by
endothelium-derived relaxing factor and related oxides of nitrogen. J Clin
Invest 91: 308 –318, 1993.
42. Swaroop M, Bradley K, Ohura T, Tahara T, Roper MD, Rosenberg
LE, Kraus JP. Rat cystathionine beta-synthase. Gene organization and
alternative splicing. J Biol Chem 267: 11455–11461, 1992.
F418 H
2
S MODULATES RENAL INSUFFICIENCY
AJP-Renal Physiol VOL 297 AUGUST 2009 www.ajprenal.org
by 10.220.33.4 on November 27, 2017http://ajprenal.physiology.org/Downloaded from
43. Tessari P, Coracina A, Kiwanuka E, Vedovato M, Vettore M, Valerio
A, Zaramella M, Garibotto G. Effects of insulin on methionine and
homocysteine kinetics in type 2 diabetes with nephropathy. Diabetes 54:
2968 –2976, 2005.
44. Tripatara P, Patel NS, Collino M, Gallicchio M, Kieswich J, Castiglia
S, Benetti E, Stewart KN, Brown PA, Yaqoob MM, Fantozzi R,
Thiemermann C. Generation of endogenous hydrogen sulfide by cysta-
thionine gamma-lyase limits renal ischemia/reperfusion injury and dys-
function. Lab Invest 88: 1038 –1048, 2008.
45. Tyagi N, Moshal KS, Sen U, Vacek TP, Kumar M, Hughes WM Jr,
Kundu S, Tyagi SC. H
2
S protects against methionine-induced oxidative
stress in brain endothelial cells. Antioxid Redox Signal 11: 25–33, 2009.
46. Wang R. Two’s company, three’s a crowd: can H
2
S be the third endog-
enous gaseous transmitter? FASEB J 16: 1792–1798, 2002.
47. Wartiovaara J, Ofverstedt LG, Khoshnoodi J, Zhang J, Makela E,
Sandin S, Ruotsalainen V, Cheng RH, Jalanko H, Skoglund U,
Tryggvason K. Nephrin strands contribute to a porous slit diaphragm
scaffold as revealed by electron tomography. J Clin Invest 114: 1475–
1483, 2004.
48. Wei HL, Zhang CY, Jin HF, Tang CS, Du JB. Hydrogen sulfide
regulates lung tissue-oxidized glutathione and total antioxidant capacity in
hypoxic pulmonary hypertensive rats. Acta Pharmacol Sin 29: 670 679,
2008.
49. Wohlgemuth SE, Taylor AC, Grieshaber MK. Ventilatory and meta-
bolic responses to hypoxia and sulphide in the lugworm Arenicola marina
(L.). J Exp Biol 203: 3177–3188, 2000.
50. Xia M, Chen L, Muh RW, Li PL, Li N. Production and actions of
hydrogen sulfide, a novel gaseous bioactive substance, in the kidneys.
J Pharmacol Exp Ther 329: 1056 –1062, 2009.
51. Yan SK, Chang T, Wang H, Wu L, Wang R, Meng QH. Effects of
hydrogen sulfide on homocysteine-induced oxidative stress in vascular
smooth muscle cells. Biochem Biophys Res Commun 351: 485– 491, 2006.
52. Yang G, Wu L, Jiang B, Yang W, Qi J, Cao K, Meng Q, Mustafa AK,
Mu W, Zhang S, Snyder SH, Wang R. H
2
S as a physiologic vasorelax-
ant: hypertension in mice with deletion of cystathionine gamma-lyase.
Science 322: 587–590, 2008.
53. Yang G, Yang W, Wu L, Wang R. H2S, endoplasmic reticulum stress,
and apoptosis of insulin-secreting beta cells. J Biol Chem 282: 16567–
16576, 2007.
54. Yao K. Effects of several unusual sulfur-containing amino acids on rat
liver cystathionine-gamma-lyase. Physiol Chem Phys 7: 401– 408,
1975.
55. Yi F, dos Santos EA, Xia M, Chen QZ, Li PL, Li N. Podocyte injury and
glomerulosclerosis in hyperhomocysteinemic rats. Am J Nephrol 27:
262–268, 2007.
56. Yi F, Li PL. Mechanisms of homocysteine-induced glomerular injury and
sclerosis. Am J Nephrol 28: 254 –264, 2008.
