1
|
Qian C, Dong G, Yang C, Zheng W, Zhong C, Shen Q, Lu Y, Zhao Y. Broadening horizons: molecular mechanisms and disease implications of endothelial-to-mesenchymal transition. Cell Commun Signal 2025; 23:16. [PMID: 39789529 PMCID: PMC11720945 DOI: 10.1186/s12964-025-02028-y] [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: 10/15/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025] Open
Abstract
Endothelial-mesenchymal transition (EndMT) is defined as an important process of cellular differentiation by which endothelial cells (ECs) are prone to lose their characteristics and transform into mesenchymal cells. During EndMT, reduced expression of endothelial adhesion molecules disrupts intercellular adhesion, triggering cytoskeletal reorganization and mesenchymal transition. Numerous studies have proved that EndMT is a multifaceted biological event driven primarily by cytokines such as TGF-β, TNF-α, and IL-1β, alongside signaling pathways like WNT, Smad, MEK-ERK, and Notch. Nevertheless, the exact roles of EndMT in complicated diseases have not been comprehensively reviewed. In this review, we summarize the predominant molecular regulatory mechanisms and signaling pathways that contribute to the development of EndMT, as well as highlight the contributions of a series of imperative non-coding RNAs in curbing the initiation of EndMT. Furthermore, we discuss the significant impact of EndMT on worsening vasculature-related diseases, including cancer, cardiovascular diseases, atherosclerosis, pulmonary vascular diseases, diabetes-associated fibrotic conditions, and cerebral cavernous malformation, providing the implications that targeting EndMT holds promise as a therapeutic strategy to mitigate disease progression.
Collapse
Affiliation(s)
- Cheng Qian
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Guanglu Dong
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chunmei Yang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Weiwei Zheng
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chongjin Zhong
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qiuhong Shen
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yang Zhao
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
2
|
Zhang R, Lu M, Ran C, Niu L, Qi Q, Wang H. Ginsenoside Rg1 improves hypoxia-induced pulmonary vascular endothelial dysfunction through TXNIP/NLRP3 pathway-modulated mitophagy. J Ginseng Res 2025; 49:80-91. [PMID: 39872289 PMCID: PMC11764820 DOI: 10.1016/j.jgr.2024.10.002] [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: 06/12/2024] [Revised: 10/15/2024] [Accepted: 10/23/2024] [Indexed: 01/30/2025] Open
Abstract
Background Vascular endothelial dysfunction (VED) is one of the main pathogenic events in pulmonary arterial hypertension (PAH). Previous studies have demonstrated that the ginsenoside Rg1 (Rg1) can ameliorate PAH, but the mechanism by which Rg1 affects pulmonary VED in hypoxia-induced PAH remains unclear. Methods Network pharmacology, molecular docking and other experiments were used to explore the mechanisms by which Rg1 affects PAH. A PAH mouse model was established via hypoxia combined with the vascular endothelial growth factor (VEGFR) inhibitor su5416 (SuHx), and a cell model was established via hypoxia. The functions of Rg1 in VED, oxidative stress, inflammation, mitophagy, and TXNIP and NLRP3 expression were examined. Results In hypoxia-induced VED, progressive exacerbation of oxidative stress, inflammation, and mitophagy were observed, and were associated with elevated TXNIP and NLRP3 expression in vivo and in vitro. Rg1 improved hypoxia-induced impaired endothelium-dependent vasodilation and increased nitric oxide (NO) and endothelial NO synthase (eNOS) expression. Rg1, SRI37330 (a TXNIP inhibitor), MCC950 (an NLRP3 inhibitor), and Liensinine (a mitophagy inhibitor) attenuated oxidative stress, inflammation, and mitophagy by reducing the expression of TXNIP and NLRP3 in mice and cells. Furthermore, the combination of SB203580 (a mitophagy agonist) with Rg1 disrupted the protective effect of Rg1 on hypoxia-induced pulmonary artery and human pulmonary artery endothelial cells (HPAECs). Conclusion Rg1 improves hypoxia-induced pulmonary vascular endothelial dysfunction through TXNIP/NLRP3 pathway-modulated oxidative stress, inflammation and mitophagy.
