1
|
Zhong YL, Xu CQ, Li J, Liang ZQ, Wang MM, Ma C, Jia CL, Cao YB, Chen J. Mitochondrial dynamics and metabolism in macrophages for cardiovascular disease: A review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156620. [PMID: 40068296 DOI: 10.1016/j.phymed.2025.156620] [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: 12/16/2024] [Revised: 02/18/2025] [Accepted: 03/05/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Mitochondria regulate macrophage function, affecting cardiovascular diseases like atherosclerosis and heart failure. Their dynamics interact with macrophage cell death mechanisms, including apoptosis and necroptosis. PURPOSE This review explores how mitochondrial dynamics and metabolism influence macrophage inflammation and cell death in CVDs, highlighting therapeutic targets for enhancing macrophage resilience and reducing CVD pathology, while examining molecular pathways and pharmacological agents involved. STUDY DESIGN This is a narrative review that integrates findings from various studies on mitochondrial dynamics and metabolism in macrophages, their interactions with the endoplasmic reticulum (ER) and Golgi apparatus, and their implications for CVDs. The review also considers the potential therapeutic effects of pharmacological agents on these pathways. METHODS The review utilizes a comprehensive literature search to identify relevant studies on mitochondrial dynamics and metabolism in macrophages, their role in CVDs, and the effects of pharmacological agents on these pathways. The selected studies are analyzed and synthesized to provide insights into the complex relationships between mitochondria, the ER, and Golgi apparatus, and their implications for macrophage function and fate. RESULTS The review reveals that mitochondrial metabolism intertwines with cellular architecture and function, particularly through its intricate interactions with the ER and Golgi apparatus. Mitochondrial-associated membranes (MAMs) facilitate Ca2+ transfer from the ER to mitochondria, maintaining mitochondrial homeostasis during ER stress. The Golgi apparatus transports proteins crucial for inflammatory signaling, contributing to immune responses. Inflammation-induced metabolic reprogramming in macrophages, characterized by a shift from oxidative phosphorylation to glycolysis, underscores the multifaceted role of mitochondrial metabolism in regulating immune cell polarization and inflammatory outcomes. Notably, mitochondrial dysfunction, marked by heightened reactive oxygen species generation, fuels inflammatory cascades and promotes cell death, exacerbating CVD pathology. However, pharmacological agents such as Metformin, Nitazoxanide, and Galanin emerge as potential therapeutic modulators of these pathways, offering avenues for mitigating CVD progression. CONCLUSION This review highlights mitochondrial dynamics and metabolism in macrophage inflammation and cell death in CVDs, suggesting therapeutic targets to improve macrophage resilience and reduce pathology, with new pharmacological agents offering treatment opportunities.
Collapse
Affiliation(s)
- Yi-Lang Zhong
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Chen-Qin Xu
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Ji Li
- Department of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Zhi-Qiang Liang
- Department of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Miao-Miao Wang
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Chao Ma
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Cheng-Lin Jia
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Yong-Bing Cao
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Jian Chen
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China; Anhui Province Rural Revitalization Collaborative Technical Service Center, Huangshan University, Huangshan 245041, China; Department of Public Health, International College, Krirk University, Bangkok, Thailand.
| |
Collapse
|
2
|
Yang Y, Chen Z, Song D, Wu J, Wang J, YouyouYan. Inhibition of ferroptosis alleviates atherosclerosis and foam cell formation by regulating lipid metabolism via AMPK activation. Int Immunopharmacol 2025; 153:114553. [PMID: 40147262 DOI: 10.1016/j.intimp.2025.114553] [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: 02/12/2025] [Revised: 03/14/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Atherosclerosis (AS) is a lipid disorder characterised by lipid accumulation in the aortic wall and foam cell formation. Recent studies have shown that excess iron accelerates AS progression and foam cell formation by inducing ferroptosis. GPx4, an anti-erroptotic protein, promotes SCARB1 expression, which inhibits macrophage foam cell formation by interacting with HDL. Thus, a complex association exists between ferroptosis and lipid metabolism. However, the underlying mechanisms remain unclear. AMPK signalling is a key regulator of metabolism and is involved in the regulation of ferroptosis. In this study, we used the ferroptosis inhibitor ferrostatin-1 (Fer-1) to assay the effect of ferroptosis inhibition on AS and foam cell formation and to investigate the underlying mechanism. Our results showed that Fer-1 alleviated AS lesions and foam cell formation both in vivo and in vitro. Additionally, Fer-1 reduced iron content and lipid accumulation in oxidized low-density lipoprotein (ox-LDL)-treated macrophages by upregulating the levels of FTH, GPx4, and SCARB1 via AMPK activation. The inhibition of AMPK reduces the effect of Fer-1 on iron and lipid accumulation in macrophages, which may contribute to a deeper understanding of the pathological process of AS and provide a therapeutic target for AS.
Collapse
Affiliation(s)
- Yunfan Yang
- Department of Cardiology, Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Zhenzhen Chen
- Department of Cardiology, Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Dandan Song
- Department of Clinical Laboratory, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun 130041, China
| | - Junduo Wu
- Department of Cardiology, Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Junnan Wang
- Department of Cardiology, Second Hospital of Jilin University, Changchun, Jilin 130041, China.
| | - YouyouYan
- Department of Cardiology, Second Hospital of Jilin University, Changchun, Jilin 130041, China.
| |
Collapse
|
3
|
Li F, Wang Y, Chen S, Liu J, Wu X, Maimati Y, Ding F, Wang X, Shen Y, Chen Q, Li Y, Shen W, Zhang R, Dai Y, Lu L. Nuclear receptor Dax1 promotes atherosclerosis by lipid transport inhibition and autophagy suppression in macrophages. Eur Heart J 2025:ehaf241. [PMID: 40259807 DOI: 10.1093/eurheartj/ehaf241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/27/2024] [Accepted: 03/25/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND AND AIMS Nuclear receptors (NRs) are involved in cardiovascular physiology and pathology. Dosage-sensitive sex reversal, adrenal hypoplasia congenita critical region on the X chromosome, gene 1 (Dax1) is a co-repressor of several protective NRs. However, whether Dax1 influences atherosclerosis remains unclear. This study aims to explore the role of Dax1 in atherogenesis and find a pharmacological approach targeting Dax1 to prevent atherosclerosis. METHODS Dax1 levels were examined in human atherosclerotic arteries. Atherosclerosis animal models were established in mice with macrophage-specific Dax1 knockdown following AAV8-PCSK9 administration and double knockout of macrophage Dax1 and ApoE to evaluate the role of Dax1. Transcriptomic and proteomic analyses were employed to decipher the underlying mechanisms. 2'-Deoxycytidine, an inhibitor of Dax1, was used to verify the effects of Dax1 in macrophages and in mice with atherosclerosis. RESULTS Dax1 mRNA level was up-regulated among NRs in atherosclerotic arteries compared to non-atherosclerotic arteries. The elevation of Dax1 was prominent in the macrophages of atherosclerotic arteries. Macrophage-specific Dax1 knockout mice had less atherosclerosis than controls. Mechanistically, Dax1 inhibited liver X receptor alpha (LXRα), and interacted directly with transcription factor EB (TFEB) to suppress autophagy, resulting in lipid accumulation and inflammation in macrophages. Additionally, 2'-deoxycytidine concentration dependently decreased Dax1 levels, enhanced autophagy, reduced lipid accumulation, and inhibited atherosclerosis in mice. CONCLUSIONS This study demonstrates that Dax1 levels are increased in atherosclerotic plaques. Dax1 promotes atherosclerosis by interacting with TFEB to suppress autophagy and inhibiting LXRα for lipid transport in macrophage, indicating that Dax1 is a potential target for atherosclerosis.
Collapse
Affiliation(s)
- Feifei Li
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Yixuan Wang
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Shuai Chen
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Jingmeng Liu
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Xinrui Wu
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Yipaerguli Maimati
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Fenghua Ding
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Xiaoqun Wang
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Ying Shen
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Qiujing Chen
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Yunqi Li
- National Research Center for Translational Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Weifeng Shen
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Yang Dai
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Lin Lu
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- National Research Center for Translational Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| |
Collapse
|
4
|
Zhang C, Wang Y, Huang F, Zhang Y, Huang M, Liu H, Liu Y, Wang Q, Liu C, Angwa L, Gao Y, Sun D, Jiang Y. Novel mechanism of fluoride induced cardiovascular system injury by regulating p53/miR200c-3p during endothelial dysfunction. ENVIRONMENTAL RESEARCH 2025; 271:121102. [PMID: 39952459 DOI: 10.1016/j.envres.2025.121102] [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: 12/03/2024] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND The impairment of the cardiovascular system by fluoride has attracted public health concern, and its toxic effects on ECs have garnered extensive research attention. However, epidemiological clues of fluoride induced cardiovascular injury are limited. The function of ECs is crucial for the early diagnosis of CVD, yet mechanisms through which fluoride disrupts endothelial function are still unclear. PURPOSE To investigate the relationship between fluoride exposure and hypertension in population by epidemiological investigation. To explore the potential mechanism of functional injury of ECs induced by fluoride. RESULT Epidemiological studies have shown that the risk of hypertension in study population increased with the increased of urinary fluoride concentration [OR = 1.565, 95%CI (1.143, 2.142)]. In rat model with fluorosis alongside a model of fluoride induced ECs injury, NaF led to anti-adhesion of ECs and barrier dysfunction. Notably, the expression levels of eNOS and NO were found to be decreased, while the expression levels of ACE, vWF, ICAM-1, VCAM-1 and ET-1 were elevated. Our findings also indicated that NaF induced oxidative stress in ECs, evidenced by significant increased in ROS and MDA levels and decreased protein expression of GPx4 and SOD activity. It was further found that NaF activated the p53/miR-200c-3p signaling axis via ROS, leading to endothelial dysfunction. CONCLUSION This study found that fluoride exposure was a risk factor for hypertension. In addition, fluoride could cause ECs dysfunction by inducing oxidative stress and activating p53/miR-200c-3p. These findings were helpful to further understand the mechanism of fluoride induced cardiovascular system injury and provide a theoretical basis for fluoride induced cardiovascular system injury.