57. Yi F, Zhang AY, Janscha JL, Li PL, Zou AP. Homocysteine activates
NADH/NADPH oxidase through ceramide-stimulated Rac GTPase activ-
ity in rat mesangial cells. Kidney Int 66: 1977–1987, 2004.
58. Yi F, Zhang AY, Li N, Muh RW, Fillet M, Renert AF, Li PL. Inhibition
of ceramide-redox signaling pathway blocks glomerular injury in hyper-
homocysteinemic rats. Kidney Int 70: 88 –96, 2006.
59. Yonezawa D, Sekiguchi F, Miyamoto M, Taniguchi E, Honjo M,
Masuko T, Nishikawa H, Kawabata A. A protective role of hydrogen
sulfide against oxidative stress in rat gastric mucosal epithelium. Toxicol-
ogy 241: 11–18, 2007.
60. Zhang X, Li H, Jin H, Ebin Z, Brodsky S, Goligorsky MS. Effects of
homocysteine on endothelial nitric oxide production. Am J Physiol Renal
Physiol 279: F671–F678, 2000.
61. Zhao W, Zhang J, Lu Y, Wang R. The vasorelaxant effect of H(2)S as
a novel endogenous gaseous K(ATP) channel opener. EMBO J 20:
6008 6016, 2001.
F419H
2
S MODULATES RENAL INSUFFICIENCY
AJP-Renal Physiol VOL 297 AUGUST 2009 www.ajprenal.org
by 10.220.33.4 on November 27, 2017http://ajprenal.physiology.org/Downloaded from
... Despite evidence that NaHS solutions are not stable, some animal studies used NaHS solutions in drinking water as an H 2 S-donating compound [15][16][17][18][19][20][21][22][23][24][25][26] , with the duration of intervention ranging from 1 to 21 weeks (Table 2). These studies refreshed NaHS solutions every 12 15,17,18,24,25 or 24 [19][20][21][22][23] h. ...
... All studies except one 21 used 30 μM of NaHS solutions in drinking water. To explain the dose used (i.e., 30 μM), some authors stated that NaHS in the aqueous phase produces exactly equal concentrations of H 2 S gas, and since the physiological range of H 2 S is between 10 and 100 μM, this dose is in the physiological range 15,16 . It has also been explained that 30 μM NaHS keeps plasma H 2 S levels in the physiological range, i.e., 5-300 μM 19,20 . ...
... In our study, NaHS administration did not change body weight; this finding is in line with other studies in male mice 22,23 and male rats 18 ; however, two studies reported that NaSH restored reduced body weight in nephrectomized rats 24,26 and other studies did not report an effect of NaSH administration on body weight [15][16][17][19][20][21]25 . In addition, in our study, NaSH administration did not affect serum urea and Cr, a finding that aligns with another report 25 . ...
Article
Full-text available
Hydrogen sulfide (H2S) has many physiological and pathological roles in the human body. Sodium hydrosulfide (NaHS) is widely used as a pharmacological tool for assessing H2S effects in biological experiments. Although H2S loss from NaHS solution is a matter of minutes, some animal studies use NaHS in solution as an H2S-donating compound in drinking water. This study addresses whether 30 μM NaHS in drinking water prepared in rat/mouse water bottles remains stable for at least 12–24 h, as presumed by some authors. In conclusion, NaHS solution prepared in drinking water can not be used for H2S donation as the solution is unstable. This route of administration exposes animals to variable and lower-than-expected amounts of NaHS.
... In a relatively recent study, we reported that ciglitazone, a PPAR agonist, was found to ameliorate DN by reducing glomerular tissue homocysteine (Hcy), which is also a precursor of H 2 S [137]. We also reported that H 2 S could prevent hyperhomocysteinemia (HHcy)-induced renal failure by regulating MMP-2, -9, and collagen in mice [138,139]. Our recent study revealed that H 2 S supplementation by GYY4137 reinstated decreased PPARγ levels and improved adverse ECM remodeling in type 1 DN [140]. ...
... Moreover, HHcy has been shown to decrease H 2 S [166] and increase MMPs, which induce the degradation of elastin [167][168][169]. In the hypertensive and diabetic mouse models, HHcy-induced activation of MMPs was shown to be normalized by oral or intraperitoneal H 2 S supplementation, leading to the prevention of renal damage [137,138,140,170]. Therefore, H 2 S treatment could be a promising therapeutic approach to prevent renovascular damage by attenuating the MMP-mediated degradation of elastin. ...