Collapse
Affiliation(s)
- Ru Zhang
- The Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Meili Lu
- The Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Chenyang Ran
- The Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Linchao Niu
- The Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Qi Qi
- The Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Hongxin Wang
- The Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
3
|
Tan W, Liang Z, Tan X, Tan G. Ginsenoside Rg1 improves cigarette smoke-induced ferroptosis in COPD by regulating PERK/ATF4 axis to inhibit endoplasmic reticulum stress. Biochem Biophys Res Commun 2024; 739:150946. [PMID: 39531905 DOI: 10.1016/j.bbrc.2024.150946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/26/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Ferroptosis plays a key role in the development of chronic obstructive pulmonary disease (COPD). Whether ginsenoside Rg1 improves cigarette smoke-induced COPD or whether ginsenoside Rg1 improves COPD by inhibiting ferroptosis remains unknown. METHODS BEAS-2B cells were exposed to cigarette solution (CSE) for 24 h and treated with ginsenoside Rg1, the ferroptosis inhibitor Fer-1, and the PERK inhibitor GSK. Cell viability, endoplasmic reticulum stress, mitochondrial morphology, membrane potential, reactive oxygen species (ROS), iron levels, and the expression of related proteins were detected using corresponding methods. A COPD mouse model was constructed using cigarette smoke (CS). Ginsenoside Rg1 and GSK were administered via tube feeding 15 days after successful modeling. Mouse lung tissues were evaluated by HE staining. The expression of inflammatory markers, ROS, iron content, and related proteins was detected using corresponding methods. RESULTS The results demonstrated that in the CSE-exposed BEAS-2B cell model and CS-induced mouse COPD model, the expression levels of endoplasmic reticulum stress (ERS)-related factors such as GRP78 were increased, while those of the antioxidant markers GPX4 and GSH were significantly decreased. Ginsenoside Rg1 improved emphysema and inflammation by inhibiting ferroptosis in vivo and in vitro. Using a PERK inhibitor, we found that ginsenoside Rg1 inhibited ferroptosis in vivo and in vitro by regulating ERS. CONCLUSION This study showed that ginsenoside Rg1 alleviates cigarette smoke-induced COPD by regulating the PERK/ATF4 axis to inhibit ERS and ferroptosis.
Collapse
Affiliation(s)
- Wei Tan
- Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Zicheng Liang
- Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Xiaoning Tan
- Department of Oncology, The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, China.
| | - Guangbo Tan
- Department of Pulmonology, The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, China.
| |
Collapse
|
4
|
Guo X, Guo S, Tian F, Gao Z, Fan Y, Wang C, Xu S. CCN1 Promotes Mesenchymal Phenotype Transition Through Activating NF-κB Signaling Pathway Regulated by S100A8 in Glioma Stem Cells. CNS Neurosci Ther 2024; 30:e70128. [PMID: 39659236 PMCID: PMC11632201 DOI: 10.1111/cns.70128] [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: 07/24/2024] [Revised: 10/28/2024] [Accepted: 11/03/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND The presence of glioma stem cells (GSCs) and the occurrence of mesenchymal phenotype transition contribute to the miserable prognosis of glioblastoma (GBM). Cellular communication network factor 1 (CCN1) is upregulated within various malignancies and associated with cancer development and progression, while the implications of CCN1 in the phenotype transition and tumorigenicity of GSCs remain unclear. METHODS Data for bioinformatic analysis were obtained from The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) databases. A range of primary GBM and GSC cell models were then used to demonstrate the regulatory role of CCN1 via the phenotype validation, tumor sphere formation assays, extreme limiting dilution assays (ELDA), and transwell assays. To screen out the downstream signaling pathway, we employed high-throughput RNA-seq. Intracranial xenograft GSC mouse models were used to investigate the role of CCN1 in vivo. RESULTS Among the CCN family members, CCN1 was highly expressed in MES-GBM/GSCs and was correlated with a poor prognosis. Both in vitro and in vivo assays indicated that knockdown of CCN1 in MES-GSCs reduced the tumor stemness, proliferation, invasion, and tumorigenicity, whereas CCN1 overexpression in PN-GSCs exhibited the opposite effects. Mechanistically, CCN1 triggered the FAK/STAT3 signaling in autocrine and paracrine manners to upregulate the expression of S100A8. Knockdown of S100A8 inactivated NF-κB/p65 pathway and significantly suppressed the tumorigenesis of MES-GSCs. CONCLUSION Our findings reveal that CCN1 may be an important factor in the enhanced invasiveness and MES phenotype transition of GSCs and highlight the potential to target CCN1 for treating GBM.
