ArticlePDF Available

Endogenous Hydrogen Sulfide Regulates Pulmonary Artery Collagen Remodeling in Rats with High Pulmonary Blood Flow

Authors:

Abstract and Figures

The mechanisms responsible for the structural remodeling of pulmonary vasculature induced by increased pulmonary blood flow are not fully understood. This study explores the effect of endogenous hydrogen sulfide (H2S), a novel gasotransmitter, on collagen remodeling of the pulmonary artery in rats with high pulmonary blood flow. Thirty-two Sprague-Dawley rats were randomly divided into sham, shunt, sham+PPG (D,L-propargylglycine, an inhibitor of cystathionine-gamma-lyase), and shunt+PPG groups. After 4 weeks of shunting, the relative medial thickness (RMT) of pulmonary arteries and H2S concentration in lung tissues were investigated. Collagen I and collagen III were evaluated by hydroxyproline assay, sirius-red staining, and immunohistochemistry. Pulmonary artery matrix metalloproteinase-13 (MMP-13), tissue inhibitor of metalloproteinase-1 (TIMP-1), and connective tissue growth factor (CTGF) were evaluated by immunohistochemistry. After 4 weeks of aortocaval shunting, resulting in an elevation of lung tissue H2S to 116.4%, rats exhibited collagen remodeling and increased CTGF expression in the pulmonary arteries. Compared with those of the shunt group, lung tissue H2S production was lowered by 23.4%, RMT of the pulmonary artery further increased by 39.5%, pulmonary artery collagen accumulation became obvious, and pulmonary artery CTGF expression elevated (P<0.01) in the shunted rats treated with PPG. However, pulmonary artery MMP-13 and TIMP-1 expressions decreased significantly in rats of shunt+PPG group (P<0.01). This study suggests that endogenous H2S exerts an important regulatory effect on pulmonary collagen remodeling induced by high pulmonary blood flow.
Content may be subject to copyright.
Endogenous Hydrogen Sulfide Regulates
Pulmonary Artery Collagen Remodeling in
Rats with High Pulmonary Blood Flow
XIAOHUI LI,* HONGFANG JIN,* GENG BIN,LIWANG,àCHAOSHU TANG,AND JUNBAO DU*
,1
*Department of Pediatrics, Peking University First Hospital and Key Laboratory of Molecular
Cardiovascular Sciences, Ministry of Education, Beijing 100034, People’s Republic of China;
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing
100034, People’s Republic of China; and àDepartment of Scientific Research, Peking University Health
Science Center, Beijing 100034, People’s Republic of China
The mechanisms responsible for the structural remodeling of
pulmonary vasculature induced by increased pulmonary blood
flow are not fully understood. This study explores the effect of
endogenous hydrogen sulfide (H
2
S), a novel gasotransmitter, on
collagen remodeling of the pulmonary artery in rats with high
pulmonary blood flow. Thirty-two Sprague-Dawley rats were
randomly divided into sham, shunt, sham1PPG (D,L-propargyl-
glycine, an inhibitor of cystathionine-c-lyase), and shunt1PPG
groups. After 4 weeks of shunting, the relative medial thickness
(RMT) of pulmonary arteries and H
2
S concentration in lung
tissues were investigated. Collagen I and collagen III were
evaluated by hydroxyproline assay, sirius-red staining, and
immunohistochemistry. Pulmonary artery matrix metalloprotein-
ase-13 (MMP-13), tissue inhibitor of metalloproteinase-1 (TIMP-
1), and connective tissue growth factor (CTGF) were evaluated
by immunohistochemistry. After 4 weeks of aortocaval shunt-
ing, resulting in an elevation of lung tissue H
2
S to 116.4%, rats
exhibited collagen remodeling and increased CTGF expression
in the pulmonary arteries. Compared with those of the shunt
group, lung tissue H
2
S production was lowered by 23.4%, RMT
of the pulmonary artery further increased by 39.5%, pulmonary
artery collagen accumulation became obvious, and pulmonary
artery CTGF expression elevated (P<0.01) in the shunted rats
treated with PPG. However, pulmonary artery MMP-13 and TIMP-
1 expressions decreased significantly in rats of shunt1PPG
group (P<0.01). This study suggests that endogenous H
2
S
exerts an important regulatory effect on pulmonary collagen
remodeling induced by high pulmonary blood flow. Exp Biol Med
234:504–512, 2009
Key words: hydrogen sulfide; collagen; pulmonary artery; inhibitor of
metalloproteinase-1 (TIMP-1); matrix metalloproteinase-13 (MMP-
13)
Introduction
Congenital heart disease (CHD) is the most common
heart disease in childhood. It is often characterized by
increased pulmonary blood flow in those with a left-to-right
shunt. This increased blood flow results in vascular injury
and pulmonary hypertension during the course of the
disease. The mechanisms responsible for structural remod-
eling induced by increased pulmonary blood flow are not
fully understood. Collagen accumulation is one of the
important factors in the development of vascular remodeling
and pulmonary hypertension. The regulation of pulmonary
hypertension has also been indicated to be associated with
the role of a novel gasotransmitter, hydrogen sulfide (H
2
S),
which plays a part similar to nitric oxide (NO) and carbon
monoxide (CO) in the relaxation of blood vessels (1–5).
Based on the above evidence, we performed the present
study to investigate the possible changes and roles of
endogenous H
2
S in collagen remodeling in rats with high
pulmonary blood flow.
Materials and Methods
Animal Model of High Pulmonary Blood Flow.
Experiments were conducted in accordance with the Guide
to The Care and Use of Experimental Animals issued by the
Ministry of Health of the People’s Republic of China. Male
Sprague-Dawley rats were provided by the Animal Research
Center of Peking University First Hospital. The rats were
This work was supported by Natural Science Foundation of Beijing, P. R. China
(7072082), National Natural Science Foundation of China (30425010, 30821001,
30801251, 30630031, 30872787), Grant of Ministry of Education, China
(20070001702), and State Major Basic Research Project of China (2006CB503807).
1
To whom correspondence should be addressed at Department of Pediatrics, Peking
University First Hospital, Xi-An Men Street No. 1, West District, Beijing, 100034,
People’s Republic of China. E-mail: junbaodu@ht.rol.cn.net
Received July 28, 2008.
Accepted January 30, 2009.
504
DOI: 10.3181/0807-RM-230
1535-3702/09/2345-0504$15.00
Copyright Ó2009 by the Society for Experimental Biology and Medicine
kept in a temperature-controlled room with a 12 hour light-
dark cycle. Tap water and rat chow were provided ad
libitum. The animal model of the left-to-right shunt was
created according to the method described by Garcia and
Diebold (6) with minor modifications (7). Briefly, 32 male
Sprague-Dawley rats, weighing 120–140 g, were randomly
divided into shunt (n¼8), shuntþpropargylglycine (PPG, an
inhibitor of cystathionine-c-lyase) (n¼8), sham (n¼8), and
shamþPPG (n¼8) groups. We anesthetized rats in the shunt
and shuntþPPG groups with 0.25% pentobarbital sodium
(40 mg/kg, intraperitoneal injection). We exposed the
abdominal aorta and inferior vena cava, and then placed a
bulldog vascular clamp across the aorta caudal to the left
renal artery. We punctured the aorta at the union of the
segment two-thirds caudal to the renal artery and one-third
cephalic to the aortic bifurcation with an 18-gauge
disposable needle. Then, the needle was slowly withdrawn
and a 9–0 silk thread was used to stitch the puncture of the
abdominal wall. In the sham and shamþPPG groups, rats
underwent the same experimental protocol as mentioned
above except for the shunting procedure. We injected rats in
the shuntþPPG and shamþPPG groups intraperitoneally
with PPG at 37.5 mg/kg/d for 4 weeks (8). We injected rats
in the shunt and sham groups with the same volume of
physiological saline.