... Our own studies revealed that supplementation of H 2 S prevents HHcy-associated renal damage by regulating MMP-2 and MMP-9 in mice [138,139]. An in vitro study also demonstrated that H 2 S supplementation marginally attenuated but could not completely normalize MMP-9 levels in hyperglycemic conditions [86]. ...
Article
Full-text available
Diabetic nephropathy (DN) remains the leading cause of vascular morbidity and mortality in diabetes patients. Despite the progress in understanding the diabetic disease process and advanced management of nephropathy, a number of patients still progress to end-stage renal disease (ESRD). The underlying mechanism still needs to be clarified. Gaseous signaling molecules, so-called gasotransmitters, such as nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S), have been shown to play an essential role in the development, progression, and ramification of DN depending on their availability and physiological actions. Although the studies on gasotransmitter regulations of DN are still emerging, the evidence revealed an aberrant level of gasotransmitters in patients with diabetes. In studies, different gasotransmitter donors have been implicated in ameliorating diabetic renal dysfunction. In this perspective, we summarized an overview of the recent advances in the physiological relevance of the gaseous molecules and their multifaceted interaction with other potential factors, such as extracellular matrix (ECM), in the severity modulation of DN. Moreover, the perspective of the present review highlights the possible therapeutic interventions of gasotransmitters in ameliorating this dreaded disease.
... 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.
... Exogenous H 2 S has been shown to be cardioprotective in various experimental models of cardiac injury [15,16], offering a potential therapeutic option where a lack of its endogenous availability has resulted in myocardial ischemia and reperfusion injuries [17][18][19]. Sodium thiosulphate (Na 2 S 2 O 3 , STS), a H 2 S donating sulphur salt [20][21][22], is an industrial compound with a long history of medical use specifically in the treatment of cyanide poisoning [23][24][25]. STS has been already explored in the treatment of acute coronary syndrome observing a therapeutic benefit in the preservation of cardiac function after myocardial ischemia [26]. ...
Article
Full-text available
Hypoxia, or insufficient oxygen availability is a common feature in the development of a myriad of cardiovascular-related conditions including ischemic disease. Hydrogen sulphide (H2S) donors, such as sodium thiosulphate (STS), are known for their cardioprotective properties. However, H2S due to its gaseous nature, is released and cleared rapidly, limiting its potential translation to clinical settings. For the first time, we developed and characterised liposome formulations encapsulating STS and explored their potential for modulating STS uptake, H2S release and the ability to retain pro-angiogenic and biological signals in a hypoxia-like environment mirroring oxygen insufficiency in vitro. Liposomes were prepared by varying lipid ratios and characterised for size, polydispersity and charge. STS liposomal encapsulation was confirmed by HPLC-UV detection and STS uptake and H2S release was assessed in vitro. To mimic hypoxia, cobalt chloride (CoCl2) was administered in conjunction with formulated and non-formulated STS, to explore pro-angiogenic and metabolic signals. Optimised liposomal formulation observed a liposome diameter of 146.42 ± 7.34 nm, a polydispersity of 0.22 ± 0.19, and charge of 3.02 ± 1.44 mV, resulting in 25% STS encapsulation. Maximum STS uptake (76.96 ± 3.08%) from liposome encapsulated STS was determined at 24 h. Co-exposure with CoCl2 and liposome encapsulated STS resulted in increased vascular endothelial growth factor mRNA as well as protein expression, enhanced wound closure and increased capillary-like formation. Finally, liposomal STS reversed metabolic switch induced by hypoxia by enhancing mitochondrial bioenergetics. These novel findings provide evidence of a feasible controlled-delivery system for STS, thus H2S, using liposome-based nanoparticles. Likewise, data suggests that in scenarios of hypoxia, liposomal STS is a good therapeutic candidate to sustain pro-angiogenic signals and retain metabolic functions that might be impaired by limited oxygen and nutrient availability.
... As the development of AF seems to be a result of the combination of a plethora of risk factors and comorbidities, oxidative stress has been associated with incident AF, while oxidative stress-induced atrial remodeling is considered the most common underlying mechanism; according to several reports, hyperhomocysteinemia has been linked with endothelial dysfunction, thus it represents an important factor for cardiovascular morbidity and mortality [93,94]. We were able to locate a total of 26 reports assessing oxidative stress as a potential mechanism of Hcy-induced cardiotoxicity [48][49][50]95]. Of these, 25 trials reported a positive correlation between Hcy and cardiomyocyte oxidative stress, while only one report failed to demonstrate oxidative stress as a cause of electron transport chain dysfunction, possibly due to an increased expression of the other protective mitochondrial proteins [41]. ...