Collapse
Affiliation(s)
- Chao Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China
| | - Yue Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China; Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, People's Republic of China
| | - Fengya Huang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China; Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, People's Republic of China
| | - Yaoyuan Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China; Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, People's Republic of China
| | - Mingyue Huang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China; Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, People's Republic of China
| | - Hui Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China
| | - Yunzhu Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China
| | - Qiaoyu Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China
| | - Chang Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China
| | - Linet Angwa
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; United State University-Africa, Department of Phamaceutics, Pharmacy Practice, and Public Health, People's Republic of China
| | - Yanhui Gao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China; Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, People's Republic of China.
| | - Dianjun Sun
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China; Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, People's Republic of China.
| | - Yuting Jiang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China; Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, People's Republic of China.
| |
Collapse
|
5
|
Li Y, Yang W, Zhang X, Ba J, Yang H, Wang W, Zhang K, Yang Z, Liang H, Li Z, Ashokkumar M, Zhang J, Gao Z, Yu Y. High-frequency ultrasound induced the preparation of oxidized low density lipoprotein. ULTRASONICS SONOCHEMISTRY 2025; 115:107303. [PMID: 40056869 PMCID: PMC11930747 DOI: 10.1016/j.ultsonch.2025.107303] [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: 12/27/2024] [Revised: 02/16/2025] [Accepted: 03/04/2025] [Indexed: 03/10/2025]
Abstract
Foam cells have been frequently used in studies related to atherosclerosis. Traditional methods for inducing oxidized low-density lipoprotein (oxLDL) involve copper ion (Cu2+) treatment, which has inherent limitations such as prolonged oxidation times and residual copper ions. This study explored high-frequency ultrasound (400 kHz) as an alternative method for LDL oxidization. The findings demonstrated that high-frequency ultrasound-oxidized LDL (U-oxLDL) exhibited no significant differences compared to copper-oxidized LDL (Cu-oxLDL) in terms of electrophoretic mobility, foam cell morphology, lipid content, and cholesterol transport proteins. Additionally, lipidomic analysis revealed that U-oxLDL was more comparable to native LDL (N-LDL). Transcriptomic profiling of bone marrow-derived macrophages (BMDMs) treated with oxLDL showed that the gene expression patterns of BMDM foam cells treated with U-oxLDL were over 90 % consistent with those treated with Cu-oxLDL. Therefore, high-frequency ultrasound oxidation method represents a green and efficient strategy for oxLDL preparation, offering potential advantages for advancing atherosclerosis research.
Collapse
Affiliation(s)
- Yuanmin Li
- Shandong Provincial Hospital Affiliated to Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, China; School of Laboratory Animal& Shandong Laboratory Animal Center, Shandong First Medical University &Shandong Academy of Medical Sciences, Jinan, Shandong 250118, China; Department of Cardiology. The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, Shandong 271000, China
| | - Wanyue Yang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, China; School of Laboratory Animal& Shandong Laboratory Animal Center, Shandong First Medical University &Shandong Academy of Medical Sciences, Jinan, Shandong 250118, China; School of Clinical & Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong 250118, China
| | - Xinyi Zhang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, China; School of Laboratory Animal& Shandong Laboratory Animal Center, Shandong First Medical University &Shandong Academy of Medical Sciences, Jinan, Shandong 250118, China; School of Clinical & Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong 250118, China
| | - Jingjing Ba
- Shandong Provincial Hospital Affiliated to Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, China; School of Laboratory Animal& Shandong Laboratory Animal Center, Shandong First Medical University &Shandong Academy of Medical Sciences, Jinan, Shandong 250118, China; Department of Cardiology. The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, Shandong 271000, China
| | - Han Yang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Wen Wang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, China; School of Laboratory Animal& Shandong Laboratory Animal Center, Shandong First Medical University &Shandong Academy of Medical Sciences, Jinan, Shandong 250118, China
| | - Ke Zhang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, China; School of Laboratory Animal& Shandong Laboratory Animal Center, Shandong First Medical University &Shandong Academy of Medical Sciences, Jinan, Shandong 250118, China
| | - Ze Yang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, China; School of Laboratory Animal& Shandong Laboratory Animal Center, Shandong First Medical University &Shandong Academy of Medical Sciences, Jinan, Shandong 250118, China; Department of Cardiology. The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, Shandong 271000, China; School of Clinical & Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong 250118, China
| | - Hui Liang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, China; School of Laboratory Animal& Shandong Laboratory Animal Center, Shandong First Medical University &Shandong Academy of Medical Sciences, Jinan, Shandong 250118, China; School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271016, China
| | - Zihan Li
- Shandong Provincial Hospital Affiliated to Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, China; School of Laboratory Animal& Shandong Laboratory Animal Center, Shandong First Medical University &Shandong Academy of Medical Sciences, Jinan, Shandong 250118, China; School of Clinical & Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong 250118, China
| | - Muthupandian Ashokkumar
- School of Chemistry and Department of Chemical Engineering, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Jiguo Zhang
- School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271016, China.
| | - Zhiliang Gao
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China; Shandong Key Laboratory: Magnetic Field-free Medicine & Functional Imaging, Research Institute of Shandong University, Magnetic Field-free Medicine & Functional Imaging, National Medicine-Engineering Interdisciplinary Industry-Education Integration Innovation Platform, Jinan, Shandong 250012, China.
| | - Yang Yu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, China; School of Laboratory Animal& Shandong Laboratory Animal Center, Shandong First Medical University &Shandong Academy of Medical Sciences, Jinan, Shandong 250118, China; School of Clinical & Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong 250118, China.
| |
Collapse
|
6
|
Liao W, Li Y, Zhao H, Lu S. The Lian-Dou-Qing-Mai Formula activates the PPARγ-LXRα-ABCA1/ABCG1 pathway by regulating IL-10, leading to the promotion of cholesterol efflux and a reduction in atherosclerotic plaques. Histol Histopathol 2025; 40:585-596. [PMID: 39315497 DOI: 10.14670/hh-18-803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
BACKGROUND To observe the effect of the Lian-Dou-Qing-Mai (LDQM) formula on lipid metabolism in mice and explore its mechanism from the perspective of regulating the PPARγ/LXRα/ABCA1 signaling pathway. METHODS THP-1 cells were induced to transform into foam cells with ox-LDL. Atherosclerosis (AS) models were constructed using a high-fat diet in ApoE-/- mice. Detection kits were used to evaluate triglyceride (TG) and total cholesterol (TC) content; TNF-α, MCP-1, MMP-9, TMP-1, PPARγ, LXRα, ABCA1, and ABCG1 mRNA and protein expression were identified using real-time PCR and western blot. Aortic plaque development and lipid deposition were seen using hematoxylin and eosin (HE) and oil red O staining, respectively. RESULTS In the cell model, LDQM could inhibit the formation of THP-1 macrophage-derived foam cells and the expression of inflammatory factors, promote macrophage cholesterol efflux, increase the expression of IL-10, and activate the PPARγ-LXRα-ABCA1/ABCG1 pathway. Additional IL-10 treatment further promotes LDQM-induced cholesterol efflux in THP-1 cells; In vivo models, LDQM inhibited the area of atherosclerotic lesions, aortic lipid deposition, and inflammation levels in ApoE-/- mice through IL-10, and activated the expression level of the PPARγ-LXRα-ABCA1/ABCG1 pathway. CONCLUSION LDQM may affect the PPARγ/LXRα/ABCA1 signaling pathway through IL-10, regulate lipid metabolism, reduce serum inflammatory expression and lipid deposition, and improve the formation of atheroplaques.
Collapse
Affiliation(s)
- Wenqi Liao
- Department of Emergency, Xuzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu Province, China
| | - You Li
- Department of Oncology, Xuzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu Province, China
| | - Haoyan Zhao
- Department of Urology, Xuzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu Province, China
| | - Shu Lu
- Department of Cardiology, WuXi Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu Province, China.
| |
Collapse
|
7
|
Xiong M, Wang M, Liu X, Luo S, Wang X, Yang L, Li K, Li Y, Wei W, Chen H, Wang Y. Quercetin inhibits oligodendrocytes ferroptosis by blocking NCOA4-mediated ferritinophagy. Int Immunopharmacol 2025; 150:114152. [PMID: 39946767 DOI: 10.1016/j.intimp.2025.114152] [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: 07/03/2024] [Revised: 11/21/2024] [Accepted: 01/21/2025] [Indexed: 03/03/2025]
Abstract
Ferritinophagy is a specific type of autophagy that maintains intracellular iron metabolic homeostasis by targeting ferritin, one of the major forms of iron storage in the human body. Previous research has demonstrated that quercetin prevents the ferroptosis of oligodendrocyte progenitor cells (OPCs) by inhibiting the Id2/transferrin pathway. Given the ability of quercetin to suppress autophagy in spinal cord injury (SCI), this study aimed to investigate whether quercetin prevents ferroptosis in an autophagy-dependent manner. In erastin-treated OPCs, quercetin significantly upregulated the protein level of ferritin heavy chain (FTH) and markedly reduced its colocalization with LysoTracker, an indicator of lysosome aggregation. Quercetin significantly reduced the ferrous iron levels, the LC3II/LC3I ratio, and the number of LC3 puncta in OPCs, whereas it increased the level of sequestosome 1 (P62) in erastin-treated OPCs. Pretreatment of OPCs with autophagy inhibitor bafilomycin A1 inhibited quercetin-mediated ferritinophagy and ferroptosis, whereas pretreatment with autophagy activator rapamycin reversed the effect of quercetin on ferritinophagy and ferroptosis of OPCs, as evidenced by reduced protein levels of ferritin heavy chain and p62, as well as increased protein levels of LC3II/LC3I and prostaglandin-endoperoxide synthase 2 (PTGS2). Compared with the erastin and quercetin treated OPCs, increased rerrous iron, lipid peroxidation production, and decreased GSH content, as well as shrunken mitochondria, were observed in OPCs treated with a combination of erastin, quercetin, and rapamycin. In vivo, quercetin significantly downregulated the nuclear receptor coactivator 4 (NCOA4) and PTGS2 protein expression, as well as the LC3II/LC3I ratio. Besides that, quercetin reduced the MDA level and the colocalization of FTH with NCOA4 in spinal cord tissues. Mechanistically, NCOA4 reversed the effect of quercetin on ferritinophagy and ferroptosis of OPCs, whereas mutation of Y71 to alanine only slightly reversed the above effect. In conclusion, our findings revealed that quercetin inhibits OPCs ferroptosis by blocking NCOA4-mediated ferritinophagy. Quercetin and ferritinophagy may be potential therapeutic agents for SCI.