Measurement of Oxygen Saturation. At 4 weeks
of the experiment, we weighed and anesthetized the animals
with pentobarbital sodium (40 mg/kg, intraperitoneal
injection). We analyzed blood samples (0.5 ml) obtained
from the pulmonary artery, external carotid artery, and
jugular vein using a GASTAT-3 Blood Gas Analysis
Apparatus. The ratio Qp/Qs was calculated as an indicator
of pulmonary and systemic blood flow. Qp/Qs was
calculated by the formula: Qp/Qs ¼[oxygen saturation of
aorta (%) oxygen saturation of jugular vena cava (%)] /
[oxygen saturation of pulmonary vein (%) oxygen
saturation of pulmonary artery (%)]. When the oxygen
saturation of the aorta was .95%, we regarded the oxygen
saturation of the pulmonary vein as 100%. When the oxygen
saturation of the aorta was ,95%, we regarded the oxygen
saturation of the pulmonary vein as 95%.
The right side of the lung tissue was removed and
quickly frozen in liquid nitrogen, then stored at 808C for
homogenate. The left lower part of lung tissue was removed
and post-fixed in 10% (wt/vol) paraformaldehyde.
Measurement of H
2
S in Lung Tissue. We
homogenized lung tissue in a 10-fold volume (w/v) of an
ice-cold potassium phosphate buffer (pH ¼6.8). The
reaction was performed in a 25-ml Erlenmeyer Pyrex flask.
We used cryovial test tubes (2 ml) as the center wells, each
containing 1 M NaOH of 0.5 ml. The reaction mixture
contained lung tissue homogenate and 1 M HCL in a ratio of
1:5. We flushed the flasks containing reaction mixture and
central wells with N
2
for 30 seconds before sealing with a
double layer of parafilm. The reaction was initiated by
transferring the flasks from ice to a shaking water bath at
378C. After incubation at 378C for 4 h, we transferred the
contents of the central wells to 10-ml beakers, each
containing 0.5 ml of antioxidant solution. Subsequently,
the solution was measured with a sulfide-sensitive electrode
(PXS-270, Shanghai, China) to calculate the lung tissue H
2
S
against a standard curve.
Morphological Analysis of Small Pulmonary
Arteries. We mounted fixed lung tissue in paraffin, and
sectioned it at 4 lm thickness. We then stained the elastic
fiber according to the modified Weigert’s method and
counterstained with Van Gieson solution. We performed
morphological analysis using a Video-Linked Microscope
Digitizing Board System (Leica Q550CW, Germany). Only
vessels showing clearly defined external and internal elastic
lamina were used in the analysis. The relative medial
thickness (RMT) was calculated according to Barth’s
methods (9).
Hydroxyproline Assay of Lung Tissue. Lung
tissue homogenate was dehydrated in 0.2 ml of 6 nmol/L
HCl (14 hours). The pH of the samples was adjusted with 6
nmol/L NaOH to 6.0, and then centrifuged at 2000 rpm for
10 min. We performed the hydroxyproline assay according
to instructions provided with a commercially available kit
(Nanjing Jiancheng Bioengineering Institute, China).
Sirius-Red Staining Analysis of Collagen I and
Collagen III. Lung sections were processed by sirius-red
staining for 1 min, following dewaxing and dehydration.
Polarized light microscopy was used to distinguish type I
and type III collagen fibers (Leica, Germany).
Immunohistochemical Analysis. Lung sections
were pretreated by 3% H
2
O
2
for 15 min, followed by the
appropriate antigen repairing treatment: digestion with
gastric enzyme for 30 min at 378C for collagen I and
collagen III; heating with microwave for 15 min at 998C for
tissue inhibitor of metalloproteinase 1 (TIMP-1), matrix
metalloproteinase-13 (MMP-13), and connective tissue
growth factor (CTGF). The slides were blocked with normal
bovine serum albumin (BSA) for 30 min at room temper-
ature. Collagen I or III (Boster Bioengineering Ltd., China),
MMP-13, TIMP-1 (Neomarkers, USA) and CTGF (Boster
Bioengineering Ltd., China) antibodies were then added at
378C for 2 h respectively in different experiment. We used
biotinylated anti-rabbit IgG and horseradish peroxidase
streptavidin (Santa Cruz, Canada) sequentially at 378C for
30 min. To develop a color product, diaminobenzidine
(DAB) was added for 1–10 min and Mayer’s hematoxylin
for 1 min. We observed labeling of the smooth muscle cells
in intrapulmonary arteries using light microscopy. The mean
percentage of intensity of antibody labeling (0%, ;50%,
and ;100%) was determined (9).
Statistical Analysis. All data are expressed as
means 6SD. We analyzed data using one-way analysis
of variance (ANOVA) followed by the Student-Newman-
Keuls tests for multiple comparisons. A value of P,0.05
was considered statistically significant.
H
2
S AND PULMONARY ARTERY COLLAGEN REMODELING 505
Results
Changes in Oxygen Saturation and H
2
S Con-
tent. In the present rat model of abdominal aorta-inferior
vena cava shunt, Qp/Qs in the shunt group and shuntþPPG
group increased significantly as compared with that of sham
and shamþPPG group (P ,0.01). The shunt and
shuntþPPG groups did not differ significantly (Fig. 1).
Compared with the sham group, the lung tissue H
2
S
content in the shunt group increased significantly (P ,
0.01). In the shuntþPPG group, lung tissue H
2
S content
decreased significantly (P ,0.01). The sham and
shamþPPG groups did not differ significantly in lung tissue
H
2
S content (Fig. 2).
RMT in Pulmonary Arteries. In contrast to the
sham group, RMT of intra-acinar pulmonary arteries in the
shunt group increased significantly (16.87 61.86% vs
12.67 61.26%, P,0.01). Compared with shunt group,
RMT of intra-acinar pulmonary arteries in the shuntþPPG
group increased significantly (23.54 63.07% vs 16.87 6
1.86%, P,0.01). RMT in sham and shamþPPG groups did
not differ significantly (12.67 61.26% vs 13.00 63.20%,
P.0.05) (Fig. 3).
Hydroxyproline Concentration and Collagen
Content of Lung Tissue. Hydroxyproline concentration
in lung tissue in the shunt group was significantly greater
than that of the sham group (P,0.01). In the shuntþPPG
group, hydroxyproline concentration was significantly
greater than that of the shunt group (P,0.01). There were
no significant differences between the sham and shamþPPG
groups (P.0.05) (Fig. 4).
Sirius-red staining showed that collagen I and collagen
III were more intensely stained in the pulmonary arteries of
the shunt group than in the sham group. The shuntþPPG
group showed more predominant collagen I and collagen III
than the shunt group (Fig. 5).
Pulmonary Artery Collagen I and Collagen
III. Collagen I and collagen III protein expressions of
intra-acinar pulmonary arteries in the shunt group were
significantly greater than the sham group (P,0.01). In the
shuntþPPG group, collagen I and collagen III protein
expressions of intra-acinar pulmonary arteries were signifi-
cantly greater than in the shunt group (P,0.01). There was
no significant difference in collagen I and collagen III
protein expressions of intra-acinar pulmonary arteries
between the sham and shamþPPG groups (P.0.05) (Figs.
