1.Ulinastatin inhibits hypoxia-induced phenotype modulation of pulmonary artery smooth muscle cells by up-regulating PPAR-γ expression.
Kun TANG ; Chang LIU ; Lin CHEN ; Jing GAO ; Chao ZHANG
Journal of Southern Medical University 2016;36(10):1322-1327
OBJECTIVETo evaluate the effect of ulinastatin on hypoxia-induced phenotype modulation of pulmonary artery smooth muscle cells (PASMCs) and explore the underlying mechanism.
METHODSCultured PASMCs from SD rats were exposed to normoxic condition, normoxia with ulinastatin treatment, hypoxia, or hypoxia with ulinastatin treatment. After 24 h of exposures, the cells were examined for SM-α-actin and caplonin expressions with immunofluorescence assay and for cell migration with CCK-8 andH-TdR assays. Western blotting was used for detecting the expressions of PPAR-γ in the cells, and PPAR-γ-responsive firefly luciferase reporter was employed for measuring the transcriptional activity of PPAR-γ. The PPAR-γ inhibitor GW9662 was used to explore the mechanism of the inhibitory effect of ulinastatin on hypoxia induced-phenotype modulation of PASMCs by measuring the changes in cell proliferation and migration.
RESULTSUlinastatin obviously enhanced the expressions of SM-α-actin and calponin (P<0.05), inhibited the proliferation and migration (P<0.05), and up-regulated the expression of PPAR-γ in PASMCs exposed to hypoxia (P<0.05). Pretreatment of the cells with GW9662 abolished the effect of ulinastatin on hypoxia-induced phenotype modulation of PASMCs and enhanced the cell proliferation and migration (P<0.05).
CONCLUSIONUlinastatin inhibits hypoxia-induced phenotype modulation of PASMCs from rats possibly by up-regulating the expression of PPAR-γ.
Actins ; metabolism ; Animals ; Calcium-Binding Proteins ; metabolism ; Cell Hypoxia ; Cell Proliferation ; Cells, Cultured ; Glycoproteins ; pharmacology ; Microfilament Proteins ; metabolism ; Myocytes, Smooth Muscle ; cytology ; drug effects ; PPAR gamma ; metabolism ; Phenotype ; Pulmonary Artery ; cytology ; Rats ; Rats, Sprague-Dawley ; Up-Regulation
2.m-Nisodipine inhibited 5-HT-induced proliferation of rat PASMCs through Rho/ROCK signal pathway.
Huan-long LIU ; Ding YU ; Zhong-ning ZHU ; Su-wen SU ; Xue-yan CHEN ; Yong-jian ZHANG
Acta Pharmaceutica Sinica 2015;50(7):824-829
This paper is to report the exploration of the activation of Rho/ROCK signal pathway in 5-HT-induced proliferation of rat pulmonary artery smooth muscle cells (PASMCs) and the inhibitory effect of m-Nis on this pathway. PASMCs were cultured with the explant technique. MTT assay was used to explore the proliferation of PASMCs after 5-HT treated for different time and the intervening effect of m-Nis. RT-PCR and Western blot were used respectively to explore the mRNA expression of RhoA, ROCK1 and the protein expression of p-MYPT1 in 5-HT-treated PASMCs and intervening effect of m-Nis. The results of MTT assay suggested that 5-HT (1 µmol · L(-1)) treatment for 12-72 h significantly induced the proliferation of rat PASMCs (P<0.05 or P < 0.01), which were inhibited by m-Nis (1 x 10(-5), 1 x 10(-6), l x 10(-7), 1 x10(-8) mol · L(-1)) in dose-dependent manners (P < 0.05 or P < 0.01). Similarly, the mRNA expression of RhoA, ROCK1 and the protein expression of p-MYPT1 were also inhibited by m-Nis in different degrees (P < 0.05 or P < 0.01). Thus, the results of this study suggested that Rho/ROCK pathway played an important role in 5-HT-induced proliferation of rat PASMCs, m-Nis inhibited 5-HT-induced proliferation obviously, which may be related to the blockage of Rho/ROCK signal pathway.