Collapse
Affiliation(s)
- Xing Guo
- Department of NeurosurgeryQilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain‐Inspired Science, Shandong UniversityJinanShandongChina
| | - Shuhua Guo
- Department of Clinical LaboratoryThe Second Hospital of Shandong UniversityJinanShandongChina
| | - Feng Tian
- Department of Clinical LaboratoryThe Second Hospital of Shandong UniversityJinanShandongChina
| | - Zijie Gao
- Department of NeurosurgeryQilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain‐Inspired Science, Shandong UniversityJinanShandongChina
| | - Yang Fan
- Department of NeurosurgeryThe First Affiliated Hospital of Shandong First Medical University &Shandong Provincial Qianfoshan HospitalJinanShandongChina
| | - Chuanxin Wang
- Department of Clinical LaboratoryThe Second Hospital of Shandong UniversityJinanShandongChina
| | - Shuo Xu
- Department of NeurosurgeryQilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain‐Inspired Science, Shandong UniversityJinanShandongChina
| |
Collapse
|
5
|
Chen J, Zhang Z, Huang M, Yan J, Gao R, Cui J, Gao Y, Ma Z. Ginsenoside Rg1 Prevents and Treats Acute Pulmonary Injury Induced by High-Altitude Hypoxia. Int J Mol Sci 2024; 25:12051. [PMID: 39596120 PMCID: PMC11593513 DOI: 10.3390/ijms252212051] [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: 10/18/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
This study aimed to investigate the protective effects of ginsenoside Rg1 on high-altitude hypoxia-induced acute lung injury (ALI) and elucidated its molecular targets and related pathways, specifically its association with the fluid shear stress pathway. Using a combination of bioinformatics analysis and both in vivo and in vitro experiments, we assessed the role of ginsenoside Rg1 in mitigating physiological and biochemical disturbances induced by hypoxia. In the in vivo experiments, we measured arterial blood gas parameters, levels of inflammatory cells and cytokines, erythrocyte and platelet parameters, and conducted histological analysis in rats. The in vitro experiments utilized human pulmonary microvascular endothelial cells (HPMECs) and A549 cells to examine cell viability, intracellular reactive oxygen species (ROS) and Ca2⁺ levels, and mitochondrial function. The results of the in vivo experiments demonstrate that ginsenoside Rg1 significantly increased arterial blood oxygen partial pressure and saturation, elevated arterial blood glucose levels, and stabilized respiratory and metabolic functions in rats. It also reduced inflammatory cells and cytokines, such as tumor necrosis factor-α and interleukin-6, and improved erythrocyte and platelet abnormalities, supporting its protective role through the regulation of the fluid shear stress pathway. Histological and ultrastructural analyses revealed that Rg1 significantly protected lung tissue structure and organelles. In vitro experiments further confirmed that Rg1 improved cell viability in HPMEC and A549 cells under hypoxic conditions, decreased intracellular ROS and Ca2⁺ levels, and enhanced mitochondrial function. These findings collectively demonstrate that ginsenoside Rg1 exerts significant protective effects against high-altitude hypoxia-induced ALI by enhancing oxygen delivery and utilization, reducing inflammatory responses, and maintaining cellular metabolism and vascular function. Notably, the protective effects of Rg1 are closely associated with the regulation of the fluid shear stress pathway, suggesting its potential for treating high-altitude hypoxia-related diseases.