6, 7). Using sirius-red staining, collagen I and collagen III in
rat pulmonary artery accumulation were also more obvious
Figure 1. Ratio of Qp/Qs in rats of different groups. #P,0.01 vs.
sham; *P,0.01 vs. shamþPPG.
Figure 2. Changes of lung tissue H
2
S content in rats of different
groups. #P,0.01 vs. sham; *P,0.01 vs. shunt.
Figure 3. RMT of pulmonary arteries in rats of different groups. #P,
0.01 vs. sham; *P,0.01 vs. shunt.
Figure 4. Changes of hydroxyproline concentration in lung tissue in
rats of different groups. #P,0.01 vs. sham; *P,0.01 vs. shunt.
506 LI ET AL
Figure 5. Sirius-red staining analysis of collagen I and collagen III in rat pulmonary artery under microscope (3200). A, sham, B, shamþPPG, C,
shunt, D, shuntþPPG. Sirius-red staining analysis of collagen I and collagen III in rat pulmonary artery under polarizing microscope (3200). E,
sham, F, shamþPPG, G, shunt, H, shuntþPPG.
H
2
S AND PULMONARY ARTERY COLLAGEN REMODELING 507
in the shunt group than in the sham group, and in the
shuntþPPG group versus the shunt group (Fig. 5).
MMP-13 and TIMP-1 in Pulmonary Arter-
ies.MMP-13 and TIMP-1 protein expressions of intra-
acinar pulmonary arteries as well as the ratio of MMP-13/
TIMP-1 in the shunt group were significantly greater than
the sham group (P,0.01). In the shuntþPPG group, MMP-
13 and TIMP-1 and the ratio of MMP-13/TIMP-1 were
significantly decreased relative to the shunt group (P,
0.01). The sham and shamþPPG groups did not differ
significantly (Fig. 6).
CTGF Protein Expression of Intra-Acinar Pul-
monary Arteries. In the shunt group, CTGF protein
expression of intra-acinar pulmonary arteries was signifi-
cantly greater than the sham group (P,0.01). In the
shuntþPPG group, CTGF was significantly greater than that
of the shunt group (P,0.01). There was no significant
difference in CTGF between the sham and shamþPPG
groups (P.0.05) (Figs. 6, 8).
Discussion
Pulmonary vascular structural remodeling and hyper-
tension due to high blood flow are common complications
of congenital heart disease observed with a left-to-right
shunt. However, the mechanisms responsible for the
remodeling induced by increased pulmonary blood flow
have been unclear. In the present study, we created a rat
model of high pulmonary blood flow by an abdominal aorta-
inferior vena cava shunting operation. We found that in the
shunt group, the ratio of pulmonary blood flow and systemic
blood flow (Qp/Qs) was significantly greater than in the
sham group, which suggests that pulmonary blood flow
increased as a result of the shunt. RMT, an indicator of
pulmonary vascular structural remodeling, was significantly
greater in the shunt group, which suggested that pulmonary
vascular structure remodeled due to high pulmonary blood
flow.
H
2
S is produced endogenously in mammalian tissues
from cystathionine metabolism mainly by 3 enzymes:
cystathionine-b-synthetase (CBS), cystathionine-c-lyase
(CSE), and 3-mercaptosulfurtransferase (MST) (10, 11).
The expression of these enzymes is tissue-specific, and CSE
mainly catalyzes H
2
S production in the cardiovascular
system. Our previous studies revealed that H
2
S plays an
important role in the pathophysiological process of some
cardio-pulmonary diseases such as spontaneous hyper-
tension (8), nitric oxide (NO) synthase inhibitor-induced
hypertension (12), hypoxia-induced pulmonary hyperten-
sion (13), septic and endotoxic shock (14), and isoproter-
enol-induced myocardial injury (15). These findings suggest
that endogenous H
2
S could be a novel gasotransmitter in the
cardiovascular system (16). In this study, we observed
markedly increased H
2
S production in a rat model of high
pulmonary blood flow.
To explore whether the CSE-H
2
S pathway contributes
to the pathogenesis of pulmonary vascular structural
remodeling induced by high pulmonary blood flow, we
applied D,L-propargylglycine (PPG), an irreversible inhib-
itor of CSE, in this experiment. CSE is a key enzyme in the
pathway of cystathionine metabolism to produce endoge-
nous H
2
S in rats. PPG was first reported by Abeles and
Walsh to inactivate rat liver cystathionase in 1973 (17).
After that, further studies showed that CSE enzyme is an
alpha 2 beta 2 tetramer where the subunits are distinguish-
able by charge but not by size and that each subunit of a
CSE tetramer became modified by PPG in an inactive CSE
(18, 19).
Although the accurate half life of PPG injected
intraperitoneally has not been reported, we found that the
content of PPG in serum reached its maximum at about 2 h
after intraperitoneal injection. Approximately 21.2% of
administered PPG was excreted into the urine within6hand
could not be detected in the urine by about 12 h after
administration (20). Moreover, we found that the activity of
CSE decreased to about 4% in rats 24 h after PPG injection
daily (21). Based on the above data and our previous study
(8), PPG was injected intraperitoneally once a day in this
experiment.
In the present study, we found that there was no
statistical difference in lung tissue H
2
S production between
sham and shamþPPG group as shown on Figure 2. As we
know, the basal endogenous lung tissue H
2
S production was
in a low level (17.35 61.76 lM) in sham group, because of
a low level of the activity of CSE at this state (22).
However, in shunt group, the endogenous lung tissue H
2
S
production was increased (37.56 62.13 lM). It is likely
that PPG plays a more obviously inhibitory role in
endogenous lung tissue H
2
S production in the shunt group
where the basal H
2
S level is relatively high compared with
the sham group, which has a low basal H
2
S level. Our study
found that in shunted animals administrated PPG for 4
weeks, the H
2
S content of lung tissue was significantly
lower than in those shunted rats without PPG treatment.
Figure 6. Changes in integral scores of collagen I, collagen III, MMP-
13, TIMP-1, and CTGF protein expressions in the pulmonary artery of
different groups. #P,0.01 vs. sham; *P,0.01 vs. shunt.
508 LI ET AL
Figure 7. Immunohistochemical analysis of collagen I protein expression in rat pulmonary artery of different groups (DAB, 3200). A, sham, B,
shamþPPG, C, shunt, D, shuntþPPG. Immunohistochemical analysis of collagen III protein expression in rat pulmonary artery of different
groups (DAB, 3200). E, sham, F, shamþPPG, G, shunt, H, shuntþPPG.
H
2
S AND PULMONARY ARTERY COLLAGEN REMODELING 509
However, the pulmonary vascular structure remodeled to an
even greater degree than in shunted animals without PPG,
which suggests that endogenous H
2
S possibly participates in
pulmonary vascular structural remodeling induced by high
pulmonary blood flow.
The present study aimed to explore the possible effect
of endogenous H
2
Soncollagenremodelingofthe
pulmonary artery in rats with high pulmonary blood flow.
Previous studies have shown that extracellular matrix
contributes a great deal to pulmonary vascular structural
remodeling. Collagen I and collagen III are the most
abundant components of the extracellular matrix of the
vascular wall. Changes in the absolute or relative contents of
collagen I and collagen III would likely result in structural
remodeling. Our previous studies showed that H
2
S reduced
the collagen remodeling of pulmonary artery under hypoxia
(23). However, it is unclear whether H
2
S exerts any
regulatory effect on the collagen accumulation in pulmonary
artery induced by high flow. Since collagen protein contains
hydroxyproline up to 10–14% by weight, hydroxyproline
concentration was measured as an index of collagen content
in this experiment (24). It was found that hydroxyproline
concentration of rat lung tissue and the expression of
collagen I and collagen III increased in the shunted animals.