Animals
;
Cell Proliferation
;
drug effects
;
Myocytes, Smooth Muscle
;
cytology
;
drug effects
;
Nisoldipine
;
pharmacology
;
Protein Phosphatase 1
;
metabolism
;
Pulmonary Artery
;
cytology
;
Rats
;
Serotonin
;
pharmacology
;
Signal Transduction
;
rho-Associated Kinases
;
metabolism
;
rhoA GTP-Binding Protein
;
metabolism
3.The Effect of Umbilical Cord Blood Derived Mesenchymal Stem Cells in Monocrotaline-induced Pulmonary Artery Hypertension Rats.
Hyeryon LEE ; Jae Chul LEE ; Jung Hyun KWON ; Kwan Chang KIM ; Min Sun CHO ; Yoon Sun YANG ; Wonil OH ; Soo Jin CHOI ; Eun Seok SEO ; Sang Joon LEE ; Tae Jun WANG ; Young Mi HONG
Journal of Korean Medical Science 2015;30(5):576-585
Pulmonary arterial hypertension (PAH) causes right ventricular failure due to a gradual increase in pulmonary vascular resistance. The purposes of this study were to confirm the engraftment of human umbilical cord blood-mesenchymal stem cells (hUCB-MSCs) placed in the correct place in the lung and research on changes of hemodynamics, pulmonary pathology, immunomodulation and several gene expressions in monocrotaline (MCT)-induced PAH rat models after hUCB-MSCs transfusion. The rats were grouped as follows: the control (C) group; the M group (MCT 60 mg/kg); the U group (hUCB-MSCs transfusion). They received transfusions via the external jugular vein a week after MCT injection. The mean right ventricular pressure (RVP) was significantly reduced in the U group after the 2 week. The indicators of RV hypertrophy were significantly reduced in the U group at week 4. Reduced medial wall thickness in the pulmonary arteriole was noted in the U group at week 4. Reduced number of intra-acinar muscular pulmonary arteries was observed in the U group after 2 week. Protein expressions such as endothelin (ET)-1, endothelin receptor A (ERA), endothelial nitric oxide synthase (eNOS) and matrix metalloproteinase (MMP)-2 significantly decreased at week 4. The decreased levels of ERA, eNOS and MMP-2 immunoreactivity were noted by immnohistochemical staining. After hUCB-MSCs were administered, there were the improvement of RVH and mean RVP. Reductions in several protein expressions and immunomodulation were also detected. It is suggested that hUCB-MSCs may be a promising therapeutic option for PAH.
Animals
;
Cytokines/metabolism
;
Disease Models, Animal
;
Endothelin-1/metabolism
;
Fetal Blood/*cytology
;
Gene Expression Regulation/drug effects
;
Hemodynamics
;
Humans
;
Hypertension, Pulmonary/chemically induced/*therapy
;
Hypertrophy, Right Ventricular/physiopathology
;
Immunohistochemistry
;
Lung/metabolism/pathology
;
Male
;
Matrix Metalloproteinase 2/metabolism
;
*Mesenchymal Stem Cell Transplantation
;
Mesenchymal Stromal Cells/*cytology/metabolism
;
Monocrotaline/toxicity
;
Nitric Oxide Synthase Type III/metabolism
;
Pulmonary Artery/pathology
;
Rats
;
Rats, Sprague-Dawley
;
Receptor, Endothelin A/metabolism
4.Effect of puerarin on hypoxia induced proliferation of PASMCs by regulating reactive oxygen.