Collapse
Affiliation(s)
- Junru Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; (J.C.); (R.G.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Z.Z.); (M.H.); (J.Y.); (J.C.)
| | - Zhuo Zhang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Z.Z.); (M.H.); (J.Y.); (J.C.)
| | - Mingyue Huang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Z.Z.); (M.H.); (J.Y.); (J.C.)
| | - Jiayi Yan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Z.Z.); (M.H.); (J.Y.); (J.C.)
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rong Gao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; (J.C.); (R.G.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Z.Z.); (M.H.); (J.Y.); (J.C.)
| | - Jialu Cui
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Z.Z.); (M.H.); (J.Y.); (J.C.)
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yue Gao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; (J.C.); (R.G.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Z.Z.); (M.H.); (J.Y.); (J.C.)
| | - Zengchun Ma
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; (J.C.); (R.G.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Z.Z.); (M.H.); (J.Y.); (J.C.)
| |
Collapse
|
6
|
Ran C, Lu M, Zhao F, Hao Y, Guo X, Li Y, Su Y, Wang H. Ginsenoside Rg1 alleviates vascular remodeling in hypoxia-induced pulmonary hypertension mice through the calpain-1/STAT3 signaling pathway. J Ginseng Res 2024; 48:405-416. [PMID: 39036731 PMCID: PMC11258379 DOI: 10.1016/j.jgr.2024.03.001] [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: 07/05/2023] [Revised: 12/30/2023] [Accepted: 03/04/2024] [Indexed: 07/23/2024] Open
Abstract
Background Hypoxic pulmonary hypertension (HPH) is the main pathological change in vascular remodeling, a complex cardiopulmonary disease caused by hypoxia. Some research results have shown that ginsenoside Rg1 (Rg1) can improve vascular remodeling, but the effect and mechanism of Rg1 on hypoxia-induced pulmonary hypertension are not clear. The purpose of this study was to discuss the potential mechanism of action of Rg1 on HPH. Methods C57BL/6 mice, calpain-1 knockout mice and Pulmonary artery smooth muscle cells (PASMCs) were exposed to a low oxygen environment with or without different treatments. The effect of Rg1 and calpain-1 silencing on inflammation, fibrosis, proliferation and the protein expression levels of calpain-1, STAT3 and p-STAT3 were determined at the animal and cellular levels. Results At the mouse and cellular levels, hypoxia promotes inflammation, fibrosis, and cell proliferation, and the expression of calpain-1 and p-STAT3 is also increased. Ginsenoside Rg1 administration and calpain-1 knockdown, MDL-28170, and HY-13818 treatment showed protective effects on hypoxia-induced inflammation, fibrosis, and cell proliferation, which may be associated with the downregulation of calpain-1 and p-STAT3 expression in mice and cells. In addition, overexpression of calpain 1 increased p-STAT3 expression, accelerating the onset of inflammation, fibrosis and cell proliferation in hypoxic PASMCs. Conclusion Ginsenoside Rg1 may ameliorate hypoxia-induced pulmonary vascular remodeling by suppressing the calpain-1/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Chenyang Ran
- The Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Meili Lu
- The Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Fang Zhao
- Institute of Innovation and Applied Research in Chinese Medicine and Department of Rheumatology of the First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yi Hao
- The Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Xinyu Guo
- The Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Yunhan Li
- The Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Yuhong Su
- The College of Food and Health of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Hongxin Wang
- The Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
7
|
