1
|
Gou Y, Zhao A, Qin T, Yang B. Identification of the Neointimal Hyperplasia-Related LncRNA-mRNA-Immune Cell Regulatory Network in a Rat Carotid Artery Balloon Injury Model. Int Heart J 2024; 65:945-955. [PMID: 39261031 DOI: 10.1536/ihj.24-062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Excessive neointimal hyperplasia (NIH) of coronary vessels in patients is the main cause of restenosis (RS) after percutaneous coronary intervention (PCI). This study aimed to identify the regulatory genes related to NIH in a rat carotid artery balloon injury model.We established a rat model and performed RNA sequencing to identify differentially expressed long non-coding RNAs (DElncRNAs) and differentially expressed message RNAs (DEmRNAs). Immune cells were analyzed using a murine Microenvironment Cell Population counter. The Pearson correlation between DEmRNAs, DElncRNAs, and immune cells was analyzed, followed by function enrichment analysis. Core DEmRNA was identified using Cytoscape. Next, a core lncRNAs-mRNAs-immune cell regulatory network was constructed. NIH-related gene sets from the Gene Expression Omnibus and GeneCards databases were used for validation.A total of 2,165 DEmRNAs and 705 DElncRNAs were identified in rat carotid artery tissue. Four key immune cells were screened out, including mast cells, vessels, endothelial cells, and fibroblasts. Based on the Pearson correlation between DEmRNAs, DElncRNAs and 4 key immune cells, 246 DEmRNAs and 93 DElncRNAs were obtained. DEmRNAs that interact with lncRNAs were mainly involved in the cell cycle, MAPK signaling pathway, and PI3K-Akt signaling pathway. A core lncRNA-mRNA-immune cell regulatory network was constructed, including 9 mRNAs, 4 lncRNAs, and fibroblasts. External datasets validation confirmed the significant correlation of both these mRNAs and lncRNAs with NIH.In this study, an lncRNA-mRNA-immune cell regulatory network related to NIH was constructed, which provided clues for exploring the potential mechanism of RS in cardiovascular diseases.
Collapse
Affiliation(s)
- Yuan Gou
- Department of Vascular Surgery, Jining Medical University Affiliated Jining No. 1 People's Hospital
| | - Anli Zhao
- Department of Cardiovascular Medicine, Affiliated Hospital of Jining Medical University
| | - Tao Qin
- Department of Vascular Surgery, Jining Medical University Affiliated Jining No. 1 People's Hospital
| | - Bin Yang
- Department of Vascular Surgery, Jining No. 1 People's Hospital
| |
Collapse
|
2
|
Kusakabe Y, Matsumoto K, Tsuyuki T, Hayashi Y, Watanabe H. Baicalin target protein, Annexin A2, is a target of new antitumor drugs. Sci Rep 2024; 14:21814. [PMID: 39294172 PMCID: PMC11410801 DOI: 10.1038/s41598-024-68528-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 07/24/2024] [Indexed: 09/20/2024] Open
Abstract
Baicalin is a flavonoid extracted from Scutellaria baicalensis Georgi. As it has significant antitumor and apoptosis-inducing effects, baicalin may be useful as a lead compound in new antitumor drug development. However, as the pharmacological actions of baicalin have yet to be elucidated, we isolated its target protein, which was successfully identified as Annexin A2. Annexin A2 forms a heterotetramer with S100A10 protein, which plays an important role in the plasminogen activator system. The heterotetramer bound to tissue plasminogen activator (tPA) activates the conversion of plasminogen to plasmin and promotes the expression of STAT-3 and NF-κB, which are target genes involved in the development of cancer. Moreover, NF-κB and STAT-3 induce the expression of cell inhibitors of apoptotic proteins and inhibit apoptosis. To examine whether these antitumor and apoptosis-inducing effects of baicalin are mediated by Annexin A2, we prepared Annexin A2 knockdown HepG2 cells. We compared mRNA expression by RT-qPCR and apoptosis by caspase-3 activity assays in Annexin A2 knockdown HepG2 cells. The results showed that the antitumor and apoptosis-inducing effects of baicalin are mediated by Annexin A2. The results of this study suggest that agents capable of inhibiting Annexin A2 may be useful candidates for the development of novel antitumor agents.
Collapse
Affiliation(s)
| | | | | | | | - Hideaki Watanabe
- Department of Dermatology, Showa University Northern Yokohama Hospital, Chigasakichuo, Tsuduki-ku, Yokohama City, Kanagawa, Japan.
| |
Collapse
|
3
|
Si L, Lai Y. Pharmacological mechanisms by which baicalin ameliorates cardiovascular disease. Front Pharmacol 2024; 15:1415971. [PMID: 39185317 PMCID: PMC11341428 DOI: 10.3389/fphar.2024.1415971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 07/29/2024] [Indexed: 08/27/2024] Open
Abstract
Baicalin is a flavonoid glycoside obtained from the dried root of Scutellaria baicalensis Georgi, which belongs to the Labiatae family. Accumulating evidence indicates that baicalin has favorable therapeutic effects on cardiovascular diseases. Previous studies have revealed the therapeutic effects of baicalin on atherosclerosis, myocardial ischemia/reperfusion injury, hypertension, and heart failure through anti-inflammatory, antioxidant, and lipid metabolism mechanisms. In recent years, some new ideas related to baicalin in ferroptosis, coagulation and fibrinolytic systems have been proposed, and new progress has been made in understanding the mechanism by which baicalin protects cardiomyocytes. However, many relevant underlying mechanisms remain unexplained, and much experimental data is lacking. Therefore, further research is needed to determine these mechanisms. In this review, we summarize the mechanisms of baicalin, which include its anti-inflammatory and antioxidant effects; inhibition of endothelial cell apoptosis; modulation of innate immunity; suppression of vascular smooth muscle cells proliferation, migration, and contraction; regulation of coagulation and fibrinolytic systems; inhibition of myocardial hypertrophy; prevention of myocardial fibrosis; and anti-apoptotic effects on cardiomyocytes.
Collapse
Affiliation(s)
- Lujia Si
- Acupunture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Lai
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
4
|
Wang H, Zhang L, Liu H, Yang Y, Lu W, Cao X, Yang X, Qin Q, Song R, Feng D, Wang S, Bai T, He J. PDZK1 confers sensitivity to sunitinib in clear cell renal cell carcinoma by suppressing the PDGFR-β pathway. Br J Cancer 2024; 131:347-360. [PMID: 38822145 PMCID: PMC11263541 DOI: 10.1038/s41416-024-02725-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND Sunitinib has emerged as the primary treatment for advanced or metastatic clear cell renal cell carcinoma (ccRCC) due to its significant improvement in patients' average survival time. However, drug resistance and adverse effects of sunitinib pose challenges to its clinical benefits. METHODS The differentially expressed genes (DEGs) associated with sunitinib sensitivity and resistance in ccRCC were investigated. Cell counting kit-8, plate colony formation, flow cytometry and subcutaneous xenograft tumor model assays were employed to explore the effects of PDZK1 on ccRCC. Further research on the molecular mechanism was conducted through western blot, co-immunoprecipitation, immunofluorescence co-localization and immunohistochemical staining. RESULTS We elucidated that PDZK1 is significantly downregulated in sunitinib-resistant ccRCC specimens, and PDZK1 negatively regulates the phosphorylation of PDGFR-β and the activation of its downstream pathways through interaction with PDGFR-β. The dysregulated low levels of PDZK1 contribute to inadequate inhibition of cell proliferation, tumor growth, and insensitivity to sunitinib treatment. Notably, our preclinical investigations showed that miR-15b antagomirs enhance sunitinib cytotoxic effects against ccRCC cells by upregulating PDZK1 levels, suggesting their potential in overcoming sunitinib resistance. CONCLUSIONS Our findings establish the miR-15b/PDZK1/PDGFR-β axis as a promising therapeutic target and a novel predictor for ccRCC patients' response to sunitinib treatment.
Collapse
MESH Headings
- Sunitinib/pharmacology
- Sunitinib/therapeutic use
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/metabolism
- Humans
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Receptor, Platelet-Derived Growth Factor beta/genetics
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/pathology
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Animals
- Drug Resistance, Neoplasm/genetics
- Mice
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Xenograft Model Antitumor Assays
- MicroRNAs/genetics
- Signal Transduction/drug effects
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Male
- Mice, Nude
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
Collapse
Affiliation(s)
- Haibo Wang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, People's Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Lijie Zhang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Hua Liu
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
| | - Yumeng Yang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
| | - Wenxiu Lu
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
| | - Xuedi Cao
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
| | - Xiaomei Yang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Qiong Qin
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Ran Song
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Duiping Feng
- Department of Interventional Radiology, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Songlin Wang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, People's Republic of China
- Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy and Tooth Regeneration, School of Stomatology, Capital Medical University, Beijing, People's Republic of China
| | - Tao Bai
- Department of Pathology, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.
| | - Junqi He
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China.
| |
Collapse
|
5
|
Cianciosi D, Forbes-Hernandez T, Armas Diaz Y, Elexpuru-Zabaleta M, Quiles JL, Battino M, Giampieri F. Manuka honey's anti-metastatic impact on colon cancer stem-like cells: unveiling its effects on epithelial-mesenchymal transition, angiogenesis and telomere length. Food Funct 2024; 15:7200-7213. [PMID: 38896046 DOI: 10.1039/d4fo00943f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Colorectal cancer often leads to metastasis, with cancer stem cells (CSCs) playing a pivotal role in this process. Two closely linked mechanisms, epithelial-mesenchymal transition and angiogenesis, contribute to metastasis and recent research has also highlighted the impact of telomere replication on this harmful tumor progression. Standard chemotherapy alone can inadvertently promote drug-resistant CSCs, posing a challenge. Combining chemotherapy with other compounds, including natural ones, shows promise in enhancing effectiveness while minimizing side effects. This study investigated the anti-metastatic potential of Manuka honey, both alone and in combination with 5-fluorouracil, using a 3D model of colonospheres enriched with CSC-like cells. In summary, it was observed that the treatment reduced migration ability by downregulating the transcription factors Slug, Snail, and Twist, which are key players in epithelial-mesenchymal transition. Additionally, Manuka honey downregulated pro-angiogenic factors and shortened CSC telomeres by downregulating c-Myc - demonstrating an effective anti-metastatic potential. This study suggests new research opportunities for studying the impact of natural compounds when combined with pharmaceuticals, with the potential to enhance effectiveness and reduce side effects.
Collapse
Affiliation(s)
- Danila Cianciosi
- Department of Clinical Sciences, Polytechnic University of Marche, Via Pietro Ranieri 65, Ancona, 60131, Italy.
| | - Tamara Forbes-Hernandez
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Centre, University of Granada, Armilla, 18016, Spain
| | - Yasmany Armas Diaz
- Department of Clinical Sciences, Polytechnic University of Marche, Via Pietro Ranieri 65, Ancona, 60131, Italy.
| | - Maria Elexpuru-Zabaleta
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, Santander, 39011, Spain
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods, Polytechnic University of Marche, Italy, Universidad Europea del Atlántico Spain and Jiangsu University, China
| | - José L Quiles
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Centre, University of Granada, Armilla, 18016, Spain
| | - Maurizio Battino
- Department of Clinical Sciences, Polytechnic University of Marche, Via Pietro Ranieri 65, Ancona, 60131, Italy.
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, Santander, 39011, Spain
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods, Polytechnic University of Marche, Italy, Universidad Europea del Atlántico Spain and Jiangsu University, China
- International Joint Research Laboratory of Intelligent Agriculture and Agri-Products Processing, Jiangsu University, Zhenjiang, 212013, China
| | - Francesca Giampieri
- Department of Clinical Sciences, Polytechnic University of Marche, Via Pietro Ranieri 65, Ancona, 60131, Italy.
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, Santander, 39011, Spain
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods, Polytechnic University of Marche, Italy, Universidad Europea del Atlántico Spain and Jiangsu University, China
- International Joint Research Laboratory of Intelligent Agriculture and Agri-Products Processing, Jiangsu University, Zhenjiang, 212013, China
| |
Collapse
|
6
|
Hamdin CD, Wu ML, Chen CM, Ho YC, Jiang WC, Gung PY, Ho HH, Chuang HC, Tan TH, Yet SF. Dual-Specificity Phosphatase 6 Deficiency Attenuates Arterial-Injury-Induced Intimal Hyperplasia in Mice. Int J Mol Sci 2023; 24:17136. [PMID: 38138967 PMCID: PMC10742470 DOI: 10.3390/ijms242417136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/29/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023] Open
Abstract
In response to injury, vascular smooth muscle cells (VSMCs) of the arterial wall dedifferentiate into a proliferative and migratory phenotype, leading to intimal hyperplasia. The ERK1/2 pathway participates in cellular proliferation and migration, while dual-specificity phosphatase 6 (DUSP6, also named MKP3) can dephosphorylate activated ERK1/2. We showed that DUSP6 was expressed in low baseline levels in normal arteries; however, arterial injury significantly increased DUSP6 levels in the vessel wall. Compared with wild-type mice, Dusp6-deficient mice had smaller neointima. In vitro, IL-1β induced DUSP6 expression and increased VSMC proliferation and migration. Lack of DUSP6 reduced IL-1β-induced VSMC proliferation and migration. DUSP6 deficiency did not affect IL-1β-stimulated ERK1/2 activation. Instead, ERK1/2 inhibitor U0126 prevented DUSP6 induction by IL-1β, indicating that ERK1/2 functions upstream of DUSP6 to regulate DUSP6 expression in VSMCs rather than downstream as a DUSP6 substrate. IL-1β decreased the levels of cell cycle inhibitor p27 and cell-cell adhesion molecule N-cadherin in VSMCs, whereas lack of DUSP6 maintained their high levels, revealing novel functions of DUSP6 in regulating these two molecules. Taken together, our results indicate that lack of DUSP6 attenuated neointima formation following arterial injury by reducing VSMC proliferation and migration, which were likely mediated via maintaining p27 and N-cadherin levels.
Collapse
Affiliation(s)
- Candra D. Hamdin
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 350401, Taiwan; (C.D.H.); (P.-Y.G.); (H.-H.H.)
