1.Bax inhibitor 1 inhibits vascular calcification in mice by activating optic atrophy 1 expression.
Wei Ren CHEN ; Hui DU ; Geng QIAN ; Yu Jie ZHOU ; Yun Dai CHEN ; Qian MA ; Xue Sha WU ; Yuan SHA
Journal of Southern Medical University 2022;42(3):330-337
OBJECTIVE:
To investigate the effects of Bax inhibitor 1 (BI- 1) and optic atrophy protein 1 (OPA1) on vascular calcification (VC).
METHODS:
Mouse models of VC were established in ApoE-deficient (ApoE-/-) diabetic mice by high-fat diet feeding for 12 weeks followed by intraperitoneal injections with Nε-carboxymethyl-lysine for 16 weeks. ApoE-/- mice (control group), ApoE-/- diabetic mice (VC group), ApoE-/- diabetic mice with BI-1 overexpression (VC + BI-1TG group), and ApoE-/- diabetic mice with BI-1 overexpression and OPA1 knockout (VC+BI-1TG+OPA1-/- group) were obtained for examination of the degree of aortic calcification using von Kossa staining. The changes in calcium content in the aorta were analyzed using ELISA. The expressions of Runt-related transcription factor 2 (RUNX2) and bone morphogenetic protein 2 (BMP-2) were detected using immunohistochemistry, and the expression of cleaved caspase-3 was determined using Western blotting. Cultured mouse aortic smooth muscle cells were treated with 10 mmol/L β-glycerophosphate for 14 days to induce calcification, and the changes in BI-1 and OPA1 protein expressions were examined using Western blotting and cell apoptosis was detected using TUNEL staining.
RESULTS:
ApoE-/- mice with VC showed significantly decreased expressions of BI-1 and OPA1 proteins in the aorta (P=0.0044) with obviously increased calcium deposition and expressions of RUNX2, BMP-2 and cleaved caspase-3 (P= 0.0041). Overexpression of BI-1 significantly promoted OPA1 protein expression and reduced calcium deposition and expressions of RUNX2, BMP-2 and cleaved caspase-3 (P=0.0006). OPA1 knockdown significantly increased calcium deposition and expressions of RUNX2, BMP-2 and cleaved caspase-3 in the aorta (P=0.0007).
CONCLUSION
BI-1 inhibits VC possibly by promoting the expression of OPA1, reducing calcium deposition and inhibiting osteogenic differentiation and apoptosis of the vascular smooth muscle cells.
Animals
;
Apolipoproteins E/metabolism*
;
Calcium/metabolism*
;
Caspase 3/metabolism*
;
Cells, Cultured
;
Core Binding Factor Alpha 1 Subunit/metabolism*
;
Diabetes Mellitus, Experimental/pathology*
;
GTP Phosphohydrolases/metabolism*
;
Membrane Proteins/metabolism*
;
Mice
;
Mice, Knockout
;
Muscle, Smooth, Vascular/pathology*
;
Myocytes, Smooth Muscle/pathology*
;
Optic Atrophy, Autosomal Dominant/pathology*
;
Osteogenesis
;
Vascular Calcification/pathology*
;
bcl-2-Associated X Protein/metabolism*
2.Research advance of ANRIL on atherosclerosis by regulating cell proliferation and apoptosis.
Juan FANG ; Zhicheng PAN ; Xiaogang GUO
Journal of Zhejiang University. Medical sciences 2020;49(1):113-117
Atherosclerosis is an important pathological basis for coronary artery disease. ANRIL is an antisense non-coding RNA located in Chr9p21 locus, which was identified as the most significant risk locus associated with atherosclerosis. ANRIL can produce multiple transcripts including linear and circular transcripts after various transcript splicing. It has been illustrated that ANRIL plays important roles in the pathology of atherosclerosis by regulating the proliferation and apoptosis of vascular cells. Linear ANRIL can regulate the proliferation of vascular smooth muscle cells (VSMCs) in plaques by chromatin modification, as well as influence the proliferation and the apoptosis of macrophages in post transcription; circular ANRIL can affect the proliferation and apoptosis of VSMCs by chromatin modification as well as interfering with rRNA maturation. In this review, we describe the ANRIL evolution, different transcripts characteristics, and their roles in the proliferation and apoptosis of vascular cells to participate in the process of atherosclerosis, for further understanding the pathogenesis of atherosclerosis and finding potential targets for diagnosis and treatment of atherosclerosis.