Zhou Z, Hu C, Cui B, You L, An R, Liang K, Wang X. Ginsenoside Rg1 Suppresses Pyroptosis via the NF-κB/NLRP3/GSDMD Pathway to Alleviate Chronic Atrophic Gastritis In Vitro and In Vivo. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 38855973 DOI: 10.1021/acs.jafc.4c01271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Chronic atrophic gastritis (CAG) is characterized by the loss of gastric glandular cells, which are replaced by the intestinal-type epithelium and fibrous tissue. Ginsenoside Rg1 (Rg1) is the prevalent ginsenoside in ginseng, with a variety of biological activities, and is usually added to functional foods. As a novel form of programmed cell death (PCD), pyroptosis has received substantial attention in recent years. Despite the numerous beneficial effects, the curative impact of Rg1 on CAG and whether its putative mechanism is partially via inhibiting pyroptosis still remain unknown. To address this gap, we conducted a study to explore the mechanisms underlying the potential anti-CAG effect of Rg1. We constructed a CAG rat model using a multifactor comprehensive method. A cellular model was developed by using 1-methyl-3-nitro-1-nitrosoguanidine (MNNG) combined with Nigericin as a stimulus applied to GES-1 cells. After Rg1 intervention, the levels of inflammatory indicators in the gastric tissue/cell supernatant were reduced. Rg1 relieved oxidative stress via reducing the myeloperoxidase (MPO) and malonaldehyde (MDA) levels in the gastric tissue and increasing the level of superoxide dismutase (SOD). Additionally, Rg1 improved MNNG+Nigericin-induced pyroptosis in the morphology and plasma membrane of the cells. Further research supported novel evidence for Rg1 in the regulation of the NF-κB/NLRP3/GSDMD pathway and the resulting pyroptosis underlying its therapeutic effect. Moreover, by overexpression and knockout of GSDMD in GES-1 cells, our findings suggested that GSDMD might serve as the key target in the effect of Rg1 on suppressing pyroptosis. All of these offer a potential theoretical foundation for applying Rg1 in ameliorating CAG.
Collapse
Affiliation(s)
- Zehua Zhou
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Cheng Hu
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Bo Cui
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lisha You
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Rui An
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Kun Liang
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xinhong Wang
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
8
|
Liu J, Fang G, Lan C, Qiu C, Yao L, Zhang Q, Hu J, Zhang Y, Yang Y, Zhang Y. Forsythoside B Mitigates Monocrotaline-Induced Pulmonary Arterial Hypertension via Blocking the NF-κB Signaling Pathway to Attenuate Vascular Remodeling. Drug Des Devel Ther 2024; 18:767-780. [PMID: 38495631 PMCID: PMC10942864 DOI: 10.2147/dddt.s444605] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/24/2024] [Indexed: 03/19/2024] Open
Abstract
Purpose Pulmonary arterial hypertension (PAH) is a devastating disease with little effective treatment. The proliferation of pulmonary artery smooth muscle cells (PASMCs) induced by the nuclear factor-κB (NF-κB) signaling activation plays a pivotal role in the pathogenesis of PAH. Forsythoside B (FTS•B) possesses inhibitory effect on NF-κB signaling pathway. The present study aims to explore the effects and mechanisms of FTS•B in PAH. Methods Sprague-Dawley rats received monocrotaline (MCT) intraperitoneal injection to establish PAH model, and FTS•B was co-treated after MCT injection. Right ventricular hypertrophy and pulmonary artery pressure were measured by echocardiography and right heart catheterization, respectively. Histological alterations were detected by H&E staining and immunohistochemistry. FTS•B's role in PASMC proliferation and migration were evaluated by CCK-8 and wound healing assay. To investigate the underlying mechanisms, Western blotting, immunofluorescence staining and ELISA were conducted. The NF-κB activator PMA was used to investigate the role of NF-κB in FTS•B's protective effects against PAH. Results FTS•B markedly alleviated MCT-induced vascular remodeling and pulmonary artery pressure, and improved right ventricular hypertrophy and survival. FTS•B also reversed PDGF-BB-induced PASMC proliferation and migration, decreased PCNA and CyclinD1 expression in vitro. The elevated levels of IL-1β and IL-6 caused by MCT were decreased by FTS•B. Mechanistically, MCT-triggered phosphorylation of p65, IκBα, IKKα and IKKβ was blunted by FTS•B. FTS•B also reversed MCT-induced nuclear translocation of p65. However, all these protective effects were blocked by PMA-mediated NF-κB activation. Conclusion FTS•B effectively attenuates PAH by suppressing the NF-κB signaling pathway to attenuate vascular remodeling. FTS•B might be a promising drug candidate with clinical translational potential for the treatment of PAH.