Interestingly, these factors increased even more in the PPG
treated-shunt group where the production of endogenous
H
2
S was inhibited. This suggests that endogenous H
2
S
probably plays an important role in the regulation of
collagen remodeling induced by high pulmonary blood
flow.
Matrix metalloproteinases (MMPs; also called collage-
nases or matrixins) are key enzymes in extracellular matrix
degradation. MMPs involve a family of zinc-dependent
endopeptidases. Collagenase MMP-13 degrades mainly
fibrillar collagens, which include collagen I and collagen
III in the vascular wall. MMP activity is regulated at
different levels, including transcriptional control, extracel-
lular activation of proenzymes, and active enzyme, tissue
Figure 8. Immunohistochemical analysis of CTGF protein expression in rat pulmonary artery of different groups (DAB, 3200). A, sham. B,
shamþPPG, C, shunt, D, shuntþPPG.
510 LI ET AL
inhibitor of metalloproteinases (TIMPs). TIMPs bind to
active and alternative sites of the activated MMP to inhibit
the activity of MMP. The major member is TIMP-1, a 30-
kDa glycoprotein that is synthesized by most cells (25). It
was reported that an imbalance between gene expression of
interstitial collagenases and gene expression of TIMP-1
contributed to extracellular matrix accumulation (26).
Connective tissue growth factor (CTGF), a potent stimulator
of collagen synthesis (27), consists of 349 amino acids with
four distinct domains and induced by mechanical shear
stress in vitro (28, 29). As mentioned above, the balance of
MMP and TIMP-1 is involved in the regulation of collagen
degradation and CTGF contributed to collagen synthesis.
So, we included MMP, TIMP-1, CTGF as well as collagen
in the present study to investigate the role of H
2
S in the
regulation of pulmonary collagen accumulation.
In the present study, it was found that the expression of
MMP-13 and TIMP-1 is greater in the pulmonary artery
after 4 weeks of left-to-right shunt in rat. When PPG was
used to inhibit the production of endogenous H
2
S in shunted
animals, the expression of MMP-13 and TIMP-1 was
significantly down-regulated, which suggests that endoge-
nous H
2
S probably reduced collagen accumulation through
increasing its degradation in the shunted group. Our present
study indicated that CTGF expression up-regulated in
animals with 4 weeks of shunting. When PPG was used
to inhibit the production of endogenous H
2
S, CTGF up-
regulation was further increased. These results suggest that
endogenous H
2
S regulates the expression of CTGF, which
might be associated with the regulation of collagen
remodeling in pulmonary artery induced by high pulmonary
blood flow.
In conclusion, this study provides the first evidence that
endogenous H
2
S exerts regulatory effects on pulmonary
artery remodeling induced by high pulmonary blood flow.
Endogenous H
2
S might participate in regulating collagen
metabolism by affecting the degradation and synthesis of
collagen. However, the molecular mechanisms by which
H
2
S regulates pulmonary artery structural remodeling will
require further study.
1. Du J, Hui Y, Cheung Y, Bin G, Jiang H, Chen X, Tang C. The possible
role of hydrogen sulfide as a smooth muscle cell proliferation inhibitor
in rat cultured cells. Heart Vessels 19:75–80, 2004.
2. Pearson RJ, Wilson T, Wang R. Endogenous hydrogen sulfide and the
cardiovascular system-what’s the smell all about? Clin Invest Med 29:
146–150, 2006.
3. Wang R. The gasotransmitter role of hydrogen sulfide. Antioxid Redox
Signal 5:493–-501, 2003.
4. Xiaohui L, Junbao D, Lin S, Jian L, Xiuying T, Jianguang Q, Bing W,
Hongfang J, Chaoshu T. Down-regulation of endogenous hydrogen
sulfide pathway in pulmonary hypertension and pulmonary vascular
structural remodeling induced by high pulmonary blood flow in rats.
Circ J 69:1418–1424, 2005.
5. 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 J20:6008–
6016, 2001.
6. Garcia R, Diebold S. Simple, rapid, and effective method of producing
aortocaval shunts in the rat. Cardiovasc Res 24:430–432, 1990.
7. Ocampo C, Ingram P, Ilbawi M, Arcilla R, Gupta M. Revisiting the
surgical creation of volume load by aorto-caval shunt in rats. Mol Cell
Biochem 251:139–143, 2003.
8. Yan H, Du J, Tang C. The possible role of hydrogen sulfide on the
pathogenesis of spontaneous hypertension in rats. Biochem Biophys
Res Commun 313:22–27, 2004.
9. Barth PJ, Kimpel C, Roy S, Wagner U. An improved mathematical
approach for the assessment of the medial thickness of pulmonary
arteries. Pathol Res Pract 189:567–76, 1993.
10. Stipanuk MH, Beck PW. Characterization of the enzymic capacity for
cysteine desulphhydration in liver and kidney of the rat. Biochem J
206:267–277, 1982.
11. Erickson PF, Maxwell IH, Su LJ, Baumann M, Glode LM. Sequence of
cDNA for rat cystathionine gamma-lyase and comparison of deduced
amino acid sequence with related Escherichia coli enzymes. Biochem J
269:335–340, 1990.
12. Zhong G, Chen F, Cheng Y, Tang C, Du J. The role of hydrogen sulfide
generation in the pathogenesis of hypertension in rats induced by
inhibition of nitric oxide synthase. J Hypertens 21:1879–1885, 2003.
13. Chunyu Z, Junbao D, Dingfang B, Hui Y, Xiuying T, Chaoshu T. The
regulatory effect of hydrogen sulfide on hypoxic pulmonary hyper-
tension in rats. Biochem Biophys Res Commun 302:810–816, 2003.
14. Hui Y, Du J, Tang C, Bin G, Jiang H. Changes in arterial hydrogen
sulfide (H(2)S) content during septic shock and endotoxin shock in rats.
J Infect 47:155–160, 2003.
15. Geng B, Chang L, Pan C, Qi Y, Zhao J, Pang Y, Du J, Tang C.
Endogenous hydrogen sulfide regulation of myocardial injury induced
by isoproterenol. Biochem Biophys Res Commun 318:756–763, 2004.
16. Chen CQ, Xin H, Zhu YZ. Hydrogen sulfide: third gaseous transmitter,
but with great pharmacological potential. Acta Pharmacol Sin 28:1709–
1716, 2007.
17. Abeles RH, Walsh CT. Acetylenic enzyme inactivators. Inactivation of
gamma-cystathionase, in vitro and in vivo, by propargylglycine. J Am
Chem Soc 95: 6124–6125, 1973.
18. Marcotte P, Walsh C. Active site-directed inactivation of cystathionine
gamma-synthetase and glutamic pyruvic transaminase by propargyl-
glycine. Biochem Biophys Res Commun 62:677–682, 1975.
19. Johnston M, Jankowski D, Marcotte P, Tanaka H, Esaki N, Soda K,
Walsh C. Suicide inactivation of bacterial cystathionine gamma-
synthase and methionine gamma-lyase during processing of L-
propargylglycine. Biochemistry18:4690–4701, 1979.