Apoptosis
;
genetics
;
Atherosclerosis
;
genetics
;
Cell Proliferation
;
genetics
;
Humans
;
Myocytes, Smooth Muscle
;
pathology
;
RNA, Long Noncoding
;
metabolism
3.Effect and its molecular mechanisms of curcumin on pulmonary artery smooth muscle cells in rat model with chronic obstructive pulmonary disease.
Xiangang LIN ; Yenong CHEN ; Zhuqing LIU
Journal of Zhejiang University. Medical sciences 2016;45(5):469-476
To investigate the effects and the underlying molecular mechanisms of curcumin on pulmonary artery smooth muscle cells in rat model with chronic obstructive pulmonary disease (COPD).A total of 75 male Wistar rats were randomly divided into control group (group CN), model group (group M), low-dose curcumin group (group CL), medium-dose curcumin group (group CM) and high-dose curcumin group (group CH). HE staining was used to observe the morphology of pulmonary artery. Proliferating cell nuclear antigen (PCNA), apoptosis-related protein Bcl-2 and Bax were detected by immunohistochemical staining. TUNEL kit was used to analyze the effects of curcumin on apoptosis of smooth muscle cells, and the protein expressions of SOCS-3/JAK2/STAT pathway in lung tissues were determined by western blot.Right ventricular systolic pressure (RVSP) and right ventricular hypertrophy index (RVMI) in group M were significantly higher than those in group CN, group CH and group CM (all<0.05). HE staining and TUNEL kit test showed that the number of pulmonary artery smooth muscle cells had a significant increase in group M, while the pulmonary artery tube became thin, and the smooth muscle cells shrinked in group CM and group CH. Immunohistochemistry showed that PCNA and Bcl-2 in group M were significantly higher than those in group CN (all<0.05), while Bax expression was significantly lower than that in group CN (<0.05). PCNA in group CM and group CH were significantly lower than that in group M (all<0.05), while Bax expression was significantly higher than that in group M (<0.05). Western blot showed that SOCS-3 protein was significantly decreased in group M, while the p-JAK2, p-STAT1, p-STAT3 were significantly increased (all<0.05). Compared with group M, SOCS-3 protein in group CM and group CH were significantly increased (all<0.05), while the p-JAK2, p-STAT3 were significantly reduced (all<0.05).Curcumin could promote the apoptosis of smooth muscle cells in rats with COPD, and improve the mean pulmonary artery pressure and RVMI through stimulating SOCS-3/JAK2/STAT signaling pathway.
Animals
;
Apoptosis
;
drug effects
;
physiology
;
Arterial Pressure
;
drug effects
;
physiology
;
Curcumin
;
pharmacology
;
Hypertrophy, Right Ventricular
;
pathology
;
physiopathology
;
Janus Kinase 2
;
drug effects
;
physiology
;
Lung
;
chemistry
;
drug effects
;
Male
;
Myocytes, Smooth Muscle
;
drug effects
;
pathology
;
Proliferating Cell Nuclear Antigen
;
drug effects
;
metabolism
;
Proto-Oncogene Proteins c-bcl-2
;
drug effects
;
metabolism
;
Pulmonary Artery
;
drug effects
;
pathology
;
Pulmonary Disease, Chronic Obstructive
;
pathology
;
physiopathology
;
Rats
;
Rats, Wistar
;
STAT Transcription Factors
;
Suppressor of Cytokine Signaling 3 Protein
;
drug effects
;
physiology
;
Ventricular Pressure
;
drug effects
;
bcl-2-Associated X Protein
;
drug effects
;
metabolism
4.Autophagy in atherosclerosis: a phenomenon found in human carotid atherosclerotic plaques.