Xiao-dan ZHANG ; Li-wei WANG ; Shu-jing WANG ; Da-ling ZHU ; Yan-nan YANG ; Jie-jing SHENG ; Sha-sha SONG
China Journal of Chinese Materia Medica 2015;40(15):3027-3033
To discuss the effect of puerarin (Pue) on the proliferation of hypoxia-induced pulmonary artery smooth muscle cells (PASMCs) and discuss whether its mechanism is achieved by regulating reactive oxygen. PASMCs of primarily cultured rats (2-5 generations) were selected in the experiment. MTT, Western blot, FCM and DCFH-DA were used to observe Pue's effect the proliferation of PASMCs. The Western blot was adopted to detect whether ROS participated in Pue's effect in inhibiting PASMC proliferation. The PASMCs were divided into five groups: the normoxia group, the hypoxia group, the hypoxia + Pue group, the hypoxia + Pue + Rotenone group and the hypoxia + Rotenone group, with Rotenone as the ROS blocker. According to the results, under the conditions of normoxia, Pue had no effect on the PASMC proliferation; But, under the conditions of hypoxia, it could inhibit the PASMC proliferation; Under the conditions of normoxia and hypoxia, Pue had no effect on the expression of the tumor necrosis factor-α (TNF-α) among PASMCs, could down-regulate the expression of hypoxia-induced cell cycle protein Cyclin A and proliferative nuclear antigen (PCNA). DCFH-DA proved Pue could reverse ROS rise caused by hypoxia. Both Rotenone and Pue could inhibit the up-regulated expressions of HIF-1α, Cyclin A, PCNA caused by anoxia, with a synergistic effect. The results suggested that Pue could inhibit the hypoxia-induced PASMC proliferation. Its mechanism may be achieved by regulating ROS.
Animals
;
Cell Cycle
;
drug effects
;
Cell Proliferation
;
drug effects
;
Cells, Cultured
;
Hypoxia
;
pathology
;
Isoflavones
;
pharmacology
;
Male
;
Myocytes, Smooth Muscle
;
drug effects
;
physiology
;
Proliferating Cell Nuclear Antigen
;
analysis
;
Pulmonary Artery
;
cytology
;
drug effects
;
Rats
;
Rats, Wistar
;
Reactive Oxygen Species
;
metabolism
5.Effect of autophagy inhibitor chloroquine on the proliferation of PASMCs induced by hypoxia.
Huan-Mian ZHU ; Ran CHEN ; Feng XUE ; Yang-Ping SHENTU ; Xiao-Fang FAN ; Yong-Sheng GONG ; Hong-Yu ZHANG ; Xiao-Xia KONG
Chinese Journal of Applied Physiology 2014;30(1):8-12
OBJECTIVETo investigate the role of autophagy inhibitor chloroquine (CQ) in the proliferation of pulmonary arterial smooth muscle cells (PASMCs) in hypoxia conditions.
METHODSThe following groups in this study were set up: control group, hypoxia group, 50 micromol/L CQ + hypoxia group, 50 micromol/L CQ group. The viability of PASMCs in every group was detected by MTT assay. Autophagic vacuoles in the cells were observed by MDC staining. Protein expression of microtubule associated protein light chain 3 (LC3) was measured by Western blot. Migration of PASMCs was detected by wound healing assay.
RESULTSCompared with control group, no effect on the viability of PASMCs was observed treated by CQ alone. In 1% hypoxia group, cell viability increased significantly compared with that in control group. The number of autophagic vacuoles and the rate of cell migration and also protein expression of LC3-II were also markedly increased. Compared with hypoxia group, addition of CQ increased the number of autophagic vacuoles and the levels of LC3-II protein, but decreased the proliferation and migration of PASMCs.
CONCLUSIONHypoxia could activates autophagy and contributes to proliferation and migration of PASMCs, and autophagy inhibitor CQ could decrease the effect of hypoxia on PASMCs through inhibiting autophagy process.
Autophagy ; drug effects ; Cell Hypoxia ; Cell Movement ; Cell Survival ; Cells, Cultured ; Chloroquine ; pharmacology ; Humans ; Microtubule-Associated Proteins ; metabolism ; Myocytes, Smooth Muscle ; drug effects ; Pulmonary Artery ; cytology
6.Inhibitory effect of taurine in hypoxia-induced rat pulmonary artery smooth muscle cell proliferation and signal transduction mechanism.