20. Zhang J, Machida Y, Sugahara K, Kodama H. Determination of D,L-
propargylglycine and N-acetylpropargylglycine in urine and several
tissues of D,L-propargylglycine-treated rats using liquid chromatog-
raphy-mass spectrometry. J Chromatogr B Biomed Appl 660:375–379,
1994.
21. Kodama H, Mikasa H, Sasaki K, Awata S, Nakayama K. Unusual
metabolism of sulfur-containing amino acids in rats treated with DL-
propargylglycine. Arch Biochem Biophys 225:25–32, 1983.
22. Li XH, Bu DF, Jin HF, Ding YG, Du JB, Li J, Tang CS. The dynamic
changes in endogenous hydrogen sulfide pathway at the early stage of
pulmonary hypertension induced by high pulmonary flow in rats. Chin
J Appl Physiol 23:75–79, 2007.
23. Zhang CY, Du JB, Yan H, Tang CS. Effect of a new gasotransmitter,
hydrogen sulfide, on collagen remodeling of pulmonary artery under
hypoxia. Zhonghua Jie He He Hu Xi Za Zhi 28:448–452, 2005.
24. Edwards GA, Orien WD. Modified assay for determination of
hydroxyproline in a tissue hydrolyzate. Clin Chim Acta 104:161–
167, 1980.
25. Pe´rez-Ramos J, Lourdes Segura-Valdez M, Vanda B, Selman M, Pardo
A. Matrix metalloproteinases 2, 9, and 13, and tissue inhibitors of
H
2
S AND PULMONARY ARTERY COLLAGEN REMODELING 511
metalloproteinases 1 and 2 in experimental lung silicosis. Am J Respir
Crit Care Med 160:1274–1282,1999.
26. Ruiz V, Ordo´n˜ez RM, Berumen J, Ramı´rez R, Uhal B, Becerril C,
Pardo A, Selman M. Unbalanced collagenases/TIMP-1 expression and
epithelial apoptosis in experimental lung fibrosis. Am J Physiol Lung
Cell Mol Physiol 285:1026–1036, 2003.
27. Twigg SM, Joly AH, Chen MM, Tsubaki J, Kim HS, Hwa V, Oh Y,
Rosenfeld RG. Connective tissue growth factor/IGF-binding protein-
related protein-2 is a mediator in the induction of fibronectin by
advanced glycosylation end-products in human dermal fibroblasts.
Endocrinology 143:1260–1269, 2002.
28. Schild C, Trueb B. Mechanical stress is required for high-level
expression of connective tissue growth factor. Exp Cell Res 274:83–91,
2002.
29. Kessler D, Dethlefsen S, Haase I, Plomann M, Hirche F, Krieg T, Eckes
B. Fibroblasts in mechanically stressed collagen lattices assume a
‘‘synthetic’’ phenotype. J Biol Chem 276:36575–36585, 2001.
512 LI ET AL
... Furthermore, NaHS decreased the area of atherosclerotic lesions in ApoE knock-out mice [6]. Endogenous H 2 S has been shown to control remodeling of the vascular wall as relative medial thickness of the pulmonary artery could be increased by CSE inhibitors in a model of rat pulmonary hypertension [7]. ...
... Other studies have also demonstrated that exogenous H 2 S sources (NaHS) led to reduction of MMP-13 and/or MMP-8 levels in human chondrocytes under inflammatory conditions [14] and in a model of rat heart disease [31]. A similar regulation of collagen accumulation has been observed by Li et al (2007Li et al ( , 2009) who demonstrated that increased CSE and endogenous H 2 S levels in lung tissues of pulmonary hypertensive rat were correlated with increased collagen I and III levels [7,33]. Thus, the down-regulation of MMP activities by endogenous H 2 S production should increase vascular wall thickness. ...
... Other studies have also demonstrated that exogenous H 2 S sources (NaHS) led to reduction of MMP-13 and/or MMP-8 levels in human chondrocytes under inflammatory conditions [14] and in a model of rat heart disease [31]. A similar regulation of collagen accumulation has been observed by Li et al (2007Li et al ( , 2009) who demonstrated that increased CSE and endogenous H 2 S levels in lung tissues of pulmonary hypertensive rat were correlated with increased collagen I and III levels [7,33]. Thus, the down-regulation of MMP activities by endogenous H 2 S production should increase vascular wall thickness. ...
Article
Full-text available
Hydrogen sulfide (H 2 S) is a mediator with demonstrated protective effects for the cardiovas-cular system. On the other hand, prostaglandin (PG)E 2 is involved in vascular wall remodel-ing by regulating matrix metalloproteinase (MMP) activities. We tested the hypothesis that endogenous H 2 S may modulate PGE 2 , MMP-1 activity and endogenous tissue inhibitors of MMPs (TIMP-1/-2). This regulatory pathway could be involved in thinning of abdominal aor-tic aneurysm (AAA) and thickening of saphenous vein (SV) varicosities. The expression of the enzyme responsible for H 2 S synthesis, cystathionine-γ-lyase (CSE) and its activity, were significantly higher in varicose vein as compared to SV. On the contrary, the endoge-nous H 2 S level and CSE expression were lower in AAA as compared to healthy aorta (HA). Endogenous H 2 S was responsible for inhibition of PGE 2 synthesis mostly in varicose veins and HA. A similar effect was observed with exogenous H 2 S and consequently decreasing active MMP-1/TIMP ratios in SV and varicose veins. In contrast, in AAA, higher levels of PGE 2 and active MMP-1/TIMP ratios were found versus HA. These findings suggest that differences in H 2 S content in AAA and varicose veins modulate endogenous PGE 2 production and consequently the MMP/TIMP ratio. This mechanism may be crucial in vascular wall remodeling observed in different vascular pathologies (aneurysm, varicosities, atheroscle-rosis and pulmonary hypertension).
... 8,26 Additionally, there is evidence that H 2 S is involved in collagen metabolism and regulation of the vascular wall. 15,27 Hydrogen sulfide treatment induces angiogenesis after cerebral ischemia and may be of value in regenerative recovery after stroke. 21 Genetic polymorphisms of the genes encoding CBS may be associated with variable enzyme activity resulting in altered homocysteine and H 2 S levels and may play a role in the pathogenesis of cerebrovascular disease. ...
... 23,34,35 Cystathionine b-synthase also catalyzes the formation of H 2 S. Endogenous H 2 S, as well as carbon monoxide (CO) and nitric oxide (NO), play a role in pulmonary artery collagen remodeling in rats with high pulmonary blood flow. 16,27,36 Hydrogen sulfide also has a regulatory effect on vascular collagen content in hypertensive rats. 38 Polymorphisms of CBS may be associated with altered H 2 S levels that subsequently impact collagen metabolism in cerebral vessels. ...
Article
OBJECTIVE Cystathionine β-synthase (CBS) is involved in homocysteine and hydrogen sulfide (H 2 S) metabolism. Both products have been implicated in the pathophysiology of cerebrovascular diseases. The impact of CBS polymorphisms on aneurysmal subarachnoid hemorrhage (aSAH) and its clinical sequelae is poorly understood. METHODS Blood samples from all patients enrolled in the CARAS (Cerebral Aneurysm Renin Angiotensin System) study were used for genetic evaluation. The CARAS study prospectively enrolled aSAH patients at 2 academic institutions in the United States from 2012 to 2015. Common CBS polymorphisms were detected using 5′exonuclease genotyping assays. Analysis of associations between CBS polymorphisms and aSAH was performed. RESULTS Samples from 149 aSAH patients and 50 controls were available for analysis. In multivariate logistic regression analysis, the insertion allele of the 844ins68 CBS insertion polymorphism showed a dominant effect on aSAH. The GG genotype of the CBS G/A single nucleotide polymorphism (rs234706) was independently associated with unfavorable functional outcome (modified Rankin Scale Score 3–6) at discharge and last follow-up, but not clinical vasospasm or delayed cerebral ischemia (DCI). CONCLUSIONS The insertion allele of the 844ins68 CBS insertion polymorphism was independently associated with aSAH while the GG genotype of rs234706 was associated with an unfavorable outcome both at discharge and last follow-up. Increased CBS activity may exert its neuroprotective effects through alteration of H 2 S levels, and independent of clinical vasospasm and DCI.