Huihui LIU ; Yongjun CAO ; Tong TONG ; Jijun SHI ; Yanlin ZHANG ; Yaping YANG ; Chunfeng LIU ;
Chinese Medical Journal 2015;128(1):69-74
BACKGROUNDAutophagy has been found to be involved in animal and cell models of atherosclerosis, but to date, it lacks general observation in human atherosclerotic plaques. Here, we investigated autophagy in smooth muscle cells (SMCs), endothelial cells (ECs), and macrophages in human atherosclerotic plaques via transmission electron microscopy (TEM), western blotting, and immunohistochemistry analysis.
METHODSThe histopathologic morphology of these plaques was observed via hematoxylin and eosin staining. The ultrastructural morphology of the SMCs, ECs, and macrophages in these plaques was observed via TEM. The localization of microtubule-associated protein 1 light chain 3 (MAP1-LC3), a relatively special maker of autophagy, in plaques was observed by double fluorescent immunochemistry and western blotting.
RESULTSAll of these human atherosclerotic plaques were considered advanced and unstable in histologically observation. By double fluorescent immunochemistry, the expression of LC3-II increased in the SMCs of the fibrous cap, the macrophages, and the microvascular ECs of the plaque shoulders. The protein level of LC3-II by western blotting significantly increased in plaques compared with normal controls. In addition, TEM observation of plaques revealed certain features of autophagy in SMCs, ECs, and macrophages including the formation of myelin figures, vacuolization, and the accumulation of inclusions in the cytosol. These results indicate that autophagy is activated in SMCs, ECs, and macrophages in human advanced atherosclerotic plaques.
CONCLUSIONSOur study is to demonstrate the existence of autophagy in human atherosclerotic plaques by different methods, which may contribute to the development of pharmacological approaches to stabilize vulnerable and rupture-prone lesions.
Atherosclerosis ; metabolism ; physiopathology ; Autophagy ; physiology ; Endothelial Cells ; pathology ; Humans ; In Vitro Techniques ; Microscopy, Electron, Transmission ; Microtubule-Associated Proteins ; metabolism ; Myocytes, Smooth Muscle ; pathology ; Plaque, Atherosclerotic ; metabolism ; physiopathology ; ultrastructure
5.The function and meaning of receptor activator of NF-κB ligand in arterial calcification.
Bin NIE ; Shao-qiong ZHOU ; Xin FANG ; Shao-ying ZHANG ; Si-ming GUAN
Journal of Huazhong University of Science and Technology (Medical Sciences) 2015;35(5):666-671
Osteoclast-like cells are known to inhibit arterial calcification. Receptor activator of NF-κB ligand (RANKL) is likely to act as an inducer of osteoclast-like cell differentiation. However, several studies have shown that RANKL promotes arterial calcification rather than inhibiting arterial calcification. The present study was conducted in order to investigate and elucidate this paradox. Firstly, RANKL was added into the media, and the monocyte precursor cells were cultured. Morphological observation and Tartrate resistant acid phosphatase (TRAP) staining were used to assess whether RANKL could induce the monocyte precursor cells to differentiate into osteoclast-like cells. During arterial calcification, in vivo and in vitro expression of RANKL and its inhibitor, osteoprotegerin (OPG), was detected by real-time PCR. The extent of osteoclast-like cell differentiation was also assessed. It was found RANKL could induce osteoclast-like cell differentiation. There was no in vivo or in vitro expression of osteoclast-like cells in the early stage of calcification. At that time, the ratio of RANKL to OPG was very low. In the late stage of calcification, a small amount of osteoclast-like cell expression coincided with a relatively high ratio of RANKL to OPG. According to the results, the ratio of RANKL to OPG was very low during most of the arterial calcification period. This made it possible for OPG to completely inhibit RANKL-induced osteoclast-like cell differentiation. This likely explains why RANKL had the ability to induce osteoclast-like cell differentiation but acted as a promoter of calcification instead.