Xiao-Dan ZHANG ; Peng SUN ; Da-Ling ZHU ; Nan XIE
China Journal of Chinese Materia Medica 2014;39(10):1902-1907
OBJECTIVETo discuss the effect of taurine (Tau) on the proliferation of hypoxia-induced pulmonary artery smooth muscle cells (PASMCs), and study whether the extracellular signal-regulated kinase 1/2 (ERK1/2) signal pathway participated in the Tau-inhibited PASMC proliferation process and the possible molecular mechanism.
METHODThe primary culture was performed for PASMCs in rats. The second to fifth generations were adopted for the experiment. The Tau concentration was 80 mmol x L(-1). The concentration of ERK1/2 blocker (PD98059) was 50 micromol x L(-1). The drug administration time was 24 h. The effect of Tau on the PASMC proliferation was detected by MTT assay, immunofluorescence staining method and western blot under different conditions. The PASMCs were growing were divided into four groups: the normoxia group, the normoxia + Tau group, the hypoxia group and the hypoxia + Tau group. The Western blot was adopted to detect whether the ERK1/2 signal pathway participated in the Tau-inhibited PASMC proliferation process. Subsequently, the PASMCs were divided into five groups: the normoxia group, the hypoxia group, the hypoxia + Tau group, the hypoxia + Tau + PD98059 group and the hypoxia + PD98059 group.
RESULTHypoxia could induce the PASMC proliferation. Under the conditions of normoxia, Tau had no effect on the PASMC proliferation. Under the conditions of normoxia and hypoxia, Tau had no effect on the expression of the tumor necrosis factor-alpha (TNF-alpha) among PASMCs. Tau could reverse the expression up-regulation of hypoxia-induced proliferative cell nuclear antigen (PCNA) (P < 0.01) and Cyclin A (Cyclin A) (P < 0. 05). Under the conditions of normoxia, Tau had no effect on the expression of phosphoryl extracellular signal-regulated kinase 1/2 (p-ERK1/2). Hypoxia could up-regulate the p-ERK1/2 expression (P < 0.01). Tau could reverse the up-regulation of the hypoxia-induced p-ERK1/2 expression(P < 0.01). Both PD98059 and Tau could inhibit the up-regulated expressions of PCNA, Cyclin A and p-ERK1/2. According to the comparison between the single addition of Tau and PD98059 under conditions of hypoxia, the hypoxia + Tau + PD98059 group showed more significant down-regulation in the expressions of PCNA, Cyclin A and p-ERK1/2.
CONCLUSIONTau could inhibit the hypoxia-induced PASMC proliferation, and may regulate it through ERK1/2 pathway.
Animals ; Cell Hypoxia ; drug effects ; Cell Proliferation ; drug effects ; Cells, Cultured ; MAP Kinase Signaling System ; drug effects ; Myocytes, Smooth Muscle ; cytology ; drug effects ; metabolism ; Oxygen ; metabolism ; Pulmonary Artery ; cytology ; drug effects ; metabolism ; physiopathology ; Rats ; Rats, Wistar ; Taurine ; pharmacology ; Tumor Necrosis Factor-alpha ; genetics ; metabolism
7.Decreased amplitude of Ca²⁺i elevation induced by menthol in pulmonary arterial smooth muscle cells of pulmonary hypertensive rats.
