1
|
Liu X, Tan X, Su Y, Zou L, Yuan W, Yu C. Anthocyanin attenuates pulmonary arterial hypertension and associated heart failure via improving mitochondrial function through Nrf2-dependent mechanism. Clin Exp Hypertens 2025; 47:2503805. [PMID: 40343792 DOI: 10.1080/10641963.2025.2503805] [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: 04/04/2025] [Revised: 04/29/2025] [Accepted: 05/04/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND Mitochondrial dysfunction of pulmonary vascular endothelial cells is one of the important pathogenesis of pulmonary arterial hypertension (PAH). Anthocyanin can protect mitochondrial function, but whether anthocyanin can prevent PAH and its related mechanism has not been reported. METHODS Using rodent PAH models induced by chronic hypoxia for 21 days, we investigated the changes of hemodynamics, histopathology and vascular endothelial function after anthocyanin treatment for 21 days. RESULTS We found that anthocyanin treatment improved pulmonary endothelial function, vascular remodeling and associated right heart failure. Mechanistically, we found that anthocyanin treatment promoted the nucleus translocation of Nrf2 and improved the impaired mitochondrial function. The beneficial effects of anthocyanin on regulation of mitochondrial function were abolished by inhibition of Nrf2. Finally, in Nrf2 deficient mice, the protective effects of anthocyanin on PAH were largely vanished. CONCLUSIONS Collectively, we showed that anthocyanin attenuated PAH and associated right heart failure via improving mitochondrial function through Nrf2-dependent mechanism, suggesting that anthocyanin may represent a novel therapeutic potential for PAH.
Collapse
Affiliation(s)
- Xiao Liu
- Department of Cardiology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoli Tan
- Department of Cardiology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Yidan Su
- Department of Cardiology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Luohao Zou
- Department of Cardiology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Wenhui Yuan
- Department of Cardiology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Changqing Yu
- Department of Cardiology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
Yang Q, Liang Y, Wang R, Zhang T, Chai R, Yan Y, Tie Y, Wang Y, Sun X, Cai Y, Zhao X. The transcription factor BMAL1 inhibits endothelial cell apoptosis by targeting STAT6 to repress its expression. Cell Signal 2025; 132:111812. [PMID: 40246133 DOI: 10.1016/j.cellsig.2025.111812] [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: 02/20/2025] [Revised: 04/08/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
Corneal transparency is critical for optimal visual function, and corneal neovascularization represents the primary cause of visual impairment globally. Recent studies have identified the transcription factor BMAL1 as a significant regulator of angiogenesis. However, its specific role and underlying mechanisms in endothelial cell apoptosis remain inadequately understood. This study seeks to elucidate the role and underlying mechanisms of BMAL1 in endothelial cell apoptosis by employing genetic modification, alkali-burned mouse corneal neovascularization models, lentiviral transfection, proteomic analysis, and other complementary methodologies. Our results showed that BMAL1 expression is significantly elevated in corneal neovascularization induced by alkali burn and removal of Bmal1 in endothelial cells resulted in the suppression of corneal neovascularization in alkali burn mouse models. In vivo experiments have demonstrated that the knockout of Bmal1 in endothelial cells leads to an increase in endothelial cell apoptosis. Complementary in vitro studies revealed that overexpression of BMAL1 in endothelial cells inhibits apoptosis, while knockdown of BMAL1 promotes apoptosis. Proteomic analysis identified STAT6 as a downstream target of BMAL1 involved in the regulation of endothelial cell apoptosis. Further cell salvage experiments confirmed that BMAL1 modulates endothelial cell apoptosis through the regulation of STAT6 expression. Finally, the results of dual-luciferase reporter assay demonstrated that BMAL1 exerts transcriptional repressive effects on the promoter bound by STAT6. This study elucidates the novel role and mechanism of BMAL1 in the regulation of angiogenesis and endothelial cell apoptosis, thereby identifying a potential therapeutic target for the treatment of vascular diseases such as corneal neovascularization.
Collapse
Affiliation(s)
- Qi Yang
- Department of Ophthalmology, General Hospital of Xinjiang Military Command, Urumqi 830000, China; Key Laboratory of Ministry of Education, Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China; Graduate School, Xinjiang Medical University, Urumqi 830054, China
| | - Ya'nan Liang
- Key Laboratory of Ministry of Education, Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Rui Wang
- Key Laboratory of Ministry of Education, Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Tongmei Zhang
- Key Laboratory of Ministry of Education, Center of Clinical Aerospace Medicine, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Ruiqing Chai
- Key Laboratory of Ministry of Education, Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China; Northwest University School of Medicine, Xi'an 710069, China
| | - Yiquan Yan
- Key Laboratory of Ministry of Education, Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China; Department of Internal Medicine, Hospital of Unit 96608, PLA, Hanzhong 723000, China
| | - Yateng Tie
- Key Laboratory of Ministry of Education, Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Yongchun Wang
- Key Laboratory of Ministry of Education, Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Xiqing Sun
- Key Laboratory of Ministry of Education, Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Yan Cai
- Department of Ophthalmology, General Hospital of Xinjiang Military Command, Urumqi 830000, China.
| | - Xingcheng Zhao
- Key Laboratory of Ministry of Education, Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
3
|
Mavaddatiyan L, Naeini S, Khodabandeh S, Hosseini F, Skelton RP, Azizi V, Talkhabi M. Exploring the association between aging, ferroptosis, and common age-related diseases. Arch Gerontol Geriatr 2025; 135:105877. [PMID: 40339241 DOI: 10.1016/j.archger.2025.105877] [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: 10/01/2024] [Revised: 04/14/2025] [Accepted: 04/26/2025] [Indexed: 05/10/2025]
Abstract
Aging is a natural biological process that is characterized by the progressive decline in physiological functions and an increased vulnerability to age-related diseases. The aging process is driven by different cell and molecular mechanisms. It has recently been shown that aging is associated with heightened vulnerability to ferroptosis (an intracellular iron-dependent form of programmed cell death). This susceptibility arises from various factors including oxidative stress, impaired antioxidant defences, and dysregulated iron homeostasis. The progressive decline in cellular antioxidant capacity and the accumulation of damaged components contribute to the increased susceptibility of aging cells to ferroptosis. Dysregulation of key regulators involved in ferroptosis, such as glutathione peroxidase 4 (GPX4), iron regulatory proteins, and lipid metabolism enzymes, further exacerbates this vulnerability. The decline in cellular defence mechanisms against ferroptosis during aging contributes to the accumulation of damaged cells and tissues, ultimately resulting in the manifestation of age-related diseases. Understanding the intricate relevance between aging and ferroptosis holds significant potential for developing strategies to counteract the detrimental effects of aging and age-related diseases. This will subsequently act to mitigate the negative consequences of aging and improving overall health in the elderly population. This review aims to clarify the relationship between aging and ferroptosis, and explores the underlying mechanisms and implications for age-related disorders, including neurodegenerative, cardiovascular, and neoplastic diseases. We also discuss the accumulating evidence suggesting that the imbalance of redox homeostasis and perturbations in iron metabolism contribute to the age-associated vulnerability to ferroptosis.
Collapse
Affiliation(s)
- Laleh Mavaddatiyan
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - SaghiHakimi Naeini
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Sara Khodabandeh
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Fatemeh Hosseini
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - RhysJ P Skelton
- Flinders Medical Centre, Department of Ophthalmology, Bedford Park, Australia
| | - Vahid Azizi
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Mahmood Talkhabi
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
4
|
Zhong C, Deng K, Lang X, Shan D, Xie Y, Pan W, Yu J. Therapeutic potential of natural flavonoids in atherosclerosis through endothelium-protective mechanisms: An update. Pharmacol Ther 2025; 271:108864. [PMID: 40274196 DOI: 10.1016/j.pharmthera.2025.108864] [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: 12/10/2024] [Revised: 02/27/2025] [Accepted: 04/20/2025] [Indexed: 04/26/2025]
Abstract
Atherosclerosis and its associated cardiovascular complications remain significant global public health challenges, underscoring the urgent need for effective therapeutic strategies. Endothelial cells are critical for maintaining vascular health and homeostasis, and their dysfunction is a key contributor to the initiation and progression of atherosclerosis. Targeting endothelial dysfunction has, therefore, emerged as a promising approach for the prevention and management of atherosclerosis. Among natural products, flavonoids, a diverse class of plant-derived phenolic compounds, have garnered significant attention for their anti-atherosclerotic properties. A growing body of evidence demonstrates that flavonoids can mitigate endothelial dysfunction, highlighting their potential as endothelial dysfunction-targeted therapeutics for atherosclerosis. In this review, we summarize current knowledge on the roles of natural flavonoids in modulating various aspects of endothelial dysfunction and their therapeutic effects on atherosclerosis, focusing on the underlying molecular mechanisms. We also discuss the challenges and future prospects of translating natural flavonoids into clinical applications for cardiovascular medicine. This review aims to provide critical insights to advance the development of novel endothelium-protective pharmacotherapies for atherosclerosis.
Collapse
Affiliation(s)
- Chao Zhong
- Center for Translational Medicine, Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Keke Deng
- Center for Translational Medicine, Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Xiaoya Lang
- Center for Translational Medicine, Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Dan Shan
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Yanfei Xie
- Center for Translational Medicine, Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Wen Pan
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi University of Chinese Medicine, Nanchang 330006, China.
| | - Jun Yu
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
5
|
Zhang X, Sha Y, Wu Y, Guan H, Yang X, Wang W, Zhang W, Liu Y, Zhu L, Li Q. Targeting endothelial cells: A novel strategy for pulmonary fibrosis treatment. Eur J Pharmacol 2025; 997:177472. [PMID: 40054716 DOI: 10.1016/j.ejphar.2025.177472] [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: 09/30/2024] [Revised: 01/10/2025] [Accepted: 03/04/2025] [Indexed: 04/12/2025]
Abstract
Endothelial cells (ECs) are a monolayer of flat cells lining the inner surfaces of blood and lymphatic vessels. They play a key role in many physiological and pathological processes. Specifically, they maintain vascular permeability and structural stability and participate in immune responses, inflammation, coagulation, and other vital functions. ECs play a decisive role in various age-related diseases; however, their involvement in pulmonary fibrosis (PF) remains poorly understood. PF refers to a group of chronic interstitial lung diseases characterised by progressive scarring of the pulmonary parenchyma, primarily caused by aberrant tissue repair mechanisms. These changes lead to irreversible loss of lung function. Although the exact pathophysiological mechanism underlying PF has not yet been elucidated, recent studies have indicated that ECs may play a pivotal role in PF. This review outlines the involvement of pulmonary vascular ECs in PF, focusing on the regulation of vascular remodelling and endothelial barrier integrity and on the maintenance of angiogenesis through EC-specific markers, such as vascular endothelial growth factor. This review also explores processes such as endothelial-to-mesenchymal transition, immune cell interactions, anti-EC antibody reactions, metabolic dysregulation, and cellular senescence. By elucidating recent advancements in understanding the role of ECs in PF and examining drugs targeting ECs for the treatment of PF, this study provides novel insights into the pathological mechanisms of PF and the development of endothelium-based therapeutic agents.
Collapse
Affiliation(s)
- Xin Zhang
- Medical College, Anhui University of Science and Technology, Huainan, Anhui, 232001, China; Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Yuxia Sha
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Yu Wu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Haiyang Guan
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Xu Yang
- Medical College, Anhui University of Science and Technology, Huainan, Anhui, 232001, China
| | - Wenjin Wang
- Medical College, Anhui University of Science and Technology, Huainan, Anhui, 232001, China
| | - Wenlong Zhang
- Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Yunyun Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Lili Zhu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Qing Li
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| |
Collapse
|
6
|
Wang LT, Yin HL, Jin YM, Hu DD, Yang XX, Sheng J, Huang YW, Wang XJ. Ellagic acid ameliorates atherosclerosis by inhibiting PCSK9 through the modulation of FoxO3 and HNF1α. Nutrition 2025; 134:112717. [PMID: 40086009 DOI: 10.1016/j.nut.2025.112717] [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: 10/29/2024] [Revised: 01/10/2025] [Accepted: 02/13/2025] [Indexed: 03/16/2025]
Abstract
INTRODUCTION Proprotein convertase subtilisin/kexin type 9 (PCSK9) hinders the clearance of low-density lipoprotein cholesterol (LDL-C) by promoting the degradation of the low-density lipoprotein receptor (LDLR), leading to the accumulation of LDL-C and thus becoming an important cause of atherosclerosis. Ellagic acid, a naturally occurring polyphenol widely present in fruits, vegetables, and nuts, has attracted significant attention due to its potential role in the prevention and treatment of cardiovascular diseases. However, the molecular mechanisms by which ellagic acid alleviates atherosclerosis by inhibiting PCSK9 are not fully understood. MATERIALS AND METHODS This study further validated the mechanism of action of ellagic acid through in vitro HepG2 cell experiments and a high-fat diet-induced ApoE-/- mouse model. RESULTS The results showed that ellagic acid significantly reduced the expression and secretion of PCSK9 while upregulating LDLR protein levels; its mechanism is related to the inhibition of hepatocyte nuclear factor 1α (HNF1α) expression and the promotion of forkhead box O3 (FoxO3) expression increase. Additionally, ellagic acid reduced aortic plaque deposition in mice induced by a high-fat diet; consistent with the in vitro experimental results, ellagic acid lowered the expression and secretion of PCSK9 and elevated LDLR protein levels by inhibiting HNF1α and increased FoxO3 expression. CONCLUSIONS In summary, this study demonstrates that ellagic acid inhibits PCSK9 by regulating HNF1α and FoxO3, thereby increasing LDLR levels and alleviating atherosclerosis. This finding not only consolidates the scientific basis of plant-based diets for preventing cardiovascular diseases but also provides an important direction for developing functional foods and nutritional intervention strategies based on natural polyphenols.