Article
Full-text available
High levels of homocysteine (Hcy) have been linked with adverse cardiovascular outcomes, such as arrhythmias and stroke. In the context of paroxysmal atrial fibrillation (PAF), hyperhomocysteinemia has been demonstrated to be an independent predictor of future events. The aim of this report was to address the potential value of Hcy levels in predicting future paroxysms of atrial fibrillation (AF), as well as to identify the potential mechanisms of action. We searched PubMed and the Cochrane Database on 16 January 2022. Keywords used were homocysteine or hyperhomocysteinemia paired with a total of 67 different keywords or phrases that have been implicated with the pathogenesis of AF. We included primary reports of clinical and non-clinical data in the English language, as well as systematic reviews with or without meta-analyses. We placed no time constraints on our search strategy, which yielded 3748 results. Following title review, 3293 reports were excluded and 455 reports were used for title and abstract review, after which 109 reports were finally used for full-text review. Our review indicates that Hcy levels seem to hold a predictive value in PAF. Herein, potential mechanisms of action are presented and special considerations are made for clinically relevant diagnostic procedures that could complement plasma levels in the prediction of future PAF events. Finally, gaps of evidence are identified and considerations for future clinical trial design are presented.
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
Full-text available
Citation: Youness, R.A.; Habashy, D.A.; Khater, N.; Elsayed, K.; Dawoud, A.; Hakim, S.; Nafea, H.; Bourquin, C.; Abdel-Kader, R.M.; Gad, M.Z. Role of Hydrogen Sulfide in Oncological and Non-Oncological Disorders and Its Regulation by Non-Coding RNAs: A Comprehensive Review. Non-Coding RNA 2024, 10, 7. https://doi. Abstract: Recently, myriad studies have defined the versatile abilities of gasotransmitters and their synthesizing enzymes to play a "Maestro" role in orchestrating several oncological and non-oncological circuits and, thus, nominated them as possible therapeutic targets. Although a significant amount of work has been conducted on the role of nitric oxide (NO) and carbon monoxide (CO) and their interrelationship in the field of oncology, research about hydrogen sulfide (H 2 S) remains in its infancy. Recently, non-coding RNAs (ncRNAs) have been reported to play a dominating role in the regulation of the endogenous machinery system of H 2 S in several pathological contexts. A growing list of microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are leading the way as upstream regulators for H 2 S biosynthesis in different mammalian cells during the development and progression of human diseases; therefore, their targeting can be of great therapeutic benefit. In the current review, the authors shed the light onto the biosynthetic pathways of H 2 S and their regulation by miRNAs and lncRNAs in various oncological and non-oncological disorders.
Chapter
Diabetic kidney disease (DKD) is a chronic renal pathology, which is currently the leading cause of end-stage renal disease. It accounts for 40% of morbidity and mortality among the diabetic population despite optimal management. Its clinical hallmark includes persistent hyperglycemia, hypercreatininemia, uremia, sustained albuminuria, renal hemodynamic changes, and elevated blood pressure. Histologically, DKD presents with excessive accumulation and deposition of extracellular matrix, leading to expansion of mesangial matrix, thickening of glomerular basement membrane, and tubulointerstitial fibrosis. At the molecular level, accumulating evidence suggests that hyperglycemia or high glucose mediates renal injury in DKD via multiple molecular mechanisms such as induction of oxidative stress, upregulation of renal transforming growth factor beta-1 expression, production of pro-inflammatory cytokines, activation of fibroblasts and renin-angiotensin-aldosterone system, and depletion of adenosine triphosphate. Moreover, existing therapies only retard the disease progression but do not prevent or reverse it. Therefore, novel modes of pharmacotherapeutic intervention are in demand to target additional disease mechanisms. A substantial body of experimental evidence demonstrates that hydrogen sulfide (H2S), a gas with a historic notorious label, has recently been established to possess important therapeutic properties that prevent and/or reverse DKD development and progression of DKD by targeting several important molecular pathways, and therefore could be considered a novel pharmacological agent for DKD treatment. The aim of this chapter is to discuss recent experimental findings on the molecular mechanisms underlying the pharmacotherapeutic effects of H2S against DKD development and progression, and its translation from bench to bedside, which could lay the foundation for its future clinical use. A section of the chapter also discusses focal segmental glomerulosclerosis as a mediator of DKD progression to end-stage renal disease, and H2S as a potential novel therapy.