... 10 H 2 S is an important endogenous vasodilator and has been confirmed as a gas opener of the K ATP channel in VSM. 11 Studies have shown that H 2 S is a powerful pulmonary artery vasodilator that can significantly reduce PH. 12 Endogenous H 2 S was found to be involved in the occurrence of hypoxic, 13 high pulmonary blood flow 14 and monocrotaline-induced 15,16 animal models of PH, and exogenous H 2 S could alleviate PH and pulmonary vascular structural remodeling. The mechanism includes relaxation of pulmonary VSM, 12 inhibition of proliferation in VSM cells, 17 induction of apoptosis in VSM cells, 18 inhibition of abnormal accumulation of extracellular matrix in VSM cells, 19 and improvement of antioxidant capacity of lung tissue, 20 etc. H 2 S is a potential dilatator of the human pulmonary artery and is an important drug for lowering pulmonary artery pressure. ...
Article
Full-text available
Objective To explore the relationship between endogenous hydrogen sulfide (H2S) and high-resolution computed tomography (HRCT) indexes in pulmonary vascular remodeling. Methods A total of 94 stable chronic obstructive pulmonary disease (COPD) patients were recruited for the study.Plasma H2S levels were measured using fluorescence probe. Fluorescence quantitative polymerase chain reaction was used to measure H2S synthase cystathionine-γ-lyase (CSE) mRNA and cystathionine-β-synthesis enzyme (CBS) mRNA. The main pulmonary artery diameter (mPAD), axial diagonal mPAD, coronal mPAD, sagittal mPAD, right pulmonary artery diameter (RPAD), left pulmonary artery diameter (LPAD), and ascending aortic diameter (AAD) and the percentage of total cross-sectional area of vessels less than 5 mm² of total lung area (%CSA <5) on HRCT were measured. Pulmonary arterial systolic pressure (PASP) of echocardiography, blood gas analysis, and routine blood tests were performed. Correlation analysis and multivariate linear regression were performed using SPSS 22.0. Results H2S was negatively correlated with mPAD, axial diagonal mPAD, and sagittal mPAD (r = −0.25~−0.32) and positively correlated with PaO2 (r = 0.35). Relative expression of CSE mRNA was positively correlated with PASP, coronal mPAD, sagittal mPAD, white blood cell count (WBC), and neutrophil count (N) (r = 0.30~0.44). The relative expression of CBS mRNA was positively correlated with PASP, WBC, and N (r = 0.34~0.41). In separate models predicting pulmonary vascular indexes, a 1μmol/L increase in H2S predicted lower pulmonary artery diameter (for axial diagonal mPAD, 0.76mm lower; for mPAD/AAD, 0.68mm lower). All P values were less than 0.05. Conclusion Endogenous H2S may be involved in pulmonary vascular remodeling, providing a new method for the diagnosis and treatment of COPD. The generation of H2S may be inhibited by hypoxia, inflammation, etc.
... Moreover, the expression of collagen-promoting molecules connective tissue growth factor (CTGF) and MMP-13 were increased after the application of D, Lpropargylglycine (PPG), whereas the expression of tissue inhibitor of metalloproteinase 1 (TIMP-1) was significantly decreased. All of the above results indicate that H2S alleviates oxidative stress injuries, thus inhibiting pulmonary vascular remodeling [130,131]. ...
Article
Full-text available
Background: Hydrogen sulfide (H2S) is considered to be the third gasotransmitter after carbon monoxide (CO) and nitric oxide (NO). It plays an important role in the regulation of vascular homeostasis. Vascular remodeling have has proved to be related to the impaired H2S generation. Aim of review: This study aimed to summarize and discuss current data about the function of H2S in vascular physiology and pathophysiology as well as the underlying mechanisms. Key scientific concepts of review: Endogenous hydrogen sulfide (H2S) as a third gasotransmitter is primarily generated by the enzymatic pathways and regulated by several metabolic pathways. H2S as a physiologic vascular regulator, inhibits proliferation, regulates its apoptosis and autophagy of vascular cells and controls the vascular tone. Accumulating evidence shows that the downregulation of H2S pathway is involved in the pathogenesis of a variety of vascular diseases, such as hypertension, atherosclerosis and pulmonary hypertension. Alternatively, H2S supplementation may greatly help to prevent the progression of the vascular diseases by regulating vascular tone, inhibiting vascular inflammation, protecting against oxidative stress and proliferation, and modulating vascular cell apoptosis, which has been verified in animal and cell experiments and even in the clinical investigation. Besides, H2S system and angiotensin-converting enzyme (ACE) inhibitors play a vital role in alleviating ischemic heart disease and left ventricular dysfunction. Notably, sulfhydryl-containing ACEI inhibitor zofenopril is superior to other ACE inhibitors due to its capability of H2S releasing, in addition to ACE inhibition. The design and application of novel H2S donors have significant clinical implications in the treatment of vascular-related diseases. However, further research regarding the role of H2S in vascular physiology and pathophysiology is required.
... 6 Under abnormal high-flow physiologic conditions, most of the pulmonary vasculature is exposed to high shear stress, resulting in pulmonary vascular remodeling. Experimental attempts to reproduce high-flow PH in animals have been made, and various shunt-related models, such as cervical, abdominal, femoral, and central shunts, [7][8][9][10][11] have been described. However, all these models produced only modest degrees of PH. ...
Article
Background: This study sought to establish a new model of high-flow pulmonary hypertension (PH) in mice. This model may be useful for studies seeking to reduce the pulmonary vascular resistance and delay the development of PH caused by congenital heart disease. Materials and methods: The right pulmonary artery was ligated via a right posterolateral thoracotomy. Pulmonary hemodynamics was evaluated by right heart catheterization immediately after ligation and at 2, 4, 8, and 12 wk postoperatively. The right ventricle (RV) and the left ventricle (LV) with septum (S) were weighed to calculate the RV/(LV + S) ratio as an index of right ventricular hypertrophy. Morphologic changes in the left lungs were analyzed, and percentages of muscularized pulmonary vessels were assessed by hematoxylin and eosin, elastica van Gieson and alpha-smooth muscle actin staining. All the study data were compared with data from a model of PH generated by hypoxic stimulation. Results: A pulmonary hypertensive state was successfully induced by 2 wk after surgery. However, the morphologic analysis demonstrated that pulmonary vascular muscularization, as evaluated using right ventricular systolic pressure and RV/(LV + S), was not significantly increased until 4 wk postoperatively. When mice from the new model and the hypoxic model were compared, no significant differences were observed in any of the evaluated indices. Conclusions: High-flow PH can be induced within 4 wk after ligation of the right pulmonary artery, which is easily performed in mice. Such mice can be used as a model of high-flow PH.