Collapse
Affiliation(s)
- Li-Tian Wang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China; Faculty of basic medical science, Kunming medical university, Kunming, China
| | - Huai-Liu Yin
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China; College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Ya-Min Jin
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China; College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Dan-Dan Hu
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China; College of Science, Yunnan Agricultural University, Kunming, China
| | - Xiang-Xuan Yang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China; College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Jun Sheng
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China; State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China.
| | - Ye-Wei Huang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China; Yunnan Plateau Characteristic Agricultural Industry Research Institute, Yunnan Agricultural University, Kunming, China.
| | - Xuan-Jun Wang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China; College of Resources, Environment, and Chemistry, Chuxiong Normal University, Chuxiong, China.
| |
Collapse
|
7
|
Zhou J, Yang F, Zhang X, Wang C, Wu Z, Gao J. Jiangniaosuan formula inhibits hyperuricemia-induced apoptosis of renal tubular epithelial cells via ROS/HIF-1α/EZH2 pathway: A network pharmacology analysis and experimental validation. Bioorg Chem 2025; 159:108363. [PMID: 40088688 DOI: 10.1016/j.bioorg.2025.108363] [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: 01/03/2025] [Revised: 02/28/2025] [Accepted: 03/09/2025] [Indexed: 03/17/2025]
Abstract
OBJECTIVE This study aimed to explore the main chemical components of Jiangniaosuan Formula (JNSF), the therapeutic effect of JNSF on hyperuricemia (HUA) mice, and the underlying mechanism by which JNSF inhibits renal tubular epithelial cell apoptosis. METHODS Ultra Performance Liquid Chromatography-Quadrupole-Time of Flight Mass Spectrometry (UPLC-Q-TOF-MS) was used to analyze the chemical composition of JNSF and its serum metabolites. Network pharmacology was performed to predict the potential target genes and pathways. In vitro and in vivo models were established to verify the lower serum uric acid (SUA) and renal protective effects. RESULTS UPLC-Q-TOF-MS identified 61 chemical compounds in JNSF and 56 metabolites in serum after oral administration. Network pharmacology suggested that Hypoxia-Inducible Factor 1-Alpha (HIF-1α), Cysteine-dependent Aspartate-specific Protease-3 (Caspase-3) and B-cell Lymphoma 2 (Bcl-2) might be the therapeutic targets of JNSF for the HUA treatment and JNSF may exert the therapeutic effect on uric acid nephropathy (UAN) through regulating HIF-1α signaling pathway and apoptosis pathway. In vivo experiments showed that JNSF could reduce SUA, protect renal function and tubular function, alleviate renal interstitial edema and fibrosis, reduce the expression of Reactive Oxygen Species (ROS), HIF-1α and Enhancer of Zeste Homolog 2 (EZH2), and inhibit cell apoptosis in HUA mice. In vitro experiments demonstrated that JNSF reversed apoptosis induced by EZH2 overexpression plasmid. Furthermore, we found that UA could promote the binding of HIF-1α to EZH2 protein and its promoter, enhancing EZH2 transcription, suggesting that JNSF could alleviate the progression of HUA-induced kidney injury by inhibiting the activation of ROS/HIF-1α/EZH2 pathway. CONCLUSION JNSF may attenuate HUA-induced renal injury by inhibiting apoptosis through the downregulation of ROS/HIF-1α/EZH2 pathway.
Collapse
Affiliation(s)
- Jiabao Zhou
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, China; TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, China; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, China; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, China
| | - Feng Yang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, China; TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, China; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, China; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, China
| | - Xuming Zhang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, China; TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, China; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, China; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, China
| | - Chuanxu Wang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, China; TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, China; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, China; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, China
| | - Zhiyuan Wu
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, China; TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, China; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, China; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, China
| | - Jiandong Gao
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, China; TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, China; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, China; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, China.
| |
Collapse
|
8
|
Hou A, Wu Y, Liu Y, Liu C, Yao H, Zeng X. Regulation of a chromenylium fluorophore with a built-in recognition site for the detection of sulfite. Org Biomol Chem 2025; 23:5148-5153. [PMID: 40304617 DOI: 10.1039/d5ob00504c] [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: 05/02/2025]
Abstract
A double charged cationic pyridinium-fused chromenylium analogue DMCQ with an acceptor-π-acceptor (A-π-A) electronic structure was designed and prepared for the sensitive and selective detection of SO32-. Undergoing a Michael addition reaction with SO32- at the built-in pyridinium moiety in the chromenylium fluorophore, the π-electron structure of the chromenylium skeleton transferred from an (A-π-A) to (A-π-D) system along with a prominent fluorescence enhancement. Based on this specific addition reaction, the DMCQ probe exhibits high selectivity for the detection of SO32- over other biologically related species within a wide pH range (4-10). As the highly electron-deficient nature of the pyridinium-fused chromenylium accelerated the reaction with SO32-, the probe presented a rapid response time (60 s), together with high sensitivity (33 nM). Besides, DMCQ could not only image SO32- in living cells but also monitor SO32- in food and water samples with a satisfactory recovery of 81.30-112.89%.
Collapse
Affiliation(s)
- Anqi Hou
- Tianjin Key Laboratory for Photoelectric Materials and Devices, School of Materials Science & Engineering, Tianjin University of Technology, Tianjin 300384, China.
- Key Laboratory of Display Materials and Photoelectric Devices, Ministry of Education, School of Materials Science & Engineering, Tianjin University of Technology, Tianjin, 300384, China
| | - Yuanyuan Wu
- Tianjin Key Laboratory for Photoelectric Materials and Devices, School of Materials Science & Engineering, Tianjin University of Technology, Tianjin 300384, China.
- Key Laboratory of Display Materials and Photoelectric Devices, Ministry of Education, School of Materials Science & Engineering, Tianjin University of Technology, Tianjin, 300384, China
| | - Yumeng Liu
- Tianjin Key Laboratory for Photoelectric Materials and Devices, School of Materials Science & Engineering, Tianjin University of Technology, Tianjin 300384, China.
- Key Laboratory of Display Materials and Photoelectric Devices, Ministry of Education, School of Materials Science & Engineering, Tianjin University of Technology, Tianjin, 300384, China
| | - Chang Liu
- Tianjin Key Laboratory for Photoelectric Materials and Devices, School of Materials Science & Engineering, Tianjin University of Technology, Tianjin 300384, China.
- Key Laboratory of Display Materials and Photoelectric Devices, Ministry of Education, School of Materials Science & Engineering, Tianjin University of Technology, Tianjin, 300384, China
| | - Huirong Yao
- Tianjin Key Laboratory for Photoelectric Materials and Devices, School of Materials Science & Engineering, Tianjin University of Technology, Tianjin 300384, China.
- Key Laboratory of Display Materials and Photoelectric Devices, Ministry of Education, School of Materials Science & Engineering, Tianjin University of Technology, Tianjin, 300384, China
| | - Xianshun Zeng
- Tianjin Key Laboratory for Photoelectric Materials and Devices, School of Materials Science & Engineering, Tianjin University of Technology, Tianjin 300384, China.
- Key Laboratory of Display Materials and Photoelectric Devices, Ministry of Education, School of Materials Science & Engineering, Tianjin University of Technology, Tianjin, 300384, China
| |
Collapse
|
9
|
Xu J, Yang C, Xiang J, Song R, Ou D, Wang R. Dihydromyricetin switches pyroptosis to apoptosis during Aeromonas hydrophila infection in Mylopharyngodon piceus. FISH & SHELLFISH IMMUNOLOGY 2025; 165:110460. [PMID: 40441665 DOI: 10.1016/j.fsi.2025.110460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 05/08/2025] [Accepted: 05/25/2025] [Indexed: 06/02/2025]
Abstract
Ampelopsis grossedentata is a traditional Chinese medicine used to treat fever, anti-inflammatory and parasitic infections. Dihydromyricetin is one of the flavonoids with high content, which has been proved to have certain antibacterial and anti-inflammatory effects. However, its antibacterial mechanism is still unclear. In this study, the mechanisms by which A. grossedentata extract and dihydromyricetin promote the organism's defence against A. hydrophila were investigated. The results showed that both A. grossedentata grape extract and dihydromyricetin (DMY) promoted the expression of IL1β in MPK cells. A. hydrophila infection resulted in a significant increase in the expression of caspase1, IL1β, and GSDME, and a significant enhancement of IL1β and LDH release, which were slowed down by DMY. Differently, on the basis of the up-regulation of A. hydrophila, DMY treatment further enhanced the expression of caspase3 and BAX and the activity of caspase3 resulting in stronger apoptosis. And at the same time, the titer of A. hydrophila was decreased by DMY treatment. In black carp (Mylopharyngodon piceus), injections of A. grossedentata extract and DMY also significantly reduced the lethality of A. hydrophila. The up-regulation of the expression of genes associated with pyroptosis due to A. hydrophila infection in splenocytes was attenuated by dihydromyricetin treatment, whereas the expression of apoptosis-associated genes was significantly enhanced. Tissue sections showed significant damage to splenic tissue following A. hydrophila infection, whereas splenic damage was alleviated and its apoptosis-associated DNA breaks were enhanced by dihydromyricetin injection. DMY may regulate multiple modes of cell death in cells. Further studies revealed that TBK1 may be a molecular target of DMY because of their high structural binding potential and as well as functional synergy and relevance. Therefore, these data implied that DMY potentially regulates TBK1 to transform A. hydrophila-induced pyroptosis into apoptosis. This provides a theoretical basis for the application of dihydromyricetin as an antibiotic alternative and feed additive in aquaculture.
Collapse
Affiliation(s)
- Jingjing Xu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410128, China
| | - Can Yang
- Hunan Fisheries Science Institute, Hunan Academy of Agricultural Sciences, Changsha, 410153, China
| | - Jing Xiang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410128, China
| | - Rui Song
- Hunan Fisheries Science Institute, Hunan Academy of Agricultural Sciences, Changsha, 410153, China
| | - Dongsheng Ou
- Hunan Fisheries Science Institute, Hunan Academy of Agricultural Sciences, Changsha, 410153, China
| | - Ruozhong Wang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410128, China.
| |
Collapse
|
10
|
Su L, Xiang S, Mao H, Fu J, Ding H, Wang F, Zhao L, Bai J, Cen Y, Zhu D, Deng M, Li Y. The protective role and mechanism of ginkgolide B in regulating mTOR signalling pathway involved in Manganese-induced apoptosis and autophagy in N27 cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 299:118362. [PMID: 40409184 DOI: 10.1016/j.ecoenv.2025.118362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 05/16/2025] [Accepted: 05/18/2025] [Indexed: 05/25/2025]
Abstract
Ginkgolide B (GB) is a diterpenoid compound found in the leaves of the traditional Chinese medicine plant Ginkgo biloba, which has been shown to have a variety of pharmacological functions. However, the role of GB in Manganese(Mn)-induced apoptosis and autophagy in neuronal cells remains poorly understood. The aim of this study is to investigate the effects of GB on Mn-induced apoptosis and autophagy in N27 cells and further elucidate the mechanism of its protective effects. N27 cells were exposed to MnCl2, GB, mTOR activator, mTOR inhibitor, and autophagy inhibitor, respectively. Cell viability and cytotoxicity were monitored by CCK8 and LDH kits, DA and TH by ELISA, apoptosis by flow cytometry and TUNEL staining, autophagy by MDC staining and mCherry-GFP-LC3B transfection, and autophagy and apoptosis-related proteins by WB. Furthermore, the lentiviral vector was adopted to construct Beclin-1 gene overexpression and knockdown models, and CO-IP was employed to detect the interaction between Beclin-1 and BCL-2. After MnCl2 treatment of N27 cells, cell viability, enhanced apoptosis and autophagy, and inhibited mTOR signalling pathway. Addition of GB to Mn-stained cells was found to effectively alleviate apoptosis and autophagy, particularly, the optimized effect was realized when GB was combined with mTOR activator. The mTOR inhibitor Rapamycin and autophagy inhibitor Bafilomycin A1 could reverse the efficacy of GB; knockdown of Beclin-1 could alleviate MnCl2-induced apoptosis and autophagy-related molecular indices, whereas Beclin-1 overexpression demonstrated the opposite way. It was observed in this study that Mn-induced apoptosis and autophagy in N27 cells were Beclin-1-dependent; GB could mitigate the apoptosis and autophagy by activating the mTOR signalling pathway.
Collapse
Affiliation(s)
- Liyu Su
- Zunyi Medical University School of Public Health, Zunyi, Guizhou 563000, China; Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes
| | - Shunyu Xiang
- Zunyi Medical University School of Public Health, Zunyi, Guizhou 563000, China; Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes
| | - Huan Mao
- Zunyi Medical University School of Public Health, Zunyi, Guizhou 563000, China; Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes
| | - Jiayu Fu
- Zunyi Medical University School of Public Health, Zunyi, Guizhou 563000, China; Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes
| | - Hongwei Ding
- Zunyi Medical University School of Public Health, Zunyi, Guizhou 563000, China; Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes
| | - Feng Wang
- Zunyi Medical University School of Public Health, Zunyi, Guizhou 563000, China; Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes
| | - Lan Zhao
- Zunyi Medical University School of Public Health, Zunyi, Guizhou 563000, China; Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes
| | - Jianwei Bai
- Zunyi Medical University School of Public Health, Zunyi, Guizhou 563000, China; Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes
| | - Yuyan Cen
- Zunyi Medical University School of Public Health, Zunyi, Guizhou 563000, China; Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes
| | - Deyu Zhu
- Zunyi Medical University School of Public Health, Zunyi, Guizhou 563000, China; Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes
| | - Meng Deng
- The Third Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China; The First People's Hospital of Zunyi, Zunyi, Guizhou 563000, China
| | - Yan Li
- Zunyi Medical University School of Public Health, Zunyi, Guizhou 563000, China; Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes.
| |
Collapse
|
11
|
Zhang Q, Ma S, Kang X, Liu Y, Ma F, Yu F, Luo X, Li G, Hao Y, Zhang H, Liu B, Jiang Y. A dual-targeting bio-liposomes nanodrug repair endothelial cell dysfunction and restore macrophage cholesterol flow homeostasis to treat early atherosclerosis. J Nanobiotechnology 2025; 23:365. [PMID: 40394654 PMCID: PMC12090647 DOI: 10.1186/s12951-025-03436-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 05/01/2025] [Indexed: 05/22/2025] Open
Abstract
Hyperhomocysteinemia (HHy) can lead to vascular endothelial cell dysfunction, progressive inflammation and lipid metabolism disorder, which finally result in the onset and development of atherosclerosis, a major contributor to cardiovascular diseases. Given the complexity of pathological process, treatments based on a single target often showed limited therapeutic efficacy against AS. Thus, developing nanodrug for enhanced multi-targets therapy is promising. In this study, we constructed a dual-targeting nanodrug (HA-ML@ES NPs) co-loaded with Shikonin (SKN) and Evolocumab (Evol). In vitro results showed that HA-ML@ES NPs could simultaneously target dysfunctional endothelial cell and inflammatory macrophage through the interaction between HA and CD44. In vivo assay indicated that HA-ML@ES NPs with long circulation and plaque accumulation efficiently attenuate endothelial cell dysfunction by inhibiting glycolysis and restore cholesterol flow homeostasis in macrophage by reprogramming macrophage phenotype, which finally attenuated the development of atherosclerosis. Collectively, these results present a highly promising dual-cell therapeutic approach based on HA-ML@ES NPs for the management of early atherosclerosis.