- National Health Research Institutes and Department of Life Sciences, National Central University Joint Ph.D. Program in Biomedicine, Zhongli District, Taoyuan 320317, Taiwan
| | - Meng-Ling Wu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (M.-L.W.); (Y.-C.H.)
| | - Chen-Mei Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 350401, Taiwan; (C.D.H.); (P.-Y.G.); (H.-H.H.)
| | - Yen-Chun Ho
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (M.-L.W.); (Y.-C.H.)
| | - Wei-Cheng Jiang
- Department of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
| | - Pei-Yu Gung
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 350401, Taiwan; (C.D.H.); (P.-Y.G.); (H.-H.H.)
| | - Hua-Hui Ho
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 350401, Taiwan; (C.D.H.); (P.-Y.G.); (H.-H.H.)
| | - Huai-Chia Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan 350401, Taiwan; (H.-C.C.); (T.-H.T.)
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, Zhunan 350401, Taiwan; (H.-C.C.); (T.-H.T.)
| | - Shaw-Fang Yet
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 350401, Taiwan; (C.D.H.); (P.-Y.G.); (H.-H.H.)
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404328, Taiwan
| |
Collapse
|
7
|
Yang X, Yang Y, Liu K, Zhang C. Traditional Chinese medicine monomers: Targeting pulmonary artery smooth muscle cells proliferation to treat pulmonary hypertension. Heliyon 2023; 9:e14916. [PMID: 37128338 PMCID: PMC10147991 DOI: 10.1016/j.heliyon.2023.e14916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 02/01/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Pulmonary hypertension (PH) is a complex multifactorial disease characterized by increased pulmonary vascular resistance and pulmonary vascular remodeling (PVR), with high morbidity, disability, and mortality. The abnormal proliferation of pulmonary artery smooth muscle cells (PASMCs) is the main pathological change causing PVR. At present, clinical treatment drugs for PH are limited, which can only improve symptoms and reduce hospitalization but cannot delay disease progression and reduce survival rate. In recent years, numerous studies have shown that traditional Chinese medicine monomers (TCMs) inhibit excessive proliferation of PASMCs resulting in alleviating PVR through multiple channels and multiple targets, which has attracted more and more attention in the treatment of PH. In this paper, the experimental evidence of inhibiting PASMCs proliferation by TCMs was summarized to provide some directions for the future development of these mentioned TCMs as anti-PH drugs in clinical.
Collapse
|
8
|
Liu L, Lan X, Chen X, Dai S, Wang Z, Zhao A, Lu L, Huang N, Chen J, Yang P, Liao Y. Multi-functional plant flavonoids regulate pathological microenvironments for vascular stent surface engineering. Acta Biomater 2023; 157:655-669. [PMID: 36436757 DOI: 10.1016/j.actbio.2022.11.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 10/23/2022] [Accepted: 11/16/2022] [Indexed: 11/27/2022]
Abstract
In-stent restenosis (ISR) and late thrombosis, usually caused by excessive smooth muscle cell (SMC) proliferation and delayed endothelial layer repair, respectively, are the main risks for the failure of vascular stent implantation. For years, modification of stents with biomolecules that could selectively inhibit SMC proliferation and support endothelial cell (EC) growth had drawn extensive attention. However, the modulatory effect of these biomolecules faces the impact of oxidative stress, inflammation, and hyperlipidemia of the pathological vascular microenvironment, which is caused by the stent implantation injury and atherosclerosis lesions. Here, we modified stents with a natural and multi-functional flavonoid, baicalin (BCL), using poly-dopamine (PDA) coating technology to combat the harmful impact of the pathological microenvironment. Stent with an appropriate BCL immobilization density (approximately 2.03 μg/cm2) successfully supported ECs growth while inhibited SMC proliferation. Furthermore, baicalin-modified surfaces regulated the oxidative stress, inflammation, and high-lipid of the pathological microenvironment to inhibit endothelial dysfunction and the oxidized low-density lipoprotein (ox-LDL)-induced macrophage foam cells formation. In vivo results showed that baicalin-modified stents exhibited significant anti-ISR, anti-inflammatory, and endothelialization-promoting functions. Our study suggests that the multi-functional baicalin with pathological microenvironment-regulation (PMR) effect has potential use in the surface engineering of cardiovascular devices. STATEMENT OF SIGNIFICANCE: Empowering vascular stents with selective modulation of smooth muscle cells and endothelial cells by surface technology has become an important research direction for stent surface engineering. However, stent coatings that can furthermodulate the pathological microenvironment of blood vessels have been rarely reported. In this study, we constructed a multifunctional coating based on a flavonoid, baicalin, which can selectively modulate vascular wall cells and improve the pathological microenvironment. This study may provide a reference for developing advanced vascular stents.
Collapse
Affiliation(s)
- Luying Liu
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Xiaorong Lan
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou 646000, China
| | - Xiao Chen
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Sheng Dai
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Zhixing Wang
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Ansha Zhao
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Lei Lu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| | - Nan Huang
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Jiang Chen
- Sichuan Key Laboratory for Disease Gene Study, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32 of the West Second Section of First Ring Road, Chengdu 610031, PR China.
| | - Ping Yang
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China.
| | - Yuzhen Liao
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China.
| |
Collapse
|
9
|
Deciphering the Mechanism of Wogonin, a Natural Flavonoid, on the Proliferation of Pulmonary Arterial Smooth Muscle Cells by Integrating Network Pharmacology and In Vitro Validation. Curr Issues Mol Biol 2023; 45:555-570. [PMID: 36661523 PMCID: PMC9858126 DOI: 10.3390/cimb45010037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Wogonin is one of the main active components of Scutellaria baicalensis, which has anti-inflammatory, anti-angiogenesis, and anti-fibrosis effects. Nevertheless, the effect of wogonin on pulmonary hypertension (PH) still lacks systematic research. This study aims to elucidate the potential mechanism of wogonin against PH through network pharmacology and further verify it through biological experiments in pulmonary arterial smooth muscle cells (PASMCs). The potential targets and pathways of wogonin against PH were predicted and analyzed by network pharmacology methods and molecular docking technology. Subsequently, the proliferation of PASMCs was induced by platelet-derived growth factor-BB (PDGF-BB). Cell viability and migration ability were examined. The method of Western blot was adopted to analyze the changes in related signaling pathways. Forty potential targets related to the effect of wogonin against PH were obtained. Based on the protein-protein interaction (PPI) network, gene-ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) enrichment, and molecular docking, it was shown that the effect of wogonin against PH is closely related to the proliferation of PASMCs and the hypoxia-inducible factor-1α (HIF-1α) pathway. A variety of results from biological experiments verified that wogonin can effectively inhibit the proliferation, migration, and phenotypic transformation of PDGF-BB-mediated PASMCs. In addition, the anti-proliferation effect of wogonin may be achieved by regulating HIF-1/ NADPH oxidase 4 (NOX4) pathway.
Collapse
|
10
|
Wang Z, Wang H, Guo C, Yu F, Zhang Y, Qiao L, Zhang H, Zhang C. Role of hsa_circ_0000280 in regulating vascular smooth muscle cell function and attenuating neointimal hyperplasia via ELAVL1. Cell Mol Life Sci 2023; 80:3. [PMID: 36477660 PMCID: PMC9729135 DOI: 10.1007/s00018-022-04602-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 09/25/2022] [Accepted: 10/07/2022] [Indexed: 12/12/2022]
Abstract
The pathological proliferation of cells in vascular smooth muscle underlies neointimal hyperplasia (NIH) development during atherosclerosis. Circular RNAs (circRNAs), which represent novel functional biomarkers and RNA-binding proteins, contribute to multiple cardiovascular diseases; however, their roles in regulating the vascular smooth muscle cell cycle remain unknown. Thus, we aimed to identify the roles of circRNAs in vascular smooth muscle during coronary heart disease (CHD). Through circRNA sequencing of CHD samples and human antigen R (ELAVL1) immunoprecipitation, we identified circRNAs that are associated with CHD and interact with ELAVL1. Our results suggested that the hsa_circ_0000280 associated with CHD inhibits cell proliferation and induces ELAVL1-dependent cell cycle arrest. Gain/loss-of-function experiments and assays in vivo indicated that hsa_circ_0000280 facilitates interactions between ELAVL1 and cyclin-dependent kinase suppressor 1 (CDKN1A) mRNA and stabilization of this complex and leads to cell cycle arrest at the G1/S checkpoint, inhibiting cell proliferation of vascular smooth muscle cells in vitro and NIH in vivo. Importantly, hsa_circ_0000280 reduced neointimal thickness and smooth muscle cell proliferation in vivo. Taken together, these findings reveal a novel pathway in which hsa_circ_0000280 facilitates the regulation of ELAVL1 on CDKN1A mRNA to inhibit NIH. Therefore, measuring and modulating their expression might represent a potential diagnostic or therapeutic strategy for CHD.
Collapse
Affiliation(s)
- Zunzhe Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan City, 250012, Shandong, China
- Department of Geriatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Huating Wang
- Department of Cardiology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, Shandong, China
| | - Chenghu Guo
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan City, 250012, Shandong, China
| | - Fangpu Yu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan City, 250012, Shandong, China
| | - Ya Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan City, 250012, Shandong, China
| | - Lei Qiao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan City, 250012, Shandong, China
| | - Haijun Zhang
- Institute of Vascular Intervention, Medical College of Tongji University, Shanghai, 200072, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan City, 250012, Shandong, China.
| |
Collapse
|
11
|
Li M, Ren C. Exploring the protective mechanism of baicalin in treatment of atherosclerosis using endothelial cells deregulation model and network pharmacology. BMC Complement Med Ther 2022; 22:257. [PMID: 36192741 PMCID: PMC9527735 DOI: 10.1186/s12906-022-03738-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 09/23/2022] [Indexed: 11/21/2022] Open
Abstract
Background Baicalin is a generally available flavonoid with potent biological activity. The present study aimed to assess the underlying mechanism of baicalin in treatment of atherosclerosis (AS) with the help of network pharmacology, molecular docking and experimental validation. Methods The target genes of baicalin and AS were identified from public databases, and the overlapping results were considered to be baicalin-AS targets. Core target genes of baicalin were obtained through the PPI network and validated by a clinical microarray dataset (GSE132651). Human aortic endothelial cells (HAECs) were treated with Lipopolysaccharide (LPS) to construct an endothelial injury model. The expression of NOX4 was examined by real-time qPCR and western blot. Flow cytometry was used to detect intracellular levels of reactive oxygen species (ROS). Furthermore, HAECs were transfected with NOX4-specific siRNA and then co-stimulated with baicalin and LPS to investigate whether NOX4 was involved in the anti-oxidative stress effects of baicalin. Results In this study, baicalin had 45 biological targets against AS. Functional enrichment analysis demonstrated that most targets were involved in oxidative stress. Using the CytoHubba plug-in, we obtained the top 10 genes in the PPI network ranked by the EPC algorithm. Molecular docking and microarray dataset validation indicated that NOX4 may be an essential target of baicalin, and its expression was significantly suppressed in AS samples compared to controls. In endothelial injury model, intervention of HAECs with baicalin increased the expression levels of NOX4 and NOS3 (eNOS), and decreased LPS-induced ROS generation. After inhibition of NOX4, the anti-ROS-generating effect of baicalin was abolished. Conclusion Collectively, we combined network pharmacology and endothelial injury models to investigate the anti-AS mechanism of baicalin. The results demonstrate that baicalin may exert anti-oxidative stress effects by targeting NOX4, providing new mechanisms and insights to baicalin for the treatment of AS. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-022-03738-3.
Collapse
Affiliation(s)
- Mingshuang Li
- grid.452858.6Taizhou Hospital, Shanghai University of Traditional Chinese Medicine, Taizhou, Zhejiang China ,grid.452858.6Taizhou Hospital of Traditional Chinese Medicine, Taizhou, Zhejiang China
| | - Conglin Ren
- grid.452858.6Taizhou Hospital, Shanghai University of Traditional Chinese Medicine, Taizhou, Zhejiang China ,grid.452858.6Taizhou Hospital of Traditional Chinese Medicine, Taizhou, Zhejiang China
| |
Collapse
|
12
|
Liu JX, Huang T, Xie D, Yu Q. Bves maintains vascular smooth muscle cell contractile phenotype and protects against transplant vasculopathy via Dusp1-dependent p38MAPK and ERK1/2 signaling. Atherosclerosis 2022; 357:20-32. [PMID: 36037759 DOI: 10.1016/j.atherosclerosis.2022.08.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/26/2022] [Accepted: 08/10/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND AIMS Vascular smooth muscle cell (VSMC) plasticity is tightly associated with the pathological process of vasculopathy. Blood vessel epicardial substance (Bves) has emerged as an important regulator of intracardiac vasculogenesis and organ homeostasis. However, the involvement and role of Bves in VSMC plasticity and neointimal lesion development remain unclear. METHODS We used an in vivo rat model of graft arteriosclerosis and in vitro PDGF-treated VSMCs and identified the novel VSMC contractile phenotype-related gene Bves using a transcriptomic analysis and literature search. In vitro knockdown and overexpression approaches were used to investigate the mechanisms underlying VSMC phenotypic plasticity. In vivo, VSMC-specific Bves overexpression in rat aortic grafts was generated to assess the physiological function of Bves in neointimal lesion development. RESULTS Here, we found that Bves expression was negatively regulated in aortic allografts in vivo and PDGF-treated VSMCs in vitro. The genetic knockdown of Bves dramatically inhibited, whereas Bves overexpression markedly promoted, the VSMC contractile phenotype. Furthermore, RNA sequencing unraveled a positive correlation between Bves and dual-specificity protein phosphatase 1 (Dusp1) expression in VSMCs. We found that Bves knockdown restrained Dusp1 expression, but enhanced p38MAPK and ERK1/2 activation, resulting in the loss of the VSMC contractile phenotype. In vivo, an analysis of a rat graft model confirmed that VSMC-specific Bves and Dusp1 overexpression in aortic allografts significantly attenuated neointimal lesion formation. CONCLUSIONS Bves maintains the VSMC contractile phenotype through Dusp1-dependent p38MAPK and ERK1/2 signaling, and protects against neointimal formation, underscoring the important role of Bves in preventing transplant vasculopathy.