Article
H 2 S belongs to the class of molecules known as gasotransmitters, which also includes nitric oxide (NO) and carbon monoxide (CO). Three enzymes are recognized as endogenous sources of H 2 S in various cells and tissues: cystathionine g-lyase (CSE), cystathionine β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (3-MST). The current article reviews the regulation of these enzymes as well as the pathways of their enzymatic and non-enzymatic degradation and elimination. The multiple interactions of H 2 S with other labile endogenous molecules (e.g. NO) and reactive oxygen species are also outlined. The various biological targets and signaling pathways are discussed, with special reference to H 2 S and oxidative posttranscriptional modification of proteins, the effect of H 2 S on channels and intracellular second messenger pathways, the regulation of gene transcription and translation and the regulation of cellular bioenergetics and metabolism. The pharmacological and molecular tools currently available to study H 2 S physiology are also reviewed, including their utility and limitations. In subsequent sections, the role of H 2 S in the regulation of various physiological and cellular functions is reviewed. The physiological role of H 2 S in various cell types and organ systems are overviewed. Finally, the role of H 2 S in the regulation of various organ functions is discussed as well as the characteristic bell-shaped biphasic effects of H 2 S. In addition, key pathophysiological aspects, debated areas, and future research and translational areas are identified A wide array of significant roles of H 2 S in the physiological regulation of all organ functions emerges from this review.
Article
Extracellular matrix (ECM) is a non-cellular three-dimensional macromolecular network that not only provides mechanical support but also transduces essential molecular signals in organ functions. ECM is constantly remodeled to control tissue homeostasis, responsible for cell adhesion, cell migration, cell-to-cell communication, and cell differentiation, etc. The dysregulation of ECM components contributes to various diseases, including cardiovascular diseases, fibrosis, cancer, and neurodegenerative diseases, etc. Aberrant ECM remodeling is initiated by various stress, such as oxidative stress, inflammation, ischemia, and mechanical stress, etc. Hydrogen sulfide (H2S) is a gasotransmitter that exhibits a wide variety of cytoprotective and physiological functions through its anti-oxidative and anti-inflammatory actions. Amounting research shows that H2S can attenuate aberrant ECM remodeling. In this review, we discussed the implications and mechanisms of H2S in the regulation of ECM remodeling in cardiovascular diseases, and highlighted the potential of H2S in the prevention and treatment of cardiovascular diseases through attenuating adverse ECM remodeling.
Chapter
Hydrogen sulfide (H2S), known as a gas signal molecule, plays an important role in the development of cardiovascular diseases (CVD) through mechanisms such as angiogenesis, vasodilation, and anti-vascular endothelial cell senescence. Current studies have shown that H2S can regulate cardiac function through epigenetic regulation. The regulation has opened up a new avenue for the study of CVD development mechanism and H2S related drug discoveries.
Article
Bronchopulmonary dysplasia (BPD), which remains a major clinical problem for preterm infants, is caused mainly by hyperoxia, mechanical ventilation and inflammation. Many approaches have been developed with the aim of decreasing the incidence of or alleviating BPD, but effective methods are still lacking. Gasotransmitters, a type of small gas molecule that can be generated endogenously, exert a protective effect against BPD-associated lung injury; nitric oxide (NO), carbon monoxide (CO) and hydrogen sulfide (H2S) are three such gasotransmitters. The protective effects of NO have been extensively studied in animal models of BPD, but the results of these studies are inconsistent with those of clinical trials. NO inhalation seems to have no effect on BPD, although side effects have been reported. NO inhalation is not recommended for BPD treatment in preterm infants, except those with severe pulmonary hypertension. Both CO and H2S decreased lung injury in BPD rodent models in preclinical studies. Another small gas molecule, hydrogen, exerts a protective effect against BPD. The nuclear factor erythroid-derived 2 (Nrf2)/heme oxygenase-1 (HO-1) axis seems to play a central role in the protective effect of these gasotransmitters on BPD. Gasotransmitters play important roles in mammals, but further clinical trials are needed to explore their effects on BPD.
Article
Lipoprotein(a) [Lp(a)] is a strong genetic risk factor for coronary heart diseases. However, the metabolism of this protein remains poorly understood. Efficient and specific drugs that can decrease high plasma levels of Lp(a) have not been developed yet. Hydrogen sulfide (H2 S), a member of the gas transmitter family, performs important biological actions, including protection against cardiovascular diseases and maintenance of the lipid metabolism equilibrium in hepatocytes and adipocytes. In this study, we investigated the possible molecular mechanism of H2 S that influences apolipoprotein(a) [apo(a)] biosynthesis. We also determined the effects of H2 S on apo(a) expression and secretion in HepG2 cells as well as the underlying mechanisms. Results showed that H2 S significantly inhibited the expression and secretion levels of apo(a). These effects were attenuated by the PKCα inhibitor and FXR siRNA. H2 S also reduced HNF4α expression and enhanced FXR expression. The Akt inhibitor partially reversed H2 S-induced inhibition of apo(a) and HNF4α expression and apo(a) secretion. This study reveals that H2 S suppressed apo(a) expression and secretion via the PKCα-FXR and PI3K/Akt-HNF4α pathways.
Article
Full-text available
A cDNA clone for cystathionine gamma-lyase was isolated from a rat cDNA library in lambda gt11 by screening with a monospecific antiserum. The identity of this clone, containing 600 bp proximal to the 3'-end of the gene, was confirmed by positive hybridization selection. Northern-blot hybridization showed the expected higher abundance of the corresponding mRNA in liver than in brain. Two further cDNA clones from a plasmid pcD library were isolated by colony hybridization with the first clone and were found to contain inserts of 1600 and 1850 bp. One of these was confirmed as encoding cystathionine gamma-lyase by hybridization with two independent pools of oligodeoxynucleotides corresponding to partial amino acid sequence information for cystathionine gamma-lyase. The other clone (estimated to represent all but 8% of the 5'-end of the mRNA) was sequenced and its deduced amino acid sequence showed similarity to those of the Escherichia coli enzymes cystathionine beta-lyase and cystathionine gamma-synthase throughout its length, especially to that of the latter.
Article
To explore the time-dependent changes of endogenous hydrogen sulfide system at the early stage of pulmonary hypertension induced by high pulmonary flow in rats. Eighty male SD rats, whose weight ranged 140 - 160 g, were randomly divided into control group (n = 40) and shunt group (n = 40). Rats in shunt group were subjected to an abdominal aorta-inferior vena cava shunt to create an animal model of high pulmonary flow. After 1 d, 3 d, 1 week, 4 week and 8 weeks of experiment, systolic pulmonary artery pressure (SPAP) of each rat, the H2S of rat lung tissue and CSEmRNA of rat lung tissue were evaluated, respectively. SPAP increased significantly as compared with those in control group in 1 week and 8 weeks of experiment. In contrast to control group, the H2S of rat lung tissue increased significantly on 3 d and in 4 weeks, respectively. Meanwhile, in contrast to control group, relative amount of CSE mRNA of lung tissues elevated significantly on 3 d and in 4 weeks, respectively. Moreover, SPAP and the H2S of rat lung tissue, the CSE mRNA of rat lung tissue correlated negatively in 1 week, 4 weeks and 8 weeks of experiment. Animal model of rats with high pulmonary blood flow exhibited pulmonary hypertension. Lung tissue H2S and CSE mRNA of rats exhibited double peaks within 8 weeks. These results revealed that endogenous H2S system might be relevant with the development of pulmonary hypertension induced by high pulmonary blood flow, and probably, it played a protective role in the regulation of pulmonary hypertension, especially, at its early stage.