Collapse
Grants
- 2023AAC005035 The Natural Science Foundation of Ningxia Hui Autonomous Region
- XJKF240301, XJKF240304, XJKF230125 and XJKF240326 Open Competition Mechanism to Select the Best Candidates for Key Research Projects of Ningxia Medical University
- XJKF240301, XJKF240304, XJKF230125 and XJKF240326 Open Competition Mechanism to Select the Best Candidates for Key Research Projects of Ningxia Medical University
- XJKF240301, XJKF240304, XJKF230125 and XJKF240326 Open Competition Mechanism to Select the Best Candidates for Key Research Projects of Ningxia Medical University
- XJKF240301, XJKF240304, XJKF230125 and XJKF240326 Open Competition Mechanism to Select the Best Candidates for Key Research Projects of Ningxia Medical University
- 2023BEG02074, 2022BFH02013, 2022BEG02054 Key Projects of the Key R&D Program of the Ning Xia Hui Autonomous Region
- U21A20343 National Natural Science Foundation of China
- 2024ZD0531200 Noncommunicable Chronic Diseases-National Science and Technology Major Project
- Key Projects of the Key R&D Program of the Ning Xia Hui Autonomous Region
Collapse
Affiliation(s)
- Qi Zhang
- School of Inspection, Ningxia Medical University, Yinchuan, 750004, China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
| | - Shengchao Ma
- School of Inspection, Ningxia Medical University, Yinchuan, 750004, China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
| | - Xue Kang
- Department of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Yi Liu
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Fei Ma
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Feifei Yu
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Xiaolan Luo
- School of Inspection, Ningxia Medical University, Yinchuan, 750004, China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
| | - Guizhong Li
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Yinju Hao
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
- Medical Experimental Center, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
| | - Huiping Zhang
- Medical Experimental Center, General Hospital of Ningxia Medical University, Yinchuan, 750004, China.
- Department of Medical Genetics, Hunan Provincial Maternal and Child Health Hospital, Changsha, 410008, China.
| | - Bin Liu
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China.
- College of Biology, Hunan University, Changsha, 410082, China.
| | - Yideng Jiang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China.
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
12
|
Zaidi SA, Fan Z, Chauhdari T, Ding Y. MicroRNA regulatory dynamic, emerging diagnostic and therapeutic frontier in atherosclerosis. Microvasc Res 2025; 160:104818. [PMID: 40368159 DOI: 10.1016/j.mvr.2025.104818] [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: 03/01/2025] [Revised: 05/07/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025]
Abstract
MicroRNAs (miRNAs), a class of non-coding RNAs, are pivotal post-transcriptional regulators of gene expression with profound implications in the pathogenesis of atherosclerosis (AS). As a progressive arterial disease driven by vascular cells dysfunction, lipid dysregulation and subsequent chronic inflammation, AS remains a leading cause of global morbidity. Recent studies have demonstrated how important miRNAs are in regulating central biological processes in the vascular wall, such as endothelial function, vascular smooth muscle cell (VSMC) phenotypic switching, and macrophage polarization. This review provides comprehensive insight into the role of miRNAs in the development and complexity of atherosclerotic plaques according to their effects on endothelial cells, macrophages, and VSMCs. We also go over the growing prospects of miRNAs as therapeutic targets and diagnostic biomarkers, providing information to be used in the study of vascular diseases. Lastly, we address recent complications and potential applications of miRNA-based approaches in clinical practice.
Collapse
Affiliation(s)
- Syeda Armana Zaidi
- College of Life Sciences, University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, PR China.
| | - Zhiyu Fan
- College of Life Sciences, University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, PR China.
| | - Talha Chauhdari
- College of Life Sciences, University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, PR China.
| | - Yongsheng Ding
- College of Life Sciences, University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, PR China.
| |
Collapse
|
13
|
Qi D, Wang F, Zhang X, Li B, Zhou W, Sheng S, Zhu R, Cao L, Zhao C, Deng X, Ouyang T, Zheng F. Epigenetic upregulation of CLEC5A contributes to monocyte/macrophage dysfunction in coronary artery disease. Int J Biol Macromol 2025; 308:142471. [PMID: 40147659 DOI: 10.1016/j.ijbiomac.2025.142471] [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: 09/23/2024] [Revised: 03/21/2025] [Accepted: 03/22/2025] [Indexed: 03/29/2025]
Abstract
Inflammatory activation and dysfunction of immune cells are essential events in coronary artery disease (CAD) pathogenesis. C-type lectin domain family 5 member A (CLEC5A) has recently been regarded as a potent modulator of inflammation, while its contribution to CAD remains undefined. This study aims to clarify the involvement of CLEC5A in atherosclerosis and explore its epigenetic regulatory mechanisms. Integrated methylome and transcriptome analyses identified CLEC5A as a DNA methylation-driven gene in CAD. Functional studies revealed that DNMT1 overexpression suppresses CLEC5A expression in THP-1 cells, with subsequent identification of the cg06744540 CpG site as the critical regulatory locus. Clinical correlation analyses demonstrated that elevated CLEC5A expression is inversely associated with hypomethylation at cg06744540 in CAD patients. Furthermore, CLEC5A overexpression significantly enhanced monocyte inflammation, migration, and adhesion, promoted macrophage polarization and lipid accumulation, and inhibited apoptosis. Mechanistic investigations revealed that CLEC5A exacerbates inflammation in monocyte/macrophage by activating the NF-κB signaling pathway. Conversely, CLEC5A knockdown resulted in opposite effects. Notably, treatment of cells with folic acid, a methylation-enhancing factor, significantly increased DNMT1 expression and declined CLEC5A expression. Consistently, folic acid reversed high-fat-induced CLEC5A expression and inflammation and suppressed the formation of atherosclerotic plaques in vivo. In conclusion, the epigenetic upregulation of CLEC5A through DNMT1-mediated cg06744540 demethylation contributed to monocyte/macrophage dysfunction, thus accelerating CAD progression.
Collapse
Affiliation(s)
- Daoxi Qi
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, China
| | - Fan Wang
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, China
| | - Xiaokang Zhang
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, China
| | - Boyu Li
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, China
| | - Wenjie Zhou
- Department of Medical Genetics, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Shuyang Sheng
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, China
| | - Ruiyang Zhu
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, China
| | - Liang Cao
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, China
| | - Chang Zhao
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, China
| | - Xinyu Deng
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, China
| | - Tanglin Ouyang
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, China
| | - Fang Zheng
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, China.
| |
Collapse
|
14
|
Li L, Che Y, Zhu L, Liu Z, Song Y, Wu Q, Chen M, Wang J. Calycosin, a Bioactive Isoflavone, Ameliorates Oxidative Stress and Inflammation in Lipopolysaccharide-Induced Intestinal Cell Damage Model via the Nrf2 and NF-κB Signaling Pathways. FASEB J 2025; 39:e70550. [PMID: 40244892 DOI: 10.1096/fj.202402387r] [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: 10/07/2024] [Revised: 03/28/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025]
Abstract
Calycosin (CA), a bioactive isoflavone derived from Astragali Radix, has garnered interest in food sciences and pharmacology due to its potential antioxidant and anti-inflammatory properties. Our previous studies found that CA induced host defense peptide production in porcine IPEC-J2 cells and alleviated hydrogen peroxide-induced cellular oxidative damage. However, it is still unknown whether CA can protect against pathogenic microorganisms or toxins that cause intestinal cell damage. In this study, we aimed to investigate the protective effects of CA against LPS-induced intestinal cell damage. Using IPEC-J2 intestinal cells to develop a damage model, we found that LPS exposure caused significant morphological damage and apoptosis. However, pretreatment with CA effectively attenuated these adverse effects. Mechanistically, CA alleviated LPS-induced oxidative stress by reducing reactive oxygen species, malondialdehyde, 8-hydroxy-2'-deoxyguanine and carbonyl production, while enhancing antioxidant enzyme activities and gene expression. Furthermore, CA restored the LPS-induced decrease in nuclear erythroid 2-related factor 2 (Nrf2) expression, and Nrf2 silencing abolished the protective effects of CA, indicating that its action is mediated through the Nrf2 pathway. Additionally, CA suppressed LPS-induced inflammation by downregulating inflammation-related gene expression and inhibiting the nuclear factor (NF)-κB pathway. Collectively, our findings demonstrate that CA protects intestinal cells from LPS-induced damage by mitigating oxidative stress and inflammation via the Nrf2 and NF-κB pathways. These results suggest that CA has potential to be developed as a feed additive to prevent intestinal injury in animals, warranting further investigation in vivo.
Collapse
Affiliation(s)
- Lu Li
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- College of Life Science and Food Engineering, Hebei University of Engineering, Handan, China
| | - Yuyan Che
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Longlong Zhu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Zeqing Liu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Yanchen Song
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Qiong Wu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Meixia Chen
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Sino-US Joint Laboratory of Animal Science, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Jing Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Sino-US Joint Laboratory of Animal Science, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| |
Collapse
|
15
|
Yu WL, Deng LW, Li HH, Wang CK, Zuo XY, Wang ZC, Meng L, Wen LX, Zeng WZ, Zhao Y, Wang XH. FBXO38 Regulates Nox1 Stability to Reduce Vascular Endothelial Damage Induced by Low Oscillatory Shear Stress. Cardiovasc Ther 2025; 2025:4506032. [PMID: 40313652 PMCID: PMC12043393 DOI: 10.1155/cdr/4506032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/28/2025] [Indexed: 05/03/2025] Open
Abstract
Oxidative stress and endothelial dysfunction are critical drivers of atherosclerosis, but the mechanisms regulating oxidative stress under disturbed flow conditions remain incompletely understood. The ubiquitin-proteasome system, particularly E3 ubiquitin ligases, may play a pivotal role in modulating these processes. FBXO38, an E3 ligase involved in proteasomal degradation, has been implicated in various physiological pathways, but its role in regulating oxidative stress in endothelial cells is unknown. We hypothesized that FBXO38 mitigates endothelial damage induced by low oscillatory shear stress (LOSS) by promoting the ubiquitin-proteasome-dependent degradation of Nox1, a major source of reactive oxygen species (ROS). Using an in vitro LOSS model in human umbilical vein endothelial cells (HUVECs) and an in vivo mouse partial carotid ligation model, we assessed the expression of FBXO38 and Nox1 through quantitative PCR, western blotting, immunofluorescence, and immunohistochemistry. LOSS significantly reduced FBXO38 protein expression (by ~60%, p < 0.0001 at 24 h), leading to increased Nox1 protein levels (approximately two-fold, p < 0.001) and apoptosis. FBXO38 overexpression markedly attenuated Nox1 accumulation (~50% reduction, p < 0.05), reduced ROS production, and improved cell viability under LOSS conditions, whereas FBXO38 knockdown exacerbated these effects. Moreover, FBXO38 directly interacted with Nox1, suggesting a ubiquitin-dependent degradation mechanism. Our results reveal that FBXO38 regulates endothelial oxidative stress by controlling Nox1 stability under disturbed shear stress conditions. Although FBXO38 emerges as a promising candidate for therapeutic targeting, further studies are necessary to validate its potential in preclinical and clinical settings.
Collapse
Affiliation(s)
- Wan-li Yu
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li-wen Deng
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huan-huan Li
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chun-kai Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiang-yi Zuo
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zi-chang Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Meng
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lan-xin Wen
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wan-zhi Zeng
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Zhao
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xue-hu Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
16
|
Shi J, Yu Y, Yuan H, Li Y, Xue Y. Mitochondrial dysfunction in AMI: mechanisms and therapeutic perspectives. J Transl Med 2025; 23:418. [PMID: 40211347 PMCID: PMC11987341 DOI: 10.1186/s12967-025-06406-5] [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: 01/03/2025] [Accepted: 03/20/2025] [Indexed: 04/13/2025] Open
Abstract
Acute myocardial infarction (AMI) and the myocardial ischemia-reperfusion injury (MI/RI) that typically ensues represent a significant global health burden, accounting for a considerable number of deaths and disabilities. In the context of AMI, percutaneous coronary intervention (PCI) is the preferred treatment option for reducing acute ischemic damage to the heart. Despite the modernity of PCI therapy, pathological damage to cardiomyocytes due to MI/RI remains an important target for intervention that affects the long-term prognosis of patients. In recent years, mitochondrial dysfunction during AMI has been increasingly recognized as a critical factor in cardiomyocyte death. Damaged mitochondria play an active role in the formation of an inflammatory environment by triggering key signaling pathways, including those mediated by cyclic GMP-AMP synthase, NOD-like receptors and Toll-like receptors. This review emphasizes the dual role of mitochondria as both contributors to and regulators of inflammation. The aim is to explore the complex mechanisms of mitochondrial dysfunction in AMI and its profound impact on immune dysregulation. Specific interventions including mitochondrial-targeted antioxidants, membrane-stabilizing peptides, and mitochondrial transplantation therapies have demonstrated efficacy in preclinical AMI models.
Collapse
Affiliation(s)
- Jingle Shi
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yiding Yu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huajing Yuan
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Yitao Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
17
|
Huang T, Zhang Y, Qiao L, Luo D, Ba L, Xu S, Meng L, Cao S, Wang T, Kou X. Active ingredients of blueberry pomace: A comprehensive review of isolation, identification, health benefits and food applications. Food Chem X 2025; 27:102459. [PMID: 40321340 PMCID: PMC12047588 DOI: 10.1016/j.fochx.2025.102459] [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: 02/12/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 05/08/2025] Open
Abstract
Blueberry pomace is a valuable source of a wide range of active compounds and nutrients and is receiving increasing research attention. However, limited attention has been paid in the literature to the nutritional properties and practical applications of blueberry pomace. In this paper, we review the methods used to extract bioactive compounds from blueberry pomace, study their health effects and explore the prospects for their application in the food industry, such as fermented beverages, nutritional supplements or natural colors. In addition, recent studies have examined the content of various active ingredients such as flavonoids, vitamins, and dietary fiber and suggested the validity of the corresponding extraction methods. In addition, these compounds have great potential in human health, such as antioxidant, hypoglycemic, improvement of cognitive impairment and gut health. This analysis also highlights the bioavailability of the active ingredients in blueberry pomace. Thus, blueberry pomace offers a wide field of scientific and technological exploration, but significant challenges must be faced in order to optimize its utilization and promote further research on extraction, applications, and innovative methods.