Collapse
Affiliation(s)
- Jin-Xin Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tong Huang
- The Eight Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Dawei Xie
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qihong Yu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China.
| |
Collapse
|
13
|
Luo Z, Qi B, Sun Y, Chen Y, Lin J, Qin H, Wang N, Shi R, Shang X, Chen S, Chen J. Engineering Bioactive M2 Macrophage-Polarized, Anti-inflammatory, miRNA-Based Liposomes for Functional Muscle Repair: From Exosomal Mechanisms to Biomaterials. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2201957. [PMID: 35802903 DOI: 10.1002/smll.202201957] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/03/2022] [Indexed: 02/05/2023]
Abstract
Severe inflammation and myogenic differentiation disorder are the major obstacles to skeletal muscle healing after injury. MicroRNAs (miRNAs) play an important role as regulatory molecules during the process of muscle healing, but the detailed mechanism of miRNA-mediated intercellular communication between myoblasts and macrophages remains unclear. Here, it is reported that myoblasts secrete miRNAs-enriched exosomes in the inflammatory environment, through which miR-224 is transferred into macrophages to inhibit M2 polarization. Further data demonstrate that WNT-9a may be a direct target of miR-224 for macrophage polarization. In turn, the secretome of M1 macrophages impairs myogenic differentiation and promotes proliferation. Single-cell integration analysis suggests that the elevation of exosome-derived miR-224 is caused by the activation of the key factor E2F1 in myoblasts and demonstrates the RB/E2F1/miR-224/WNT-9a axis. In vivo results show that treatment with antagomir-224 or liposomes containing miR-224 inhibitors suppresses fibrosis and improves muscle recovery. These findings indicate the importance of the crosstalk between myoblasts and macrophages via miRNA-containing exosomes in the regulation of macrophage polarization and myogenic differentiation/proliferation during muscle healing. This study provides a strategy for treating muscle injury through designing an M2 polarization-enabling anti-inflammatory and miRNA-based bioactive material.
Collapse
Affiliation(s)
- Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Beijie Qi
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yaying Sun
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yisheng Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Jinrong Lin
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Haocheng Qin
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai, 200433, P. R. China
| | - Ning Wang
- Department of Ophthalmology, Shanghai 9th People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Runjie Shi
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai ninth people's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Xiliang Shang
- Department of Ophthalmology, Shanghai 9th People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Shiyi Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Jiwu Chen
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200080, P. R. China
| |
Collapse
|
14
|
Zhang D, Cao Y, Liu D, Zhang J, Guo Y. The Etiology and Molecular Mechanism Underlying Smooth Muscle Phenotype Switching in Intimal Hyperplasia of Vein Graft and the Regulatory Role of microRNAs. Front Cardiovasc Med 2022; 9:935054. [PMID: 35966541 PMCID: PMC9365958 DOI: 10.3389/fcvm.2022.935054] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Mounting evidence suggests that the phenotypic transformation of venous smooth muscle cells (SMCs) from differentiated (contractile) to dedifferentiated (proliferative and migratory) phenotypes causes excessive proliferation and further migration to the intima leading to intimal hyperplasia, which represents one of the key pathophysiological mechanisms of vein graft restenosis. In recent years, numerous miRNAs have been identified as specific phenotypic regulators of vascular SMCs (VSMCs), which play a vital role in intimal hyperplasia in vein grafts. The review sought to provide a comprehensive overview of the etiology of intimal hyperplasia, factors affecting the phenotypic transformation of VSMCs in vein graft, and molecular mechanisms of miRNAs involved in SMCs phenotypic modulation in intimal hyperplasia of vein graft reported in recent years.
Collapse
Affiliation(s)
- Dengshen Zhang
- Department of Cardiovascular Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yiran Cao
- Department of Cardiovascular Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Daxing Liu
- Department of Cardiovascular Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jian Zhang
- Department of Cardiovascular Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yingqiang Guo
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Yingqiang Guo,
| |
Collapse
|
15
|
Mechanistic and therapeutic perspectives of baicalin and baicalein on pulmonary hypertension: A comprehensive review. Biomed Pharmacother 2022; 151:113191. [PMID: 35643068 DOI: 10.1016/j.biopha.2022.113191] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/18/2022] [Accepted: 05/22/2022] [Indexed: 11/20/2022] Open
Abstract
Pulmonary hypertension (PH) is a chronic and fatal disease, for which new therapeutic drugs and approaches are needed urgently. Baicalein and baicalin, the active compounds of the traditional Chinese medicine, Scutellaria baicalensis Georgi, exhibit a wide range of pharmacological activities. Numerous studies involving in vitro and in vivo models of PH have revealed that the treatment with baicalin and baicalein may be effective. This review summarizes the potential mechanisms driving the beneficial effects of baicalin and baicalein treatment on PH, including anti-inflammatory response, inhibition of pulmonary smooth muscle cell proliferation and endothelial-to-mesenchymal transformation, stabilization of the extracellular matrix, and mitigation of oxidative stress. The pharmacokinetics of these compounds have also been reviewed. The therapeutic potential of baicalin and baicalein warrants their continued study as natural treatments for PH.
Collapse
|
16
|
Lu LY, Pan N, Huang ZH, Wang JS, Tang YB, Sun HS, Han H, Yang HY, Zhu JZ, Guan YY, Zhang B, Li DZ, Wang GL. CFTR Suppresses Neointimal Formation Through Attenuating Proliferation and Migration of Aortic Smooth Muscle Cells. J Cardiovasc Pharmacol 2022; 79:914-924. [PMID: 35266910 PMCID: PMC9162269 DOI: 10.1097/fjc.0000000000001257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 02/06/2022] [Indexed: 12/14/2022]
Abstract
ABSTRACT Cystic fibrosis transmembrane conductance regulator (CFTR) plays important roles in arterial functions and the fate of cells. To further understand its function in vascular remodeling, we examined whether CFTR directly regulates platelet-derived growth factor-BB (PDGF-BB)-stimulated vascular smooth muscle cells (VSMCs) proliferation and migration, as well as the balloon injury-induced neointimal formation. The CFTR adenoviral gene delivery was used to evaluate the effects of CFTR on neointimal formation in a rat model of carotid artery balloon injury. The roles of CFTR in PDGF-BB-stimulated VSMC proliferation and migration were detected by mitochondrial tetrazolium assay, wound healing assay, transwell chamber method, western blot, and qPCR. We found that CFTR expression was declined in injured rat carotid arteries, while adenoviral overexpression of CFTR in vivo attenuated neointimal formation in carotid arteries. CFTR overexpression inhibited PDGF-BB-induced VSMC proliferation and migration, whereas CFTR silencing caused the opposite results. Mechanistically, CFTR suppressed the phosphorylation of PDGF receptor β, serum and glucocorticoid-inducible kinase 1, JNK, p38 and ERK induced by PDGF-BB, and the increased mRNA expression of matrix metalloproteinase-9 and MMP2 induced by PDGF-BB. In conclusion, our results indicated that CFTR may attenuate neointimal formation by suppressing PDGF-BB-induced activation of serum and glucocorticoid-inducible kinase 1 and the JNK/p38/ERK signaling pathway.
Collapse
Affiliation(s)
- Liu-Yi Lu
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| | - Ni Pan
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| | - Ze-Han Huang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jing-Song Wang
- Vascular surgery department, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, Guangzhou, China; and
| | - Yong-Bo Tang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Hong-Shuo Sun
- Departments of Surgery, Physiology and Pharmacology, Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Hui Han
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Han-Yan Yang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jun-Zhen Zhu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yong-Yuan Guan
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Bin Zhang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Dong-Zhi Li
- Prenatal Diagnostic Center, Guangzhou Women and Children Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Guan-Lei Wang
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
17
|
Evening Primrose Extracts Inhibit PDGF-BB-Induced Vascular Smooth Muscle Cell Proliferation and Migration by Regulating Cell-Cycle-Related Proteins. Curr Issues Mol Biol 2022; 44:1928-1940. [PMID: 35678660 PMCID: PMC9164085 DOI: 10.3390/cimb44050131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 04/23/2022] [Accepted: 04/25/2022] [Indexed: 11/30/2022] Open
Abstract
The proliferation and migration of vascular smooth muscle cells (VSMCs) are important factors in the occurrence of cardiovascular diseases, such as blood flow abnormalities, stroke and atherosclerosis. Evening primrose, known as Oenothera biennis, is a plant native to Korea that exerts physiological activities, such as antioxidant effects, the inhibition of lipid accumulation and the prevention of muscle atrophy. However, the function of evening primrose stem (EVP) in the regulation of VSMC proliferation and migration and the underlying mechanisms have not been identified. In this study, the effect of EVP on the platelet-derived growth factor (PDGF)-induced proliferation and migration of VSMCs was investigated. The results show that PDGF-BB-induced proliferation of VSMCs was inhibited by EVP at concentrations of 25, 50 or 100 μg/mL in a concentration-dependent manner, and a migration assay showed that EVP inhibited cell migration. Cell cycle analysis was performed to confirm the mechanism by which cell proliferation and migration was inhibited. The results indicate that proteins involved in the cell cycle, such as cyclin, CDK and phosphorylated Rb, were downregulated by EVP at concentrations of 100 μg/mL, thereby increasing the proportion of cells in the G0/G1 phase and inhibiting cell cycle progression. In the PDGF receptor (PDGFR) signaling pathway, phosphorylation of the PDGFR was inhibited by EVP at concentrations of 100 μg/mL, and PLCγ phosphorylation was also decreased. The PDGF-BB-induced effect of EVP on the proliferation of VSMCs involved the inhibition of Akt phosphorylation and the reduction in the phosphorylation of MAPK proteins such as ERK, P38 and JNK. In conclusion, the results demonstrate that EVP inhibited PDGF-BB-induced VSMC proliferation and migration by regulating cell-cycle-related proteins.
Collapse
|
18
|
Borkar NA, Ambhore NS, Kalidhindi RSR, Pabelick CM, Prakash YS, Sathish V. Kisspeptins inhibit human airway smooth muscle proliferation. JCI Insight 2022; 7:152762. [PMID: 35420998 DOI: 10.1172/jci.insight.152762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 04/12/2022] [Indexed: 11/17/2022] Open
Abstract
Sex/gender disparity in asthma is recognized, and suggests a modulatory role for sex-steroids, particularly estrogen. However, studies including our own show a dichotomous role for estrogen in airway remodeling, making it unclear whether sex hormones are protective or detrimental in asthma, and suggesting a need to explore mechanisms upstream or independent of estrogen. We hypothesize that Kisspeptin (Kp)/KISS1R signaling serves this role. Airway smooth muscle (ASM) is a key structural cell type that contributes to remodeling in asthma. We explored the role of Kp/KISS1R in regulating ASM proliferation. We report novel data that Kp and KISS1R are expressed in human airways, especially ASM, with lower expression in ASM from females compared to males, and asthmatics showing lowest expression compared to non-asthmatics. Proliferation studies showed that cleaved forms of Kp, particularly Kp-10 mitigates PDGF-induced ASM proliferation. Pharmacological inhibition and shRNA knockdown of KISS1R increased basal ASM proliferation, further amplified by PDGF. The anti-proliferative effect of Kp-10 in ASM was found to be mediated by inhibition of MAPK-ERK-Akt pathways, with altered expression of PCNA, C/EBP-alpha, Ki-67, Cyclin-D1, and Cyclin-E leading to cell-cycle arrest at G0/G1 phase. Overall, we demonstrate the importance of Kp/KISS1R signaling in regulating ASM proliferation and a potentially novel therapeutic avenue to blunt remodeling in asthma.
Collapse
Affiliation(s)
- Niyati A Borkar
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, United States of America
| | - Nilesh Sudhakar Ambhore
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, United States of America
| | | | - Christina M Pabelick
- Department of Anesthesiology and Physiology, Mayo Clinic, Rochester, United States of America
| | - Y S Prakash
- Department of Anesthesiology and Physiology, Mayo Clinic, Rochester, United States of America
| | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, United States of America
| |
Collapse
|
19
|
Cao SH, Chen ZH, Ma RY, Yue L, Jiang HM, Dong LH. Dynamics and Functional Interplay of Nonhistone Lysine Crotonylome and Ubiquitylome in Vascular Smooth Muscle Cell Phenotypic Remodeling. Front Cardiovasc Med 2022; 9:783739. [PMID: 35369347 PMCID: PMC8964401 DOI: 10.3389/fcvm.2022.783739] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundThe crotonylation of histones is discovered of late as one of the post-translational modifications (PTMs) that can regulate gene expression. However, the function of crotonylation on nonhistone proteins in vascular smooth muscle cells (VSMCs) is unclear. Here, we aim to find the cellular characteristics of crotonylated nonhistone proteins and the cross talk with ubiquitinated proteins in VSMC phenotypic remodeling using the modified omics and proteomic analysis.MethodsWe performed the modified omics and proteomic analysis of VSMCs before and after the stimulation with platelet-derived growth factor-BB (PDGF-BB). The crotonylated and ubiquitinated pan-antibody was used to enrich proteins and then subjected to a high-throughput mass spectrometry analysis. The enrichment analysis was performed within differentially modified proteins in regard to GO terms, KEGG, and protein domains.ResultsAs a result, there were 2,138 crotonylation sites in 534 proteins and 1,359 ubiquitination sites corresponding to 657 proteins. These crotonylated proteins detected after PDGF-BB stimulation might be involved in various vital cellular pathways and carry out important functions in VSMCs. Some of them closely took part in significant physiological processes of VSMC phenotypic remodeling, including glycolysis/gluconeogenesis, vascular smooth muscle contraction, and the PI3K-Akt signaling pathway. Furthermore, the KEGG pathway enrichment analysis showed the involvement of ubiquitinated proteins in the physiological processes of VSMC phenotypic remodeling, including glycolysis/gluconeogenesis, vascular smooth muscle contraction, RAS signaling pathway, or the PI3K-Akt signaling pathway. A cross talk analysis showed that there were 199 sites within the 177 proteins modified by crotonylation and ubiquitination simultaneously. Protein–protein interaction (PPI) network analysis indicated that crotonylated and ubiquitinated proteins play an important role in cellular bioprocess commonly and possibly have a synergistic effect.ConclusionIn summary, our bioinformatics analysis shows that the crotonylation and ubiquitination of nonhistone proteins play an essential role in VSMC phenotypic transformation induced by PDGF-BB stimulation. The cross talk between crotonylation and ubiquitination in glycolysis is possibly a novel mechanism during VSMC phenotypic remodeling.