Collapse
Affiliation(s)
- Man Xiong
- Guangzhou University of Chinese Medicine, Guangzhou 510000 China
| | - Mingsen Wang
- Department of Orthopedic, Chaoshan Guoyu Hospital of Jieyang, Jieyang 515300 China
| | - Xinfang Liu
- Orthopedic Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510000 China
| | - Sidong Luo
- Orthopedic Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510000 China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510000 China
| | - Xu Wang
- School of Biomedicine and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006 China
| | - Lei Yang
- Orthopedic Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510000 China
| | - Kaifan Li
- Orthopedic Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510000 China
| | - Yongsheng Li
- Guangdong Cord Blood Bank, Guangzhou 510663 China; Guangzhou Municipality Tianhe Nuoya Bio-engineering Co., Ltd., Guangzhou 510663 China
| | - Wei Wei
- Guangdong Cord Blood Bank, Guangzhou 510663 China; Guangzhou Municipality Tianhe Nuoya Bio-engineering Co., Ltd., Guangzhou 510663 China
| | - Hongdong Chen
- Department of No.1 General Surgery, Guangdong Second Provincial General Hospital, Guangzhou 510000 China
| | - Yeyang Wang
- Orthopedic Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510000 China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510000 China; Orthopaedic Center, Guangdong Second Provincial General Hospital, Guangzhou 510000 China; Zhaoqing Central People's Hospital, Zhaoqing, Guangdong 526000, China.
| |
Collapse
|
8
|
Khoso MA, Liu H, Zhao T, Zhao W, Huang Q, Sun Z, Dinislam K, Chen C, Kong L, Zhang Y, Liu X. Impact of plant-derived antioxidants on heart aging: a mechanistic outlook. Front Pharmacol 2025; 16:1524584. [PMID: 40191425 PMCID: PMC11969199 DOI: 10.3389/fphar.2025.1524584] [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: 11/07/2024] [Accepted: 03/07/2025] [Indexed: 04/09/2025] Open
Abstract
Heart aging involves a complex interplay of genetic and environmental influences, leading to a gradual deterioration of cardiovascular integrity and function. Age-related physiological changes, including ventricular hypertrophy, diastolic dysfunction, myocardial fibrosis, increased arterial stiffness, and endothelial dysfunction, are influenced by key mechanisms like autophagy, inflammation, and oxidative stress. This review aims to explore the therapeutic potential of plant-derived bioactive antioxidants in mitigating heart aging. These compounds, often rich in polyphenols, flavonoids, and other phytochemicals, exhibit notable antioxidant, anti-inflammatory, and cardioprotective properties. These substances have intricate cardioprotective properties, including the ability to scavenge ROS, enhance endogenous antioxidant defenses, regulate signaling pathways, and impede fibrosis and inflammation-promoting processes. By focusing on key molecular mechanisms linked to cardiac aging, antioxidants produced from plants provide significant promise to reduce age-related cardiovascular decline and improve general heart health. Through a comprehensive analysis of preclinical and clinical studies, this work highlights the mechanisms associated with heart aging and the promising effects of plant-derived antioxidants. The findings may helpful for researchers in identifying specific molecules with therapeutic and preventive potential for aging heart.
Collapse
Affiliation(s)
- Muneer Ahmed Khoso
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Heng Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Tong Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Wenjie Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Qiang Huang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Zeqi Sun
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Khuzin Dinislam
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Chen Chen
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Lingyi Kong
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Yong Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Xin Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| |
Collapse
|
9
|
Pei X, Cui F, Chen Y, Yang Z, Xie Z, Wen Y. miR-214-3p Promotes ox-LDL-Induced Macrophages Ferroptosis and Inflammation via GPX4. J Inflamm Res 2025; 18:3937-3950. [PMID: 40125091 PMCID: PMC11927573 DOI: 10.2147/jir.s507076] [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: 12/04/2024] [Accepted: 03/04/2025] [Indexed: 03/25/2025] Open
Abstract
Purpose Atherosclerosis (AS) is a chronic inflammatory disease caused by the dysregulation of lipid metabolism. It has been established that oxidized low-density lipoprotein (ox-LDL)-induced macrophage inflammation and ferroptosis play important roles in AS. However, the mechanism by which ox-LDL induces inflammation in macrophages requires further investigation. Materials and Methods A foam cell model derived from ox-LDL-induced macrophages was constructed to study its mechanism of action. The levels of interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α were evaluated using an Enzyme-Linked Immunosorbent Assay (ELISA). Oil Red O staining was used to detect intracellular lipid accumulation levels. Lactate dehydrogenase (LDH), malondialdehyde (MDA), reactive oxygen species (ROS), and Fe2+ levels were assessed. Dual-luciferase and RNA-binding protein immunoprecipitation (RIP) experiments validated the binding relationship between microRNA (miR)-214-3p and glutathione peroxidase 4 (GPX4). Results The levels of IL-6, IL-1β, and TNF-α were significantly increased in ox-LDL-induced macrophages, accompanied by increased lipid accumulation, indicating the promotion of foam cell formation. Additionally, ox-LDL increased LDH, MDA, ROS, and Fe2+. The expression of miR-214-3p positively correlated with ox-LDL concentration in macrophages. Treatment with an miR-214-3p inhibitor reduces lipid accumulation, inflammatory responses, and ferroptosis in macrophages. Dual-luciferase and RIP experiments confirmed that miR-214-3p regulates GPX4 transcription. Silenced GPX4 reversed the inflammatory effects of the miR-214-3p inhibitor on ox-LDL-induced macrophages. Low GPX4 expression also increased lipid accumulation and ferroptosis in macrophages. Conclusion miR-214-3p promotes macrophage ferroptosis and inflammation induced by ox-LDL. This mechanism may be associated with miR-214-3p-induced GPX4 expression, which underscores the therapeutic significance of targeting macrophage inflammation and ferroptosis in addressing AS.
Collapse
Affiliation(s)
- Xueliang Pei
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, People’s Republic of China
| | - Facai Cui
- Clinical Laboratory, Henan Provincial People’s Hospital, Zhengzhou, Henan, People’s Republic of China
| | - Yu Chen
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Zhiyuan Yang
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, People’s Republic of China
| | - Zhouliang Xie
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, People’s Republic of China
| | - Yongjin Wen
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, People’s Republic of China
| |
Collapse
|
10
|
Li L, Lai L, Qiu D, Ding Y, Yu M, Zhang T, Wang Z, Wang S. P2Y 6 receptor: A promising therapeutic target for atherosclerosis. Eur J Pharmacol 2025; 998:177513. [PMID: 40097133 DOI: 10.1016/j.ejphar.2025.177513] [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: 11/17/2024] [Revised: 02/18/2025] [Accepted: 03/14/2025] [Indexed: 03/19/2025]
Abstract
Atherosclerosis is induced by lipid accumulation, inflammation, and endothelial dysfunction, and is the leading cause of death from cardiovascular disease worldwide. The P2Y6 receptor can be activated by the extracellular release of UDP. The evidence from the last decade has highlighted its critical therapeutic effect in atherosclerosis, yet with unclear mechanisms. This review introduced the P2Y6 receptor in atherosclerosis, and its mechanisms of atherosclerosis-promoting in macrophages, endothelial cells, and vascular smooth muscle cells. Finally, we discussed the development and potential of P2Y6 receptor antagonists in treating atherosclerosis.
Collapse
Affiliation(s)
- Lixia Li
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Liting Lai
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Dan Qiu
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yang Ding
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Meiling Yu
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Tingyu Zhang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zongbao Wang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Shuzhi Wang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
11
|
Palumbo M, Ugolotti M, Zimetti F, Adorni MP. Anti-atherosclerotic effects of natural compounds targeting lipid metabolism and inflammation: Focus on PPARs, LXRs, and PCSK9. ATHEROSCLEROSIS PLUS 2025; 59:39-53. [PMID: 39877131 PMCID: PMC11773090 DOI: 10.1016/j.athplu.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025]
Abstract
A large body of evidence has shown that modulation of the nuclear receptors peroxisome proliferator-activated receptors (PPARs), the liver X receptors (LXRs), the proprotein convertase subtilisin/kexin type 9 (PCSK9) and inflammatory processes by natural compounds has hypolipidemic and anti-atherosclerotic effects. These beneficial outcomes are certainly related to the crucial function of these targets in maintaining cholesterol homeostasis and regulating systemic inflammation. Currently, the therapeutic scenario for cardiovascular diseases (CVD) offers a plethora of widely validated and functional pharmacological treatments to improve the health status of patients. However, patients are increasingly sceptical of pharmacological treatments which are often associated with moderate to severe side effects. The aim of our review is to provide a collection of the most recent scientific evidence on the most common phytochemicals, used for centuries in the Mediterranean diet and traditional chinese medicine that act on these key regulators of cholesterol homeostasis and systemic inflammation, which could constitute important tools for CVD management.