Collapse
Affiliation(s)
- Jiying Liu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, Sichuan, 610083, People’s Republic of China
- Department of Cardiology, The Third People’s Hospital of Yibin, Yibin, Sichuan, 644000, People’s Republic of China
| | - Guangyao Fang
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, Sichuan, 610083, People’s Republic of China
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610083, People’s Republic of China
| | - Cong Lan
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, Sichuan, 610083, People’s Republic of China
| | - Chenming Qiu
- Department of Burn and Plastic Surgery, General Hospital of Western Theater Command, Chengdu, Sichuan, 610083, People’s Republic of China
| | - Li Yao
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, Sichuan, 610083, People’s Republic of China
| | - Qian Zhang
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, Sichuan, 610083, People’s Republic of China
| | - Jingtang Hu
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, Sichuan, 610083, People’s Republic of China
| | - Yaolei Zhang
- Basic Medical Laboratory, General Hospital of Western Theater Command, Chengdu, Sichuan, 610083, People’s Republic of China
| | - Yongjian Yang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, Sichuan, 610083, People’s Republic of China
| | - Yan Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, Sichuan, 610083, People’s Republic of China
| |
Collapse
|
9
|
Ba L, E M, Wang R, Wu N, Wang R, Liu R, Feng X, Qi H, Sun H, Qiao G. Triptolide attenuates cardiac remodeling by inhibiting pyroptosis and EndMT via modulating USP14/Keap1/Nrf2 pathway. Heliyon 2024; 10:e24010. [PMID: 38293551 PMCID: PMC10825440 DOI: 10.1016/j.heliyon.2024.e24010] [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: 08/04/2023] [Revised: 12/21/2023] [Accepted: 01/02/2024] [Indexed: 02/01/2024] Open
Abstract
Background Cardiac remodeling is a common pathological feature in many cardiac diseases, characterized by cardiac hypertrophy and fibrosis. Triptolide (TP) is a natural compound derived from Tripterygium wilfordii Hook F. However, the related mechanism of it in cardiac remodeling has not been fully understood. Methods and results Transverse aortic constriction (TAC)-induced cardiac hypertrophic mouse model and angiotensin II (Ang II)-induced cardiomyocytes hypertrophic model were performed. Firstly, the results indicate that TP can improve cardiac function, decreased cardiomyocyte surface area and fibrosis area, as well as lowered the protein expressions of brain natriuretic peptide (BNP), β-major histocompatibility complex (β-MHC), type I and III collagen (Col I and III). Secondly, TP suppressed cardiac pyroptosis, and decreased the levels of Interleukin-1β (IL-1β), Interleukin-18 (IL-18) by Enzyme-linked immunosorbent assay (ELISA), and pyroptosis-associated proteins. Furthermore, TP enhanced the expressions of Nuclear factor erythroid 2-related factor 2 (Nrf2) and Heme oxygenase 1 (HO-1). Interestingly, when Nrf2 was silenced by siRNA, TP lost its properties of reducing pyroptosis and cardiac hypertrophy. In addition, in the Transforming Growth Factor β1 (TGF-β1)-induced primary human coronary artery endothelial cells (HCAEC) model, TP was found to inhibit the process of endothelial-to-mesenchymal transition (EndMT), characterized by the loss of endothelial-specific markers and the gain of mesenchymal markers. This was accompanied by a suppression of Slug, Snail, and Twist expression. Meanwhile, the inhibitory effect of TP on EndMT was weakened when Nrf2 was silenced by siRNA. Lastly, potential targets of TP were identified through network pharmacology analysis, and found that Ubiquitin-Specific Protease 14 (USP14) was one of them. Simultaneously, the data indicated that decrease the upregulation of USP14 and Kelch-like ECH-Associated Protein 1 (Keap1) caused by cardiac remodeling. However, Keap1 was decreased and Nrf2 was increased when USP14 was silenced. Furthermore, CoIP analysis showed that USP14 directly interacts with Keap1. Conclusion TP can observably reduce pyroptosis and EndMT by targeting the USP14/Keap1/Nrf2 pathway, thereby significantly attenuating cardiac remodeling.