Collapse
|
20
|
Jiang H, Zhao Y, Feng P, Liu Y. Sulfiredoxin-1 Inhibits PDGF-BB-Induced Vascular Smooth Muscle Cell Proliferation and Migration by Enhancing the Activation of Nrf2/ARE Signaling. Int Heart J 2022; 63:113-121. [PMID: 35034915 DOI: 10.1536/ihj.21-213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Sulfiredoxin1 (Srxn1), an endogenous antioxidant protein, is involved in cardiovascular diseases. In this study, we aimed to investigate the role of Srxn1 in VSMCs and its molecular mechanism. The murine vascular smooth muscle cells MOVAS were treated with different doses of platelet-derived growth factor-BB (PDGF-BB); then, Srxn1 expression was detected using reverse transcription-quantitative polymerase chain reaction and western blot analysis. MTT and wound healing assay were used to examine the effect of Srxn1 on MOVAS cell proliferation and migration. Reactive oxygen species (ROS) production, malondialdehyde (MDA) level, and superoxide dismutase (SOD) activity in MOVAS cells were detected using corresponding commercial kits. Moreover, the expression of proliferating cell nuclear antigen (PCNA), matrix metalloproteinase 2 (MMP-2), and nuclear factor erythroid-2-related factor 2 (Nrf2) /antioxidant response element (ARE) signaling-related proteins was detected using western blot analysis. In our study, PDGF-BB dose-dependently increased Srxn1 expression in MOVAS cells, and Srxn1 expression was increased with time dependence in PDGF-BB-treated MOVAS cells. The knockdown of Srxn1 increased PDGF-BB-induced the proliferation, migration, ROS production, MDA level, and the protein expression of PCNA and MMP-2, as well as decreased SOD activity and the expression of Nrf2/ARE signaling-related proteins in PDGF-BB-stimulated MOVAS cells. However, the overexpression of Srxn1 showed the opposite results to those of knockdown of Srxn1. Moreover, the inhibitory effects of Srxn1 overexpression on PDGF-BB induced proliferation, migration, ROS production, and MDA level and the promotion of Srxn1 overexpression on PDGF-BB induced SOD activity were partially reversed by the knockdown of Nrf2. Srxn1 inhibited PDGF-BB-induced proliferation, migration, and oxidative stress through activating Nrf2/ARE signaling.
Collapse
Affiliation(s)
- Haijie Jiang
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University
| | - Yueyan Zhao
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University
| | - Panyang Feng
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University
| | - Yan Liu
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University
| |
Collapse
|
21
|
Hu Z, Guan Y, Hu W, Xu Z, Ishfaq M. An overview of pharmacological activities of baicalin and its aglycone baicalein: New insights into molecular mechanisms and signaling pathways. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:14-26. [PMID: 35656442 PMCID: PMC9118284 DOI: 10.22038/ijbms.2022.60380.13381] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/27/2021] [Indexed: 11/28/2022]
Abstract
The flavonoids, baicalin, and its aglycone baicalein possess multi-fold therapeutic properties and are mainly found in the roots of Oroxylum indicum (L.) Kurz and Scutellaria baicalensis Georgi. These flavonoids have been reported to possess various pharmacological properties, including antibacterial, antiviral, anticancer, anticonvulsant, anti-oxidant, hepatoprotective, and neuroprotective effects. The pharmacological properties of baicalin and baicalein are due to their abilities to scavenge reactive oxygen species (ROS) and interaction with various signaling molecules associated with apoptosis, inflammation, autophagy, cell cycle, mitochondrial dynamics, and cytoprotection. In this review, we summarized the molecular mechanisms underlying the chemopreventive and chemotherapeutic applications of baicalin and baicalein in the treatment of cancer and inflammatory diseases. In addition, the preventive effects of baicalin and baicalein on mitochondrial dynamics and functions were highlighted with a particular emphasis on their anti-oxidative and cytoprotective properties. The current review highlights could be useful for future prospective studies to further improve the pharmacological applications of baicalein and baicalin. These studies should define the threshold for optimal drug exposure, dose optimization and focus on therapeutic drug monitoring, objective disease markers, and baicalin/baicalein drug levels.
Collapse
Affiliation(s)
- Zhihua Hu
- College of Computer Science, Huanggang Normal University, Huanggang 438000, China.,These authors contributed equally to this work
| | - Yurong Guan
- College of Computer Science, Huanggang Normal University, Huanggang 438000, China.,These authors contributed equally to this work
| | - Wanying Hu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, P. R. China
| | - Zhiyong Xu
- Hubei Zhiying Medical Imaging Center, Radiology Department of Huanggang Hospital of Traditional Chinese Medicine, China
| | - Muhammad Ishfaq
- College of Computer Science, Huanggang Normal University, Huanggang 438000, China
| |
Collapse
|
22
|
Liu H, Cheng Y, Chu J, Wu M, Yan M, Wang D, Xie Q, Ali F, Fang Y, Wei L, Yang Y, Shen A, Peng J. Baicalin attenuates angiotensin II-induced blood pressure elevation and modulates MLCK/p-MLC signaling pathway. Biomed Pharmacother 2021; 143:112124. [PMID: 34492423 DOI: 10.1016/j.biopha.2021.112124] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/18/2021] [Accepted: 08/24/2021] [Indexed: 01/05/2023] Open
Abstract
Scutellaria baicalensis Georgi is an extensively used medicinal herb for the treatment of hypertension in traditional Chinese medicine. Baicalin, is an important flavonoid in Scutellaria baicalensis Georgi extracts, which exhibits therapeutic effects on anti-hypertension, but its underlying mechanisms remain to be further explored. Therefore, we investigated the effects and molecular mechanisms of Baicalin on anti-hypertension. In vivo studies revealed that Baicalin treatment significantly attenuated the elevation in blood pressure, the pulse propagation and thickening of the abdominal aortic wall in C57BL/6 mice infused with Angiotensin II (Ang II). Moreover, RNA-sequencing and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses identified 537 differentially expressed transcripts and multiple enriched signaling pathways (including vascular smooth muscle contraction and calcium signaling pathway). Consistently, we found that Baicalin pretreatment significantly alleviated the Ang II induced constriction of abdominal aortic ring, while promoted NE pre-contracted vasodilation of abdominal aortic ring at least partly dependent on L-type calcium channel. In addition, Ang II stimulation significantly increased cell viability and PCNA expression, while were attenuated after Baicalin treatment. Moreover, Baicalin pretreatment attenuated Ang II-induced intracellular Ca2+ release, Angiotensin II type 1 receptor (AT1R) expression and activation of MLCK/p-MLC pathway in vascular smooth muscle cells (VSMCs). The present work further addressed the pharmacological and mechanistic insights on anti-hypertension of Baicalin, which may help better understand the therapeutic effect of Scutellaria baicalensis Georgi on anti-hypertension.
Collapse
MESH Headings
- Angiotensin II
- Animals
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/enzymology
- Aorta, Abdominal/physiopathology
- Blood Pressure/drug effects
- Calcium Signaling/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Disease Models, Animal
- Flavonoids/pharmacology
- Hypertension/chemically induced
- Hypertension/enzymology
- Hypertension/physiopathology
- Hypertension/prevention & control
- Hypoglycemic Agents/pharmacology
- Male
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myosin Light Chains/metabolism
- Myosin-Light-Chain Kinase/metabolism
- Phosphorylation
- Rats, Wistar
- Mice
- Rats
Collapse
Affiliation(s)
- Huixin Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Chen Keji Academic Thought Inheritance Studio, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Ying Cheng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Jianfeng Chu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Chen Keji Academic Thought Inheritance Studio, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Meizhu Wu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Chen Keji Academic Thought Inheritance Studio, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Mengchao Yan
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Chen Keji Academic Thought Inheritance Studio, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Di Wang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Chen Keji Academic Thought Inheritance Studio, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Qiurong Xie
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Chen Keji Academic Thought Inheritance Studio, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Farman Ali
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Chen Keji Academic Thought Inheritance Studio, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Yi Fang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Chen Keji Academic Thought Inheritance Studio, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Lihui Wei
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Chen Keji Academic Thought Inheritance Studio, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Yanyan Yang
- Laboratory Animal Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Aling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Chen Keji Academic Thought Inheritance Studio, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China.
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China; Chen Keji Academic Thought Inheritance Studio, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China.
| |
Collapse
|
23
|
Zhang Y, Ma A, Xi H, Chen N, Wang R, Yang C, Chen J, Lv P, Zheng F, Kang W. Antrodia cinnamomea ameliorates neointimal formation by inhibiting inflammatory cell infiltration through downregulation of adhesion molecule expression in vitro and in vivo. FOOD SCIENCE AND HUMAN WELLNESS 2021. [DOI: 10.1016/j.fshw.2021.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
24
|
BOP1 Knockdown Attenuates Neointimal Hyperplasia by Activating p53 and Inhibiting Nascent Protein Synthesis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5986260. [PMID: 33510838 PMCID: PMC7826231 DOI: 10.1155/2021/5986260] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 11/18/2020] [Accepted: 12/26/2020] [Indexed: 12/12/2022]
Abstract
The rate of ribosome biogenesis plays a vital role in cell cycle progression and proliferation and is strongly connected with coronary restenosis and atherosclerosis. Blocking of proliferation 1 (BOP1) has been found as an evolutionarily conserved gene and a pivotal regulator of ribosome biogenesis and cell proliferation. However, little is known about its role in neointimal formation and its relationship with vascular smooth muscle cell (VSMC) proliferation and migration. The present study mainly explores the effect of BOP1 on VSMCs, the progression of neointimal hyperplasia, and the pathogenic mechanism. The expression of BOP1 was found to be significantly elevated during neointimal formation in human coronary samples and the rat balloon injury model. BOP1 knockdown inspires the nucleolus stress, which subsequently activates the p53-dependent stress response pathway, and inhibits the nascent protein synthesis, which subsequently inhibits the proliferation and migration of VSMCs. Knockdown ribosomal protein L11 (RPL11) by transfecting with siRNA or inhibiting p53 by pifithrin-α (PFT-α) partly reserved the biological effects induced by BOP1 knockdown. The present study revealed that BOP1 deletion attenuates VSMC proliferation and migration by activating the p53-dependent nucleolus stress response pathway and inhibits the synthesis of nascent proteins. BOP1 may become a novel biological target for neointimal hyperplasia.
Collapse
|
25
|
Xu Z, Liu X, Li Y, Gao H, He T, Zhang C, Hao W, Teng X. Shuxuetong injection simultaneously ameliorates dexamethasone-driven vascular calcification and osteoporosis. Exp Ther Med 2021; 21:197. [PMID: 33488806 PMCID: PMC7812579 DOI: 10.3892/etm.2021.9630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 11/12/2020] [Indexed: 12/15/2022] Open
Abstract
Osteoporosis (OP) and vascular calcification (VC) share a number of common risk factors, pathophysiological mechanisms and etiology, which are known as bone-vascular axis. The present study aimed to investigate the effects of Shuxuetong (SXT) injection on VC and osteoporosis. A rat model of VC and osteoporosis was induced by dexamethasone (DEX; 1 mg/kg/day for 4 weeks, intramuscularly). Simultaneously, 0.6 ml/kg/day SXT was intraperitoneally injected. Compared with control rats, DEX induced significantly more VC and OP, as determined by increased calcium deposition and alkaline phosphatase activity in the aorta, disturbed structure, decreased levels of cortical bone thickness and trabecular bone area, and increased apoptosis in the bone. SXT injection ameliorated DEX-induced VC and osteoporosis; furthermore, the osteoblastic differentiation of vascular smooth muscle cells and the activation of endoplasmic reticulum stress in the DEX group was also prevented by SXT injection. Compared with control rats, protein expression levels of sclerostin, a crucial crosslink of the bone-vascular axis, were significantly increased in the aorta and bone of rats with DEX, which was also attenuated by SXT injection. Thus, the present study suggested that SXT injection could ameliorate both VC and OP, and may be mediated by the regulation of sclerostin. The present study may provide the basis a novel strategy for the prevention and treatment of VC and OP, which emerge as side-effects of glucocorticoids.
Collapse
Affiliation(s)
- Zhe Xu
- Department of Anesthesiology, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, Hebei 050011, P.R. China
| | - Xiaoguang Liu
- Department of Anesthesiology, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, Hebei 050011, P.R. China
| | - Yanqing Li
- Department of Gynecology, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, Hebei 050011, P.R. China
| | - Hongliang Gao
- Department of Anesthesiology, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, Hebei 050011, P.R. China
| | - Tao He
- Department of Anesthesiology, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, Hebei 050011, P.R. China
| | - Chunlei Zhang
- Department of Anesthesiology, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, Hebei 050011, P.R. China
| | - Wei Hao
- Department of Anesthesiology, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, Hebei 050011, P.R. China
| | - Xu Teng
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China.,Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
26
|
Hou Y, Pi C, Feng X, Wang Y, Fu S, Zhang X, Zhao L, Wei Y. Antitumor Activity In Vivo and Vitro of New Chiral Derivatives of Baicalin and Induced Apoptosis via the PI3K/Akt Signaling Pathway. Mol Ther Oncolytics 2020; 19:67-78. [PMID: 33072864 PMCID: PMC7533372 DOI: 10.1016/j.omto.2020.08.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/27/2020] [Indexed: 01/07/2023] Open
Abstract
In this study, a pair of chiral baicalin (BA) derivatives were synthesized by combining BA with phenylalanine methyl ester based on molecular docking technology, namely BAD and BAL. Cell cytotoxicity trails showed that the cell growth inhibitory effects of both BAD and BAL were increased by 8- to 12-fold compared with BA on A549 cells. Flow cytometry showed that the apoptotic rates of 50 μg/mL BA, BAD, and BAL to A549 cells for 48 h were 17.94%, 24.32%, and 39.69%, respectively. Western blotting analysis showed that BAD and BAL could promote Bax, caspase-3, and caspase-9 expression and inhibit Bcl-2 expression by inhibiting the expression of p-Akt. The tumor inhibition rates of BA, BAD, and BAL in nude mice of tumor-bearing experiment lasting for 24 days were 35.01%, 53.30%, and 59.35%, respectively. These results in vitro and in vivo showed that BAL had higher antitumor activity than did BAD and BA, which were related to promotion of the apoptosis of tumor cells by inhibiting the expression of p-Akt on PI3K/Akt pathway. This study provides an experimental basis for the development of a new configuration of BA for the treatment of cancer.