Collapse
Affiliation(s)
| | | | | | - Maria Pia Adorni
- Department of Medicine and Surgery, Unit of Neuroscience, University of Parma, Italy
| |
Collapse
|
12
|
Zheng Y, Kou J, Gao X, Guo J, Liu Q, Ren H, Gao T, Wang Q, Zhao Y, Wang Y, Li H, Yang L. Berberine Inhibited SASP-Related Inflammation through RXRα/PPARγ/NEDD4 Pathway in Atherosclerosis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2025; 53:251-283. [PMID: 39829230 DOI: 10.1142/s0192415x25500107] [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: 01/22/2025]
Abstract
The accumulation of aging cells significantly contributes to chronic inflammatory diseases such as atherosclerosis. Human carotid artery single-cell sequencing has shown that large numbers of aging foam cells are present in the plaques of human patients. Berberine (BBR) has been shown to inhibit cell senescence, however, the mechanisms involved in its treatment of atherosclerotic senescence have not yet been determined. Changes in plaque morphology and blood chemistry were observed in ApoE[Formula: see text] mice fed with a high-fat diet before and after BBR treatment. Inflammatory proteins linked to the senescence-associated secretory phenotypes (SASP) were detected in RAW264.7 and peritoneal macrophage-derived foam cells. Smart-seq analysis was used to explore the pathways associated with BBR therapy for atherosclerosis. Finally, the effect of lentivirus-mediated knockdown of RXRα in macrophages in plaques on atherosclerosis treatment with BBR was determined. We found that BBR reduced inflammation linked to SASP in atherosclerosis through the RXRα/PPARγ/NEDD4 signaling pathway. BBR increased GATA4 binding to p62, promoted ubiquitination, and inhibited SASP-associated protein production in RAW264.7 and peritoneal macrophage-derived foam cells. Mechanistically, according to the Smart-seq results, BBR activated RXRα and PPARγ, synergistically increased NEDD4 transcription levels, and promoted ubiquitination-mediated degradation of the GATA4/p62 complex. Additionally, the anti-aging impact of BBR on atherosclerosis was negated when macrophage-specific RXRα was knocked down using lentivirus (pLVCD68-shRNA RXRα) in ApoE[Formula: see text] mice. BBR activated PPARγ through RXRα-PPARγ immune complex in macrophage-derived foam cells, increased NEDD4 transcriptional activity, promoted ubiquitination of GATA4-p62 complex, and inhibited SASP-related inflammation. These findings suggest the potential of BBR as a novel approach to addressing SASP-associated inflammation in atherosclerosis.
Collapse
Affiliation(s)
- Yinghong Zheng
- Department of Pharmacology, Tianjin Medical University 22 Qixiangtai Road, Heping District, Tianjin 300070, P. R. China
- Department of Pathophysiology, Harbin Medical University 157 Baojian Road, Nangang District, Harbin 150081, P. R. China
| | - Jiayuan Kou
- Department of Biochemistry and Molecular Biology, Harbin Medical University 157 Baojian Road, Nangang District, Harbin 150081, P. R. China
| | - Xi Gao
- Department of Pathophysiology, Harbin Medical University 157 Baojian Road, Nangang District, Harbin 150081, P. R. China
| | - Jinxiang Guo
- Department of Pathophysiology, Harbin Medical University 157 Baojian Road, Nangang District, Harbin 150081, P. R. China
| | - Qian Liu
- Department of Pharmacology, Tianjin Medical University 22 Qixiangtai Road, Heping District, Tianjin 300070, P. R. China
| | - Huiwen Ren
- Department of Pharmacology, Tianjin Medical University 22 Qixiangtai Road, Heping District, Tianjin 300070, P. R. China
| | - Tielei Gao
- Department of Forensic Medicine, Harbin Medical University 157 Baojian Road, Nangang District, Harbin 150081, P. R. China
| | - Qianbing Wang
- Department of Pathophysiology, Harbin Medical University 157 Baojian Road, Nangang District, Harbin 150081, P. R. China
| | - Yajie Zhao
- Department of Pathophysiology, Harbin Medical University 157 Baojian Road, Nangang District, Harbin 150081, P. R. China
| | - Yuqin Wang
- Department of Pathophysiology, Harbin Medical University 157 Baojian Road, Nangang District, Harbin 150081, P. R. China
| | - Hong Li
- Department of Pathophysiology, Harbin Medical University 157 Baojian Road, Nangang District, Harbin 150081, P. R. China
| | - Liming Yang
- Department of Pathophysiology, Harbin Medical University 157 Baojian Road, Nangang District, Harbin 150081, P. R. China
- Department of Cardiology The Second Affiliated Hospital of Harbin Medical University Harbin, P. R. China
| |
Collapse
|
13
|
Alharbi HOA, Alshebremi M, Babiker AY, Rahmani AH. The Role of Quercetin, a Flavonoid in the Management of Pathogenesis Through Regulation of Oxidative Stress, Inflammation, and Biological Activities. Biomolecules 2025; 15:151. [PMID: 39858545 PMCID: PMC11763763 DOI: 10.3390/biom15010151] [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: 12/12/2024] [Revised: 01/05/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Quercetin, a flavonoid found in vegetables and fruits, has been extensively studied for its health benefits and disease management. Its role in the prevention of various pathogenesis has been well-documented, primarily through its ability to inhibit oxidative stress, inflammation, and enhance the endogenous antioxidant defense mechanisms. Electronic databases such as Google Scholar, Scopus, PubMed, Medline, and Web of Science were searched for information regarding quercetin and its role in various pathogeneses. The included literature comprised experimental studies, randomized controlled trials, and epidemiological studies related to quercetin, while editorials, case analyses, theses, and letters were excluded. It has been reported to have a wide range of health benefits including hepatoprotective, antidiabetic, anti-obesity, neuroprotective, cardioprotective, wound healing, antimicrobial, and immunomodulatory effects, achieved through the modulation of various biological activities. Additionally, numerous in vitro and in vivo studies have shown that quercetin's efficacies in cancer management involve inhibiting cell signaling pathways, such as inflammation, cell cycle, and angiogenesis, activating cell signaling pathways including tumor suppressor genes, and inducing apoptosis. This review aims to provide a comprehensive understanding of the health benefits of quercetin in various pathogeneses. Additionally, this review outlines the sources of quercetin, nanoformulations, and its applications in health management, along with key findings from important clinical trial studies. Limited clinical data regarding quercetin's safety and mechanism of action are available. It is important to conduct more clinical trials to gain a deeper understanding of the disease-preventive potential, mechanisms of action, safety, and optimal therapeutic dosages. Furthermore, more research based on nanoformulations should be performed to minimize/overcome the hindrance associated with bioavailability, rapid degradation, and toxicity.
Collapse
Affiliation(s)
| | | | | | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| |
Collapse
|
14
|
Huang M, Xie X, Yuan R, Xin Q, Ma S, Guo H, Miao Y, Hu C, Zhu Y, Cong W. The multifaceted anti-atherosclerotic properties of herbal flavonoids: A comprehensive review. Pharmacol Res 2025; 211:107551. [PMID: 39701504 DOI: 10.1016/j.phrs.2024.107551] [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: 09/25/2024] [Revised: 12/12/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
Atherosclerosis (AS) is a major etiological factor underpinning a spectrum of cardiovascular diseases, leading to cerebral infarction, coronary artery disease, and peripheral vascular disease. The chronic progression of AS, spanning from initial plaque formation to the occurrence of acute cardiovascular events, underscores the complexity of AS and the challenges it presents in terms of treatment. Currently, the clinical management of AS relies predominantly on statins and proprotein convertase subtilisin/kexin type 9 inhibitors, which primarily aim to reduce low-density lipoprotein levels and have demonstrated some therapeutic efficacy. Nevertheless, due to their potential side effects, there is a pressing need to actively investigate alternative treatment approaches. Researches on natural compounds derived from herbal medicines, such as flavonoids, hold significant promise in combating AS by regulating lipid metabolism, reducing oxidative stress and inflammation, inhibiting the proliferation of vascular smooth muscle cells, modulating autophagy and additional pathways. Various targets participate in these physiological processes, encompassing acyl-CoA: cholesterol acyltransferase (ACAT), ATP citrate lyase (ACLY), nuclear factor erythroid 2-related factor 2 (Nrf2), krüppel-like factor 2 (KLF2), NOD-like receptor protein 3 (NLRP3), transcription factor EB (TFEB) and so on. This comprehensive review endeavors to synthesize and analyse the most recent findings on herbal flavonoids, shedding light on their anti-atherosclerotic potential and the underlying protective mechanisms and related-targets, which might pave the way for the development of novel drug candidates or the optimization of flavonoid-based therapies.
Collapse
Affiliation(s)
- Meiwen Huang
- School of Pharmacy, Macau University of Science and Technology, Macau 999078, China; Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Xuena Xie
- School of Pharmacy, Macau University of Science and Technology, Macau 999078, China; Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Rong Yuan
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Qiqi Xin
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Shudong Ma
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Hongai Guo
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yu Miao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Chunyu Hu
- Department of Teaching Quality Construction, Graduate School, China Academy of Chinese Medical Sciences, 100700, China
| | - Yizhun Zhu
- School of Pharmacy, Macau University of Science and Technology, Macau 999078, China
| | - Weihong Cong
- School of Pharmacy, Macau University of Science and Technology, Macau 999078, China; Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China.
| |
Collapse
|
15
|
Miyajima C, Nagasaka M, Aoki H, Toriuchi K, Yamanaka S, Hashiguchi S, Morishita D, Aoyama M, Hayashi H, Inoue Y. The Hippo Signaling Pathway Manipulates Cellular Senescence. Cells 2024; 14:13. [PMID: 39791714 PMCID: PMC11719916 DOI: 10.3390/cells14010013] [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: 10/28/2024] [Revised: 12/21/2024] [Accepted: 12/24/2024] [Indexed: 01/12/2025] Open
Abstract
The Hippo pathway, a kinase cascade, coordinates with many intracellular signals and mediates the regulation of the activities of various downstream transcription factors and their coactivators to maintain homeostasis. Therefore, the aberrant activation of the Hippo pathway and its associated molecules imposes significant stress on tissues and cells, leading to cancer, immune disorders, and a number of diseases. Cellular senescence, the mechanism by which cells counteract stress, prevents cells from unnecessary damage and leads to sustained cell cycle arrest. It acts as a powerful defense mechanism against normal organ development and aging-related diseases. On the other hand, the accumulation of senescent cells without their proper removal contributes to the development or worsening of cancer and age-related diseases. A correlation was recently reported between the Hippo pathway and cellular senescence, which preserves tissue homeostasis. This review is the first to describe the close relationship between aging and the Hippo pathway, and provides insights into the mechanisms of aging and the development of age-related diseases. In addition, it describes advanced findings that may lead to the development of tissue regeneration therapies and drugs targeting rejuvenation.