Collapse
Affiliation(s)
- Tianyu Huang
- School of Food Science and Engineering, Guiyang University, Guizhou Province 550005, China
| | - Yu Zhang
- School of Food Science and Engineering, Guiyang University, Guizhou Province 550005, China
| | - Linxiang Qiao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Donglan Luo
- School of Biological and Environmental Engineering, Guiyang University, Guiyang 550005, China
| | - Liangjie Ba
- School of Food Science and Engineering, Guiyang University, Guizhou Province 550005, China
| | - Su Xu
- School of Food Science and Engineering, Guiyang University, Guizhou Province 550005, China
| | - Lingshuai Meng
- School of Food Science and Engineering, Guiyang University, Guizhou Province 550005, China
| | - Sen Cao
- School of Food Science and Engineering, Guiyang University, Guizhou Province 550005, China
| | - Tao Wang
- Guizhou Light Industry Technical College; Guiyang, 550025, China
| | - Xiaohong Kou
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| |
Collapse
|
18
|
Wang P, Chen W, Zhang J, Pan C, Lv Y, Sun Y, Wang Y, Ma X, Gao C, Chen T, Wu A, Zheng J. Advances in the treatment of atherosclerotic plaque based on nanomaterials. Nanomedicine (Lond) 2025; 20:869-881. [PMID: 40109186 PMCID: PMC11988221 DOI: 10.1080/17435889.2025.2480049] [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: 01/01/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
Atherosclerosis is the leading cause of cardiovascular disease worldwide, posing not only a significant threat to cardiovascular health but also impairing the function of multiple organs, with severe cases potentially being life-threatening. Consequently, the effective treatment of atherosclerosis is of paramount importance in reducing the mortality associated with cardiovascular diseases. With the advancement of nanomedicine and a deeper understanding of the pathological mechanisms underlying atherosclerosis, nanomaterials have emerged as promising platforms for precise diagnosis and targeted therapeutic strategies. These materials offer notable advantages, including targeted drug delivery, enhanced bioavailability, improved drug stability, and controlled release. This review provides an overview of the mechanisms underlying atherosclerotic plaque development and examines nanomaterial-based therapeutic approaches for managing atherosclerotic plaques, including therapies targeting cholesterol metabolism, anti-inflammatory strategies, macrophage clearance, and immunotherapy. Additionally, the paper discusses the current technical challenges associated with the clinical transformation of these therapies. Finally, the potential future integration of nanomaterials, smart nanomaterials, and artificial intelligence in the treatment of atherosclerosis is also explored.
Collapse
Affiliation(s)
- Pengyu Wang
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo, China
- Department of Radiology, Ningbo No.2 Hospital, Ningbo, China
| | - Weiwei Chen
- Traditional Chinese Medicine Department, Minglou Street Community Health Service Center, Yingzhou, Ningbo, China
| | - Jingfeng Zhang
- Department of Radiology, Ningbo No.2 Hospital, Ningbo, China
| | - Chunshu Pan
- Department of Radiology, Ningbo No.2 Hospital, Ningbo, China
| | - Yagui Lv
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Yanzi Sun
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Yanan Wang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Xuehua Ma
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Changyong Gao
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Tianxiang Chen
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Aiguo Wu
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Jianjun Zheng
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo, China
- Department of Radiology, Ningbo No.2 Hospital, Ningbo, China
| |
Collapse
|
19
|
Wu X, Pan X, Kang J, Huang Y, Ren J, Pan J, Yu K, Li Y. Ferulic acid inhibits ox-LDL-induced ferroptosis and apoptosis in RAW 264.7 cells via the HIF-1 signaling pathway. Front Pharmacol 2025; 16:1524736. [PMID: 40170728 PMCID: PMC11958962 DOI: 10.3389/fphar.2025.1524736] [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: 11/19/2024] [Accepted: 03/03/2025] [Indexed: 04/03/2025] Open
Abstract
Objective Ferulic acid (FA) has shown potential in treating atherosclerosis (AS) by improving lipid metabolism and exerting anti-hypoxic effects. This study aimed to validate the mechanism of FA in AS through in vitro experiments. Methods Network analysis was employed to predict the mechanisms underlying the therapeutic effects of FA on AS. An in vitro foam cell model was established using RAW 264.7 cells treated with ox-LDL. Cellular lipid accumulation was detected using Oil Red O staining; cell viability was assessed by cell counting kit-8; mitochondrial morphology and function were evaluated by transmission electron microscopy and JC-1 staining; apoptosis levels were detected by TUNEL and DAPI staining; mitochondrial Fe2+ content was measured by Mito-FerroGreen; and Western blot was performed to determine the protein expression levels of HIF-1α, Bax, Bcl2, GPX4, and EGFR. Results Network analysis suggested that FA may exert its therapeutic effects on AS through the HIF-1 signaling pathway and is closely associated with the regulation of ferroptosis and apoptosis. FA upregulated the expression of ALOX5, BCL2, ERN1, GPX4, NOS3, and SLC2A1 mRNA and downregulated the expression of BAX, CYCS, EGFR, FLT1, HIF1A, NFKB1, NOS2, PARP1, and STAT3 mRNA. In vitro experiments demonstrated that FA reduces lipid accumulation, increases cell viability, improves mitochondrial function, and decreases reactive oxygen species content. Additionally, FA inhibited ferroptosis and apoptosis by suppressing the HIF-1 signaling pathway, up-regulating the expression of GPX4 and Bcl2, and down-regulating the expression of HIF-1α and Bax protein. HIF-1 agonists reversed these effects by activating the HIF-1 signaling pathway. Conclusion FA improves mitochondrial function and suppresses ferroptosis and apoptosis by inhibiting the HIF-1 signaling pathway, thereby treating AS.
Collapse
Affiliation(s)
- Xize Wu
- The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Xue Pan
- College of Traditional Chinese Medicine, Dazhou Vocational College of Chinese Medicine, Dazhou, Sichuan, China
| | - Jian Kang
- The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Yuxi Huang
- The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Jiaqi Ren
- The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Jiaxiang Pan
- Department of Cardiology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Kaifeng Yu
- Department of Cardiology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Yue Li
- Department of Cardiology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
| |
Collapse
|
20
|
He SQ, Huang B, Xu F, Yang JJ, Li C, Liu FR, Yuan LQ, Lin X, Liu J. Functions and application of circRNAs in vascular aging and aging-related vascular diseases. J Nanobiotechnology 2025; 23:216. [PMID: 40098005 PMCID: PMC11917153 DOI: 10.1186/s12951-025-03199-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
Circular RNAs (circRNAs), constituting a novel class of endogenous non-coding RNAs generated through the reverse splicing of mRNA precursors, possess the capacity to regulate gene transcription and translation. Recently, the pivotal role of circRNAs in controlling vascular aging, as well as the pathogenesis and progression of aging-related vascular diseases, has garnered substantial attention. Vascular aging plays a crucial role in the increased morbidity and mortality of the elderly. Endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) are crucial components of the intima and media layers of the vascular wall, respectively, and are closely involved in the mechanisms underlying vascular aging and aging-related vascular diseases. The review aims to provide a comprehensive exploration of the connection between circRNAs and vascular aging, as well as aging-related vascular diseases. Besides, circRNAs, as potential diagnostic markers or therapeutic targets for vascular aging and aging-related vascular diseases, will be discussed thoroughly, along with the challenges and limitations of their clinical application. Investigating the role and molecular mechanisms of circRNAs in vascular aging and aging-related vascular diseases will provide a novel insight into early diagnosis and therapy, and even effective prognosis assessment of these conditions.
Collapse
Affiliation(s)
- Sha-Qi He
- Department of Radiology, the Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Bei Huang
- Department of Radiology, the Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Feng Xu
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, the Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Jun-Jie Yang
- Department of Radiology, the Second Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830054, China
| | - Cong Li
- Department of Radiology, the Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Feng-Rong Liu
- Department of Anesthesiology, the Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Ling-Qing Yuan
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, the Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Xiao Lin
- Department of Radiology, the Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| | - Jun Liu
- Department of Radiology, the Second Xiangya Hospital of Central South University, Changsha, 410011, China.
- Clinical Research Center for Medical Imaging in Hunan Province, Quality Control Center in Hunan Province, Changsha, 410011, China.
| |
Collapse
|
21
|
Ding K, Bao Q, He J, Wang J, Wang H. Tetrahydropalmatine improves mitochondrial function in vascular smooth muscle cells of atherosclerosis in vitro by inhibiting Ras homolog gene family A/Rho-associated protein kinase-1 signaling pathway. Open Med (Wars) 2025; 20:20241059. [PMID: 40109328 PMCID: PMC11920760 DOI: 10.1515/med-2024-1059] [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: 05/10/2024] [Revised: 08/27/2024] [Accepted: 09/17/2024] [Indexed: 03/22/2025] Open
Abstract
Background Tetrahydropalmatine (THP) regulates mitochondrial function in vascular smooth muscle cells (VSMCs) to prevent or alleviate atherosclerosis (AS), with unclear specific mechanism. Methods AS models were constructed by oxidized low-density lipoprotein (ox-LDL)-treated VSMCs. Cell counting kit-8 for cell viability, wound scratch assay for cell migration, and flow cytometry for cell cycle, intracellular reactive oxygen species, and mitochondrial membrane potential (MMP) were performed. Malondialdehyde (MDA) and superoxide dismutase (SOD) levels by biochemical kits, oxygen consumption rate (OCR) by seahorse apparatus, apoptosis by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay (TUNEL) staining, and apoptosis-related expression by western blot were detected. Ras homolog gene family A/Rho-associated protein kinase-1 (RhoA/ROCK1) levels were measured by western blot and ELISA. The RhoA agonist, U46619, was employed to validate mechanism of THP. Results THP suppressed cell cycle progression and cell migration whereas alleviating cell viability and oxidative stress, as reduced MDA and enhanced SOD levels in ox-LDL-incubated VSMCs. THP protected mitochondrial function by higher MMP levels and OCR values. Additionally, THP decreased TUNEL-positive cells, Bax, Caspase-3, RhoA, ROCK1, and osteopontin expression, while increased Bcl-2 and smooth muscle myosin heavy chain levels. Furthermore, U46619 intervention antagonized effects of THP. Conclusion THP improved mitochondrial function in VSMCs of AS by inhibiting RhoA/ROCK1 signaling pathway.
Collapse
Affiliation(s)
- Ke Ding
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University, No. 54, Youdian Road, Hangzhou, 310006, Zhejiang, China
| | - Qiying Bao
- Department of Pharmacy, Hangzhou Fuyang Hospital of TCM Orthopedics, Hangzhou, 311499, Zhejiang, China
| | - Jiaqi He
- Traditional Chinese Medicine Dispensary, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, Zhejiang, China
| | - Jiahong Wang
- Traditional Chinese Medicine Dispensary, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, Zhejiang, China
| | - Hui Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 548, Binwen Road, Hangzhou, 310053, Zhejiang, China
| |
Collapse
|
22
|
Xing A, Wang F, Liu J, Zhang Y, He J, Zhao B, Sun B. The prospect and underlying mechanisms of Chinese medicine in treating periodontitis. Chin J Nat Med 2025; 23:269-285. [PMID: 40122658 DOI: 10.1016/s1875-5364(25)60842-9] [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: 03/22/2024] [Revised: 06/18/2024] [Accepted: 07/13/2024] [Indexed: 03/25/2025]
Abstract
Inflammation represents a critical immune response triggered by cellular activities and inflammatory mediators following tissue damage. It plays a central role in the pathological progression of diverse diseases, including psychiatric disorders, cancer, and immunological conditions, rendering it an essential target for therapeutic intervention. Periodontitis, a prevalent oral inflammatory disease, is a leading cause of tooth loss and poses significant health challenges globally. Traditionally, inflammatory diseases such as periodontitis have been treated with systemic administration of synthetic chemicals. However, recent years have witnessed challenges, including drug resistance and microbial dysbiosis associated with these treatments. In contrast, natural products derived from Chinese medicine offer numerous benefits, such as high safety profiles, minimal side effects, innovative pharmacological mechanisms, ease of extraction, and multiple targets, rendering them viable alternatives to conventional antibiotics for treating inflammatory conditions. Numerous effective anti-inflammatory natural products have been identified in traditional Chinese medicine (TCM), including alkaloids, flavonoids, terpenoids, lignans, and other natural products that exhibit inhibitory effects on inflammation and are potential therapeutic agents. Several studies have confirmed the substantial anti-inflammatory and immunomodulatory properties of these compounds. This comprehensive review examines the literature on the anti-inflammatory effects of TCM-derived natural products from databases such as PubMed, Web of Science, and CNKI, focusing on terms like "inflammation", "periodontitis", "pharmacology", and "traditional Chinese medicine". The analysis systematically summarizes the molecular pharmacology, chemical composition, and biological activities of these compounds in inflammatory responses, alongside their mechanisms of action. This research seeks to deepen understanding of the mechanisms and biological activities of herbal extracts in managing inflammatory diseases, potentially leading to the development of promising new anti-inflammatory drug candidates. Future applications could extend to the treatment of various inflammatory conditions, including periodontitis.
Collapse
Affiliation(s)
- Aili Xing
- Oral and Maxillofacial Surgery, Hospital of Stomatologyl, Jilin University, Changchun 130021, China
| | - Feng Wang
- Oral and Maxillofacial Surgery, Hospital of Stomatologyl, Jilin University, Changchun 130021, China
| | - Jinzhong Liu
- Preventive Dentistry, Hospital of Stomatologyl, Jilin University, Changchun 130021, China
| | - Yuan Zhang
- Oral and Maxillofacial Surgery, Hospital of Stomatologyl, Jilin University, Changchun 130021, China
| | - Jingya He
- Oral and Maxillofacial Surgery, Hospital of Stomatologyl, Jilin University, Changchun 130021, China
| | - Bin Zhao
- Periodontics, Hospital of Stomatologyl, Jilin University, Changchun 130021, China.
| | - Bin Sun
- Oral and Maxillofacial Surgery, Hospital of Stomatologyl, Jilin University, Changchun 130021, China.
| |
Collapse
|
23
|
Cui XY, Zhan JK. Capsaicin and TRPV1: A Novel Therapeutic Approach to Mitigate Vascular Aging. Aging Dis 2025:AD.2024.1292. [PMID: 39965247 DOI: 10.14336/ad.2024.1292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/08/2025] [Indexed: 02/20/2025] Open
Abstract
Vascular aging and its associated diseases represent a principal cause of mortality among the global elderly population, making the mitigation of vascular aging a significant aspiration for humanity. This article explores the intersection of nature and health, focusing on the role of the natural plant, pepper, and its principal bioactive compound, capsaicin, in combating vascular aging. By examining molecular and cellular mechanisms as well as phenotypic alterations in blood vessels, we offer a comprehensive review of the effects of capsaicin and its receptor, transient receptor potential vanilloid 1 (TRPV1), within vascular aging. We propose that capsaicin may serve as the medication with the potential to slow the progress of vascular aging and could constitute a new strategy to treat vascular aging related disease.
Collapse
Affiliation(s)
- Xing-Yu Cui
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, China
| | - Jun-Kun Zhan
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, China
| |
Collapse
|
24
|
Fang Y, Pan J, Wang P, Wang R, Liang S. A comprehensive review of Schisandrin B's preclinical antitumor activity and mechanistic insights from network pharmacology. Front Pharmacol 2025; 16:1528533. [PMID: 39995410 PMCID: PMC11847788 DOI: 10.3389/fphar.2025.1528533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/21/2025] [Indexed: 02/26/2025] Open
Abstract
As an active constituent in the extract of dried fruits of Schisandra chinensis, Schisandrin B exhibits diverse pharmacological effects, including liver protection, anti-inflammatory and anti-oxidant. Numerous studies have demonstrated that Schisandrin B exhibits significant antitumor activity against various malignant tumors in preclinical studies, which is achieved by inhibiting cell proliferation and metastasis and promoting apoptosis. As a potential antitumor agent, Schisandrin B holds broad application prospects. This review systematically elaborates on the antitumor effect of Schisandrin B and the related molecular mechanism, and preliminarily predicts its antitumor targets by network pharmacology, thereby pave the way for further research, development, and clinical application.