Chapter
Chronic kidney disease (CKD) is a common global health challenge characterized by irreversible pathological processes that reduce kidney function and culminate in the development of end-stage renal disease. It is associated with increased morbidity and mortality in addition to increased caregiver burden and higher financial cost. A central player in CKD pathogenesis and progression is renal hypoxia. Renal hypoxia stimulates induction of oxidative and endoplasmic reticulum stress, inflammation, and tubulointerstitial fibrosis, which in turn promote cellular susceptibility and further aggravate hypoxia, thus forming a pathological vicious cycle in CKD progression. Although the importance of CKD is widely appreciated, including improvements in the quality of existing therapies such as dialysis and transplantation, new therapeutic options are limited, as there is still increased morbidity, mortality, and poor quality of life among CKD patients. Growing evidence indicates that hydrogen sulfide (H2S), a small gaseous signaling molecule with an obnoxious smell, accumulates in the renal medulla under hypoxic conditions and functions as an oxygen sensor that restores oxygen balance and increases medullary flow. Moreover, plasma H2S level has been recently reported to be markedly reduced in CKD patients and animal models. Also, H2S has been established to possess potent antioxidant, anti-inflammatory, and anti-fibrotic properties in several experimental models of kidney diseases, suggesting that its supplementation could protect against CKD and retard its progression. The purpose of this chapter is to discuss current clinical and experimental developments regarding CKD, its pathophysiology, and potential cellular and molecular mechanisms of protection by H2S in experimental models of CKD. A section of the chapter also discusses hyperhomocysteinemia and autosomal dominant polycystic kidney disease, which are forms of CKD, and H2S as an additional/alternative agent for pharmacological treatment or management of these conditions.
Article
Significance: As a new important gas signaling molecule like NO and CO, hydrogen sulfide (H2S), which can be produced by endogenous H2S-producing enzymes through L-cysteine metabolism in mammalian cells, has attracted wide attention for long. H2S has been proved to play an important regulatory role in numerous physiological and pathophysiological processes. However, the deep mechanisms of those different functions of H2S still remain uncertain. A better understanding of the mechanisms can help us develop novel therapeutic strategies. Recent advances: H2S can play a regulating role through various mechanisms, such as regulating epigenetic modification, protein expression levels, protein activity, protein localization, redox microenvironment, and interaction with other gas signaling molecules like nitric oxide (NO) and carbon dioxide (CO). In addition to discussing the molecular mechanisms of H2S from the above perspectives, this paper will also review the regulation of H2S on common signaling pathways in the cells, including the PI3K/Akt, MAPK, JAK/STAT signaling pathway, etc. Critical issues: Although there are many studies on the mechanism of H2S, little is known about its direct target molecules. This paper will also review the existing reports about them. Furthermore, the interaction between direct target molecules of H2S and the downstream signaling pathways involved also needs to be clarified. Future directions: An in-depth discussion of the mechanism of H2S and the direct target molecules will help us achieving a deeper understanding of the physiological and pathophysiological processes regulated by H2S, and lay a foundation for developing new clinical therapeutic drugs in the future.
Article
Full-text available
We previously reported that increase in plasma homocysteine (Hcys) levels by a 6-week methionine treatment produced remarkable glomerular injury. However, the mechanism by which hyperhomocysteinemia (hHcys) produces glomerular injury remains unknown. The present study was to observe when glomerular injury happens during hHcys and to explore the possible role of podocyte injury in the progression of glomerulosclerosis associated with hHcys. Uninephrectomized Sprague-Dawley rats treated with methionine were used to examine the time course of glomerular injury induced by hHcys. Creatinine clearance was not different until rats were treated with methionine for 6 weeks, although plasma Hcys levels significantly increased at the 1st week of methionine treatment. However, urinary albumin excretion increased at the 2nd week of methionine treatment. Morphological examinations showed that mesangial expansion occurred at the 2nd week and podocyte effacement was also observed as processed glomerular damage during hHcys. Immunofluorescence analyses demonstrated that podocin and nephrin expressions were reduced, while alpha-actinin-4 increased during hHcys. Increased plasma Hcys level is an important pathogenic factor resulting in glomerular injury even in the very early time of hHcys. These pathogenic effects of Hcys are associated with podocyte injury and changed expression and distribution of podocyte-associated proteins.