Collapse
Affiliation(s)
- Chiharu Miyajima
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; (M.N.); (S.Y.); (S.H.); (D.M.); (H.H.)
| | - Mai Nagasaka
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; (M.N.); (S.Y.); (S.H.); (D.M.); (H.H.)
- Department of Experimental Chemotherapy, Cancer Chemotherapy Center of JFCR, Tokyo 135-8550, Japan
| | - Hiromasa Aoki
- Department of Pathobiology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; (H.A.); (K.T.); (M.A.)
| | - Kohki Toriuchi
- Department of Pathobiology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; (H.A.); (K.T.); (M.A.)
| | - Shogo Yamanaka
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; (M.N.); (S.Y.); (S.H.); (D.M.); (H.H.)
| | - Sakura Hashiguchi
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; (M.N.); (S.Y.); (S.H.); (D.M.); (H.H.)
| | - Daisuke Morishita
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; (M.N.); (S.Y.); (S.H.); (D.M.); (H.H.)
| | - Mineyoshi Aoyama
- Department of Pathobiology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; (H.A.); (K.T.); (M.A.)
| | - Hidetoshi Hayashi
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; (M.N.); (S.Y.); (S.H.); (D.M.); (H.H.)
| | - Yasumichi Inoue
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; (M.N.); (S.Y.); (S.H.); (D.M.); (H.H.)
| |
Collapse
|
16
|
Shao R, Chen R, Zheng Q, Yao M, Li K, Cao Y, Jiang L. Oxidative stress disrupts vascular microenvironmental homeostasis affecting the development of atherosclerosis. Cell Biol Int 2024; 48:1781-1801. [PMID: 39370593 DOI: 10.1002/cbin.12239] [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: 02/11/2024] [Revised: 05/20/2024] [Accepted: 06/10/2024] [Indexed: 10/08/2024]
Abstract
Atherosclerosis is primarily an inflammatory reaction of the cardiovascular system caused by endothelial damage, leading to progressive thickening and hardening of the vessel walls, as well as extensive necrosis and fibrosis of the surrounding tissues, the most necessary pathological process causing cardiovascular disease. When the body responds to harmful internal and external stimuli, excess oxygen free radicals are produced causing oxidative stress to occur in cells and tissues. Simultaneously, the activation of inflammatory immunological processes is followed by an elevation in oxygen free radicals, which directly initiates the release of cytokines and chemokines, resulting in a detrimental cycle of vascular homeostasis abnormalities. Oxidative stress contributes to the harm inflicted upon vascular endothelial cells and the decrease in nitric oxide levels. Nitric oxide is crucial for maintaining vascular homeostasis and is implicated in the development of atherosclerosis. This study examines the influence of oxidative stress on the formation of atherosclerosis, which is facilitated by the vascular milieu. It also provides an overview of the pertinent targets and pharmaceutical approaches for treating this condition.
Collapse
Affiliation(s)
- Ruifei Shao
- Medical School, Center for Translational Research in Clinical Medicine, Kunming University of Science and Technology, Kunming, China
| | - Rui Chen
- Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Qiang Zheng
- Medical School, Center for Translational Research in Clinical Medicine, Kunming University of Science and Technology, Kunming, China
| | - Mengyu Yao
- Medical School, Center for Translational Research in Clinical Medicine, Kunming University of Science and Technology, Kunming, China
| | - Kunlin Li
- Department of General Surgery II, The First People's Hospital of Yunnan Province, Kunming, China
| | - Yu Cao
- Yunnan Key Laboratory of Innovative Application of Traditional Chinese Medicine, Department of Cardiovascular Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Lihong Jiang
- Medical School, Center for Translational Research in Clinical Medicine, Kunming University of Science and Technology, Kunming, China
- Yunnan Key Laboratory of Innovative Application of Traditional Chinese Medicine, Department of Cardiovascular Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
17
|
Siew WS, Tang YQ, Goh BH, Yap WH. The senescent marker p16INK4a enhances macrophage foam cells formation. Mol Biol Rep 2024; 51:1021. [PMID: 39331194 DOI: 10.1007/s11033-024-09946-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND The senescence marker p16INK4a, which constitutes part of the genome 9p21.3 cardiovascular disease (CVD) risk allele, is believed to play a role in foam cells formation. This study aims to unravel the role of p16INK4a in mediating macrophage foam cells formation, cellular senescence, and autophagy lysosomal functions. METHODS The mammalian expression plasmid pCMV-p16INK4a was used to induce p16INK4a overexpression in THP-1 macrophages. Next, wild-type and p16INK4a-overexpressed macrophages were incubated with oxidized LDL to induce foam cells formation. Lipids accumulation was evaluated using Oil-red-O staining and cholesterol efflux assay, as well as expression of scavenger receptors CD36 and LOX-1. Cellular senescence in macrophage foam cells were determined through analysis of senescence-associated β-galactosidase activity and other SASP factors expression. Meanwhile, autophagy induction was assessed through detection of autophagosome formation and LC3B/p62 markers expression. RESULTS The findings showed that p16INK4a enhanced foam cells formation with increased scavenger receptors CD36 and LOX-1 expression and reduced cholesterol efflux in THP-1 macrophages. Besides, β-galactosidase activity was enhanced, and SASP factors such as IL-1α, TNF-α, and MMP9 were up-regulated. In addition, p16INK4a is also shown to induce autophagy, as well as increasing autophagy markers LC3B and p62 expression. CONCLUSIONS This study provides insights on p16INK4a in mediating macrophages foam cells formation, cellular senescence, and foam cells formation.
Collapse
Affiliation(s)
- Wei Sheng Siew
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, 47500, Malaysia
| | - Yin Quan Tang
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, 47500, Malaysia
| | - Bey Hing Goh
- Sunway Biofunctional Molecules Discovery Centre, School of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500, Selangor, Malaysia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, Australia
| | - Wei Hsum Yap
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, 47500, Malaysia.
| |
Collapse
|
18
|
侯 永, 李 霞, 王 建, 刘 振, 韩 明, 刘 烝, 金 卫. [Protective Effect and Mechanism of miR-328-3p on Coronary Artery Endothelial Cell Injury Induced by Oxidized Low-density Lipoprotein]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:1210-1216. [PMID: 39507982 PMCID: PMC11536260 DOI: 10.12182/20240960601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Indexed: 11/08/2024]
Abstract
Objective To investigate the protective effect of miR-328-3p on oxidized low-density lipoprotein (ox-LDL)-induced coronary artery endothelial cell injury and the potentially relevant mechanisms. Methods Human coronary artery endothelial cells (HCAECs) were induced with ox-LDL, and the cells were divided into a control group consisting of normal cells, an ox-LDL group receiving ox-LDL treatment, an ox-LDL+miR-NC group transfected with miR-NC and treated with ox-LDL, an ox-LDL+miR-328-3p group transfected with miR-328-3p and treated with ox-LDL, and ox-LDL+miR-328-3p+pcDNA group co-transfected miR-328-3p and pcDNA and treated with ox-LDL, and an ox-LDL+miR-328-3p+insulin-like growth factor 2 (IGF2) group co-transfected miR-328-3p and IGF2 and treated with ox-LDL. The expression level of miR-328-3p was determined with RT-qPCR. Cell proliferation was determined by MTT. Cell apoptosis was measured by flow cytometry. Western blot was conducted to examine the protein expression levels of cleaved cas-3 and IGF2. ELISA was performed to determine the levels of tumor necrosis factor α (TNF-α), interleukin (IL)-6, and IL-1β. Dual luciferase reporter experiment was performed to verify the targeting relationship between miR-328-3p and IGF2. Results Compared with those of the control group, miR-328-3p expression level and cell activity were significantly reduced in the ox-LDL group (P<0.05), while the apoptotic rate, the protein expression levels of cleaved cas-3, IGF2, Bax, and Bcl-2, and the levels of TNF-α, IL-6, and IL-1β were significantly increased (P<0.05). Compared with those of the ox-LDL+miR-NC group, miR-328-3p expression level and cell activity significantly increased in the ox-LDL+miR-328-3p group (P<0.05), while the apoptosis rate, the protein expression levels of cleaved cas-3 and IGF2, and the levels of TNF-α, IL-6, and IL-1β were significantly reduced. IGF2 was a functional target of miR-328-3p. Compared with those of the ox-LDL+miR-328-3p+pcDNA co-transfection group, the IGF2 protein level was significantly increased (P<0.05) and cell activity was significantly decreased (P<0.05) in the ox-LDL+miR-328-3p+IGF2 co-transfection group, while the apoptosis rate, cleaved cas-3 protein level, and the levels of TNF-α, IL-6, and IL-1β were significantly elevated (P<0.05). Conclusion miR-328-3p inhibits ox-LDL-induced apoptosis and inflammatory in coronary artery endothelial cell injury through targeted negative regulation of IGF2.