Collapse
Affiliation(s)
- Yanhua Fang
- The Key Laboratory of biomarker high throughput screening and target translation of breast and gastrointestinal tumor, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Juan Pan
- The Key Laboratory of biomarker high throughput screening and target translation of breast and gastrointestinal tumor, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Piao Wang
- The Key Laboratory of biomarker high throughput screening and target translation of breast and gastrointestinal tumor, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
- Department of Oncology, Central Hospital of Liwan, Guangzhou, China
| | - Ruoyu Wang
- The Key Laboratory of biomarker high throughput screening and target translation of breast and gastrointestinal tumor, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Shanshan Liang
- The Key Laboratory of biomarker high throughput screening and target translation of breast and gastrointestinal tumor, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| |
Collapse
|
25
|
Dong Q, Dai G, Quan N, Tong Q. Role of natural products in cardiovascular disease. Mol Cell Biochem 2025; 480:733-745. [PMID: 38879838 DOI: 10.1007/s11010-024-05048-3] [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: 04/19/2024] [Accepted: 06/09/2024] [Indexed: 01/03/2025]
Abstract
As the world's aging population increases, cardiovascular diseases (CVDs) associated with aging deserve increasing attention. CVD in association with age is considered a major cause of morbidity and mortality worldwide. In this review, we provide an overview of the key molecular pathways, cellular processes such as autophagy, oxidative stress, inflammatory responses, myocardial remodeling and ischemia-refocused injury that accompany CVD as well as the natural products of targeting these mechanisms and some of the dietary habits that have been studied in cardiovascular-related diseases. The potential preventive and therapeutic avenues resulting from these dietary habits and natural products related to animal models and clinical studies can help us to better understand the processes involved in cardiovascular diseases and provide recommendations to reduce the cardiovascular burden associated with aging heart.
Collapse
Affiliation(s)
- Qi Dong
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Gaoying Dai
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Nanhu Quan
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Qian Tong
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
26
|
Xie J, Yang Q, Zeng X, Zeng Q, Xiao H. Dihydromyricetin inhibits injury caused by ischemic stroke through the lncRNA SNHG17/miR-452-3p/CXCR4 axis. PeerJ 2025; 13:e18876. [PMID: 39897488 PMCID: PMC11786715 DOI: 10.7717/peerj.18876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/27/2024] [Indexed: 02/04/2025] Open
Abstract
Ischemic stroke (IS) is an important cause of death worldwide. Dihydromyricetin (DHM) has been reported to have neuroprotective potential, but its role and mechanism in IS have not been fully elucidated. The MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay was used to determine the safe dose of DHM in BV2 microglia and its applicability in OGD/R-treated cells. The mechanism of action of DHM was explored by RT-qPCR, ELISA, luciferase reporter gene assay and western blotting. DHM dose-dependently enhanced BV2 cell viability post-OGD/R and attenuated inflammation and oxidative stress. The protective effects of DHM were found to be mediated through the downregulation of SNHG17, which in turn modulated miR-452-3p expression. miR-452-3p was identified as a negative regulator of pro-inflammatory CXCR4, a direct target whose expression was inversely affected by SNHG17. The interaction between SNHG17 and miR-452-3p was further confirmed by RNA pull-down assays. Furthermore, manipulation of the SNHG17/miR-452-3p/CXCR4 axis was shown to modulate the NF-κB signaling pathway as evidenced by changes in phosphorylation levels. In conclusion, our findings elucidate a novel DHM-mediated neuroprotective mechanism in microglial cells involving the SNHG17/miR-452-3p/CXCR4 regulatory axis. This axis attenuates OGD/R-induced inflammatory and oxidative stress, suggesting a therapeutic potential for DHM in conditions characterized by such pathological processes.
Collapse
Affiliation(s)
- Jiacheng Xie
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Qiuyue Yang
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xueliang Zeng
- Department of Pharmacology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Qi Zeng
- Department of Ultrasound, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Hai Xiao
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
27
|
Jiang X, Huang H. The therapeutic potential of apigenin against atherosclerosis. Heliyon 2025; 11:e41272. [PMID: 39811295 PMCID: PMC11732486 DOI: 10.1016/j.heliyon.2024.e41272] [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: 09/10/2024] [Revised: 12/02/2024] [Accepted: 12/15/2024] [Indexed: 01/16/2025] Open
Abstract
Apigenin is a natural flavonoid abundantly found in fruits, vegetables, and medicinal plants. It possesses protective effects against cancer, metabolic syndrome, dyslipidemia, etc. Atherosclerosis, a chronic immune-mediated inflammatory disease, is the underlying cause of coronary heart disease, stroke, and myocardial infarction. Numerous in vivo and in vitro studies have shown a protective effect of apigenin against atherosclerosis, attributed to its antioxidant and anti-inflammatory properties, as well as its antihypertensive effect and regulation of lipid metabolism. This study aimed to review the effects and mechanisms of apigenin against atherosclerosis for the first time. Apigenin displays encouraging results, and this review confirms the potential value of apigenin as a candidate medication for atherosclerosis.
Collapse
Affiliation(s)
- Xueqiang Jiang
- Sinopharm Dongfeng General Hospital, Hubei Clinical Research Center of Hypertension, Hubei University of Medicine, Shiyan, 442008, China
| | - Huimin Huang
- Sinopharm Dongfeng General Hospital, Hubei Clinical Research Center of Hypertension, Hubei University of Medicine, Shiyan, 442008, China
- Department of Pharmacy, Xi'an Jiaotong University, Xi'an, 710003, China
| |
Collapse
|
28
|
Zhou W, Li X, Zhou H, Hu Y, Chen Y, Guo D. TNF-α/IL-1β/IL-1α/IL-12 inflammatory cytokine axes coupled with TLR1/TLR3/TLR5/MYD88 immune signaling pathway over-activation contribute to simultaneous carotid and coronary artery and occlusion in elderly patients. Cytokine 2025; 185:156808. [PMID: 39556941 DOI: 10.1016/j.cyto.2024.156808] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/22/2024] [Accepted: 11/10/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND It remains difficult to evaluate the risk factors for concomitant carotid artery as well as coronary artery diseases in elderly patients. The aim of this research was to determine the TNF-α/IL-1β/IL-1α/IL-12 axes-TLR1/TLR3/TLR5/MYD88 immune signaling pathway interactions in coexistent carotid artery occlusion and coronary artery occlusion in elderly patients. METHODS Elderly patients, who underwent carotid ultrasonography and coronary computed tomography angiography, were consecutively included in this research. The analyzed groups consisted of those with coexistent carotid artery occlusion and coronary artery occlusion as well as healthy individuals were enrolled as control group. The circulating levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-1α (IL-1α), interleukin-12 (IL-12), toll-like receptor 1 (TLR1), toll-like receptor 3 (TLR3), toll-like receptor 5 (TLR5) and myeloid differentiation factor 88 (MYD88) were measured. RESULTS The biomarkers (TNF-α, IL-1β, IL-1α, IL-12, TLR1, TLR3, TLR5 and MYD88) were significantly increased in carotid artery occlusion + left circumflex coronary artery occlusion group when compared with control group and carotid artery occlusion + right coronary artery occlusion group, respectively (P < 0.001), and were further elevated in carotid artery occlusion + left anterior descending coronary artery occlusion group when compared to carotid artery occlusion + right coronary artery occlusion group and carotid artery occlusion + left circumflex coronary artery occlusion group, respectively (P < 0.001). CONCLUSION This research demonstrated that the TNF-α/IL-1β/IL-1α/IL-12 axes and TLR1/TLR3/TLR5/MYD88 immune signaling pathway implicated in the pathogenesis of carotid artery occlusion with coronary artery occlusion in elderly patients.
Collapse
Affiliation(s)
- Wenhang Zhou
- Xiamen Road Branch Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223005, China
| | - Xia Li
- Xiamen Road Branch Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223005, China
| | - Hualan Zhou
- Department of Geriatrics, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Youdong Hu
- Department of Geriatrics, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Ying Chen
- Department of Geriatrics, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Dianxuan Guo
- Xiamen Road Branch Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223005, China.
| |
Collapse
|
29
|
Yin Y, Xu J, Ilyas I, Xu S. Bioactive Flavonoids in Protecting Against Endothelial Dysfunction and Atherosclerosis. Handb Exp Pharmacol 2025; 287:1-31. [PMID: 38755351 DOI: 10.1007/164_2024_715] [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] [Indexed: 05/18/2024]
Abstract
Atherosclerosis is a common cardiovascular disease closely associated with factors such as hyperlipidaemia and chronic inflammation. Among them, endothelial dysfunction serves as a major predisposing factor. Vascular endothelial dysfunction is manifested by impaired endothelium-dependent vasodilation, enhanced oxidative stress, chronic inflammation, leukocyte adhesion and hyperpermeability, endothelial senescence, and endothelial-mesenchymal transition (EndoMT). Flavonoids are known for their antioxidant activity, eliminating oxidative stress induced by reactive oxygen species (ROS), thereby preventing the oxidation of low-density lipoprotein (LDL) cholesterol, reducing platelet aggregation, alleviating ischemic damage, and improving vascular function. Flavonoids have also been shown to possess anti-inflammatory activity and to protect the cardiovascular system. This review focuses on the protective effects of these naturally-occuring bioactive flavonoids against the initiation and progression of atherosclerosis through their effects on endothelial cells including, but not limited to, their antioxidant, anti-inflammatory, anti-thrombotic, and lipid-lowering properties. However, more clinical evidences are still needed to determine the exact role and optimal dosage of these compounds in the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yanjun Yin
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Jingjing Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Iqra Ilyas
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Suowen Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
30
|
Shao CL, Meng WT, Wang YC, Liu JJ, Ning K, Hou XX, Guo HD. Regulating NETs contributes to a novel antiatherogenic effect of MTHSWD via inhibiting endothelial injury and apoptosis. Int Immunopharmacol 2024; 143:113368. [PMID: 39418732 DOI: 10.1016/j.intimp.2024.113368] [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: 08/13/2024] [Revised: 10/05/2024] [Accepted: 10/06/2024] [Indexed: 10/19/2024]
Abstract
Neutrophil extracellular traps (NETs) are implicated in the occurrence and progression of atherosclerosis (AS), which can result in adverse cardiovascular events. We investigated the potential mechanism of action of Modified Taohong Siwu Decoction (MTHSWD) against AS based on its effect on NETs. A model of unstable plaque in AS was established by tandem stenosis (TS) of the right common carotid artery in ApoE-/- mice combined with a western diet (WD). The research found that MTHSWD reduced the weight of mice with AS to varying degrees, and significantly decreased the levels of plasma total cholesterol (TC) and triglycerides (TG). Meanwhile, we found that MTHSWD not only significantly improved cardiac EF, FS, cardiac hypertrophy, and ventricular remodeling, but also ameliorated the silent and depressed hypoactivity state caused by AS in ApoE-/- mice. Additionally, the study revealed that MTHSWD improved the severity of AS, protected the vascular structure, increased plaque stability and vessel patency. It also significantly reduced vascular cell apoptosis, platelet aggregation, and the presence of inflammatory cells such as neutrophils (NEUs), as well as the expression of neutrocyte elastase (NE) and myeloperoxidase (MPO), which are components of NETs. Subsequently, NEUs studies have shown that MTHSWD not only significantly reduces the dsDNA content of NETs, but also lowers the expression of NETs components NE and citH3. NETs treating the human umbilical vein endothelial cells (HUVECs) demonstrated that NETs differentially increased the protein expression of endothelial inflammatory adhesion factors CD62P, VCAM-1 and ICAM-1, while significantly decreasing the viability of HUVECs. Pharmacological treatment discovered that MTHSWD significantly improved HUVECs viability impaired by NETs, and promoted the growth and proliferation of endothelial cells. Furthermore, it significantly reduced early and late apoptosis of HUVECs caused by NETs, decreased the expression of pro-apoptotic proteins BAX and Cleaved-Caspase-3, and increased the expression of anti-apoptotic protein Bcl-2. Thus, study suggests that MTHSWD may improve body weight, lipid levels, cardiac function, vigour, and the severity of AS in ApoE-/- AS mice. The novel effect of MTHSWD against AS may be attributed to the inhibition of endothelial injury and apoptosis through the regulation of NETs. This, in turn, reduces the levels of platelets, inflammatory cells, and components of NETs in AS plaques, achieving a benign cycle that protects endothelial cells and vascular structure and function. This result provides some clues and evidence for studying the mechanism of action and clinical application of MTHSWD and its active ingredients against AS.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Atherosclerosis/drug therapy
- Atherosclerosis/pathology
- Extracellular Traps/drug effects
- Extracellular Traps/metabolism
- Humans
- Male
- Mice
- Human Umbilical Vein Endothelial Cells
- Mice, Inbred C57BL
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Neutrophils/drug effects
- Neutrophils/immunology
- Apolipoproteins E/genetics
- Mice, Knockout
- Disease Models, Animal
- Plaque, Atherosclerotic/drug therapy
- Plaque, Atherosclerotic/pathology
- Mice, Knockout, ApoE
- Cells, Cultured
Collapse
Affiliation(s)
- Chang-le Shao
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wan-Ting Meng
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ya-Chao Wang
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jia-Jia Liu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ke Ning
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xin-Xin Hou
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Hai-Dong Guo
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
31
|
Wu S, Dong R, Xie Y, Chen W, Liu W, Weng Y. CO-loaded hemoglobin/EGCG nanoparticles functional coatings for inflammation modulation of vascular implants. Regen Biomater 2024; 12:rbae148. [PMID: 39886364 PMCID: PMC11781197 DOI: 10.1093/rb/rbae148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/08/2024] [Accepted: 12/12/2024] [Indexed: 02/01/2025] Open
Abstract
During the implantation process of cardiovascular implants, vascular damage caused by inflammation occurs, and the inflammatory process is accompanied by oxidative stress. Currently, carbon monoxide (CO) has been demonstrated to exhibit various biological effects including vasodilatation, antithrombotic, anti-inflammatory, apoptosis-inducing and antiproliferative properties. In this study, hemoglobin/epigallocatechin-3-gallate (EGCG) core-shell nanoparticle-containing coating on stainless steel was prepared for CO loading and inflammation modulation. Inspired by strong coordination ability with CO, hemoglobin nanoparticle was first prepared and encapsulated into EGCG metal-phenolic networks. A polydopamine (PDA) linking layer was then coated on 316 stainless steel, and the hemoglobin/EGCG nanoparticles were loaded with the subsequent PDA deposition. It showed that the maximum release amount of CO by the coating was 17.0 nmol/cm2 in 48 h. In vitro evaluations conducted in a simulated inflammatory environment revealed that the coating, which released CO from hemoglobin/EGCG nanoparticles, effectively mitigated the lipopolysaccharide-induced inflammatory response in macrophages. Specifically, it decreased the expression of tumor necrosis factor-α, increased the expression of interleukin-10, suppressed the polarization of macrophages toward the M1 phenotype and reduced intracellular reactive oxygen species (ROS). Furthermore, under simulated oxidative stress conditions, the coating decreased the apoptosis of endothelial cells induced by oxidative stress and down-regulated intracellular ROS levels. In vivo implantation results further confirmed that the coating, with its hemoglobin/EGCG nanoparticles and CO release capabilities, reduced macrophage-mediated inflammatory responses and modulated the polarization phenotype of macrophages.