Collapse
Affiliation(s)
- Yi Hou
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, No. 3-5, Zhongshan Road, Jiangyang District, Luzhou, Sichuan 646000, P.R. China
| | - Chao Pi
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, No. 3-5, Zhongshan Road, Jiangyang District, Luzhou, Sichuan 646000, P.R. China
| | - Xianhu Feng
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, No. 3-5, Zhongshan Road, Jiangyang District, Luzhou, Sichuan 646000, P.R. China
| | - Yuanyuan Wang
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, No. 3-5, Zhongshan Road, Jiangyang District, Luzhou, Sichuan 646000, P.R. China
| | - Shaozhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, No. 25, Taiping Street, Luzhou, Sichuan 646000, P.R. China
| | - Xiaomei Zhang
- Institute of Chinese Medicine and Pharmaceutical Chemistry, Chongqing Academy of Chinese Materia Medica, No. 34, Nanshan Road, Nanshan Street, Nan’an District, Chongqing 400065, P.R. China
| | - Ling Zhao
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, No. 3-5, Zhongshan Road, Jiangyang District, Luzhou, Sichuan 646000, P.R. China
| | - Yumeng Wei
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, No. 3-5, Zhongshan Road, Jiangyang District, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
27
|
Ma Y, Yu X, Zhang L, Liu J, Shao X, Li YX, Wang YL. Uterine decidual niche modulates the progressive dedifferentiation of spiral artery vascular smooth muscle cells during human pregnancy†. Biol Reprod 2020; 104:624-637. [PMID: 33336235 DOI: 10.1093/biolre/ioaa208] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/13/2019] [Accepted: 11/04/2020] [Indexed: 02/06/2023] Open
Abstract
Uterine spiral artery (SPA) remodeling is a crucial event during pregnancy to provide enough blood supply to maternal-fetal interface and meet the demands of the growing fetus. Along this process, the dynamic change and the fate of spiral artery vascular smooth muscle cells (SPA-VSMCs) have long been debatable. In the present study, we analyzed the cell features of SPA-VSMCs at different stages of vascular remodeling in human early pregnancy, and we demonstrated the progressively morphological change of SPA-VSMCs at un-remodeled (Un-Rem), remodeling, and fully remodeled (Fully-Rem) stages, indicating the extravillous trophoblast (EVT)-independent and EVT-dependent phases of SPA-VSMC dedifferentiation. In vitro experiments in VSMC cell line revealed the efficient roles of decidual stromal cells, decidual natural killer cells (dNK), decidual macrophages, and EVTs in inducing VSMCs dedifferentiation. Importantly, the potential transformation of VSMC toward CD56+ dNKs was displayed by immunofluorescence-DNA in-situ hybridization-proximity ligation and chromatin immunoprecipitation assays for H3K4dime modification in the myosin heavy chain 11 (MYH11) promoter region. The findings clearly illustrate a cascade regulation of the progressive dedifferentiation of SPA-VSMCs by multiple cell types in uterine decidual niche and provide new evidences to reveal the destination of SPA-VSMCs during vascular remodeling.
Collapse
Affiliation(s)
- Yeling Ma
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xin Yu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lanmei Zhang
- Department of Gynecology and Obstetrics, The 306 Hospital of PLA, Beijing, China
| | - Juan Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xuan Shao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yu-Xia Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yan-Ling Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
28
|
Jasemi SV, Khazaei H, Aneva IY, Farzaei MH, Echeverría J. Medicinal Plants and Phytochemicals for the Treatment of Pulmonary Hypertension. Front Pharmacol 2020; 11:145. [PMID: 32226378 PMCID: PMC7080987 DOI: 10.3389/fphar.2020.00145] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/04/2020] [Indexed: 12/18/2022] Open
Abstract
Background Pulmonary hypertension (PH) is a progressive disease that is associated with pulmonary arteries remodeling, right ventricle hypertrophy, right ventricular failure and finally death. The present study aims to review the medicinal plants and phytochemicals used for PH treatment in the period of 1994 – 2019. Methods PubMed, Cochrane and Scopus were searched based on pulmonary hypertension, plant and phytochemical keywords from August 23, 2019. All articles that matched the study based on title and abstract were collected, non-English, repetitive and review studies were excluded. Results Finally 41 studies remained from a total of 1290. The results show that many chemical treatments considered to this disease are ineffective in the long period because they have a controlling role, not a therapeutic one. On the other hand, plants and phytochemicals could be more effective due to their action on many mechanisms that cause the progression of PH. Conclusion Studies have shown that herbs and phytochemicals used to treat PH do their effects from six mechanisms. These mechanisms include antiproliferative, antioxidant, antivascular remodeling, anti-inflammatory, vasodilatory and apoptosis inducing actions. According to the present study, many of these medicinal plants and phytochemicals can have effects that are more therapeutic than chemical drugs if used appropriately.
Collapse
Affiliation(s)
- Seyed Vahid Jasemi
- Department of Internal Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hosna Khazaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ina Yosifova Aneva
- Institute of Biodiversity and Ecosystem Research, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Javier Echeverría
- Departamento de Ciencias del Ambiente, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| |
Collapse
|
29
|
Li L, Liu M, He L, Wang S, Cui S. Baicalin relieves TNF-α-evoked injury in human aortic endothelial cells by up-regulation of miR-145. Phytother Res 2019; 34:836-845. [PMID: 31793706 DOI: 10.1002/ptr.6566] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/04/2019] [Accepted: 11/08/2019] [Indexed: 12/18/2022]
Abstract
Hypertension is recognized to be associated with low-grade inflammation. Baicalin (BAI) is reported to possess various pharmacological including anti-inflammatory activities. This research explored the molecular mechanism by which BAI functions in human aortic endothelial cells (HAECs). HAECs were pretreated with BAI. Cell viability, apoptosis, and expressions of crucial proteins were respectively evaluated using cell counting kit-8 assay, flow cytometry, and western blot. Productions of cytokines were respectively assessed employing quantitative real-time polymerase chain reaction and enzyme-linked immunosorbent assay. Cell transfection was utilized to alter miR-145 expression. The expressions of proteins participated in JNK and p38MAPK pathways were analyzed utilizing western blot. TNF-α inducement successfully evoked inflammatory injury in HAECs, exhibiting as prominently suppressed viability, while facilitated apoptosis and productions of cytokines. However, BAI pretreatment significantly ameliorated TNF-α-triggered inflammatory injuries. Besides, miR-145 expression was markedly inhibited by TNF-α inducement, while notably elevated by BAI pretreatment. Although miR-145 overexpression had no significant influence on apoptosis, miR-145 silence observably reversed BAI pretreatment-evoked protective influences on TNF-α-induced HAECs, as well as the inhibited impacts on the levels of key proteins involved in JNK and p38MAPK pathways. This investigation illustrated that BAI relieved TNF-α-triggered injuries through upregulating miR-145 via suppressing JNK and p38MAPK pathways.
Collapse
Affiliation(s)
- Ling Li
- Department of Hypertension, Henan Provincial People's Hospital, Zhengzhou, China
| | - Min Liu
- Department of Hypertension, Henan Provincial People's Hospital, Zhengzhou, China
| | - Lianman He
- Department of Hypertension, Henan Provincial People's Hospital, Zhengzhou, China
| | - Shanshan Wang
- Department of Hypertension, Henan Provincial People's Hospital, Zhengzhou, China
| | - Shuxian Cui
- Department of Hypertension, Henan Provincial People's Hospital, Zhengzhou, China
| |
Collapse
|
30
|
Liu Y, Hong Z, Chen P, Wang J, Zhou Y, Huang J. Baicalin inhibits growth and induces apoptosis of human osteosarcoma cells by suppressing the AKT pathway. Oncol Lett 2019; 18:3188-3194. [PMID: 31452795 PMCID: PMC6676451 DOI: 10.3892/ol.2019.10617] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 06/06/2019] [Indexed: 02/06/2023] Open
Abstract
Osteosarcoma (OS) is one of the most prevalent types of bone malignancies with poor overall prognosis, and is reported mainly in children and adolescents. Therefore, the investigation of novel and efficient treatment strategies for patients with OS is required. Baicalin exhibits potential anticancer effects, including in OS. However, its therapeutic effect against OS and the underlying mechanisms have not been fully evaluated. In the present study, the effect of baicalin on the proliferation and apoptosis of OS cells and its underlying mechanism of AKT pathway activation was explored. Cell confluence and cell number counts revealed suppressed the growth of OS cells that were treated with baicalin. Analysis of cell viability, cell survival and cell cycle, as well as cell apoptosis revealed decreased cell viability and survival, induced cell cycle arrest and apoptosis of treated cells. Western blot analysis demonstrated significantly decreased ratios of phosphorylated-AKT/AKT and Bcl-2/Bax, and decreased protein levels of cyclin D1 and CDK4 in cells treated with baicalin. Thus, the findings from the present study suggest that the suppression of the AKT pathway may be the underlying mechanism of the antitumor effect of baicalin in OS cells.
Collapse
Affiliation(s)
- Yu Liu
- Fujian University of Traditional Chinese Medicine, Key Laboratory of Orthopedics and Traumatology of Traditional Chinese Medicine and Rehabilitation, Ministry of Education, Fuzhou, Fujian 350122, P.R. China
| | - Zhenqiang Hong
- Fujian University of Traditional Chinese Medicine, Key Laboratory of Orthopedics and Traumatology of Traditional Chinese Medicine and Rehabilitation, Ministry of Education, Fuzhou, Fujian 350122, P.R. China
| | - Peng Chen
- Fujian University of Traditional Chinese Medicine, Key Laboratory of Orthopedics and Traumatology of Traditional Chinese Medicine and Rehabilitation, Ministry of Education, Fuzhou, Fujian 350122, P.R. China
| | - Jinzhao Wang
- Fujian University of Traditional Chinese Medicine, Key Laboratory of Orthopedics and Traumatology of Traditional Chinese Medicine and Rehabilitation, Ministry of Education, Fuzhou, Fujian 350122, P.R. China
| | - Yimin Zhou
- Fujian University of Traditional Chinese Medicine, Key Laboratory of Orthopedics and Traumatology of Traditional Chinese Medicine and Rehabilitation, Ministry of Education, Fuzhou, Fujian 350122, P.R. China
| | - Jianyun Huang
- Department of Orthopedics, Third Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| |
Collapse
|
31
|
RTA 408 Inhibits Interleukin-1β-Induced MMP-9 Expression via Suppressing Protein Kinase-Dependent NF-κB and AP-1 Activation in Rat Brain Astrocytes. Int J Mol Sci 2019; 20:ijms20112826. [PMID: 31185608 PMCID: PMC6600142 DOI: 10.3390/ijms20112826] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/03/2019] [Accepted: 06/07/2019] [Indexed: 12/20/2022] Open
Abstract
Neuroinflammation is characterized by the elevated expression of various inflammatory proteins, including matrix metalloproteinases (MMPs), induced by various pro-inflammatory mediators, which play a critical role in neurodegenerative disorders. Interleukin-1β (IL-1β) has been shown to induce the upregulation of MMP-9 through nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX)-reactive oxygen species (ROS)-dependent signaling pathways. N-(2-cyano-3,12-dioxo-28-noroleana-1,9(11)-dien-17-yl)-2-2-difluoropropanamide (RTA 408), a novel synthetic triterpenoid, has been shown to possess anti-oxidant and anti-inflammatory properties in various types of cells. Here, we evaluated the effects of RTA 408 on IL-1β-induced inflammatory responses by suppressing MMP-9 expression in a rat brain astrocyte (RBA-1) line. IL-1β-induced MMP-9 protein and mRNA expression, and promoter activity were attenuated by RTA 408. The increased level of ROS generation in RBA-1 cells exposed to IL-1β was attenuated by RTA 408, as determined by using 2′,7′-dichlorodihydrofluorescein diacetate (DCFH-DA) and CellROX. In addition, the inhibitory effects of RTA 408 on MMP-9 expression resulted from the suppression of the IL-1β-stimulated activation of Pyk2 (proline-rich tyrosine kinase), platelet-derived growth factor receptor β (PDGFRβ), Akt, ROS, and mitogen-activated protein kinases (MAPKs). Pretreatment with RTA 408 attenuated the IL-1β-induced c-Jun phosphorylation, mRNA expression, and promoter activity. IL-1β-stimulated nuclear factor-κB (NF-κB) p65 phosphorylation, translocation, and promoter activity were also attenuated by RTA 408. Furthermore, IL-1β-induced glial fibrillary acidic protein (GFAP) protein and mRNA expression, and cell migration were attenuated by pretreatment with RTA 408. These results provide new insights into the mechanisms by which RTA 408 attenuates IL-1β-mediated inflammatory responses and exerts beneficial effects for the management of brain diseases.