Acid Phosphatase
;
genetics
;
metabolism
;
Animals
;
Aorta
;
drug effects
;
metabolism
;
pathology
;
Cell Differentiation
;
Coculture Techniques
;
Gene Expression Regulation
;
Isoenzymes
;
genetics
;
metabolism
;
Male
;
Monocytes
;
cytology
;
drug effects
;
metabolism
;
Myocytes, Smooth Muscle
;
drug effects
;
metabolism
;
pathology
;
Osteoclasts
;
drug effects
;
metabolism
;
pathology
;
Osteoprotegerin
;
genetics
;
metabolism
;
RANK Ligand
;
genetics
;
metabolism
;
pharmacology
;
Rats
;
Rats, Sprague-Dawley
;
Signal Transduction
;
Tartrate-Resistant Acid Phosphatase
;
Vascular Calcification
;
genetics
;
metabolism
;
pathology
6.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
7.Inhibitory effects of roscovitine on proliferation and migration of vascular smooth muscle cells in vitro.
Shuang-shuang ZHANG ; Wei WANG ; Chong-qiang ZHAO ; Min-jie XIE ; Wen-yu LI ; Xiang-li YANG ; Jia-gao LV
Journal of Huazhong University of Science and Technology (Medical Sciences) 2014;34(6):791-795
Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) are the major cause of in-stent restenosis (ISR). Intervention proliferation and migration of VSMCs is an important strategy for antirestenotic therapy. Roscovitine, a second-generation cyclin-dependent kinase inhibitor, can inhibit cell cycle of multiple cell types. We studied the effects of roscovitine on cell cycle distribution, proliferation and migration of VSMCs in vitro by flow cytometry, BrdU incorporation and wound healing assay, respectively. Our results showed that roscovitine increased the proportion of G0/G1 phase cells after 12 h (69.57±3.65 vs. 92.50±1.68, P=0.000), 24 h (80.87±2.24 vs. 90.25±0.79, P=0.000) and 48 h (88.08±3.86 vs. 88.87±2.43, P=0.427) as compared with control group. Roscovitine inhibited proliferation and migration of VSMCs in a concentration-dependent way. With the increase of concentration, roscovitine showed increased capacity for growth and migration inhibition. Roscovitine (30 μmol/L) led to an almost complete VSMCs growth and migration arrest. Combined with its low toxicity and selective inhibition to ISR-VSMCs, roscovitine may be a potential drug in the treatment of vascular stenosis diseases and particularly useful in the prevention and treatment of ISR.
Animals
;
Cell Cycle
;
drug effects
;
Cell Line
;
Cell Movement
;
drug effects
;
Graft Occlusion, Vascular
;
drug therapy
;
metabolism
;
pathology
;
Muscle, Smooth, Vascular
;
metabolism
;
pathology
;
Myocytes, Smooth Muscle
;
metabolism
;
pathology
;
Protein Kinase Inhibitors
;
pharmacology
;
Purines
;
pharmacology
;
Rats
8.Effect of compound Danshen dripping pills combined with atorvastatin on restenosis after angioplasty in rabbits.
Jieli SONG ; Jinpei ZENG ; Yongxia ZHANG ; Pengfei LI ; Lihong ZHANG ; Cibin CHEN
Journal of Southern Medical University 2014;34(9):1337-1341
OBJECTIVETo study the effect of compound Danshen dripping pills and atorvastatin on restenosis after abdominal aorta angioplasty in rabbits.
METHODSRabbit models of abdominal aorta restenosis after angioplasty were established and treated with saline (group A), compound Danshen dripping pills (group B), atorvastatin (group C), or compound Danshen dripping pills plus atorvastatin (group D). HE staining was used to determine the thickness of arterial intimal hyperplasia and assess the morphological changes of the narrowed artery. Immunohistochemistry was employed to detect the expression of nuclear factor-κB (NF-κB) and monocyte chemoattractant protein-1 (MCP-1).