Collapse
Affiliation(s)
- 永兰 侯
- 新乡市中心医院 心血管内科 (新乡 453000)Department of Cardiovascular Medicine, Xinxiang Central Hospital, Xinxiang 453000, China
| | - 霞 李
- 新乡市中心医院 心血管内科 (新乡 453000)Department of Cardiovascular Medicine, Xinxiang Central Hospital, Xinxiang 453000, China
| | - 建美 王
- 新乡市中心医院 心血管内科 (新乡 453000)Department of Cardiovascular Medicine, Xinxiang Central Hospital, Xinxiang 453000, China
| | - 振 刘
- 新乡市中心医院 心血管内科 (新乡 453000)Department of Cardiovascular Medicine, Xinxiang Central Hospital, Xinxiang 453000, China
| | - 明磊 韩
- 新乡市中心医院 心血管内科 (新乡 453000)Department of Cardiovascular Medicine, Xinxiang Central Hospital, Xinxiang 453000, China
| | - 烝昊 刘
- 新乡市中心医院 心血管内科 (新乡 453000)Department of Cardiovascular Medicine, Xinxiang Central Hospital, Xinxiang 453000, China
| | - 卫东 金
- 新乡市中心医院 心血管内科 (新乡 453000)Department of Cardiovascular Medicine, Xinxiang Central Hospital, Xinxiang 453000, China
| |
Collapse
|
19
|
Wang YM, Chu TJ, Wan RT, Niu WP, Bian YF, Li J. Quercetin ameliorates atherosclerosis by inhibiting inflammation of vascular endothelial cells via Piezo1 channels. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155865. [PMID: 39004029 DOI: 10.1016/j.phymed.2024.155865] [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: 12/26/2023] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Natural antioxidants, exemplified by quercetin (Qu), have been shown to exert a protective effect against atherosclerosis (AS). However, the precise pharmacological mechanisms of Qu also remain elusive. PURPOSE Here, we aimed to uncover the anti-atherosclerotic mechanisms of Qu. METHODS/STUDY DESIGNS The inflammatory cytokine expression, activity of NLRP3 inflammasome and NF-κB, as well as mechanically activated currents and intracellular calcium levels were measured in endothelial cells (ECs). In addition, to explore whether Qu inhibited atherosclerotic plaque formation via Piezo1 channels, Ldlr-/- and Piezo1 endothelial-specific knockout mice (Piezo1△EC) were established. RESULTS Our findings revealed that Qu significantly inhibited Yoda1-evoked calcium response in human umbilical vein endothelial cells (HUVECs), underscoring its role as a selective modulator of Piezo1 channels. Additionally, Qu effectively reduced mechanically activated currents in HUVECs. Moreover, Qu exhibited a substantial inhibitory effect on inflammatory cytokine expression and reduced the activity of NF-κB/NLRP3 in ECs exposed to ox-LDL or mechanical stretch, and these effects remained unaffected after Piezo1 genetic depletion. Furthermore, our study demonstrated that Qu substantially reduced the formation of atherosclerotic plaques, and this effect remained consistent even after Piezo1 genetic depletion. CONCLUSION These results collectively provide compelling evidence that Qu ameliorates atherosclerosis by inhibiting the inflammatory response in ECs by targeting Piezo1 channels. In addition, Qu modulated atherosclerosis via inhibiting Piezo1 mediated NFκB/IL-1β and NLRP3/caspase1/ IL-1β axis to suppress the inflammation. Overall, this study reveals the potential mechanisms by which natural antioxidants, such as Qu, protect against atherosclerosis.
Collapse
Affiliation(s)
- Yu-Man Wang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road Changqing District, Ji'nan, Shandong 250355, China
| | - Tian-Jiao Chu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road Changqing District, Ji'nan, Shandong 250355, China
| | - Ren-Tao Wan
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wei-Pin Niu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road Changqing District, Ji'nan, Shandong 250355, China
| | - Yi-Fei Bian
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road Changqing District, Ji'nan, Shandong 250355, China.
| | - Jing Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road Changqing District, Ji'nan, Shandong 250355, China.
| |
Collapse
|
20
|
Wai KW, Low LE, Goh BH, Yap WH. Nrf2 Connects Cellular Autophagy and Vascular Senescence in Atherosclerosis: A Mini-Review. J Lipid Atheroscler 2024; 13:292-305. [PMID: 39355399 PMCID: PMC11439754 DOI: 10.12997/jla.2024.13.3.292] [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: 12/09/2024] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 10/03/2024] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2), a transcriptional factor that maintains intracellular redox equilibrium, modulates the expression of antioxidant genes, scavenger receptors, and cholesterol efflux transporters, all of which contribute significantly to foam cell development and plaque formation. Nrf2 has recently emerged as a key regulator that connects autophagy and vascular senescence in atherosclerosis. Autophagy, a cellular mechanism involved in the breakdown and recycling of damaged proteins and organelles, and cellular senescence, a state of irreversible growth arrest, are both processes implicated in the pathogenesis of atherosclerosis. The intricate interplay of these processes has received increasing attention, shedding light on their cumulative role in driving the development of atherosclerosis. Recent studies have revealed that Nrf2 plays a critical role in mediating autophagy and senescence in atherosclerosis progression. Nrf2 activation promotes autophagy, which increases lipid clearance and prevents the development of foam cells. Meanwhile, the activation of Nrf2 also inhibits cellular senescence by regulating the expression of senescence markers to preserve cellular homeostasis and function and delay the progression of atherosclerosis. This review provides an overview of the molecular mechanisms through which Nrf2 connects cellular autophagy and vascular senescence in atherosclerosis. Understanding these mechanisms can provide insights into potential therapeutic strategies targeting Nrf2 to modulate cellular autophagy and vascular senescence, thereby preventing the progression of atherosclerosis.
Collapse
Affiliation(s)
- Kai Wen Wai
- School of Biosciences, Taylor’s University, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Liang Ee Low
- Chemical Engineering Discipline, School of Engineering, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya, Selangor, Malaysia
- Monash-Industry Plant Oils Research Laboratory (MIPO), Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya, Selangor, Malaysia
- Advanced Engineering Platform, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya, Selangor, Malaysia
| | - Bey Hing Goh
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor Darul Ehsan, Malaysia
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, No.5 Jalan Universiti, Bandar Sunway, Subang Jaya, Selangor Darul Ehsan, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Wei Hsum Yap
- School of Biosciences, Taylor’s University, Subang Jaya, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
21
|
Zuo C, Cai L, Li Y, Ding C, Liu G, Zhang C, Wang H, Zhang Y, Ji M. The Molecular Mechanism of Radix Paeoniae Rubra.-Cortex Moutan. Herb Pair in the Treatment of Atherosclerosis: A Work Based on Network Pharmacology and In Vitro Experiments. Cardiovasc Toxicol 2024; 24:800-817. [PMID: 38951468 DOI: 10.1007/s12012-024-09881-2] [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: 03/12/2024] [Accepted: 06/10/2024] [Indexed: 07/03/2024]
Abstract
Radix Paeoniae Rubra. (Chishao, RPR) and Cortex Moutan. (Mudanpi, CM) are a pair of traditional Chinese medicines that play an important role in the treatment of atherosclerosis (AS). The main objective of this study was to identify potential synergetic function and underlying mechanisms of RPR-CM in the treatment of AS. The main active ingredients, targets of RPR-CM and AS-related genes were obtained from public databases. A Venn diagram was utilized to screen the common targets of RPR-CM in treating AS. The protein-protein interaction network was established based on STRING database. Biological functions and pathways of potential targets were analyzed through Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses. Cytoscape was used to construct the drug-compound-target-signal pathway network. Molecular docking was performed to verify the binding ability of the bioactive ingredients and the target proteins. The endothelial inflammation model was constructed with human umbilical vein endothelial cells stimulated with ox-LDL, and the function of RPR-CM in treating AS was verified by CCK-8 assay, enzyme-linked immunosorbent assay, and qPCR. In this study, 12 active components and 401 potential target genes of RPR-CM were identified, among which quercetin, kaempferol and baicalein were considered to be the main active components. A total of 1903 AS-related genes were identified through public databases and four GEO datasets (GSE57691, GSE72633, GSE6088 and GSE199819). There are 113 common target genes of RPR-CM in treating AS. PPI network analysis identified 17 genes in cluster 1 as the core targets. Bioinformatics analysis showed that RPR-CM in AS treatment was associated with multiple downstream biological processes and signal pathways. PTGS2, JUN, CASP3, TNF, IL1B, IL6, FOS, STAT1 were identified as the core targets of RPR-CM, and molecular docking showed that the main bioactive components of RPR-CM had good binding ability with the core targets. RPR-CM extract significantly inhibited the levels of inflammatory factors TNF-α, IL-6, IL-1β, MCP-1, VCAM-1 and ICAM-1 in HUVECs, and inhibited endothelial inflammation. This study revealed the active ingredients of RPR-CM, and identified the key downstream targets and signaling pathways in the treatment of AS, providing theoretical basis for the application of RPR-CM in prevention and treatment of AS.
Collapse
Affiliation(s)
- Caojian Zuo
- Department of Cardiology, Lianshui People's Hospital, Kangda College of Nanjing Medical University, Lianshui County, No 6, Hongri East Avenue, Huai'an, 223400, Jiangsu, China.