Collapse
|
32
|
Li J, Wang H, Shi X, Zhao L, Lv T, Yuan Q, Hao W, Zhu J. Anti-proliferative and anti-migratory effects of Scutellaria strigillosa Hemsley extracts against vascular smooth muscle cells. JOURNAL OF ETHNOPHARMACOLOGY 2019; 235:155-163. [PMID: 30763696 DOI: 10.1016/j.jep.2019.02.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/30/2019] [Accepted: 02/09/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The abnormal increase in vascular smooth muscle cell (VSMC) proliferation and migration are critical events in the pathogenesis of cardiovascular diseases (CVDs) including restenosis and atherosclerosis. The dried roots of Scutellaria baicalensis Georgi (common name: Huangqin in China) have been confirmed to possess beneficial effects on CVD by clinical and modern pharmacological studies. Flavonoids in Huangqin exert anti-proliferative and anti-migratory effects. Similar to Huangqin, Scutellaria strigillosa Hemsley (SSH) has been used to clear heat and damp and is especially rich in flavonoids including wogonin, wogonoside, baicalein, and baicalin. However, there have been few of reports about pharmacological activities of SSH. AIM OF THE STUDY To investigate the anti-proliferative and anti-migratory properties of Scutellaria strigillosa Hemsley extract (SSHE) in vitro and in vivo and explore its possible mechanism of action. MATERIALS AND METHODS The chemical constituents of SSHE were analyzed by ultra-high performance liquid chromatography coupled with triple time-of-flight mass spectrometry (UPLC-Triple-TOF-MS/MS). Cell proliferation and migration were investigated using BrdU incorporation assay and cell scratch test, respectively. The protein expression was determined by western blotting. In vivo, we established an artery ligation model of C57BL/6 mice and orally administered them with 50 or 100 mg/kg/day of SSHE. The carotid arteries were harvested and the intima-media thickness was examined 28 days post-ligation. RESULTS Twelve compounds were identified and tentatively characterized. SSHE significantly inhibited the VSMC proliferation and migration stimulated by PDGF-BB and decreased the relative protein expression of regulatory signaling intermediates. Furthermore, the expression of SM22α was significantly elevated in SSHE-pretreated VSMCs, whereas knockdown of SM22α impaired the PDGF-BB-induced proliferation and migration arrest. Meanwhile, both ROS generation and the phosphorylation of ERK decreased in SSHE-pretreated VSMCs. In carotid artery ligation mice model, SSHE treatment significantly inhibited neointimal hyperplasia. CONCLUSIONS SSHE significantly inhibited the PDGF-BB-induced VSMC proliferation, migration, and neointimal hyperplasia of carotid artery caused by ligation. Upregulation of SM22α expression, inhibition of ROS generation and ERK phosphorylation were, at least, partly responsible for the effects of SSHE on VSMCs.
Collapse
MESH Headings
- Animals
- Becaplermin/administration & dosage
- Carotid Intima-Media Thickness
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Chromatography, High Pressure Liquid
- Dose-Response Relationship, Drug
- Male
- Mice
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Plant Extracts/administration & dosage
- Plant Extracts/pharmacology
- Rats
- Scutellaria/chemistry
- Tandem Mass Spectrometry
Collapse
Affiliation(s)
- Jiankun Li
- The Forth Affiliated Hospital of Hebei Medical University, No. 12 Health Road, Shijiazhuang 050011, PR China.
| | - Hairong Wang
- Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang 050017, PR China.
| | - Xiaowei Shi
- Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang 050017, PR China.
| | - Lili Zhao
- Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang 050017, PR China.
| | - Tao Lv
- Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang 050017, PR China.
| | - Qi Yuan
- Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang 050017, PR China.
| | - Wenyang Hao
- Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang 050017, PR China.
| | - Jing Zhu
- Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang 050017, PR China.
| |
Collapse
|
33
|
Wu JH, Zhou YF, Hong CD, Chen AQ, Luo Y, Mao L, Xia YP, He QW, Jin HJ, Huang M, Li YN, Hu B. Semaphorin-3A protects against neointimal hyperplasia after vascular injury. EBioMedicine 2018; 39:95-108. [PMID: 30579864 PMCID: PMC6355729 DOI: 10.1016/j.ebiom.2018.12.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/12/2018] [Accepted: 12/12/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Neointimal hyperplasia is a prominent pathological event during in-stent restenosis. Phenotype switching of vascular smooth muscle cells (VSMCs) from a differentiated/contractile to a dedifferentiated/synthetic phenotype, accompanied by migration and proliferation of VSMCs play an important role in neointimal hyperplasia. However, the molecular mechanisms underlying phenotype switching of VSMCs have yet to be fully understood. METHODS The mouse carotid artery ligation model was established to evaluate Sema3A expression and its role during neointimal hyperplasia in vivo. Bioinformatics analysis, chromatin immunoprecipitation (ChIP) assays and promoter-luciferase reporter assays were used to examine regulatory mechanism of Sema3A expression. SiRNA transfection and lentivirus infection were performed to regulate Sema3A expression. EdU assays, Wound-healing scratch experiments and Transwell migration assays were used to assess VSMC proliferation and migration. FINDINGS In this study, we found that semaphorin-3A (Sema3A) was significantly downregulated in VSMCs during neointimal hyperplasia after vascular injury in mice and in human atherosclerotic plaques. Meanwhile, Sema3A was transcriptionally downregulated by PDGF-BB via p53 in VSMCs. Furthermore, we found that overexpression of Sema3A inhibited VSMC proliferation and migration, as well as increasing differentiated gene expression. Mechanistically, Sema3A increased the NRP1-plexin-A1 complex and decreased the NRP1-PDGFRβ complex, thus inhibiting phosphorylation of PDGFRβ. Moreover, we found that overexpression of Sema3A suppressed neointimal hyperplasia after vascular injury in vivo. INTERPRETATION These results suggest that local delivery of Sema3A may act as a novel therapeutic option to prevent in-stent restenosis.
Collapse
Affiliation(s)
- Jie-Hong Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi-Fan Zhou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Can-Dong Hong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - An-Qi Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Luo
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Mao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan-Peng Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Quan-Wei He
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui-Juan Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Huang
- Department of Neurology, the People's Hospital of China Three Gorges University, Institute of Translational Neuroscience, Three Gorges University College of Medicine, Yichang 443002, China
| | - Ya-Nan Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
34
|
Liu Y, Jia L, Min D, Xu Y, Zhu J, Sun Z. Baicalin inhibits proliferation and promotes apoptosis of vascular smooth muscle cells by regulating the MEG3/p53 pathway following treatment with ox‑LDL. Int J Mol Med 2018; 43:901-913. [PMID: 30535498 PMCID: PMC6317676 DOI: 10.3892/ijmm.2018.4009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 11/26/2018] [Indexed: 11/15/2022] Open
Abstract
Atherosclerosis (AS) is a systemic disease associated with lipid metabolic disorders and abnormal proliferation of smooth muscle cells. Baicalin is a flavonoid compound isolated from the dry roots of Scutellaria baicalensis Georgi and exerts anti-proliferative effects in various types of cells. However, the effect of baicalin on AS remains unclear. In the present study, serum samples were collected from patients with AS and an in vitro model of AS was established using oxidized low-density lipoprotein (ox-LDL)-treated human aorta vascular smooth muscle cells (HA-VSMCs). The siRNA transfection and overexpression efficiency of endogenous maternally expressed gene 3 (MEG3) and the expression level of MEG3 were analyzed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The effects of alterations in expression levels of MEG3 were assessed by MTT assay, bromodeoxyuridine incorporation assay, 5-ethynyl-2′-deoxyuridine staining, wound healing assay, immunofluorescence and western blotting in HA-VSMCs. qPCR indicated that the expression of MEG3 was reduced in serum samples from patients with AS and ox-LDL-treated HA-VSMCs, compared with serum samples from healthy patients and untreated HA-VSMCs, respectively. Further experiments indicated that ox-LDL-induced decrease of MEG3 expression was reversed by treatment with baicalin in a concentration-dependent manner. Following treatment with ox-LDL, decreased expression of MEG3 promoted proliferation and migration, and suppressed apoptosis in HA-VSMCs. Furthermore, treatment with baicalin reversed these effects on proliferation and apoptosis in ox-LDL-treated HA-VSMCs. The current study indicated that downregulated expression of MEG3 increased cell cycle-associated protein expression. However, treatment with baicalin inhibited the expression of cell-cycle associated proteins in HA-VSMCs with MEG3 knockdown. In addition, baicalin activated the p53 signaling pathway and promoted the expression and transport of p53 from the cytoplasm to nucleus following MEG3 knockdown in ox-LDL-treated HA-VSMCs. Baicalin inhibited proliferation and promoted apoptosis by regulating the expression of MEG3/p53, indicating that baicalin may serve a role in AS by activating the MEG3/p53 signaling pathway. The present study suggested a potential mechanism underlying the protective role of baicalin in the in vitro model of AS, and these results may be used to develop novel therapeutic approaches for the affected patients.
Collapse
Affiliation(s)
- Yun Liu
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| | - Lianqun Jia
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110847, P.R. China
| | - Dongyu Min
- Traditional Chinese Medicine Experimental Center, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110032, P.R. China
| | - Yi Xu
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| | - Jinquan Zhu
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| | - Zengxian Sun
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| |
Collapse
|
35
|
Ambhore NS, Katragadda R, Raju Kalidhindi RS, Thompson MA, Pabelick CM, Prakash YS, Sathish V. Estrogen receptor beta signaling inhibits PDGF induced human airway smooth muscle proliferation. Mol Cell Endocrinol 2018; 476:37-47. [PMID: 29680290 PMCID: PMC6120801 DOI: 10.1016/j.mce.2018.04.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/09/2018] [Accepted: 04/18/2018] [Indexed: 02/07/2023]
Abstract
Airway smooth muscle (ASM) cell hyperplasia driven by persistent inflammation is a hallmark feature of remodeling in asthma. Sex steroid signaling in the lungs is of considerable interest, given epidemiological data showing more asthma in pre-menopausal women and aging men. Our previous studies demonstrated that estrogen receptor (ER) expression increases in asthmatic human ASM; however, very limited data are available regarding differential roles of ERα vs. ERβ isoforms in human ASM cell proliferation. In this study, we evaluated the effect of selective ERα and ERβ modulators on platelet-derived growth factor (PDGF)-stimulated ASM proliferation and the mechanisms involved. Asthmatic and non-asthmatic primary human ASM cells were treated with PDGF, 17β-estradiol, ERα-agonist and/or ERβ-agonist and/or G-protein-coupled estrogen receptor 30 (GPR30/GPER) agonist and proliferation was measured using MTT and CyQuant assays followed by cell cycle analysis. Transfection of small interfering RNA (siRNA) ERα and ERβ significantly altered the human ASM proliferation. The specificity of siRNA transfection was confirmed by Western blot analysis. Gene and protein expression of cell cycle-related antigens (PCNA and Ki67) and C/EBP were measured by RT-PCR and Western analysis, along with cell signaling proteins. PDGF significantly increased ASM proliferation in non-asthmatic and asthmatic cells. Treatment with PPT showed no significant effect on PDGF-induced proliferation, whereas WAY interestingly suppressed proliferation via inhibition of ERK1/2, Akt, and p38 signaling. PDGF-induced gene expression of PCNA, Ki67 and C/EBP in human ASM was significantly lower in cells pre-treated with WAY. Furthermore, WAY also inhibited PDGF-activated PCNA, C/EBP, cyclin-D1, and cyclin-E. Overall, we demonstrate ER isoform-specific signaling in the context of ASM proliferation. Activation of ERβ can diminish remodeling in human ASM by inhibiting pro-proliferative signaling pathways, and may point to a novel perception for blunting airway remodeling.
Collapse
Affiliation(s)
| | - Rathnavali Katragadda
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA
| | | | - Michael A Thompson
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Christina M Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA; Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
36
|
Teliewubai J, Ji H, Lu Y, Bai B, Yu S, Chi C, Xu Y, Zhang Y. SFRP5 serves a beneficial role in arterial aging by inhibiting the proliferation, migration and inflammation of smooth muscle cells. Mol Med Rep 2018; 18:4682-4690. [PMID: 30221661 DOI: 10.3892/mmr.2018.9467] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 07/17/2018] [Indexed: 11/05/2022] Open
Abstract
Secreted frizzled-related protein 5 (SFRP5) is one of the anti-inflammatory adipokines secreted from white adipose tissue. However, little is known about the effect of SFRP5 on the cardiovascular system. The aim of the present study was to determine the effect of SFRP5 on smooth muscle cell (SMC) proliferation, migration and inflammation. The plasma levels of SFRP5 were evaluated in a cohort‑based elderly population using ELISA, and the expression of SFRP5 in Sprague‑Dawley rat aortas was detected using immunohistochemistry. SMC proliferation and migration were evaluated in vitro using 5‑ethynyl‑2'‑deoxyuridine cell proliferation and wound‑healing assays, respectively, while reactive oxygen species (ROS) production and cell signaling were assessed using a 2',7'‑dichlorodihydrofluorescein diacetate assay and immunoblotting, respectively. The results revealed that plasma levels of SFRP5 were positively correlated with age in the elderly Chinese cohort. Similarly, aorta SFRP5 expression was significantly higher in 15‑month‑old rats compared with 6‑month‑old rats. In vitro, SFRP5 significantly inhibited rat aortic SMC proliferation and migration that were induced by platelet‑derived growth factor (PDGF)‑BB, as well as inhibiting ROS generation. Compared with the effect of PDGF‑BB on SMCs, SFRP5 at 100 and 200 ng/ml significantly decreased SMC proliferation by 31.5 and 34.8%, respectively (P<0.05). SFRP5 at 100 and 200 ng/ml also inhibited the migration of SMCs by 24.9 and 28.4%, respectively, when compared with the effects of PDGF‑BB. SFRP5 attenuated the PDGF‑BB‑induced expression of β‑catenin and proliferating cell nuclear antigen, while p38 phosphorylation was significantly attenuated. Together, the present results suggested that SFRP5 may inhibit SMC proliferation, migration and inflammation by suppressing the Wnt/β‑catenin and p38/mitogen‑activated protein kinase signaling pathways.