Collapse
Affiliation(s)
- Lina Ba
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, 163319, China
| | - Mingyao E
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, 163319, China
- Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Ruixuan Wang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, 163319, China
| | - Nan Wu
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, 163319, China
| | - Rui Wang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, 163319, China
| | - Renling Liu
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, 163319, China
| | - Xiang Feng
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, 163319, China
| | - Hanping Qi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, 163319, China
| | - Hongli Sun
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, 163319, China
| | - Guofen Qiao
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| |
Collapse
|
10
|
Zhao F, Pan C, Zhang Y, Yang J, Xing X. Polyphyllin VII alleviates pulmonary hypertension by inducing miR-205-5p to target the β-catenin pathway. Biomed Pharmacother 2023; 167:115516. [PMID: 37717533 DOI: 10.1016/j.biopha.2023.115516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/01/2023] [Accepted: 09/12/2023] [Indexed: 09/19/2023] Open
Abstract
OBJECTIVE This study aims to investigate the impact of Polyphyllin VII (PP7) on pulmonary hypertension (PH) and elucidate the underlying mechanism involving microRNA (miR)-205-5p/β-catenin. METHODS The PH rat model was induced through hypoxia exposure. The effects of intraperitoneal injection of PP7 on pulmonary artery tissue pathology, hemodynamics, miR-205-5p expression and β-catenin protein levels were assessed. In vitro, pulmonary arterial smooth muscle cells (PASMCs) were subjected to hypoxic conditions. Moreover, miR-205-5p and/or β-catenin were overexpressed through transfection. PASMCs were pre-cultured in 20 μM PP7, and subsequent measurements included proliferation, apoptosis and vascular remodeling protein expression. RESULTS PP7 ameliorated PH symptoms in rats, upregulated miR-205-5p expression and inhibited β-catenin protein expression. Furthermore, miR-205-5p upregulation inhibited β-catenin expression in PASMCs. The overexpression of β-catenin aggravated hypoxia-induced proliferation, inhibited apoptosis and further augmented VEGF and α-SMA protein expression. Additionally, miR-205-5p overexpression alleviated the hypoxia-induced PASMC proliferation and apoptosis by inhibiting β-catenin protein expression. Under hypoxic conditions, PP7 significantly elevated miR-205-5p while downregulating β-catenin protein expression. Furthermore, inhibiting miR-205-5p counteracted the inhibitory effect of PP7 on β-catenin, consequently blocking the regulatory role of PP7 in PASMC proliferation and apoptosis. CONCLUSION PP7 likely modulates β-catenin protein levels by promoting miR-205-5p expression, thereby alleviating PH, vascular remodeling and airway smooth muscle remodeling.
Collapse
Affiliation(s)
- Fangyun Zhao
- Department of Pharmacy, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Chunhong Pan
- Department of Pharmacy, The First People's Hospital of Kunming City & Calmette Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yue Zhang
- Department of Pharmacy, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Jiao Yang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Xiqian Xing
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Yunnan University, Kunming, China.