Article
L-Propargylglycine, a naturally occurring gamma, delta-acetylenic alpha-amino acid, induces mechanism-based inactivation of two pyridoxal phosphate dependent enzymes of methionine metabolism: (1) cystathionine gamma-synthease, which catalyzes a gamma-replacement reaction in methionine biosynthesis, and (2) methionine gamma-lyase, which catalyzes a gamma-elimination reaction in methionine breakdown. Biphasic pseudo-first-order inactivation kinetics were observed for both enzymes. Complete inactivation is achieved with a minimum molar ratio ([propargylglycine]/[enzyme monomer]) of 4:1 for cystathionine gamma-synthase and of 8:1 for methionine gamma-lyase, consistent with a small number of turnovers per inactivation event. Partitioning ratios were determined directly from observed primary kinetic isotope effects. [alpha-2H]Propargylglycine displays kH/kD values of about 3 on inactivation half-times. [alpha-3H]-Propargylglycine gives release of tritium to solvent nominally stoichiometric with inactivation but, on correction for the calculated tritium isotope discrimination, partition ratios of four and six turnovers per monomer inactivated are indicated for cystathionine gamma-synthase and methionine gamma-lyase, respectively. The inactivation stoichiometry, using [alpha-14C]-propargylglycine, is four labels per tetramer of cystathionine gamma-synthase but usually only two labels per tetramer of methionine gamma-lyase (half-of-the-sites reactivity). Two-dimensional urea isoelectrofocusing/NaDodSO4 electrophoresis suggests (1) that both native enzymes are alpha 2 beta 2 tetramers where the subunits are distinguishable by charge but not by size and (2) that, while each subunit of a cystathionine gamma-synthase tetramer becomes modified by propargylglycine, only one alpha and one beta subunit may be labeled in an inactive alpha 2 beta 2 tetramer of methionine gamma-lyase. Steady-state spectroscopic analyses during inactivation indicated that modified cystathionine gamma-synthase may reprotonate C2 of the enzyme--inactivator adduct, so that the cofactor is still in the pyridoxaldimine oxidation state. Fully inactivated methionine gamma-lyase has lambda max values at 460 and 495 nm, which may represent conjugated pyridoximine paraquinoid that does not reprotonate at C2 of the bound adduct. Either species could arise from Michael-type addition of an enzymic nucleophile to an electrophilic 3,4-allenic paraquinoid intermediate, generated initially by propargylic rearrangement upon a 4,5-acetylenic pyridoximine structure, as originally proposed for propargylglycine inactivation of gamma-cystathionase [Abeles, R., & Walsh, C. (1973) J. Am. Chem. Soc. 95, 6124]. It is reasonable that cystathionine gamma-synthase is the major in vivo target for this natural acetylenic toxin, the growth-inhibitory effects of which are reversed by methionine.
Article
The acetylenic amino acid propargylglycine (2-amino-4-pentynoate) irreversibly inactivates two pyridoxal-P dependent enzymes which generate substrate-derived beta carbanions during catalysis.
Article
Study objective – The aim of the study was to develop a new procedure to produce abdominal aortocaval shunts in the rat without vascular microsurgery. Procedure – The inferior vena cava and abdominal aorta were exposed by laparotomy. The aorta was punctured caudal to the left renal artery with an 18 gauge disposable needle which was advanced into the vessel, perforating the adjacent wall between aorta and vena cava and penetrating the latter. A bulldog vascular clamp was placed across the aorta cephalic to the puncture, the needle was withdrawn, and the aortic puncture point was sealed with a drop of cyanoacrylate glue. The clamp was removed 30 s later. Patency of the shunt was verified visually by swelling of the vena cava and admixture of arterial and venous blood. No local haemorrhages were seen. The laporatomy was then closed. The procedure takes less than 10 min. Results – Of 11 rats which received this procedure, only one died within 24 h. All the other animals were killed 4 weeks after operation. Nine of these 10 animals had developed cardiac hypertrophy of about the same magnitude. There were no changes in sham operated controls. Conclusions – This is a reproducible, simple and rapid method of developing high output heart failure and cardiac hypertrophy in the rat which could be useful in many laboratories.
Article
S-(2-Hydroxy-2-carboxyethyl)homocysteine, S-(3-hydroxy-3-carboxy-n-propyl)-cysteine, N-acylated S-(beta-carboxyethyl)cysteine, and N-acylated S-(3-hydroxy-3-carboxy-n-propyl) cysteine were excreted in the urine after DL-propargylglycine treatment. Cystathionine was also accumulated in several tissues of DL-propargylglycine-treated rats. N-Monoacetylcystathione was found in the liver of rats and was also detected in the kidney and serum. Cystathionine gamma-lyase activity in liver decreased to about 4% of that of control rats 24 h after the DL-propargylglycine injection, and alanine aminotransferase activity decreased to about 35% of that of control rats. On the other hand, aspartate aminotransferase and cystathionine beta-synthese activity did not show significant changes from those of control rats. The ability of normal tissues to synthesize cystathionine utilizing cystathionine beta-synthase was 1.98 +/- 0.40 mumol/min/g in liver, 0.61 +/- 0.13 in kidney, and 0.18 +/- 0.015 in brain. The maximal contents of cystathionine in rat tissues and the administered amounts of DL-propargylglycine agreed well with the ability to synthesize cystathionine in each tissue.
Article
The contribution of cystathionine gamma-lyase, cystathionine beta-synthase and cysteine aminotransferase coupled to 3-mercaptopyruvate sulphurtransferase to cysteine desulphhydration in rat liver and kidney was assessed with four different assay systems. Cystathionine gamma-lyase and cystathionine beta-synthase were active when homogenates were incubated with 280 mM-L-cysteine and 3 mM-pyridoxal 5'-phosphate at pH 7.8. Cysteine aminotransferase in combination with 3-mercaptopyruvate sulphurtransferase catalysed essentially all of the H2S production from cysteine at pH 9.7 with 160 mM-L-cysteine, 2 mM-pyridoxal 5'-phosphate, 3 mM-2-oxoglutarate and 3 mM-dithiothreitol. At more-physiological concentrations of cysteine (2 mM) cystathionine gamma-lyase and cystathionine beta-synthase both appeared to be active in cysteine desulphhydration, whereas the aminotransferase pathway did not. The effect of inhibition of cystathionine gamma-lyase by a suicide inactivator, propargylglycine, in the intact rat was also investigated; there was no significant effect of propargylglycine administration on the urinary excretion of total 35S, 35SO4(2-) or [35S]taurine formed from labelled dietary cysteine.
Article
A modified assay for the determination of hydroxyproline in tissue is presented. The modifications greatly reduce the time required for analysis of excised tissue as first introduced by Stegemann and Stalder [1]. These modifications include a change in the technique for tissue hydrolysis and a change in the preparation of the hydroxyproline oxidizing agent. The analysis utilizes the standard addition technique, eliminating the need for correction of matrix effects between the specimen and standard. This paper attempts to give a complete detailed description of the assay such that the procedure may be repeated without requiring additional reference material.
Article
An experimental animal model with cystathioninuria was obtained by the injection of D,L-propargylglycine into rats. The concentrations of D,L-propargylglycine in urine, several tissues and serum at different times after the injection were measured by liquid chromatography-mass spectrometry. The propargylglycine accumulated rapidly in several tissues and serum of the rats, and reached its maximum level at about 2 h after the injection. Approximately 21.2% of the administered propargylglycine was excreted in urine. N-Acetylpropargylglycine was identified as a new metabolite of propargylglycine in urine. The concentration of propargylglycine was 100 times that of N-acetylpropargylglycine in urine.