RESULTSCompared with group A, the 3 treatment groups showed significant increased vascular cavity area and reduced intimal area and percentage of intimal hyperplasia (P<0.05). The vascular cavity area, intimal area and percentage of intimal hyperplasia levels differed significantly between group D and groups B and C (P<0.05). Immunohistochemistry showed a significant reduction of the expression rate of NF-κB and MCP-1 in the 3 treatment groups compared with group A (P<0.05), and the reduction was especially obvious in group D (P<0.05).
CONCLUTIONSCompound danshen dripping pills combined with atorvastatin produces better effects than the drugs used alone in inhibiting vascular smooth muscle cell proliferation in rabbits after abdominal aorta angioplasty possibly due to a decreased expression of MCP-1 as a result of NF-κB inhibition.
Angioplasty ; Animals ; Aorta ; pathology ; Atorvastatin Calcium ; Cell Proliferation ; Chemokine CCL2 ; metabolism ; Drugs, Chinese Herbal ; pharmacology ; Heptanoic Acids ; pharmacology ; Hyperplasia ; Myocytes, Smooth Muscle ; drug effects ; NF-kappa B ; metabolism ; Phenanthrolines ; Pyrroles ; pharmacology ; Rabbits ; Salvia miltiorrhiza ; chemistry ; Tunica Intima
9.Exogenous hydrogen sulfide reduces vascular aging in D-galactose-induced subacute aging rats.
Wei-Li QIAO ; Department of PATHOLOGY ; Wen-Xue YANG ; Lei LIU ; Yue SHI ; Jie CUI ; Hong LIU ; Chang-Dong YAN
Acta Physiologica Sinica 2014;66(3):276-282
The present study was aimed to observe the protective effect of exogenous hydrogen sulfide (H₂S) on vascular structural and functional changes of aorta in D-galactose-induced subacute aging rats. Adult male SD rats were randomly divided to five groups: the vehicle group, the D-galactose (D-gal) group, and the three NaHS groups treated with low (1 μmol·kg⁻¹·d⁻¹), middle (10 μmol·kg⁻¹·d⁻¹) or high (100 μmol·kg⁻¹·d⁻¹) dose of NaHS respectively. The D-gal group rats were given subcutaneously injection of 125 mg/kg D-gal per day for eight weeks to induce subacute aging model. In the NaHS group, D-gal was administered as above but with NaHS intraperitoneally injected at a dosage of 1, 10, 100 μmol·kg⁻¹·d⁻¹ respectively. Equivalent volumes of saline were administered per day for eight weeks in vehicle group. Morphological changes of aorta were observed by HE and Masson staining. The level of H₂S in serum, the activity of superoxide dismutase (SOD) and the content of malondialdehyde (MDA), as well as anti-superoxide anions in vascular tissue were determined by spectrophotometry. Angiotensin II (AngII) levels in plasma were measured using competitive enzyme immunoassay. The expression of angiotensin II type 1 receptor (AT1R) in aorta was determined by Western blot. The results showed that the aging aortic morphologic changes in model rats were ameliorated in NaHS groups. Decreased vascular endothelial exfoliative cells and vascular smooth muscle cell (SMC) proliferation were shown in NaHS groups by HE staining. Masson staining analysis showed reduced relative contents of collagen fibers (P < 0.05) and SMC (P < 0.05) in NaHS groups. Compared to vehicle group, serum concentration of H₂S in D-gal group was decreased, while it was increased in NaHS groups after treatment with NaHS (P < 0.05). In the D-gal group, the concentration of AngII in plasma was significantly increased compared with that in vehicle group, while it was decreased in NaHS groups (P < 0.05). Moreover, levels of vascular tissue anti-superoxide anion and the activity of SOD were obviously higher, MDA was significantly lower in all NaHS treated groups than those in the D-gal group respectively (P < 0.05). Western blot analysis showed that the expression of AT1R was increased in D-gal group compared with that in vehicle group, while it was decreased after treatment with NaHS compared with that in D-gal group (P < 0.05). These results suggest that exogenous H₂S can ameliorate the age-related changes of aortic morphology, decrease the concentration of AngII in plasma, down-regulate the expression of AT1R in vascular tissue, and mitigate the level of oxidative stress. These changes delay the vascular aging in aging rats ultimately.