Collapse
Affiliation(s)
- Sui Wu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Ruichen Dong
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Yinhong Xie
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Wenhao Chen
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Wei Liu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Yajun Weng
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| |
Collapse
|
32
|
Du L, Wang Y, Ma H, Fan J, Wang S, Liu J, Wang X. Exploring novel markers for coronary heart disease associated with systemic lupus erythematosus: A review. Medicine (Baltimore) 2024; 103:e40773. [PMID: 39686502 PMCID: PMC11651451 DOI: 10.1097/md.0000000000040773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/13/2024] [Indexed: 12/18/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune condition that is characterized by the production of autoantibodies and sustained inflammatory damage. Coronary heart disease (CHD) is a common complication of SLE, significantly increases CHD-related mortality in SLE patients. Despite conventional risk factors, the mechanisms contributing to a higher CHD risk require further investigation, with the immune and inflammatory aspects of SLE playing a significant role. Endothelial cell damage and dysfunction are key factors in the progression of coronary atherosclerosis in SLE patients. This review specifically focuses on endothelial dysfunction and the role of specific microRNAs in the context of SLE and CHD. In addition, we discuss the effects and functions of oxidative stress markers, endothelial progenitor cells, and circulating endothelial cells in individuals with both SLE and CHD. We also explored the typical inflammatory markers associated with SLE and CHD, addressing their clinical significance and limitations.
Collapse
Affiliation(s)
- Linping Du
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Weifang, China
- Shandong Second Medical University, Weifang, China
| | - Yuqun Wang
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Weifang, China
- Shandong Second Medical University, Weifang, China
| | - Honglei Ma
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Weifang, China
- Shandong Second Medical University, Weifang, China
| | - Jiaheng Fan
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Weifang, China
- Shandong Second Medical University, Weifang, China
| | - Shiqi Wang
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Weifang, China
- Shandong Second Medical University, Weifang, China
| | - Junhong Liu
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Weifang, China
- Shandong Second Medical University, Weifang, China
| | - Xiaodong Wang
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Weifang, China
| |
Collapse
|
33
|
Pourmousavi L, Asadi RH, Zehsaz F, Jadidi RP. Potential therapeutic effects of crocin. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7395-7420. [PMID: 38758225 DOI: 10.1007/s00210-024-03131-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 04/29/2024] [Indexed: 05/18/2024]
Abstract
Crocin, a natural bioactive compound derived from saffron (Crocus sativus) and other Crocus genera, has gained significant attention recently due to its potential therapeutic properties. The multifaceted nature of crocin's biological effects has piqued the interest of researchers and health enthusiasts, prompting further investigations into its mechanisms of action and therapeutic applications. This review article comprehensively explores the emerging evidence supporting crocin's role as a promising ally in protecting against metabolic disorders. The review covers the molecular mechanisms underlying crocin's beneficial effects and highlights its potential applications in preventing and treating diverse pathological conditions. Understanding the mechanisms through which crocin exerts its protective effects could advance scientific knowledge and offer potential avenues for developing novel therapeutic interventions. As we uncover the potential of crocin as a valuable ally in the fight against disorders, it becomes evident that nature's palette holds remarkable solutions for enhancing our health.
Collapse
Affiliation(s)
- Laleh Pourmousavi
- Department of Sport Sciences, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | | | - Farzad Zehsaz
- Department of Sport Sciences, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | | |
Collapse
|
34
|
Surówka A, Żołnierczuk M, Prowans P, Grabowska M, Kupnicka P, Markowska M, Szlosser Z, Kędzierska-Kapuza K. The Effect of Chronic Immunosuppressive Regimen Treatment on Apoptosis in the Heart of Rats. Pharmaceuticals (Basel) 2024; 17:1188. [PMID: 39338354 PMCID: PMC11435130 DOI: 10.3390/ph17091188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/29/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Chronic immunosuppressive therapy is currently the only effective method to prevent acute rejection of a transplanted organ. Unfortunately, the expected effect of treatment brings a number of grave side effects, one of the most serious being cardiovascular complications. In our study, we wanted to investigate how treatment with commonly used immunosuppressive drugs affects the occurrence of programmed cardiac cell death. For this purpose, five groups of rats were treated with different triple immunosuppressive regimens. Cardiac tissue fragments were subjected to the TUNEL assay to visualize apoptotic cells. The expression of Bcl-2 protein, Bax protein, caspase 3 and caspase 9 was also assessed. This study indicates that all immunosuppressive protocols used chronically at therapeutic doses result in an increased percentage of cells undergoing apoptosis in rat heart tissue. The greatest changes were recorded in the TMG (rats treated with tacrolimus, mycophenolate mofetil and glucocorticosteroids) and CMG (rats treated with cyclosporin A, mycophenolate mofetil and glucocorticosteroids) groups. The TRG (rats treated with rapamycin, tacrolimus and glucocorticosteroids) group showed the lowest percentage of apoptotic cells. The internal apoptosis pathway was confirmed only in the TMG group; in the remaining groups, the results indicate programmed cell death via the receptor pathway.
Collapse
Affiliation(s)
- Anna Surówka
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland (Z.S.)
| | - Michał Żołnierczuk
- Department of Vascular Surgery, General Surgery and Angiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Piotr Prowans
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland (Z.S.)
| | - Marta Grabowska
- Department of Histology and Developmental Biology, Faculty of Health Sciences, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Marta Markowska
- Department of Plastic and Reconstructive Surgery, 109 Military Hospital, 71-422 Szczecin, Poland;
| | - Zbigniew Szlosser
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland (Z.S.)
| | - Karolina Kędzierska-Kapuza
- Department of Gastroenterological Surgery and Transplantology, National Medical Institute, Ministry of Interior Affairs and Administration, 02-507 Warsaw, Poland;
| |
Collapse
|
35
|
Zeng LC, Zhang SH, Fu N, Gao FJ, Ren NF, Zheng W, Lin BX, Chen H. miR-3202 inhibits bronchopulmonary dysplasia-mediated apoptosis and oxidative stress in bronchial epithelial cells via targeting RAG1. Pathol Res Pract 2024; 261:155482. [PMID: 39067173 DOI: 10.1016/j.prp.2024.155482] [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] [Received: 01/31/2024] [Revised: 07/04/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND BPD is a refractory disease affecting preterm infants with alveolar dysplasia and declined pulmonary function. However, the molecular mechanism underlying BPD is largely unknown. To explore the pathogenic mechanism of BPD and to facilitate better diagnosis and treatment of this disease. METHOD The DEMs and DEGs in BPD vs. Control samples from the miRNA expression data in GSE108754 and mRNA expression data in the GSE108755 were screened, followed by the construction of the miRNA-mRNA regulatory network. DEGs PPI network and hub DEGs analysis were constructed by using the STRING database and Cytoscape software. Functional and pathway enrichment analyses were then performed for these DEGs and DEMs based on the ClusterProfiler package in the R and the miRWalk database. The k-mean algorithm is used to perform clustering analysis of DEGs. Cellular experiments (flow cytometry, western blot, RT-PCR, dual-luciferase reporter assay) were used to validate the results of bioinformatics. RESULTS We obtained 20 DEMs and 262 DEGs. A 15 DEMs-11 DEGs regulatory network was constructed. miR-3202-RAG1 is a core sub-network. Hyperoxia induced a cell model of BPD. The upregulation of RAG1 and downregulation of miR-3202 were observed in BPD cells. Furthermore, siRNA targeting RAG1 was transfected into BEAS-2B cells to inhibit its expression and miR-3202 mimics was transfected into the cells to increase its expression. Inhibition of RAG1 and elevation of miR-3202 inhibit cell apoptosis and reduce ROS level caused by hyperoxia. A double-luciferase reporter assay revealed that miR-3202 directly targets RAG1. CONCLUSION The miRNA-3202/RAG1 axis contributes into BPD-induced cell apoptosis and ROS production. The present study provides a probable target for the treatment of BPD.
Collapse
Affiliation(s)
- Li-Chun Zeng
- Neonatology Department, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong 518133, China
| | - Shu-Hui Zhang
- Neonatology Department, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong 518133, China
| | - Na Fu
- Neonatology Department, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong 518133, China
| | - Feng-Jiao Gao
- Neonatology Department, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong 518133, China
| | - Na-Fang Ren
- Neonatology Department, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong 518133, China
| | - Wei Zheng
- Neonatology Department, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong 518133, China
| | - Bai-Xing Lin
- Neonatology Department, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong 518133, China
| | - Hui Chen
- Neonatology Department, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong 518133, China.
| |
Collapse
|
36
|
Liang L, Yi X, Wang C, Su L, Wei G. The Impact of miR-34a on Endothelial Cell Viability and Apoptosis in Ischemic Stroke: Unraveling the MTHFR-Homocysteine Pathway. CLIN INVEST MED 2024; 47:27-37. [PMID: 39325580 DOI: 10.3138/cim-2024-2711] [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] [Indexed: 09/28/2024]
Abstract
INTRODUCTION Ischemic stroke (IS) is a global health concern, often tied to dyslipidemia and vascular endothelial dysfunction. MicroRNA-34a (miR-34a) was reported to be up-regulated in the blood samples of patients with IS, but the specific role of miR-34a and methylenetetrahydrofolate reductase (MTHFR) in IS remains to be elucidated. METHODS We studied 143 subjects: 71 IS patients, and 72 healthy controls. Human umbilical vein endothelial cells (HUVECs) were cultured and transfected with a miR-34a mimic, inhibitor, or negative control. The miR-34a expression in serum and HUVECs was quantified via quantitative reverse transcription polymerase chain reaction (qRT-PCR). Viability and apoptosis of HUVECs were assessed using CCK-8 assay and flow cytometry. The expression levels of bcl-2, bax, cyt-c, cleaved caspase 3, MTHFR, and homocysteine were measured by Western blot or enzyme-linked immunosorbent assay (ELISA). The relationship between miR-34a and MTHFR was verified by luciferase reporter assay. The levels of MTHFR and homocysteine in serum were examined by ELISA. RESULTS MiR-34a expression was increased in IS patients and inhibited viability of HUVECs while promoting their apoptosis. Overexpression of miR-34a up-regulated pro-apoptotic proteins (bax, cyt-c and cleaved caspase 3) and down-regulated anti-apoptotic protein bcl-2 in HUVECs. MTHFR was identified as the downstream target of miR-34a and its expression was reduced by miR-34a overexpression, while homocysteine levels increased. Consistently, MTHFR levels were lower and homocysteine levels were higher in IS patients compared with controls. DISCUSSION Our results suggest that up-regulated miR-34a plays a role in the pathogenesis of IS, potentially through inhibiting MTHFR expression and increasing homocysteine in endothelial cells. Therefore, miR-34a might be a therapeutic target for IS.
Collapse
Affiliation(s)
- Lina Liang
- Center for Medical Laboratory Science, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi, 533000, China
- Baise Key Laboratory for Research and Development on Clinical Molecular Diagnosis for High-Incidence Diseases, Guangxi, 533000, China
- *These authors contributed equally
| | - Xueli Yi
- Center for Medical Laboratory Science, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi, 533000, China
- Baise Key Laboratory for Research and Development on Clinical Molecular Diagnosis for High-Incidence Diseases, Guangxi, 533000, China
- *These authors contributed equally
| | - Chunfang Wang
- Center for Medical Laboratory Science, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi, 533000, China
- Baise Key Laboratory for Research and Development on Clinical Molecular Diagnosis for High-Incidence Diseases, Guangxi, 533000, China
| | - Li Su
- Department of Neurology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, 533000, China
| | - Guijiang Wei
- Center for Medical Laboratory Science, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi, 533000, China
- Baise Key Laboratory for Research and Development on Clinical Molecular Diagnosis for High-Incidence Diseases, Guangxi, 533000, China
| |
Collapse
|
37
|
Bayyurt B, Akın Ş, Özbilüm Şahin N, Yelkuvan İ. Association between NKILA and some apoptotic gene expression in atherosclerosis. PeerJ 2024; 12:e17915. [PMID: 39184397 PMCID: PMC11344533 DOI: 10.7717/peerj.17915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/23/2024] [Indexed: 08/27/2024] Open
Abstract
Oxidized light-density lipoprotein (ox-LDL) causes endothelial dysfunction, which is an important determinant of atherogenesis, and subsequently leads to apoptosis. Atherosclerosis is one of the most significant cardiovascular diseases (CVDs) threatening human health and causes death worldwide. Recently, long noncoding RNAs (lncRNAs) have been suggested to involved in vascular biology. Ox-LDL activates nuclear factor kappa-B (NF-κB), and NF-κB interacting lncRNA (NKILA) inhibits NF-κB signaling. In this study, the hypothesis is that NKILA may regulate endothelial cell (EC) apoptosis and, therefore, play a role in the pathogenesis of atherosclerosis. This hypothesis is based on the knowledge that EC apoptosis contributes to atherosclerosis development and that NKILA has become a prominent lncRNA in CVDs. The expression of Bcl-2-associated X protein (BAX), caspase 9 (CASP9), cytochrome c (Cyt c, CYCS), apoptotic protease activating factor 1 (APAF1), and B-cell lymphoma 2 (BCL-2) genes in human umbilical vein endothelial cells (HUVEC) treated with ox-LDL and transfected with NKILA siRNA was analyzed using quantitative reverse transcription polymerase chain reaction (RT-qPCR). BAX, CASP9, CYCS, APAF1, and BCL-2 gene expression was downregulated in ox-LDL and NKILA siRNA-treated HUVEC. In addition, when threshold/quantification cycle (Cq) values of NKILA gene expression increased, Cq values of BAX, CASP9, APAF1, and BCL-2 gene expression increased statistics significantly. The expression detection of all these genes, resulting from NKILA gene silencing, may provide guidance for epigenetic studies on EC apoptosis in atherosclerosis.