- Department of Cardiology, Shanghai Deji Hospital, Qingdao University, Shanghai, 200331, China.
| | - Lidong Cai
- Department of Cardiology, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Ya Li
- Department of Cardiology, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Chencheng Ding
- Department of Cardiology, Shanghai Deji Hospital, Qingdao University, Shanghai, 200331, China
| | - Guiying Liu
- Department of Cardiology, Shanghai Deji Hospital, Qingdao University, Shanghai, 200331, China
| | - Changmei Zhang
- Department of Cardiology, Shanghai Deji Hospital, Qingdao University, Shanghai, 200331, China
| | - Hexiang Wang
- Department of Cardiology, Shanghai Deji Hospital, Qingdao University, Shanghai, 200331, China
| | - Yang Zhang
- Department of Cardiology, Shanghai Deji Hospital, Qingdao University, Shanghai, 200331, China
| | - Mingyue Ji
- Department of Cardiology, Lianshui People's Hospital, Kangda College of Nanjing Medical University, Lianshui County, No 6, Hongri East Avenue, Huai'an, 223400, Jiangsu, China
| |
Collapse
|
22
|
Tan M, Wang J, Chen Z, Xie X. Exploring global research trends in Chinese medicine for atherosclerosis: a bibliometric study 2012-2023. Front Cardiovasc Med 2024; 11:1400130. [PMID: 38952541 PMCID: PMC11216286 DOI: 10.3389/fcvm.2024.1400130] [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: 03/13/2024] [Accepted: 05/15/2024] [Indexed: 07/03/2024] Open
Abstract
Background While Traditional Chinese Medicine (TCM) boasts an extensive historical lineage and abundant clinical expertise in addressing atherosclerosis, this field is yet to be penetrated adequately by bibliometric studies. This study is envisaged to evaluate the contemporary scenario of TCM in conjunction with atherosclerosis over the preceding decade while also identifying forthcoming research trends and emerging topics via the lens of bibliometric analysis. Methods Literature pertaining to TCM and atherosclerosis, circulated between January 1, 2012 and November 14, 2023, was garnered for the purpose of this research. The examination embraced annual publications, primary countries/regions, engaged institutions and authors, scholarly journals, references, and keywords, utilizing analytical tools like Bibliometrix, CiteSpace, ScimagoGraphica, and VOSviewer present in the R package. Result This field boasts a total of 1,623 scholarly articles, the majority of which have been contributed by China in this field, with significant contributions stemming from the China Academy of Traditional Chinese Medicine and the Beijing University of Traditional Chinese Medicine. Moreover, this field has received financial support from both the National Natural Science Foundation of China and the National Key Basic Research Development Program. Wang Yong tops the list in terms of publication count, while Xu Hao's articles take the lead for the total number of citations, positioning them at the core of the authors' collaborative network. The Journal of Ethnopharmacology leads with the most publications and boasts the greatest total number of citations. Principal research foci within the intersection of Chinese Medicine and Atherosclerosis encompass disease characteristics and pathogenic mechanisms, theoretical underpinnings and syndrome-specific treatments in Chinese medicine, potentialities of herbal interventions, and modulation exerted by Chinese medicines on gut microbiota. Conclusion This analysis offers a sweeping survey of the contemporary condition, principal foci, and progressive trends in worldwide research related to Traditional Chinese Medicine (TCM) and atherosclerosis. It further delves into an in-depth dissection of prominent countries, research institutions, and scholars that have made noteworthy strides in this discipline. Additionally, the report analyzes the most cited articles, research developments, and hotspots in the field, providing a reference for future research directions for clinical researchers and practitioners.
Collapse
Affiliation(s)
- Moye Tan
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Jiuyuan Wang
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Zhengxin Chen
- College of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuejiao Xie
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
23
|
Bai X, Wang S, Shu L, Cao Q, Hu H, Zhu Y, Chen C. Hawthorn leaf flavonoids alleviate the deterioration of atherosclerosis by inhibiting SCAP-SREBP2-LDLR pathway through sPLA2-ⅡA signaling in macrophages in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 327:118006. [PMID: 38442806 DOI: 10.1016/j.jep.2024.118006] [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: 01/04/2024] [Revised: 02/23/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hawthorn leaves are a combination of the dried leaves of the Rosaceae plants, i.e., Crataegus pinnatifida Bge. or Crataegus pinnatifida Bge. var. major N. E. Br., is primarily cultivated in East Asia, North America, and Europe. hawthorn leaf flavonoids (HLF) are the main part of extraction. The HLF have demonstrated potential in preventing hypertension, inflammation, hyperlipidemia, and atherosclerosis. However, the potential pharmacological mechanism behind its anti-atherosclerotic effect has yet to be explored. AIM OF THE STUDY The in vivo and in vitro effects of HLF on lipid-mediated foam cell formation were investigated, with a specific focus on the levels of secreted phospholipase A2 type IIA (sPLA2-II A) in macrophage cells. MATERIALS AND METHODS The primary constituents of HLF were analyzed using ultra-high performance liquid chromatography and liquid chromatography-tandem mass spectrometry. In vivo, HLF, at concentrations of 5 mg/kg, 20 mg/kg, and 40 mg/kg, were administered to apolipoprotein E knockout mice (ApoE-/-) fed by high-fat diet (HFD) for 16 weeks. Aorta and serum samples were collected to identify lesion areas and lipids through mass spectrometry analysis to dissect the pathological process. RAW264.7 cells were incubated with oxidized low-density lipoprotein (ox-LDL) alone, or ox-LDL combined with different doses of HLF (100, 50, and 25 μg/ml), or ox-LDL plus 24-h sPLA2-IIA inhibitors, for cell biology analysis. Lipids and inflammatory cytokines were detected using biochemical analyzers and ELISA, while plaque size and collagen content of plaque were assessed by HE and the Masson staining of the aorta. The lipid deposition in macrophages was observed by Oil Red O staining. The expression of sPLA2-IIA and SCAP-SREBP2-LDLR was determined by RT-qPCR and Western blot analysis. RESULTS The chemical profile of HLF was studied using UPLC-Q-TOF-MS/MS, allowing the tentative identification of 20 compounds, comprising 1 phenolic acid, 9 flavonols and 10 flavones, including isovitexin, vitexin-4″-O-glucoside, quercetin-3-O-robibioside, rutin, vitexin-2″-O-rhamnoside, quercetin, etc. HLF decreased total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), and non-high-density lipoprotein cholesterol (non-HDL-C) levels in ApoE-/- mice (P < 0.05), reduced ox-LDL uptake, inhibited level of inflammatory factors, such as IL-6, IL-8, TNF-α, and IL-1ꞵ (P < 0.001), and alleviated aortic plaques with a thicker fibrous cap. HLF effectively attenuated foam cell formation in ox-LDL-treated RAW264.7 macrophages, and reduced levels of intracellular TC, free cholesterol (FC), cholesteryl ester (CE), IL-6, TNF-α, and IL-1β (P < 0.001). In both in vivo and in vitro experiments, HLF significantly downregulated the expression of sPLA2-IIA, SCAP, SREBP2, LDLR, HMGCR, and LOX-1 (P < 0.05). Furthermore, sPLA2-IIA inhibitor effectively mitigated inflammatory release in RAW264.7 macrophages and regulated SCAP-SREBP2-LDLR signaling pathway by inhibiting sPLA2-IIA secretion (P < 0.05). CONCLUSION HLF exerted a protective effect against atherosclerosis through inhibiting sPLA2-IIA to diminish SCAP-SREBP2-LDLR signaling pathway, to reduce LDL uptake caused foam cell formation, and to slow down the progression of atherosclerosis in mice.
Collapse
Affiliation(s)
- Xufeng Bai
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Shuwen Wang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Limei Shu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Qingyu Cao
- College of Pharmacy, Nanchang Medical College, Nanchang, Jiangxi, 330052, China
| | - Huiming Hu
- College of Pharmacy, Nanchang Medical College, Nanchang, Jiangxi, 330052, China; Key Laboratory of Pharmacodynamics and Quality Evaluation on Anti-Inflammatory Chinese Herbs, Jiangxi Administration of Traditional Chinese Medicine, Jiangxi, 330052, China; School of Biomedical Sciences, University of Queensland, St Lucia, Brisbane, Queensland, 4072, Australia.
| | - Yanchen Zhu
- College of Computer Science, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, St Lucia, Brisbane, Queensland, 4072, Australia.
| |
Collapse
|
24
|
Xu Q, Yu Z, Zhang M, Feng T, Song F, Tang H, Wang S, Li H. Danshen-Shanzha formula for the treatment of atherosclerosis: ethnopharmacological relevance, preparation methods, chemical constituents, pharmacokinetic properties, and pharmacological effects. Front Pharmacol 2024; 15:1380977. [PMID: 38910885 PMCID: PMC11190183 DOI: 10.3389/fphar.2024.1380977] [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: 02/02/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
Danshen-Shanzha Formula (DSF) is a well-known herbal combination comprising Radix Salvia Miltiorrhiza (known as Danshen in Chinese) and Fructus Crataegi (known as Shanzha in Chinese), It has been documented to exhibit considerable benefits for promoting blood circulation and removing blood stasis, and was used extensively in the treatment of atherosclerotic cardiac and cerebral vascular diseases over decades. Despite several breakthroughs achieved in the basic research and clinical applications of DSF over the past decades, there is a lack of comprehensive reviews summarizing its features and research, which hinders further exploration and exploitation of this promising formula. This review aims to provide a comprehensive interpretation of DSF in terms of its ethnopharmacological relevance, preparation methods, chemical constituents, pharmacokinetic properties and pharmacological effects. The related information on Danshen, Shanzha, and DSF was obtained from internationally recognized online scientific databases, including Web of Science, PubMed, Google Scholar, China National Knowledge Infrastructure, Baidu Scholar, ScienceDirect, ACS Publications, Online Library, Wan Fang Database as well as Flora of China. Data were also gathered from documentations, printed works and classics, such as the Chinese Pharmacopoeia, Chinese herbal classics, etc. Three essential avenues for future studies were put forward as follows: a) Develop and unify the standard preparation method of DSF as to achieve optimized pharmacological properties. b) Elucidate the functional mechanisms as well as the rationality and rule for the compatibility art of DSF by focusing on the clinic syndromes together with the subsequent development of preclinic study system in vitro and in vivo with consistent pathological features, pharmacokinetical behaviour and biomarkers. c) Perform more extensive clinical studies towards the advancement of mechanism-based on evidence-based medicine on the safety application of DSF. This review will provide substantial data support and broader perspective for further research on the renowned formula.