Gai-Ying CHEN ; Hai-Xia JIAO ; Ming-Yue WANG ; Rui-Xing WANG ; Mo-Jun LIN
Acta Physiologica Sinica 2014;66(3):267-275
The study was designed to explore the alteration of intracellular calcium concentration ([Ca²⁺]i), induced by transient receptor potential melastatin 8 (TRPM8) channel-specific agonist menthol, in pulmonary arterial smooth muscle cells (PASMCs) between control and pulmonary hypertensive (PH) rats. PH rat models were established by means of chronic hypoxia (CH) and monocrotaline (MCT) injection, respectively. PASMCs from control and PH rats were cultured. The change of [Ca²⁺]i in PASMCs induced by menthol, and the effect of TRPM8 channel-specific antagonist BCTC on the change of [Ca²⁺]i, were observed. Cellular localization of TRPM8 was examined by using immunohistochemistry. Results showed that menthol increased [Ca²⁺]i in the control PASMCs both in Ca²⁺ -normal and Ca²⁺ - free Tyrode's solutions, and at the same time BCTC could inhibit these two kinds of elevations. Compared with the control group, elevations of [Ca²⁺]i were decreased notably in CH- and MCT-pretreated PASMCs superfused with 2 mmol/L Ca²⁺ - or 0 Ca²⁺ -Tyrode's solutions. Immunohistochemical localization experiments showed that the whole PASMCs were dyed brown except for the nucleus. This study verified that TRPM8 exists both in membrane and sarcoplasmic reticulum of PASMCs. In addition, CH- and MCT-pretreatment could independently down-regulate the Ca²⁺ influx and Ca²⁺ release mediated by TRPM8 channel.
Animals
;
Calcium
;
metabolism
;
Cells, Cultured
;
Menthol
;
pharmacology
;
Myocytes, Smooth Muscle
;
drug effects
;
metabolism
;
Pulmonary Artery
;
cytology
;
Rats
;
Sarcoplasmic Reticulum
;
metabolism
;
TRPM Cation Channels
;
metabolism
8.Calcineurin/NFAT signaling pathway mediates endothelin-1-induced pulmonary artery smooth muscle cell proliferation by regulating phosphodiesterase-5.
Jiamei LU ; Xiaochuang WANG ; Xinming XIE ; Dong HAN ; Shaojun LI ; Manxiang LI
Journal of Southern Medical University 2013;33(1):26-29
OBJECTIVETo examine whether calcineurin/NFAT signaling pathway mediates endothelin-1 (ET-1)-induced proliferation of pulmonary artery smooth muscle cells (PASMCs) by regulating phosphodiesterase-5 (PDE5) and the effect of the selective calcineurin inhibitor cyclosporine A and PDE5 inhibitor sildenafil on ET-1-induced PASMC proliferation.
METHODSPASMCs were treated with ET-1 to stimulate their proliferation with or without prior treatment of the cells with CsA or sildenafil. Calcineurin activity in the cells was measured using a calcineurin activity assay kit, PDE5 expression examined using immunoblotting, and cGMP level detected using a cGMP direct immunoassay kit. PASMC proliferation following the treatments was determined using [(3)H]thymidine incorporation assay.
RESULTSET-1 caused a 2.05-fold increase in the cellular calcineurin activity, a 1.80-fold increase in PDE5 expression, and a 3.20-fold increase in the DNA synthesis rate, and reduced the cGMP level by 67%. Pretreatment of the cells with Cyclosporine blocked the effects of ET-1, and PDE5 inhibition by sildenafil pretreatment also abolished ET-1-induced reduction of cGMP level in the cells. Both Cyclosporine and sildenafil suppressed ET-1-stimulated PASMC proliferation.
CONCLUSIONActivation of calcineurin/NFAT signaling pathway mediates ET-1-induced PASMC proliferation by stimulating PDE5 expression, which further degrades cGMP. Both Cyclosporine and sildenafil can suppress ET-1-stimulated PASMC proliferation in vitro.
Animals ; Calcineurin ; metabolism ; Cell Proliferation ; drug effects ; Cells, Cultured ; Cyclic GMP ; metabolism ; Cyclic Nucleotide Phosphodiesterases, Type 5 ; metabolism ; Cyclosporine ; DNA ; biosynthesis ; Endothelin-1 ; pharmacology ; Muscle, Smooth, Vascular ; cytology ; Myocytes, Smooth Muscle ; cytology ; enzymology ; NFATC Transcription Factors ; metabolism ; Piperazines ; Pulmonary Artery ; cytology ; Purines ; Rats ; Rats, Sprague-Dawley ; Signal Transduction ; Sildenafil Citrate ; Sulfones
9.Fluoxetine Protects against Big Endothelin-1 Induced Anti-Apoptosis by Rescuing Kv1.5 Channels in Human Pulmonary Arterial Smooth Muscle Cells.