Article
Full-text available
Hydrogen sulfide (H(2)S), a novel endogenous gaseous bioactive substance, has recently been implicated in the regulation of cardiovascular and neuronal functions. However, its role in the control of renal function is unknown. In the present study, incubation of renal tissue homogenates with L-cysteine (L-Cys) (as a substrate) produced H(2)S in a concentration-dependent manner. This H(2)S production was completely abolished by inhibition of both cystathionine beta-synthetase (CBS) and cystathionine gamma-lyase (CGL), two major enzymes for the production of H(2)S, using amino-oxyacetic acid (AOAA), an inhibitor of CBS, and propargylglycine (PPG), an inhibitor of CGL. However, inhibition of CBS or CGL alone induced a small decrease in H(2)S production. In anesthetized Sprague-Dawley rats, intrarenal arterial infusion of an H(2)S donor (NaHS) increased renal blood flow, glomerular filtration rate (GFR), urinary sodium (U(Na) x V), and potassium (U(K) x V) excretion. Consistently, infusion of both AOAA and PPG to inhibit the endogenous H(2)S production decreased GFR, U(Na) x V, and U(K) x V, and either one of these inhibitors alone had no significant effect on renal functions. Infusion of L-Cys into renal artery to increase the endogenous H(2)S production also increased GFR, U(Na) x V, and U(K) x V, which was blocked by AOAA plus PPG. It was shown that H(2)S had both vascular and tubular effects and that the tubular effect of H(2)S might be through inhibition of Na(+)/K(+)/2Cl(-) cotransporter and Na(+)/K(+)/ATPase activity. These results suggest that H(2)S participates in the control of renal function and increases urinary sodium excretion via both vascular and tubular actions in the kidney.
Article
Full-text available
Hydrogen sulfide (H2S) is an endogenously gaseous mediator, regulating many pathophysiological functions in mammalian cells. H2S has been shown to inhibit myocardial ischaemia-reperfusion (I/R) injury. However, little is known about whether H2S could modulate intestinal I/R injury. This study aimed to investigate the effect of H2S on intestinal I/R injury and potential mechanism(s) underlying the action of H2S in regulating the development of intestinal I/R injury in rats. Following surgical induction of intestinal I/R injury for 1 h, groups of Sprague-Dawley rats were treated with, or without, tetramethylpyrazine (8 mg/kg), or sodium hydrosulfide (NaHS, an H2S donor at 7 or 14 micromol/kg) 30 min after occlusion. All rats were sacrificed immediately after the reperfusion. Their intestinal injury, together with that of sham-control rats, was histologically examined and their sera and intestinal malondialdehyde (MDA), superoxide dismutase (SOD), peroxidase (GSH-Px) activities were characterized by biochemical analysis. The results showed that NaHS significantly reduced intestinal I/R injury and the levels of sera and intestinal MDA activity, and dramatically increased the levels of serum and intestinal SOD and GSH-Px activity. The results suggest that H2S protects from intestinal I/R injury in rats, which is associated with increase in the activity of antioxidant enzymes.
Article
Full-text available
Studies of nitric oxide over the past two decades have highlighted the fundamental importance of gaseous signaling molecules in biology and medicine. The physiological role of other gases such as carbon monoxide and hydrogen sulfide (H2S) is now receiving increasing attention. Here we show that H2S is physiologically generated by cystathionine γ-lyase (CSE) and that genetic deletion of this enzyme in mice markedly reduces H2S levels in the serum, heart, aorta, and other tissues. Mutant mice lacking CSE display pronounced hypertension and diminished endothelium-dependent vasorelaxation. CSE is physiologically activated by calcium-calmodulin, which is a mechanism for H2S formation in response to vascular activation. These findings provide direct evidence that H2S is a physiologic vasodilator and regulator of blood pressure.