| |
Collapse
|
11
|
Liu H, Sun M, Wu N, Liu B, Liu Q, Fan X. TGF-β/Smads signaling pathway, Hippo-YAP/TAZ signaling pathway, and VEGF: Their mechanisms and roles in vascular remodeling related diseases. Immun Inflamm Dis 2023; 11:e1060. [PMID: 38018603 PMCID: PMC10629241 DOI: 10.1002/iid3.1060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 10/03/2023] [Accepted: 10/11/2023] [Indexed: 11/30/2023] Open
Abstract
Vascular remodeling is a basic pathological process in various diseases characterized by abnormal changes in the morphology, structure, and function of vascular cells, such as migration, proliferation, hypertrophy, and apoptosis. Various growth factors and pathways are involved in the process of vascular remodeling. The transforming growth factor-β (TGF-β) signaling pathway, which is mainly mediated by TGF-β1, is an important factor in vascular wall enhancement during vascular development and regulates the vascular response to injury by promoting the accumulation of intimal tissue. Vascular endothelial growth factor (VEGF) has an important effect on initiating the formation of blood vessels. The Hippo-YAP/TAZ signaling pathway also plays an important role in angiogenesis. In addition, studies have shown that there is a certain interaction between the TGF-β/Smads signaling pathway, Hippo-YAP/TAZ signaling pathway, and VEGF. Many studies have shown that in the development of atherosclerosis, hypertension, aneurysm, vertebrobasilar dolichoectasia, pulmonary hypertension, restenosis after percutaneous transluminal angioplasty, and other diseases, various inflammatory reactions lead to changes in vascular structure and vascular microenvironment, which leads to vascular remodeling. The occurrence of vascular remodeling changes the morphology of blood vessels and thus changes the hemodynamics, which is the cause of further development of the disease process. Vascular remodeling can cause vascular smooth muscle cell dysfunction and vascular homeostasis regulation. This review aims to explore the mechanisms of the TGF-β/Smads signaling pathway, Hippo-YAP/TAZ signaling pathway, and vascular endothelial growth factor in vascular remodeling and related diseases. This paper is expected to provide new ideas for research on the occurrence and development of related diseases and provide a new direction for research on the treatment of related diseases.
Collapse
Affiliation(s)
- Hui Liu
- Department of NeurologyBinzhou Medical University HospitalBinzhouChina
| | - Mingyue Sun
- Department of NeurologyBinzhou Medical University HospitalBinzhouChina
| | - Nan Wu
- Department of NeurologyBinzhou Medical University HospitalBinzhouChina
| | - Bin Liu
- Institute for Metabolic & Neuropsychiatric DisordersBinzhou Medical University HospitalBinzhouChina
| | - Qingxin Liu
- Department of NeurologyBinzhou Medical University HospitalBinzhouChina
| | - Xueli Fan
- Department of NeurologyBinzhou Medical University HospitalBinzhouChina
| |
Collapse
|
12
|
Li Q, Zhang H. Bioinformatics analysis to identify potential biomarkers for the pulmonary artery hypertension associated with the basement membrane. Open Life Sci 2023; 18:20220730. [PMID: 37772261 PMCID: PMC10523280 DOI: 10.1515/biol-2022-0730] [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: 06/15/2023] [Revised: 08/07/2023] [Accepted: 08/26/2023] [Indexed: 09/30/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rapidly progressing cardiopulmonary disease. It is characterized by increased pulmonary artery pressure and vascular resistance. The most notable histopathological characteristic is vascular remodeling. The changes in the basement membrane (BM) are believed to be related to vascular remodeling. It is crucial to identify potential biomarkers associated with the BM in PAH, to guide its treatment. The microarray datasets GSE117261 and GSE113439 were downloaded from the Gene Expression Omnibus. Two data sets were examined to identify genes associated with the BM by analyzing gene expression changes. Next, we analyzed the relevant genes in the Kyoto Encyclopedia of Genes and Genomes using Gene Ontology and Disease Ontology annotationand conducted pathway enrichment analysis. We conducted a protein-protein interaction network analysis on the genes related to BMs and used the cell cytoHubba plug-in to identify the hub genes. Furthermore, we conducted an immune infiltration analysis and implemented a histogram model. Finally, we predicted and analyzed potential therapeutic drugs for PAH and set up a miRNA network of genetic markers. Six candidate genes related to BMs, namely Integrin Subunit Alpha V, Integrin Subunit Alpha 4, ITGA2, ITGA9, Thrombospondin 1, and Collagen Type IV Alpha 3 Chain, were identified as potential modulators of the immune process in PAH. Furthermore, ginsenoside Rh1 was found to significantly impact drug targeting based on its interactions with the six BM-related genes identified earlier. A novel biomarker related to the BM, which plays a crucial role in the development of PAH, has been identified.
Collapse
Affiliation(s)
- Qian Li
- Department of Cardiac Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming650000, China
| | - Hu Zhang
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming650000, China
| |
Collapse
|