Feifeng DAI ; Zhifu MAO ; Jun XIA ; Shaoping ZHU ; Zhiyong WU
Yonsei Medical Journal 2012;53(4):842-848
PURPOSE: Pulmonary Kv channels are thought to play a crucial role in the regulation of cell proliferation and apoptosis. Previous studies have shown that fluoxetine upregulated the expression of Kv1.5 and prevented pulmonary arterial hypertension in monocrotaline-induced or hypoxia-induced rats and mice. The current study was designed to test how fluoxetine regulates Kv1.5 channels, subsequently promoting apoptosis in human PASMCs cultured in vitro. MATERIALS AND METHODS: Human PASMCs were incubated with low-serum DMEM, ET-1, and fluoxetine with and without ET-1 separately for 72 h. Then the proliferation, apoptosis, and expression of TRPC1 and Kv1.5 were detected. RESULTS: In the ET-1 induced group, the upregulation of TRPC1 and down regulation of Kv1.5 enhanced proliferation and anti-apoptosis, which was reversed when treated with fluoxetine. The decreased expression of TRPC1 increased the expression of Kv1.5, subsequently inhibiting proliferation while promoting apoptosis. CONCLUSION: The results from the present study suggested that fluoxetine protects against big endothelin-1 induced anti-apoptosis and rescues Kv1.5 channels in human pulmonary arterial smooth muscle cells, potentially by decreasing intracellular concentrations of Ca2+.
Apoptosis/drug effects/genetics
;
Blotting, Western
;
Cell Proliferation/drug effects
;
Cells, Cultured
;
Endothelin-1/*pharmacology
;
Flow Cytometry
;
Fluoxetine/*pharmacology
;
Humans
;
Kv1.5 Potassium Channel/genetics/*metabolism
;
Muscle, Smooth, Vascular/*cytology/drug effects
;
Pulmonary Artery/*cytology
;
Reverse Transcriptase Polymerase Chain Reaction
10.Hypoxic pulmonary hypertension and novel ATP-sensitive potassium channel opener: the new hope on the horizon.
Yu JIN ; Wei-Ping XIE ; Hong WANG
Chinese Journal of Applied Physiology 2012;28(6):510-523
Hypoxic pulmonary hypertension (HPH) is a syndrome characterized by the increase of pulmonary vascular tone and the structural remodeling of peripheral pulmonary arteries. The aim of specific therapies for hypoxic pulmonary hypertension is to reduce pulmonary vascular resistance, reverse pulmonary vascular remodeling, and thereby improving right ventricular function. Iptakalim, a lipophilic para-amino compound with a low molecular weight, has been demonstrated to be a new selective ATP-sensitive potassium (K(ATP)) channel opener via pharmacological, electrophysiological, biochemical studies, and receptor binding tests. In hypoxia-induced animal models, iptakalim decreases the elevated mean pressure in pulmonary arteries, and attenuates remodeling in the right ventricle, pulmonary arteries and airways. Furthermore, iptakalim has selective antihypertensive effects, selective vasorelaxation effects on smaller arteries, and protective effects on endothelial cells, but no effects on the central nervous, respiratory, digestive or endocrine systems at therapeutic dose. Our previous studies demonstrated that iptakalim inhibited the effects of endothelin-1, reduced the intracellular calcium concentration and inhibited the proliferation of pulmonary artery smooth muscle cells. Since iptakalim has been shown safe and effective in both experimental animal models and phase I clinical trials, it can be a potential candidate of HPH in the future.
Animals
;
Antihypertensive Agents
;
therapeutic use
;
Calcium
;
metabolism
;
Disease Models, Animal
;
Endothelin-1
;
metabolism
;
Hypertension, Pulmonary
;
drug therapy
;
Hypoxia
;
drug therapy
;
KATP Channels
;
drug effects
;
Myocytes, Smooth Muscle
;
cytology
;
drug effects
;
Propylamines
;
therapeutic use
;
Pulmonary Artery
;
drug effects

Result Analysis
Print
Save
E-mail