Article
Our previous studies demonstrated an increased reac-tive oxygen species (ROS) production, as well as transforming growth factor-β1 (TGF-β1) expression in the rat kidney with aging. In the present study, we examined the effect of aging on extracellular matrix (ECM) accumulation and the effects of treatment with angiotensin-converting enzyme inhibitors (captopril and lisinopril) and taurine, an antioxidant amino acid. Age-related increases in types I and IV collagen and fibronectin mRNA expression were found at 24 and 30 mo of age. In contrast, type III collagen only increased in 30-mo-old rats. Captopril-, lisinopril-, and taurine-treated animals showed a statistically significant decrease in ECM protein expression at both ages. Moreover, treatment with taurine reduced the TGF-β1 mRNA levels in 24- and 30-mo-old rats by 40%. Taurine also completely blocked increases in type I and type IV collagen expression in mesangial cells in response to TGF-β1. Our results demonstrate a protective role from both converting enzyme inhibitors and taurine in the age-related progressive renal sclerosis. In addition, taking into account that taurine is considered as an antioxidant amino acid, present data suggest a role for ROS in age-related progressive renal fibrosis, perhaps through interactions with the TGF-β1 pathway.
Article
A protein determination method which involves the binding of Coomassie Brilliant Blue G-250 to protein is described. The binding of the dye to protein causes a shift in the absorption maximum of the dye from 465 to 595 nm, and it is the increase in absorption at 595 nm which is monitored. This assay is very reproducible and rapid with the dye binding process virtually complete in approximately 2 min with good color stability for 1 hr. There is little or no interference from cations such as sodium or potassium nor from carbohydrates such as sucrose. A small amount of color is developed in the presence of strongly alkaline buffering agents, but the assay may be run accurately by the use of proper buffer controls. The only components found to give excessive interfering color in the assay are relatively large amounts of detergents such as sodium dodecyl sulfate, Triton X-100, and commercial glassware detergents. Interference by small amounts of detergent may be eliminated by the use of proper controls.
Article
Background: Homocysteine (Hcy) is an independent cardiovascular risk factor; however, in diabetes, the role of tissue Hcy leading to cardiac dysfunction is unclear. Aims: To determine whether tissue Hcy caused endothelial-myocyte uncoupling and ventricular dysfunction in diabetes. Methods: Diabetes was created in C57BL/6J male mice by injecting 65 mg/kg alloxan. To reverse diabetic complications, ciglitazone (CZ) was administered in the drinking water. Plasma glucose, Hcy, left ventricular (LV) tissue levels of Hcy and nitric oxide (NO) were measured. Glomerular filtration rate (GFR) was measured by inulin-FITC. Endothelial-myocyte coupling was measured in cardiac rings. In vivo diastolic relaxation and LV diameters were measured by a Millar catheter in LV and by M-mode echocardiography, respectively. Results: Plasma glucose, GFR and LV tissue Hcy were increased in diabetic mice and were normalized after CZ treatment; whereas, elevated plasma Hcy level remained unchanged with or without CZ treatment. NO levels in the LV were found inversely related to tissue Hcy levels. Attenuated endothelial-myocyte function in diabetic mice was ameliorated by CZ treatment. Cardiac relaxation, the ratio of LV wall thickness to LV diameter was decreased in diabetes, and normalized after CZ treatment. Conclusion: CZ normalized LV tissue levels of Hcy and ameliorated endothelial-myocyte coupling; therefore, specifically suggest the association of LV tissue Hcy levels with impair endothelial-myocyte function in diabetes.
Article
Hyperhomocysteinemia (HHcy) is an independent risk factor for cardiovascular disease, including ischemic heart disease, stroke, and peripheral vascular disease. Mutations in the enzymes responsible for homocysteine metabolism, particularly cystathionine beta-synthase (CBS) or 5,10-methylenetetrahydrofolate reductase (MTHFR), result in severe forms of HHcy. Additionally, nutritional deficiencies in B vitamin cofactors required for homocysteine metabolism, including folic acid, vitamin B6 (pyridoxal phosphate), and/or B12 (methylcobalamin), can induce HHcy. Studies using animal models of genetic- and diet-induced HHcy have recently demonstrated a causal relationship between HHcy, endothelial dysfunction, and accelerated atherosclerosis. Dietary enrichment in B vitamins attenuates these adverse effects of HHcy. Although oxidative stress and activation of proinflammatory factors have been proposed to explain the atherogenic effects of HHcy, recent in vitro and in vivo studies demonstrate that HHcy induces endoplasmic reticulum (ER) stress, leading to activation of the unfolded protein response (UPR). This review summarizes the current role of HHcy in endothelial dysfunction and explores the cellular mechanisms, including ER stress, that contribute to atherothrombosis.