Collapse
Affiliation(s)
- Burcu Bayyurt
- Department of Medical Biology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| | - Şeyda Akın
- Department of Medical Biology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| | - Nil Özbilüm Şahin
- Department of Molecular Biology and Genetics, Faculty of Science, Sivas Cumhuriyet University, Sivas, Turkey
| | - İzzet Yelkuvan
- Department of Medical Biology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| |
Collapse
|
38
|
Zhou Z, Liu Y, Xie P, Yin Z. A ROS-responsive multifunctional targeted prodrug micelle for atherosclerosis treatment. Int J Pharm 2024; 660:124352. [PMID: 38901540 DOI: 10.1016/j.ijpharm.2024.124352] [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: 01/31/2024] [Revised: 06/10/2024] [Accepted: 06/15/2024] [Indexed: 06/22/2024]
Abstract
Atherosclerosis is a chronic multifactorial cardiovascular disease. To combat atherosclerosis effectively, it is necessary to develop precision and targeted therapy in the early stages of plaque formation. In this study, a simvastatin (SV)-containing prodrug micelle SPCPV was developed by incorporating a peroxalate ester bond (PO). SPCPV could specifically target VCAM-1 overexpressed at atherosclerotic lesions. SPCPV contains a carrier (CP) composed of cyclodextrin (CD) and polyethylene glycol (PEG). At the lesions, CP and SV exerted multifaceted anti-atherosclerotic effects. In vitro studies demonstrated that intracellular reactive oxygen species (ROS) could induce the release of SV from SPCPV. The uptake of SPCPV was higher in inflammatory cells than in normal cells. Furthermore, in vitro experiments showed that SPCPV effectively reduced ROS levels, possessed anti-inflammatory properties, inhibited foam cell formation, and promoted cholesterol efflux. In vivo studies using atherosclerotic rats showed that SPCPV reduced the thickness of the vascular wall and low-density lipoprotein (LDL). This study developed a drug delivery strategy that could target atherosclerotic plaques and treat atherosclerosis by integrating the carrier with SV. The findings demonstrated that SPCPV possessed high stability and safety and had great therapeutic potential for treating early-stage atherosclerosis.
Collapse
Affiliation(s)
- Zishuo Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yaxue Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Pei Xie
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zongning Yin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
39
|
Chen S, Wu S, Lin B. The potential therapeutic value of the natural plant compounds matrine and oxymatrine in cardiovascular diseases. Front Cardiovasc Med 2024; 11:1417672. [PMID: 39041001 PMCID: PMC11260750 DOI: 10.3389/fcvm.2024.1417672] [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: 04/15/2024] [Accepted: 06/17/2024] [Indexed: 07/24/2024] Open
Abstract
Matrine (MT) and Oxymatrine (OMT) are two natural alkaloids derived from plants. These bioactive compounds are notable for their diverse pharmacological effects and have been extensively studied and recognized in the treatment of cardiovascular diseases in recent years. The cardioprotective effects of MT and OMT involve multiple aspects, primarily including antioxidative stress, anti-inflammatory actions, anti-atherosclerosis, restoration of vascular function, and inhibition of cardiac remodeling and failure. Clinical pharmacology research has identified numerous novel molecular mechanisms of OMT and MT, such as JAK/STAT, Nrf2/HO-1, PI3 K/AKT, TGF-β1/Smad, and Notch pathways, providing new evidence supporting their promising therapeutic potential against cardiovascular diseases. Thus, this review aims to investigate the potential applications of MT and OMT in treating cardiovascular diseases, encompassing their mechanisms, efficacy, and safety, confirming their promise as lead compounds in anti-cardiovascular disease drug development.
Collapse
Affiliation(s)
| | | | - Bin Lin
- Department of Cardiovascular Medicine, Wenzhou Central Hospital, Wenzhou, China
| |
Collapse
|
40
|
Liu Y, Long Y, Fang J, Liu G. Advances in the Anti-Atherosclerotic Mechanisms of Epigallocatechin Gallate. Nutrients 2024; 16:2074. [PMID: 38999821 PMCID: PMC11243004 DOI: 10.3390/nu16132074] [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: 05/29/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/14/2024] Open
Abstract
Atherosclerosis (AS) is a common clinical sickness and the major pathological basis of ischemic cardiocerebrovascular diseases (CCVDs). The pathogenesis of AS involves a variety of risk factors, and there is a lack of effective preventive and curative drugs that can completely treat AS. In recent years, with the improvement of people's living standards and changes in dietary habits, the morbidity and mortality rates of AS are on the rise, and the age of onset tends to be younger. The formation of AS is closely related to a variety of factors, and the main factors include lipid metabolism disorders, endothelial damage, inflammation, unstable plaques, etc. Epigallocatechin gallate (EGCG), as one of the main components of catechins, has a variety of pharmacological effects, and its role in the prevention of AS and the protection of cardiovascular and cerebral blood vessels has been highly valued. Recent epidemiological investigations and various in vivo and ex vivo experiments have shown that EGCG is capable of resisting atherosclerosis and reducing the morbidity and mortality of AS. In this paper, we reviewed the anti-AS effects of EGCG and its mechanisms in recent years, including the regulation of lipid metabolism, regulation of intestinal flora disorders, improvement of vascular endothelial cell functions, inhibition of inflammatory factors expression, regulation of inflammatory signaling pathways, inhibition of matrix metalloproteinase (MMP) expression, and inhibition of platelet aggregation, which are helpful for the prevention of cardiocerebrovascular diseases.
Collapse
Affiliation(s)
- Yihui Liu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Yiling Long
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Jun Fang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Gang Liu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
41
|
Chen J, He Q, Jin J. Targeting dendritic cell activation: the therapeutic impact of paeoniflorin in cortosteroid-dependent dermatitis management. Arch Dermatol Res 2024; 316:348. [PMID: 38849562 DOI: 10.1007/s00403-024-03002-3] [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: 04/11/2024] [Revised: 04/11/2024] [Accepted: 04/26/2024] [Indexed: 06/09/2024]
Abstract
This study investigates the mechanism through which paeoniflorin inhibits TSLP expression to regulate dendritic cell activation in corticosteroid-dependent dermatitis treatment. Utilizing databases like TCMSP, we identified paeoniflorin's components, targets, and constructed networks. Molecular docking and gene enrichment analysis helped pinpoint key targets and pathways affected by paeoniflorin. In vitro and in vivo models were used to study CD80, CD86, cytokines, T-cell activation, skin lesions, histopathological changes, TSLP, CD80, and CD86 expression. Our study revealed paeoniflorin's active constituent targeting IL-6 in corticosteroid-dependent dermatitis. In vitro experiments demonstrated reduced TSLP expression, CD80, CD86, and cytokine secretion post-paeoniflorin treatment. In vivo, paeoniflorin significantly decreased skin lesion severity, cytokine levels, TSLP, CD80, and CD86 expression. The study highlights paeoniflorin's efficacy in inhibiting TSLP expression and suppressing dendritic cell activation in corticosteroid-dependent dermatitis, suggesting its potential as a therapeutic intervention. Additionally, it offers insights into the complex molecular mechanisms underlying paeoniflorin's anti-inflammatory properties in treating corticosteroid-dependent dermatitis.
Collapse
Affiliation(s)
- Jinjin Chen
- Department of Dermatology, Hubei Provincial Hospital of Traditional Chinese Medicine, No. 4 Huayuanshan, Wuchang District, Wuhan, Hubei Province, 430061, China
| | - Qi He
- Department of Dermatology, Hubei Provincial Hospital of Traditional Chinese Medicine, No. 4 Huayuanshan, Wuchang District, Wuhan, Hubei Province, 430061, China
| | - Jing Jin
- Department of Dermatology, Hubei Provincial Hospital of Traditional Chinese Medicine, No. 4 Huayuanshan, Wuchang District, Wuhan, Hubei Province, 430061, China.
| |
Collapse
|
42
|
Wang J, Zou J, Shi Y, Zeng N, Guo D, Wang H, Zhao C, Luan F, Zhang X, Sun J. Traditional Chinese medicine and mitophagy: A novel approach for cardiovascular disease management. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155472. [PMID: 38461630 DOI: 10.1016/j.phymed.2024.155472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 02/06/2024] [Accepted: 02/20/2024] [Indexed: 03/12/2024]
Abstract
BACKGROUND Cardiovascular disease (CVD) remains the leading cause of morbidity and mortality worldwide, imposing an enormous economic burden on individuals and human society. Laboratory studies have identified several drugs that target mitophagy for the prevention and treatment of CVD. Only a few of these drugs have been successful in clinical trials, and most studies have been limited to animal and cellular models. Furthermore, conventional drugs used to treat CVD, such as antiplatelet agents, statins, and diuretics, often result in adverse effects on patients' cardiovascular, metabolic, and respiratory systems. In contrast, traditional Chinese medicine (TCM) has gained significant attention for its unique theoretical basis and clinical efficacy in treating CVD. PURPOSE This paper systematically summarizes all the herbal compounds, extracts, and active monomers used to target mitophagy for the treatment of CVD in the last five years. It provides valuable information for researchers in the field of basic cardiovascular research, pharmacologists, and clinicians developing herbal medicines with fewer side effects, as well as a useful reference for future mitophagy research. METHODS The search terms "cardiovascular disease," "mitophagy," "herbal preparations," "active monomers," and "cardiac disease pathogenesis" in combination with "natural products" and "diseases" were used to search for studies published in the past five years until January 2024. RESULTS Studies have shown that mitophagy plays a significant role in the progression and development of CVD, such as atherosclerosis (AS), heart failure (HF), myocardial infarction (MI), myocardial ischemia/reperfusion injury (MI/RI), cardiac hypertrophy, cardiomyopathy, and arrhythmia. Herbal compound preparations, crude extracts, and active monomers have shown potential as effective treatments for these conditions. These substances protect cardiomyocytes by inducing mitophagy, scavenging damaged mitochondria, and maintaining mitochondrial homeostasis. They display notable efficacy in combating CVD. CONCLUSION TCM (including herbal compound preparations, extracts, and active monomers) can treat CVD through various pharmacological mechanisms and signaling pathways by inducing mitophagy. They represent a hotspot for future cardiovascular basic research and a promising candidate for the development of future cardiovascular drugs with fewer side effects and better therapeutic efficacy.
Collapse
Affiliation(s)
- Jinhui Wang
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi, PR China
| | - Junbo Zou
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi, PR China
| | - Yajun Shi
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi, PR China
| | - Nan Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, PR China
| | - Dongyan Guo
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi, PR China
| | - He Wang
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi, PR China
| | - Chongbo Zhao
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi, PR China
| | - Fei Luan
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi, PR China.
| | - Xiaofei Zhang
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi, PR China.
| | - Jing Sun
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi, PR China.
| |
Collapse
|
43
|
Li JN, Wang MY, Tan YR, Wang LL. Multidirectional Intervention of Chinese Herbal Medicine in the Prevention and Treatment of Atherosclerosis: From Endothelial Protection to Immunomodulation. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:925-947. [PMID: 38798151 DOI: 10.1142/s0192415x24500381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Atherosclerosis is a significant risk factor for developing cardiovascular disease and a leading cause of death worldwide. The occurrence of atherosclerosis is closely related to factors such as endothelial injury, lipid deposition, immunity, and inflammation. Conventional statins, currently used in atherosclerosis treatment, have numerous adverse side effects that limit their clinical utility, prompting the urgent need to identify safer and more effective therapeutic alternatives. Growing evidence indicates the significant potential of Chinese herbs in atherosclerosis treatment. Herbal monomer components, such as natural flavonoid compounds extracted from herbs like Coptis chinensis and Panax notoginseng, have been utilized for their lipid-lowering and inflammation-inhibiting effects in atherosclerosis treatment. These herbs can be used as single components in treating diseases and with other Chinese medicines to form herbal combinations. This approach targets the disease mechanism in multiple ways, enhancing the therapeutic effects. Thus, this review examines the roles of Chinese herbal medicine monomers and Chinese herbal compounds in inhibiting atherosclerosis, including regulating lipids, improving endothelial function, reducing oxidative stress, regulating inflammation and the immune response, and apoptosis. By highlighting these roles, our study offers new perspectives on atherosclerosis treatment with Chinese herbs and is anticipated to contribute to advancements in related research fields.
Collapse
Affiliation(s)
- Jia-Ni Li
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Xiangya Road 88, Changsha 410078, Hunan, P. R. China
| | - Meng-Yu Wang
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Xiangya Road 88, Changsha 410078, Hunan, P. R. China
| | - Yu-Rong Tan
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Xiangya Road 88, Changsha 410078, Hunan, P. R. China
| | - Li-Li Wang
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Xiangya Road 88, Changsha 410078, Hunan, P. R. China
| |
Collapse
|
44
|
Zhou ZY, Wu L, Liu YF, Tang MY, Tang JY, Deng YQ, Liu L, Nie BB, Zou ZK, Huang L. IRE1α: from the function to the potential therapeutic target in atherosclerosis. Mol Cell Biochem 2024; 479:1079-1092. [PMID: 37310588 DOI: 10.1007/s11010-023-04780-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/03/2023] [Indexed: 06/14/2023]
Abstract
Inositol requiring enzyme 1 (IRE1) is generally thought to control the most conserved pathway in the unfolded protein response (UPR). Two isoforms of IRE1, IRE1α and IRE1β, have been reported in mammals. IRE1α is a ubiquitously expressed protein whose knockout shows marked lethality. In contrast, the expression of IRE1β is exclusively restricted in the epithelial cells of the respiratory and gastrointestinal tracts, and IRE1β-knockout mice are phenotypically normal. As research continues to deepen, IRE1α was showed to be tightly linked to inflammation, lipid metabolism regulation, cell death and so on. Growing evidence also suggests an important role for IRE1α in promoting atherosclerosis (AS) progression and acute cardiovascular events through disrupting lipid metabolism balance, facilitating cells apoptosis, accelerating inflammatory responses and promoting foam cell formation. In addition, IRE1α was recognized as novel potential therapeutic target in AS prevention. This review provides some clues about the relationship between IRE1α and AS, hoping to contribute to further understanding roles of IRE1α in atherogenesis and to be helpful for the design of novel efficacious therapeutics agents targeting IRE1α-related pathways.