Collapse
Affiliation(s)
- Jiadela Teliewubai
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Hongwei Ji
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Yuyan Lu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Bin Bai
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Shikai Yu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Chen Chi
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Yi Zhang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| |
Collapse
|
37
|
Yang J, Zeng P, Yang J, Liu X, Ding J, Wang H, Chen L. MicroRNA-24 regulates vascular remodeling via inhibiting PDGF-BB pathway in diabetic rat model. Gene 2018; 659:67-76. [PMID: 29559348 DOI: 10.1016/j.gene.2018.03.056] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/11/2018] [Accepted: 03/16/2018] [Indexed: 12/26/2022]
Abstract
PURPOSE Hyperglycemia is the high risk factor of vascular remodeling induced by angioplasty, and neointimal hyperplasia is strongly implicated in the pathogenesis of vascular remodeling caused by carotid artery balloon injury. Studies have shown that MicroRNA 24 (miR-24) plays an important role in angiocardiopathy, However, the role of miR-24 is far from thorough research. In this study, we investigate whether up-regulation of miR-24 by using miR-24 recombinant adenovirus (Ad-miR-24-GFP) can inhibit PDGF-BB signaling pathway and attenuate vascular remodeling in the diabetic rat model. METHODS Male Sprague-Dawley rats (n = 60) were randomly divided into 5 groups and fed with high sugar and high fat diet (Sham, Saline, Scramble, Ad-miR-24 groups), or ordinary diet (Control group). The front four groups were treated with streptozotocin (STZ) four weeks later and the blood glucose level was closely monitored. After the successful establishment of diabetic rats, the external carotid artery was injured by pressuring balloon 1.5 after internal carotid artery ligation, then the blood vessels were harvested 14 days later and indexes were detected including the following: HE staining for the level of vascular intima thickness, immunohistochemical detection for PCNA and P27 to test the proliferative degree of vascular smooth muscle cells (VSMCs), qRT-PCR for the level of miR-24, RAS,PDGF-R, western blot for the protein levels of JNK1/2, p- JNK1/2, ERK1/2, p-ERK1/2, RAS, PDGF-R, AP-1,P27 and PCNA. Serological detection was conducted for TNF-α, IL-6, IL-8. RESULTS The delivery of Ad-miR-24 into balloon injury site has significantly increased the level of miR-24. Up-regulation of miR-24 could regulate vascular remodeling effectively, lower the level of inflammatory factors, inhibit the expression of mRNA and protein levels of JNK1/2, ERK1/2, RAS, PDGF-R, AP-1, P27, PCNA. CONCLUSION miR-24 can inhibit the expression of AP-1 via the inhibition of PDGF-BB signaling pathway, thus inhibit VSMCs proliferation and vascular remodeling.
Collapse
MESH Headings
- Animals
- Becaplermin
- Cell Proliferation
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/pathology
- Diet, High-Fat/adverse effects
- Genetic Therapy
- Male
- MicroRNAs/genetics
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Neointima/genetics
- Neointima/prevention & control
- Proto-Oncogene Proteins c-sis
- Random Allocation
- Rats
- Rats, Sprague-Dawley
- Signal Transduction
- Streptozocin
- Vascular Remodeling
Collapse
Affiliation(s)
- Jian Yang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, China; Key Laboratory of Ischemic Cardiovascular and Cerebrovascular Disease Translational Medicine, Three Gorges University, China.
| | - Ping Zeng
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, China
| | - Jun Yang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, China; Key Laboratory of Ischemic Cardiovascular and Cerebrovascular Disease Translational Medicine, Three Gorges University, China
| | - Xiaowen Liu
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, China; Key Laboratory of Ischemic Cardiovascular and Cerebrovascular Disease Translational Medicine, Three Gorges University, China
| | - Jiawang Ding
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, China; Key Laboratory of Ischemic Cardiovascular and Cerebrovascular Disease Translational Medicine, Three Gorges University, China
| | - Huibo Wang
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, China
| | - Lihua Chen
- Department of Optometry and Ophthalmology, Yichang Central People's Hospital, Yichang, China
| |
Collapse
|
38
|
The Signaling Pathways Involved in the Antiatherosclerotic Effects Produced by Chinese Herbal Medicines. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5392375. [PMID: 30009170 PMCID: PMC6020658 DOI: 10.1155/2018/5392375] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/15/2018] [Accepted: 05/21/2018] [Indexed: 12/19/2022]
Abstract
Cardiovascular diseases (CVDs) are considered to be the predominant cause of death in the world. Chinese herb medicines (CHMs) have been widely used for the treatment of CVDs in Asian countries for thousands of years. One reason of high efficacy of CHMs in treating CVDs is attributed to their inhibition in atherosclerosis (AS) development, a critical contributor to CVDs occurrence. Cumulative studies have demonstrated that CHMs alleviate atherogenesis via mediating pathophysiologic events involved in AS. However, there is deficiency in the summaries regarding antiatherogenic signal pathways regulated by CHMs. In this review, we focus on the signal cascades by which herb medicines and relevant extractives, derivatives, and patents improve proatherogenic processes including endothelium dysfunction, lipid accumulation, and inflammation. We mainly elaborate the CHMs-mediated signaling pathways in endothelial cells, macrophages, and vascular smooth muscle cells of each pathogenic event. Moreover, we briefly describe the other AS-related factors such as thrombosis, autophagy, immune response, and noncoding RNAs and effects of CHMs on them in the way of cascade regulation, which is helpful to further illustrate the molecular mechanisms of AS initiation and progression and discover newly effective agents for AS management.
Collapse
|
39
|
Gao C, Zhou Y, Li H, Cong X, Jiang Z, Wang X, Cao R, Tian W. Antitumor effects of baicalin on ovarian cancer cells through induction of cell apoptosis and inhibition of cell migration in vitro. Mol Med Rep 2017; 16:8729-8734. [PMID: 29039573 PMCID: PMC5779949 DOI: 10.3892/mmr.2017.7757] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 08/14/2017] [Indexed: 12/15/2022] Open
Abstract
Baicalin, an active flavone isolated from Scutellaria baicalensis Georgi, has been demonstrated to induce various beneficial biochemical effects such as anti‑inflammatory, anti‑viral, and antitumor effects. However, the antitumor mechanism of baicalin is not well understood. In the present study, baicalin was demonstrated to inhibit the viability and migration of a widely used ovarian cancer cell line, A2780, in a dose‑dependent manner. MTT assays revealed that cell viability significantly decreased in ovarian cancer cells treated with baicalin compared with untreated cells, without effect on normal ovarian cells. Flow cytometric analysis indicated that baicalin suppressed cell proliferation by inducing apoptosis. The underlying mechanisms involved were indicated to be downregulation of the anti‑apoptotic protein B‑cell lymphoma 2 apoptosis regulator and activation of caspase‑3 and ‑9. In addition, wound healing and transwell assays revealed that cell migratory potential and expression of matrix metallopeptidase (MMP)‑2 and MMP‑9 were significantly inhibited when cells were exposed to baicalin, compared with untreated cells. The present study therefore suggested that baicalin has the potential to be used in novel anti‑cancer therapeutic formulations for treatment of ovarian cancer.
Collapse
Affiliation(s)
- Chen Gao
- College of Animal Science and Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong 266109, P.R. China
| | - Yinglu Zhou
- College of Animal Science and Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong 266109, P.R. China
| | - Huatao Li
- College of Animal Science and Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong 266109, P.R. China
| | - Xia Cong
- College of Animal Science and Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong 266109, P.R. China
| | - Zhongling Jiang
- College of Animal Science and Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong 266109, P.R. China
| | - Xin Wang
- College of Animal Science and Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong 266109, P.R. China
| | - Rongfeng Cao
- College of Animal Science and Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong 266109, P.R. China
| | - Wenru Tian
- College of Animal Science and Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong 266109, P.R. China
| |
Collapse
|
40
|
Park YG, Choi J, Jung HK, Kim B, Kim C, Park SY, Seol JW. Baicalein inhibits tumor progression by inhibiting tumor cell growth and tumor angiogenesis. Oncol Rep 2017; 38:3011-3018. [PMID: 29048641 DOI: 10.3892/or.2017.6007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/18/2017] [Indexed: 11/06/2022] Open
Abstract
Baicalein, a herbal medicine, is a natural flavonoid isolated from the roots of Scutellaria baicalensis Georgi. It is known for its anticancer, anti-inflammatory and neuroprotective properties. Despite these well-known properties, it is not yet clear what effect baicalein has on tumor progression. Therefore, in the present study, we used B16F10 cells, Lewis lung carcinoma (LLC) cells, and human umbilical vein endothelial cells (HUVECs) to investigate the effect of baicalein on cell proliferation and viability, migration and tube formation in vitro. In addition, an experimental animal model was used to observe the growth rate and metastasis of tumors and tumor vessel formation in vivo. Our results showed that baicalein decreased the proliferation and migration and induced tumor cell death via caspase-3 activation in the B16F10 and LLC cells, and strongly inhibited tube formation and cell migration in HUVECs. Furthermore, mouse models showed that baicalein reduced the tumor volume and greatly reduced the tumor growth rate in the early stages of tumor progression, and the baicalein-treated groups had significantly reduced expression of CD31 (endothelial cell marker) and α-SMA (mural cell marker) in the tumors, indicating that baicalein inhibits tumor angiogenesis by disrupting tumor vasculature development. Comparison of the lymph node and lung samples collected from the baicalein-treated group, and the untreated group showed that baicalein reduced metastasis of the tumor to these tissues. In summary, baicalein reduced tumor progression and metastasis, directly induced tumor cell death, and inhibited tumor angiogenesis. Our results strongly demonstrate that baicalein is a potential chemotherapeutic agent.
Collapse
Affiliation(s)
- Yang-Gyu Park
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksana, Republic of Korea
| | - Jawun Choi
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksana, Republic of Korea
| | - Hye-Kang Jung
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksana, Republic of Korea
| | - Bumseok Kim
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksana, Republic of Korea
| | - Chan Kim
- CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Sang-Youel Park
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksana, Republic of Korea
| | - Jae-Won Seol
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksana, Republic of Korea
| |
Collapse
|
41
|
Baicalin attenuates chronic hypoxia-induced pulmonary hypertension via adenosine A 2A receptor-induced SDF-1/CXCR4/PI3K/AKT signaling. J Biomed Sci 2017; 24:52. [PMID: 28774332 PMCID: PMC5543745 DOI: 10.1186/s12929-017-0359-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 07/28/2017] [Indexed: 11/28/2022] Open
Abstract
Background Baicalin, an important flavonoid in Scutellaria baicalensis Georgi extracts, exerts a variety of pharmacological effects. In this study, we explored the effects of baicalin on chronic hypoxia-induced pulmonary arterial hypertension (PAH) and investigated the mechanism underlying these effects. Moreover, we examined whether the inflammatory response was mediated by the A2A receptor (A2AR) and stromal cell-derived factor-1 (SDF-1)/C-X-C chemokine receptor type 4 (CXCR4)-induced phosphatidyl inositol-3-kinase (PI3K) signaling in vivo. Methods We established a hypoxia-induced pulmonary hypertension (HPH) mouse model by subjecting wild-type (WT) and A2AR knockout (A2AR−/−) animals to chronic hypoxia, and we examined the effects of a 4-week treatment with baicalin or the A2AR agonist CGS21680 in these animals. Invasive hemodynamic parameters, the right ventricular hypertrophy index, pulmonary congestion, the pulmonary arterial remodeling index, blood gas parameters, A2AR expression, and the expression of SDF-1/CXCR4/PI3K/protein kinase B (PKB; AKT) signaling components were measured. Results Compared with WT mice, A2AR−/− mice exhibited increased right ventricular systolic pressure (RVSP), right ventricle-to-left ventricle plus septum [RV/(LV + S)] ratio, RV weight-to-body weight (RV/BW) ratio, and lung wet weight-to-body weight (Lung/BW) ratio in the absence of an altered mean carotid arterial pressure (mCAP). These changes were accompanied by increases in pulmonary artery wall area and thickness and reductions in arterial oxygen pressure (PaO2) and hydrogen ion concentration (pH). In the HPH model, A2AR−/− mice displayed increased CXCR4, SDF-1, phospho-PI3K, and phospho-AKT expression compared with WT mice. Treating WT and A2AR−/− HPH mice with baicalin or CGS21680 attenuated the hypoxia-induced increases in RVSP, RV/(LV + S) and Lung/BW, as well as pulmonary arterial remodeling. Additionally, baicalin or CGS21680 alone could reverse the hypoxia-induced increases in CXCR4, SDF-1, phospho-PI3K, and phospho-AKT expression. Moreover, baicalin improved the hypoxemia induced by 4 weeks of hypoxia. Finally, we found that A2AR levels in WT lung tissue were enhanced by hypoxia and that baicalin up-regulated A2AR expression in WT hypoxic mice. Conclusions Baicalin exerts protective effects against clinical HPH, which are partly mediated through enhanced A2AR activity and down-regulated SDF-1/CXCR4-induced PI3K/AKT signaling. Therefore, the A2AR may be a promising target for baicalin in treating HPH.
Collapse
|
42
|
Zhang Z, Zhang L, Sun C, Kong F, Wang J, Xin Q, Jiang W, Li K, Chen O, Luan Y. Baicalin attenuates monocrotaline-induced pulmonary hypertension through bone morphogenetic protein signaling pathway. Oncotarget 2017; 8:63430-63441. [PMID: 28969002 PMCID: PMC5609934 DOI: 10.18632/oncotarget.18825] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 06/01/2017] [Indexed: 11/25/2022] Open
Abstract
Baicalin, a flavonoid compound extracted from roots of Scutellaria baicalensis Georgi (huang qin), it has been shown to effectively attenuates pulmonary hypertension (PH), however, the potential mechanism remains unexplored. In this study, we investigated the potential mechanism of baicalin on monocrotaline (MCT)-induced PH in rats. The results showed that baicalin attenuated lung damage in PH rat model through inhibiting the pulmonary arterial smooth muscle cell proliferation and induction of cells apoptosis. Furthermore, we demonstrated that baicalin inhibition the expression of nuclear factor-κB (NF-κB) p65 and bone morphogenetic protein (BMP) antagonists gremlin-1, but increased the expression of inhibitor of NF-κB (I-κBα), BMPR2, BMP-4, BMP-9 and Smad1/5/8. Additionally, baicalin suppression endothelial-to-mesenchymal transition in PH lung tissue. Collectively, we confirmed that baicalin via inhibition of NF-κB signaling to further activation of BMP signaling to have a therapeutic effect on PH and providing a promising therapeutic strategy for PH.