Aging
;
drug effects
;
Angiotensin II
;
metabolism
;
Animals
;
Aorta
;
pathology
;
Cell Proliferation
;
Endothelial Cells
;
metabolism
;
Galactose
;
adverse effects
;
Hydrogen Sulfide
;
pharmacology
;
Male
;
Malondialdehyde
;
metabolism
;
Myocytes, Smooth Muscle
;
metabolism
;
Oxidative Stress
;
Rats
;
Rats, Sprague-Dawley
;
Receptor, Angiotensin, Type 2
;
metabolism
;
Sulfides
;
pharmacology
;
Superoxide Dismutase
;
metabolism
10.PPARgamma modulates vascular smooth muscle cell phenotype via a protein kinase G-dependent pathway and reduces neointimal hyperplasia after vascular injury.
Han Mo YANG ; Baek Kyung KIM ; Ju Young KIM ; Yoo Wook KWON ; Sooryeonhwa JIN ; Joo Eun LEE ; Hyun Jai CHO ; Hae Young LEE ; Hyun Jae KANG ; Byung Hee OH ; Young Bae PARK ; Hyo Soo KIM
Experimental & Molecular Medicine 2013;45(11):e65-
Vascular smooth muscle cells (VSMCs) undergo phenotypic changes in response to vascular injury such as angioplasty. Protein kinase G (PKG) has an important role in the process of VSMC phenotype switching. In this study, we examined whether rosiglitazone, a peroxisome proliferator-activated receptor (PPAR)-gamma agonist, could modulate VSMC phenotype through the PKG pathway to reduce neointimal hyperplasia after angioplasty. In vitro experiments showed that rosiglitazone inhibited the phenotype change of VSMCs from a contractile to a synthetic form. The platelet-derived growth factor (PDGF)-induced reduction of PKG level was reversed by rosiglitazone treatment, resulting in increased PKG activity. This increased activity of PKG resulted in phosphorylation of vasodilator-stimulated phosphoprotein at serine 239, leading to inhibited proliferation of VSMCs. Interestingly, rosiglitazone did not change the level of nitric oxide (NO) or cyclic guanosine monophosphate (cGMP), which are upstream of PKG, suggesting that rosiglitazone influences PKG itself. Chromatin immunoprecipitation assays for the PKG promoter showed that the activation of PKG by rosiglitazone was mediated by the increased binding of Sp1 on the promoter region of PKG. In vivo experiments showed that rosiglitazone significantly inhibited neointimal formation after balloon injury. Immunohistochemistry staining for calponin and thrombospondin showed that this effect of rosiglitazone was mediated by modulating VSMC phenotype. Our findings demonstrate that rosiglitazone is a potent modulator of VSMC phenotype, which is regulated by PKG. This activation of PKG by rosiglitazone results in reduced neointimal hyperplasia after angioplasty. These results provide important mechanistic insight into the cardiovascular-protective effect of PPARgamma.
Animals
;
Aorta/injuries/metabolism/*pathology
;
Calcium-Binding Proteins/genetics/metabolism
;
Cell Proliferation
;
Cyclic GMP/metabolism
;
Cyclic GMP-Dependent Protein Kinases/genetics/*metabolism
;
Hyperplasia/metabolism
;
Microfilament Proteins/genetics/metabolism
;
Muscle, Smooth, Vascular/metabolism/pathology
;
Myocytes, Smooth Muscle/drug effects/*metabolism
;
Nitric Oxide/metabolism
;
PPAR gamma/agonists/*metabolism
;
Promoter Regions, Genetic
;
Rats
;
Rats, Sprague-Dawley
;
Sp1 Transcription Factor/metabolism
;
Thiazolidinediones/pharmacology
;
Thrombospondins/genetics/metabolism
;
Tunica Intima/metabolism/*pathology
;
Vascular System Injuries/*metabolism/pathology

Result Analysis
Print
Save
E-mail