Collapse
Affiliation(s)
- Zheng-Yang Zhou
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Li Wu
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Yi-Fan Liu
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Mu-Yao Tang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Jing-Yi Tang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Anaesthesiology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Ya-Qian Deng
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Lei Liu
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Bin-Bin Nie
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Zi-Kai Zou
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Liang Huang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China.
| |
Collapse
|
45
|
Di T, He L, Shi Q, Chen L, Zhu L, Zhao S, Zhang C. Emodin Blocks mPTP Opening and Improves LPS-Induced HMEC-1 Cell Injury by Upregulation of ATP5A1. Chem Biodivers 2024; 21:e202301916. [PMID: 38511277 DOI: 10.1002/cbdv.202301916] [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: 11/29/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND Emodin has been shown to exert anti-inflammatory and cytoprotective effects. Our study aimed to identify a novel anti-inflammatory mechanism of emodin. METHODS An LPS-induced model of microvascular endothelial cell (HMEC-1) injury was constructed. Cell proliferation was examined using a CCK-8 assay. The effects of emodin on reactive oxygen species (ROS), cell migration, the mitochondrial membrane potential (MMP), and the opening of the mitochondrial permeability transition pore (mPTP) were evaluated. Actin-Tracker Green was used to examine the relationship between cell microfilament reconstruction and ATP5A1 expression. The effects of emodin on the expression of ATP5A1, NALP3, and TNF-α were determined. After treatment with emodin, ATP5A1 and inflammatory factors (TNF-α, IL-1, IL-6, IL-13 and IL-18) were examined by Western blotting. RESULTS Emodin significantly increased HMEC-1 cell proliferation and migration, inhibited the production of ROS, increased the mitochondrial membrane potential, and blocked the opening of the mPTP. Moreover, emodin could increase ATP5A1 expression, ameliorate cell microfilament remodeling, and decrease the expression of inflammatory factors. In addition, when ATP5A1 was overexpressed, the regulatory effect of emodin on inflammatory factors was not significant. CONCLUSION Our findings suggest that emodin can protect HMEC-1 cells against inflammatory injury. This process is modulated by the expression of ATP5A1.
Collapse
Affiliation(s)
- Tietao Di
- Department of Trauma Orthopedics, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550000, Guizhou, China
| | - Limin He
- Department of Trauma Orthopedics, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550000, Guizhou, China
| | - Qing Shi
- Department of Nutrition, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550000, Guizhou, China
| | - Lu Chen
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550000, Guizhou, China
| | - Lei Zhu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550000, Guizhou, China
| | - Sisi Zhao
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550000, Guizhou, China
| | - Chunling Zhang
- Department of Nutrition, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550000, Guizhou, China
| |
Collapse
|
46
|
Wang S, He H, Mao Y, Zhang Y, Gu N. Advances in Atherosclerosis Theranostics Harnessing Iron Oxide-Based Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308298. [PMID: 38368274 PMCID: PMC11077671 DOI: 10.1002/advs.202308298] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/06/2024] [Indexed: 02/19/2024]
Abstract
Atherosclerosis, a multifaceted chronic inflammatory disease, has a profound impact on cardiovascular health. However, the critical limitations of atherosclerosis management include the delayed detection of advanced stages, the intricate assessment of plaque stability, and the absence of efficacious therapeutic strategies. Nanotheranostic based on nanotechnology offers a novel paradigm for addressing these challenges by amalgamating advanced imaging capabilities with targeted therapeutic interventions. Meanwhile, iron oxide nanoparticles have emerged as compelling candidates for theranostic applications in atherosclerosis due to their magnetic resonance imaging capability and biosafety. This review delineates the current state and prospects of iron oxide nanoparticle-based nanotheranostics in the realm of atherosclerosis, including pivotal aspects of atherosclerosis development, the pertinent targeting strategies involved in disease pathogenesis, and the diagnostic and therapeutic roles of iron oxide nanoparticles. Furthermore, this review provides a comprehensive overview of theranostic nanomedicine approaches employing iron oxide nanoparticles, encompassing chemical therapy, physical stimulation therapy, and biological therapy. Finally, this review proposes and discusses the challenges and prospects associated with translating these innovative strategies into clinically viable anti-atherosclerosis interventions. In conclusion, this review offers new insights into the future of atherosclerosis theranostic, showcasing the remarkable potential of iron oxide-based nanoparticles as versatile tools in the battle against atherosclerosis.
Collapse
Affiliation(s)
- Shi Wang
- State Key Laboratory of Digital Medical EngineeringJiangsu Key Laboratory for Biomaterials and DevicesSchool of Biological Sciences & Medical EngineeringSoutheast UniversityNanjing210009P. R. China
| | - Hongliang He
- State Key Laboratory of Digital Medical EngineeringJiangsu Key Laboratory for Biomaterials and DevicesSchool of Biological Sciences & Medical EngineeringSoutheast UniversityNanjing210009P. R. China
| | - Yu Mao
- School of MedicineNanjing UniversityNanjing210093P. R. China
| | - Yu Zhang
- State Key Laboratory of Digital Medical EngineeringJiangsu Key Laboratory for Biomaterials and DevicesSchool of Biological Sciences & Medical EngineeringSoutheast UniversityNanjing210009P. R. China
| | - Ning Gu
- School of MedicineNanjing UniversityNanjing210093P. R. China
| |
Collapse
|
47
|
Zhang NR, Wen Y, Li J, Zheng WJ, Jin SQ. Regular transient limb ischemia improves endothelial function and inhibits endothelial cell apoptosis to prevent atherosclerosis in rabbit. BMC Cardiovasc Disord 2024; 24:209. [PMID: 38627625 PMCID: PMC11020181 DOI: 10.1186/s12872-024-03869-0] [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: 12/18/2023] [Accepted: 03/30/2024] [Indexed: 04/19/2024] Open
Abstract
AIMS Regular transient limb ischemia (RTLI) can prevent atherosclerosis (AS) progression in hypercholesterolemic rabbits. This study aimed to investigate the minimum effective intensity and possible mechanisms of RTLI for preventing atherosclerosis. METHODS Eighty rabbits were divided into eight groups: normal (N), high cholesterol (H), three RTLI [three RTLI cycles every other day (R3qod), three RTLI cycles daily (R3qd), and six RTLI cycles daily (R6qd), each cycle of RTLI included 5 min of limb ischemia followed by 5 min limb reperfusion], and three correlated sham RTLI [sham ischemia for 30 min once every other day (S3qod), sham ischemia for 30 min once daily (S3qd), and sham ischemia for 60 min once daily (S6qd)]. Rabbits in group N were kept normally, while the others were fed 1% cholesterol diet for 12 weeks. The RTLI and sham RTLI groups were received RTLI or sham RTLI procedure, respectively. The plaque area in the thoracic aorta was determined by oil red O staining, and quantifying the ratio of plaque area to intimal area (PA/IA). Endothelium-dependent and -independent relaxation were also determined. Endothelial cell were isolated from abdominal aorta of rabbits, and the apoptosis ratio was detected using flow cytometry. RESULTS The PA/IA and early apoptotic cell ratio was significantly lower as well as the endothelium-dependent relaxation response was higher in group R6qd than those in groups H and S6qd, while those in the R3qod group was not significantly different from those in groups H and S3qod, as well as those in the R3qd group showed no significant difference compared to those in groups H and S3qd. CONCLUSIONS Six cycles of RTLI daily was the optimal effective intensity to prevent AS progression in rabbits. Endothelial function improvement and apoptosis inhibition might contribute to the anti-AS effects.
Collapse
Affiliation(s)
- Nan-Rong Zhang
- Department of Anesthesia, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510005, Guangdong, China
| | - Yi Wen
- Department of Anesthesia, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
| | - Jing Li
- Department of Anesthesia, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
| | - Wan-Jun Zheng
- Department of Anesthesia, Guangzhou Women and Children's Medical Center, Guangzhou, 510623, Guangdong, China
| | - San-Qing Jin
- Department of Anesthesia, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510005, Guangdong, China.
| |
Collapse
|
48
|
Liu Y, Gong J, Wang Q, Wei N, Zhao L, Wu Z. Activation of the Nrf2/HO-1 axis by glutaredoxin 2 overexpression antagonizes vascular endothelial cell oxidative injury and inflammation under LPS exposure. Cytotechnology 2024; 76:167-178. [PMID: 38495299 PMCID: PMC10940561 DOI: 10.1007/s10616-023-00606-x] [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: 04/24/2023] [Accepted: 11/03/2023] [Indexed: 03/19/2024] Open
Abstract
Atherosclerosis constitutes a proverbial pathogenic mechanism for cardio-cerebrovascular disease that accounts for the most common cause of disability and morbidity for human health worldwide. Endothelial dysfunction and inflammation are the key contributors to the progression of atherosclerosis. Glutaredoxin 2 (GLRX2) is abundantly existed in various tissues and possesses a range of pleiotropic efficacy including anti-oxidative and anti-inflammatory responses. However, its role in atherosclerosis is still undefined. Here, down-regulation of GLRX2 was validated in lipopolysaccha (LPS)-induced vascular endothelial cells (HUVECs). Moreover, elevation of GLRX2 reversed the inhibition of cell viability in LPS-treated HUVECs and decreased LPS-induced increases in cell apoptosis and caspase-3 activity. Additionally, enhancement of GLRX2 expression antagonized oxidative stress in HUVECs under LPS exposure by inhibiting ROS, lactate dehydrogenase and malondialdehyde production and increased activity of anti-oxidative stress superoxide dismutase. Notably, GLRX2 abrogated LPS-evoked transcripts and releases of pro-inflammatory cytokine (TNF-α, IL-6, and IL-1β), chemokine MCP-1 and adhesion molecule ICAM-1 expression. Furthermore, the activation of Nrf2/HO-1 signaling was demonstrated in LPS-stimulated HUVECs. Importantly, blockage of the Nrf2 pathway counteracted the protective roles of GLRX2 in LPS-triggered endothelial cell injury, oxidative stress and inflammatory response. Thus, these data reveal that GLRX2 may alleviate the progression of atherosclerosis by regulating vascular endothelial dysfunction and inflammation via the activation of the Nrf2 signaling, supporting a promising therapeutic approach for atherosclerosis and its complications. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-023-00606-x.
Collapse
Affiliation(s)
- Yuna Liu
- Department of Clinical Laboratory, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, No. 3 Yongding Road East Street, Beijing, 100039 People’s Republic of China
| | - Jinlin Gong
- Department of Medical Technology Support, Jingxi Medical District, Chinese PLA General Hospital, Beijing, 100097 People’s Republic of China
| | - Qing Wang
- Department of Clinical Laboratory, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, No. 3 Yongding Road East Street, Beijing, 100039 People’s Republic of China
| | - Na Wei
- Department of Clinical Laboratory, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, No. 3 Yongding Road East Street, Beijing, 100039 People’s Republic of China
| | - Lei Zhao
- Department of Clinical Laboratory, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, No. 3 Yongding Road East Street, Beijing, 100039 People’s Republic of China
| | - Zhenan Wu
- Department of Clinical Laboratory, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, No. 3 Yongding Road East Street, Beijing, 100039 People’s Republic of China
| |
Collapse
|
49
|
Zhang H, Lin L, Yang A, Liang Y, Huang B. Scutellarin alleviates tensile stress-induced proliferation and migration of venous smooth muscle cells via mediating the p38 MAPK pathway. Tissue Cell 2024; 87:102300. [PMID: 38211409 DOI: 10.1016/j.tice.2024.102300] [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: 07/04/2023] [Revised: 12/08/2023] [Accepted: 01/01/2024] [Indexed: 01/13/2024]
Abstract
OBJECTIVE Abnormal proliferation and migration of biomechanical force-induced venous smooth muscle cells (VSMCs) is a major cause to limit the efficacy of coronary artery bypass grafting (CABG) for coronary heart disease (CHD). Scutellarin is the main active ingredient of Erigeron Breviscapus, and has broad-spectrum pharmacological effects. Therefore, the present study was proposed to investigate the effect of Scutellarin on VSMCs under tensile stress. METHODS After interfering with VSMCs at different tensile stresses, the optimal tensile stress was screened. In a tensile stress environment, 100 μM Scutellarin and Hesperetin (p38 MAPK pathway activator) was used to treatment with VSMCs. CCK-8, EDU, Wound healing, flow cytometry and western blotting assays were used to detect cell proliferation, migration, apoptosis, and the expression of apoptosis-related proteins (Caspase3, Bcl2 and Bax). RESULTS Tensile stress with 10% significantly enhanced the activity, wound-healing ratio, and EDU+ cells of VSMCs, and decreased their apoptosis ratio. Moreover, it upregulated Bcl2 expression, and downregulated cleaved-Caspase3 and Bax expression of VSMCs. Hence, 10% tensile stress was selected to creates a tensile stress environment for VSMCs. Interestingly, 100 μM Scutellarin alleviated the effect of 10% tensile stress on the phenotype of VSMCs. Notably, 10% tensile stress increased the phosphorylation level of p38 MAPK (Thr180 +Tyr182) in VSMCs, which was restricted by Scutellarin. Further, Hesperetin restored the effect of Scutellarin on the phenotype of VSMCs. CONCLUSION Scutellarin alleviates tension stress-induced proliferation and migration of VSMCs via suppressing p38 MAPK pathway. Scutellarin may be used as an adjunctive strategy for future GABG treatment in CHD patients.
Collapse
Affiliation(s)
- Hu Zhang
- Departments of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Ling Lin
- Departments of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Ailing Yang
- Departments of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Yasha Liang
- Departments of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Bo Huang
- Operating Room, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China.
| |
Collapse
|
50
|
Hart DA. The Heterogeneity of Post-Menopausal Disease Risk: Could the Basis for Why Only Subsets of Females Are Affected Be Due to a Reversible Epigenetic Modification System Associated with Puberty, Menstrual Cycles, Pregnancy and Lactation, and, Ultimately, Menopause? Int J Mol Sci 2024; 25:3866. [PMID: 38612676 PMCID: PMC11011715 DOI: 10.3390/ijms25073866] [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: 02/24/2024] [Revised: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
For much of human evolution, the average lifespan was <40 years, due in part to disease, infant mortality, predators, food insecurity, and, for females, complications of childbirth. Thus, for much of evolution, many females did not reach the age of menopause (45-50 years of age) and it is mainly in the past several hundred years that the lifespan has been extended to >75 years, primarily due to public health advances, medical interventions, antibiotics, and nutrition. Therefore, the underlying biological mechanisms responsible for disease risk following menopause must have evolved during the complex processes leading to Homo sapiens to serve functions in the pre-menopausal state. Furthermore, as a primary function for the survival of the species is effective reproduction, it is likely that most of the advantages of having such post-menopausal risks relate to reproduction and the ability to address environmental stresses. This opinion/perspective will be discussed in the context of how such post-menopausal risks could enhance reproduction, with improved survival of offspring, and perhaps why such risks are preserved. Not all post-menopausal females exhibit risk for this set of diseases, and those who do develop such diseases do not have all of the conditions. The diseases of the post-menopausal state do not operate as a unified complex, but as independent variables, with the potential for some overlap. The how and why there would be such heterogeneity if the risk factors serve essential functions during the reproductive years is also discussed and the concept of sets of reversible epigenetic changes associated with puberty, pregnancy, and lactation is offered to explain the observations regarding the distribution of post-menopausal conditions and their potential roles in reproduction. While the involvement of an epigenetic system with a dynamic "modification-demodification-remodification" paradigm contributing to disease risk is a hypothesis at this point, validation of it could lead to a better understanding of post-menopausal disease risk in the context of reproduction with commonalities may also lead to future improved interventions to control such risk after menopause.
Collapse
Affiliation(s)
- David A Hart
- Department of Surgery, Faculty of Kinesiology, and McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|