Collapse
Affiliation(s)
- Zhaohua Zhang
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, China
| | - Luan Zhang
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, China
| | - Chao Sun
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Feng Kong
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Jue Wang
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Qian Xin
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Wen Jiang
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Kaili Li
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Ou Chen
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Yun Luan
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
43
|
Protective effects of aloperine on monocrotaline-induced pulmonary hypertension in rats. Biomed Pharmacother 2017; 89:632-641. [DOI: 10.1016/j.biopha.2017.02.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 02/05/2017] [Accepted: 02/10/2017] [Indexed: 01/10/2023] Open
|
44
|
Dinda B, Dinda S, DasSharma S, Banik R, Chakraborty A, Dinda M. Therapeutic potentials of baicalin and its aglycone, baicalein against inflammatory disorders. Eur J Med Chem 2017; 131:68-80. [PMID: 28288320 DOI: 10.1016/j.ejmech.2017.03.004] [Citation(s) in RCA: 370] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/20/2017] [Accepted: 03/03/2017] [Indexed: 02/08/2023]
Abstract
The flavonoids, baicalin (5,6-dihydroxy-2-phenyl-4H-1-benzopyran-4-one-7-O-d-β-glucuronic acid) 1 and its aglycone, baicalein 2 are found in edible medicinal plants, Scutellaria baicalensis Georgi and Oroxylum indicum (L.) Kurz in abundant quantities. The antioxidant and anti-inflammatory effects of these flavonoids have been demonstrated in various disease models, including diabetes, cardiovascular diseases, inflammatory bowel diseases, gout and rheumatoid arthritis, asthma, neurodegenerative-, liver- and kidney diseases, encephalomyelitis, and carcinogenesis. These flavonoids have almost no toxicity to human normal epithelial, peripheral and myeloid cells. Their antioxidant and anti-inflammatory activities are largely due to their abilities to scavenge the reactive oxygen species (ROS) and improvement of antioxidant status by attenuating the activity of NF-κB and suppressing the expression of several inflammatory cytokines and chemokines including monocyte chemotactic protein-1 (MCP-1), nitric oxide synthase, cyclooxygenases, lipoxygenases, cellular adhesion molecules, tumor necrosis factor and interleukins. In this review, we summarize the antioxidant and anti-inflammatory effects of baicalin and baicalein with molecular mechanisms for their chemopreventive and chemotherapeutic applications in the treatment of inflammatory-related diseases.
Collapse
Affiliation(s)
- Biswanath Dinda
- Department of Chemistry, Tripura University, Suryamaninagar 799022, Tripura, India.
| | - Subhajit Dinda
- Department of Chemistry, Dasaratha Deb Memorial College, Khowai 799201, Tripura, India
| | - Saikat DasSharma
- Department of Chemistry, Dasaratha Deb Memorial College, Khowai 799201, Tripura, India
| | - Rajarshi Banik
- Department of Chemistry, National Institute of Technology, Agartala 799055, Tripura, India
| | - Ankita Chakraborty
- Department of Chemistry, Tripura University, Suryamaninagar 799022, Tripura, India
| | - Manikarna Dinda
- Department of Biochemistry and Molecular Genetics, University of Virginia, School of Medicine, Charlottesville, 1300 Jefferson Park Ave, VA22908, USA
| |
Collapse
|
45
|
Roscovitine Protects From Arterial Injury by Regulating the Expressions of c-Jun and p27 and Inhibiting Vascular Smooth Muscle Cell Proliferation. J Cardiovasc Pharmacol 2017; 69:161-169. [PMID: 28009720 DOI: 10.1097/fjc.0000000000000453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
46
|
Liao CC, Day YJ, Lee HC, Liou JT, Chou AH, Liu FC. ERK Signaling Pathway Plays a Key Role in Baicalin Protection Against Acetaminophen-Induced Liver Injury. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2017; 45:105-121. [DOI: 10.1142/s0192415x17500082] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Acetaminophen (APAP) overdose causes hepatocytes necrosis and acute liver failure. Baicalin (BA), a major flavonoid of Scutellariae radix, has potent hepatoprotective properties in traditional medicine. In the present study, we investigated the protective effects of BA on a APAP-induced liver injury in a mouse model. The mice received an intraperitoneal hepatotoxic dose of APAP (300[Formula: see text]mg/kg) and after 30[Formula: see text]min, were treated with BA at concentrations of 0, 15, 30, or 60[Formula: see text]mg/kg. After 16[Formula: see text]h of treatment, the mice were sacrificed for further analysis. APAP administration significantly elevated the serum alanine transferase (ALT) enzyme levels and hepatic myeloperoxidase (MPO) activity when compared with control animals. Baicalin treatment significantly attenuated the elevation of liver ALT levels, as well as hepatic MPO activity in a dose- dependent manner (15–60[Formula: see text]mg/kg) in APAP-treated mice. The strongest beneficial effects of BA were seen at a dose of 30[Formula: see text]mg/kg. BA treatment at 30[Formula: see text]mg/kg after APAP overdose reduced elevated hepatic cytokine (TNF-[Formula: see text] and IL-6) levels, and macrophage recruitment around the area of hepatotoxicity in immunohistochemical staining. Significantly, BA treatment can also decrease hepatic phosphorylated extracellular signal-regulated kinase (ERK) expression, which is induced by APAP overdose. Our data suggests that baicalin treatment can effectively attenuate APAP-induced liver injury by down-regulating the ERK signaling pathway and its downstream effectors of inflammatory responses. These results support that baicalin is a potential hepatoprotective agent.
Collapse
Affiliation(s)
- Chia-Chih Liao
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Yuan-Ji Day
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Hung-Chen Lee
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Jiin-Tarng Liou
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - An-Hsun Chou
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Fu-Chao Liu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
47
|
Jang MA, Lee SJ, Baek SE, Park SY, Choi YW, Kim CD. α-Iso-Cubebene Inhibits PDGF-Induced Vascular Smooth Muscle Cell Proliferation by Suppressing Osteopontin Expression. PLoS One 2017; 12:e0170699. [PMID: 28114367 PMCID: PMC5256966 DOI: 10.1371/journal.pone.0170699] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 01/09/2017] [Indexed: 12/31/2022] Open
Abstract
α-Iso-cubebene (ICB) is a dibenzocyclooctadiene lignin contained in Schisandra chinensis (SC), a well-known medicinal herb that ameliorates cardiovascular symptoms. Thus, we examined the effect of ICB on vascular smooth muscle cell (VSMC) proliferation, a key feature of diverse vascular diseases. When VSMCs primary cultured from rat thoracic aorta were stimulated with PDGF (1-10 ng/ml), cell proliferation and osteopontin (OPN) expression were concomitantly up-regulated, but these effects were attenuated when cells were treated with MPIIIB10, a neutralizing monoclonal antibody for OPN. In aortic tissues exposed to PDGF, sprouting VSMC numbers increased, which was attenuated in tissues from OPN-deficient mice. Furthermore, VSMC proliferation and OPN expression induced by PDGF were attenuated dose-dependently by ICB (10 or 30 μg/ml). Reporter assays conducted using OPN promoter-luciferase constructs showed that the promoter region 538-234 bp of the transcription start site was responsible for transcriptional activity enhancement by PDGF, which was significantly inhibited by ICB. Putative binding sites for AP-1 and C/EBPβ in the indicated promoter region were suggested by TF Search, and increased binding of AP-1 and C/EBPβ in PDGF-treated VSMCs was demonstrated using a ChIP assay. The increased bindings of AP-1 and C/EBPβ into OPN promoter were attenuated by ICB. Moreover, the PDGF-induced expression of OPN was markedly attenuated in VSMCs transfected with siRNA for AP-1 and C/EBPβ. These results indicate that ICB inhibit VSMC proliferation by inhibiting the AP-1 and C/EBPβ signaling pathways and thus downregulating OPN expression.
Collapse
Affiliation(s)
- Min A. Jang
- Department of Pharmacology, School of Medicine, Pusan National University, Gyeongnam, Republic of Korea
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Gyeongnam, Republic of Korea
| | - Seung Jin Lee
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Seung Eun Baek
- Department of Pharmacology, School of Medicine, Pusan National University, Gyeongnam, Republic of Korea
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Gyeongnam, Republic of Korea
| | - So Youn Park
- Department of Pharmacology, School of Medicine, Pusan National University, Gyeongnam, Republic of Korea
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Gyeongnam, Republic of Korea
| | - Young Whan Choi
- College of Natural Resources & Life Sciences, Pusan National University, Gyeongnam, Republic of Korea
| | - Chi Dae Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Gyeongnam, Republic of Korea
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Gyeongnam, Republic of Korea
| |
Collapse
|
48
|
Minami A, Ogino M, Nakano N, Ichimura M, Nakanishi A, Murai T, Kitagishi Y, Matsuda S. Roles of oncogenes and tumor-suppressor genes in osteoclastogenesis (Review). Int J Mol Med 2017; 39:261-267. [DOI: 10.3892/ijmm.2017.2847] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 12/28/2016] [Indexed: 11/06/2022] Open
|
49
|
Lv P, Zhang F, Yin YJ, Wang YC, Gao M, Xie XL, Zhao LL, Dong LH, Lin YL, Shu YN, Zhang DD, Liu GX, Han M. SM22α inhibits lamellipodium formation and migration via Ras-Arp2/3 signaling in synthetic VSMCs. Am J Physiol Cell Physiol 2016; 311:C758-C767. [PMID: 27629412 DOI: 10.1152/ajpcell.00033.2016] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 09/07/2016] [Indexed: 02/03/2023]
Abstract
We previously demonstrated that smooth muscle (SM) 22α promotes the migration activity in contractile vascular smooth muscle cells (VSMCs). Based on the varied functions exhibited by SM22α in different VSMC phenotypes, we investigated the effect of SM22α on VSMC migration under pathological conditions. The results demonstrated that SM22α overexpression in synthetic VSMCs inhibited platelet-derived growth factor (PDGF)-BB-induced cell lamellipodium formation and migration, which was different from its action in contractile cells. The results indicated two distinct mechanisms underlying inhibition of lamellipodium formation by SM22α, increased actin dynamic stability and decreased Ras activity via interference with interactions between Ras and guanine nucleotide exchange factor. The former inhibited actin cytoskeleton rearrangement in the cell cortex, while the latter significantly disrupted actin nucleation activation of the Arp2/3 complex. Baicalin, a herb-derived flavonoid compound, inhibited VSMC migration via upregulation of SM22α expression in vitro and in vivo. These data suggest that SM22α regulates lamellipodium formation and cell migration in a phenotype-dependent manner in VSMCs, which may be a new therapeutic target for vascular lesion formation.
Collapse
Affiliation(s)
- Pin Lv
- Key Laboratory of Medical Biotechnology of Hebei Province, Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Fan Zhang
- Key Laboratory of Medical Biotechnology of Hebei Province, Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Ya-Juan Yin
- Key Laboratory of Medical Biotechnology of Hebei Province, Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Yu-Can Wang
- Key Laboratory of Medical Biotechnology of Hebei Province, Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Min Gao
- Key Laboratory of Medical Biotechnology of Hebei Province, Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Xiao-Li Xie
- Key Laboratory of Medical Biotechnology of Hebei Province, Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Li-Li Zhao
- Key Laboratory of Medical Biotechnology of Hebei Province, Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Li-Hua Dong
- Key Laboratory of Medical Biotechnology of Hebei Province, Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Yan-Ling Lin
- Key Laboratory of Medical Biotechnology of Hebei Province, Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Ya-Nan Shu
- Key Laboratory of Medical Biotechnology of Hebei Province, Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Dan-Dan Zhang
- Key Laboratory of Medical Biotechnology of Hebei Province, Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Gui-Xia Liu
- Key Laboratory of Medical Biotechnology of Hebei Province, Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Mei Han
- Key Laboratory of Medical Biotechnology of Hebei Province, Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
50
|
Inhibitory effect of NBL1 on PDGF-BB-induced human PASMC proliferation through blockade of PDGFβ-p38MAPK pathway. Biosci Rep 2016; 36:BSR20160199. [PMID: 27474499 PMCID: PMC5006314 DOI: 10.1042/bsr20160199] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 07/28/2016] [Indexed: 12/26/2022] Open
Abstract
Pulmonary artery remodelling is a key feature in the pathological progress of pulmonary arterial hypertension (PAH). Moreover, excessive proliferation of pulmonary arterial smooth muscle cells (PASMCs) plays a critical role in the pathogenesis of pulmonary artery remodelling. Neuroblastoma suppressor of tumorigenicity 1 (NBL1) has been previously shown to induce growth inhibition in tumour cells. However, the effect of NBL1 in the regulation of human PASMC proliferation remains unclear. In cultured human PASMCs, we observed a dose-dependent inhibitory effect of NBL1 on platelet derived growth factor (PDGF)-BB-induced cell growth, DNA synthesis and proliferating cell nuclear antigen (PCNA) expression, as measured by MTS assay, 5-ethynil-2-deoxyuridine (EdU) analysis and western blots respectively. We also detected the expression and activities of cell-cycle positive regulators (cyclin D1, cyclin E, CDK2, CDK4 and CDK6) and negative regulators (p21 and p27) in human PASMCs by western blots and co-immuoprecipitation (IP). Our results show that NBL1-induced growth suppression is associated with the decreased activity of cyclin D1–CDK4 and the decreased phosphorylation of p27 in PDGF-BB-treated human PASMCs. By western blots using the phosphor-specific antibodies, we further demonstrated that NBL1 induced growth suppression is mediated by blockade of the up-stream PDGF-receptor β (PDGFRβ)-p38 mitogen-activated protein kinase (MAPK). In conclusion, our results suggest that NBL1 could inhibit PDGF-BB-induced human PASMC proliferation, and the underlying mechanism is associated with the decreased cyclin D1–CDK4 activity and up-regulated p27 by decreasing the phosphorylation of p27 via blockade of PDGFRβ-p38MAPK signal cascade. Our findings may provide a potential therapeutic target for PAH.
Collapse
|