Collapse
Affiliation(s)
- Qiong Xu
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an, China
| | - Zhe Yu
- Department of Pharmaceutical Analysis, School of Pharmacy, Air Force Medical University, Xi’an, China
| | - Meng Zhang
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an, China
- School of Graduate Studies, Air Force Medical University, Xi’an, China
| | - Tian Feng
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an, China
| | - Fan Song
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an, China
| | - Haifeng Tang
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an, China
| | - Siwang Wang
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an, China
| | - Hua Li
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an, China
| |
Collapse
|
25
|
Pukhalskaia TV, Yurakova TR, Bogdanova DA, Demidov ON. Tumor-Associated Senescent Macrophages, Their Markers, and Their Role in Tumor Microenvironment. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:839-852. [PMID: 38880645 DOI: 10.1134/s0006297924050055] [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: 12/21/2023] [Revised: 04/27/2024] [Accepted: 04/27/2024] [Indexed: 06/18/2024]
Abstract
Tumor-associated macrophages (TAMs) are an important component of the tumor microenvironment (TME) and the most abundant population of immune cells infiltrating a tumor. TAMs can largely determine direction of anti-tumor immune response by promoting it or, conversely, contribute to formation of an immunosuppressive TME that allows tumors to evade immune control. Through interactions with tumor cells or other cells in the microenvironment and, as a result of action of anti-cancer therapy, macrophages can enter senescence. In this review, we have attempted to summarize information available in the literature on the role of senescent macrophages in tumors. With the recent development of senolytic therapeutic strategies aimed at removing senescent cells from an organism, it seems important to discuss functions of the senescent macrophages and potential role of the senolytic drugs in reprogramming TAMs to enhance anti-tumor immune response and improve efficacy of cancer treatment.
Collapse
Affiliation(s)
- Tamara V Pukhalskaia
- Sirius University of Science and Technology, Federal Territory Sirius, 354340, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia
| | - Taisiya R Yurakova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Daria A Bogdanova
- Sirius University of Science and Technology, Federal Territory Sirius, 354340, Russia
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia
| | - Oleg N Demidov
- Sirius University of Science and Technology, Federal Territory Sirius, 354340, Russia.
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia
- INSERM UMR1231, Université de Bourgogne, Dijon, 21000, France
| |
Collapse
|
26
|
Jiang X, Lei Y, Yin Y, Ma F, Zheng M, Liu G. Fisetin Suppresses Atherosclerosis by Inhibiting Ferroptosis-Related Oxidative Stress in Apolipoprotein E Knockout Mice. Pharmacology 2024; 109:169-179. [PMID: 38583431 DOI: 10.1159/000538535] [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: 01/11/2024] [Accepted: 03/22/2024] [Indexed: 04/09/2024]
Abstract
INTRODUCTION Fisetin has been demonstrated to inhibit the occurrence of atherosclerosis; however, the mechanism of fisetin suppressing atherosclerosis remains elusive. METHODS The function of fisetin in the inhibition of atherosclerosis was evaluated by hematoxylin and eosin and Oil Red O staining in ApoE-/- mice. Molecular biomarkers of atherosclerosis progression were detected by Western blot and qPCR. Moreover, the inhibition of atherosclerosis on oxidative stress and ferroptosis was evaluated by immunofluorescence staining, qPCR, and Western blot assays. RESULTS The obtained results showed that serum lipid was attenuated and consequentially the formation of atherosclerosis was also suppressed by fisetin in ApoE-/- mice. Exploration of the mechanism revealed that molecular biomarkers of atherosclerosis were decreased under fisetin treatment. The level of reactive oxygen species and malondialdehyde declined, while the activity of superoxide dismutases and glutathione peroxidase was increased under the fisetin treatment. Additionally, the suppressor of ferroptosis, glutathione peroxidase 4 proteins, was elevated. The ferritin was decreased in the aortic tissues treated with fisetin. CONCLUSIONS In summary, fisetin attenuated the formation of atherosclerosis through the inhibition of oxidative stress and ferroptosis in the aortic tissues of ApoE-/- mice.
Collapse
Affiliation(s)
- Xiufang Jiang
- Department of Medical Affairs, The First Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang, China
| | - Yanling Lei
- Department of Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yajuan Yin
- Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang, China
- Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, China
| | - Fangfang Ma
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Mingqi Zheng
- Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang, China
- Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, China
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Engineering Research Center of Intelligent Medical Clinical Application, Shijiazhuang, China
- Hebei International Joint Research Center for Structural Heart Disease, Shijiazhuang, China
| | - Gang Liu
- Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang, China
- Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, China
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Engineering Research Center of Intelligent Medical Clinical Application, Shijiazhuang, China
- Hebei International Joint Research Center for Structural Heart Disease, Shijiazhuang, China
| |
Collapse
|
27
|
Makinde E, Ma L, Mellick GD, Feng Y. A High-Throughput Screening of a Natural Products Library for Mitochondria Modulators. Biomolecules 2024; 14:440. [PMID: 38672457 PMCID: PMC11048375 DOI: 10.3390/biom14040440] [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: 02/21/2024] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Mitochondria, the energy hubs of the cell, are progressively becoming attractive targets in the search for potent therapeutics against neurodegenerative diseases. The pivotal role of mitochondrial dysfunction in the pathogenesis of various diseases, including Parkinson's disease (PD), underscores the urgency of discovering novel therapeutic strategies. Given the limitations associated with available treatments for mitochondrial dysfunction-associated diseases, the search for new potent alternatives has become imperative. In this report, we embarked on an extensive screening of 4224 fractions from 384 Australian marine organisms and plant samples to identify natural products with protective effects on mitochondria. Our initial screening using PD patient-sourced olfactory neurosphere-derived (hONS) cells with rotenone as a mitochondria stressor resulted in 108 promising fractions from 11 different biota. To further assess the potency and efficacy of these hits, the 11 biotas were subjected to a subsequent round of screening on human neuroblastoma (SH-SY5Y) cells, using 6-hydroxydopamine to induce mitochondrial stress, complemented by a mitochondrial membrane potential assay. This rigorous process yielded 35 active fractions from eight biotas. Advanced analysis using an orbit trap mass spectrophotometer facilitated the identification of the molecular constituents of the most active fraction from each of the eight biotas. This meticulous approach led to the discovery of 57 unique compounds, among which 12 were previously recognized for their mitoprotective effects. Our findings highlight the vast potential of natural products derived from Australian marine organisms and plants in the quest for innovative treatments targeting mitochondrial dysfunction in neurodegenerative diseases.
Collapse
Affiliation(s)
- Emmanuel Makinde
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia; (E.M.); (L.M.); (G.D.M.)
| | - Linlin Ma
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia; (E.M.); (L.M.); (G.D.M.)
- School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia
| | - George D. Mellick
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia; (E.M.); (L.M.); (G.D.M.)
- School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia
| | - Yunjiang Feng
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia; (E.M.); (L.M.); (G.D.M.)
- School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia
| |
Collapse
|
28
|
Li Y, Pan J, Yu JJJ, Wu X, Yang G, Pan X, Sui G, Wang M, Cheng M, Zhu S, Tai H, Xiao H, Xu L, Wu J, Yang Y, Tang J, Gong L, Jia L, Min D. Huayu Qutan Recipe promotes lipophagy and cholesterol efflux through the mTORC1/TFEB/ABCA1-SCARB1 signal axis. J Cell Mol Med 2024; 28:e18257. [PMID: 38526033 PMCID: PMC10962127 DOI: 10.1111/jcmm.18257] [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: 08/17/2023] [Revised: 02/01/2024] [Accepted: 02/14/2024] [Indexed: 03/26/2024] Open
Abstract
This study aims to investigate the mechanism of the anti-atherosclerosis effect of Huayu Qutan Recipe (HYQT) on the inhibition of foam cell formation. In vivo, the mice were randomly divided into three groups: CTRL group, MOD group and HYQT group. The HYQT group received HYQT oral administration twice a day (20.54 g/kg/d), and the plaque formation in ApoE-/- mice was observed using haematoxylin-eosin (HE) staining and oil red O (ORO) staining. The co-localization of aortic macrophages and lipid droplets (LDs) was examined using fluorescent labelling of CD11b and BODIPY fluorescence probe. In vitro, RAW 264.7 cells were exposed to 50 μg/mL ox-LDL for 48 h and then treated with HYQT for 24 h. The accumulation of LDs was evaluated using ORO and BODIPY. Cell viability was assessed using the CCK-8 assay. The co-localization of LC3b and BODIPY was detected via immunofluorescence and fluorescence probe. LysoTracker Red and BODIPY 493/503 were used as markers for lysosomes and LDs, respectively. Autophagosome formation were observed via transmission electron microscopy. The levels of LC3A/B II/LC3A/B I, p-mTOR/mTOR, p-4EBP1/4EBP1, p-P70S6K/P70S6K and TFEB protein level were examined via western blotting, while SQSTM1/p62, Beclin1, ABCA1, ABCG1 and SCARB1 were examined via qRT-PCR and western blotting. The nuclear translocation of TFEB was detected using immunofluorescence. The components of HYQT medicated serum were determined using Q-Orbitrap high-resolution MS analysis. Molecular docking was employed to identify the components of HYQT medicated serum responsible for the mTOR signalling pathway. The mechanism of taurine was illustrated. HYQT has a remarkable effect on atherosclerotic plaque formation and blood lipid level in ApoE-/- mice. HYQT decreased the co-localization of CD11b and BODIPY. HYQT (10% medicated serum) reduced the LDs accumulation in RAW 264.7 cells. HYQT and RAPA (rapamycin, a mTOR inhibitor) could promote cholesterol efflux, while chloroquine (CQ, an autophagy inhibitor) weakened the effect of HYQT. Moreover, MHY1485 (a mTOR agonist) also mitigated the effects of HYQT by reduced cholesterol efflux. qRT-PCR and WB results suggested that HYQT improved the expression of the proteins ABCA1, ABCG1 and SCARB1.HYQT regulates ABCA1 and SCARB1 protein depending on the mTORC1/TFEB signalling pathway. However, the activation of ABCG1 does not depend on this pathway. Q-Orbitrap high-resolution MS analysis results demonstrated that seven core compounds have good binding ability to the mTOR protein. Taurine may play an important role in the mechanism regulation. HYQT may reduce cardiovascular risk by promoting cholesterol efflux and degrading macrophage-derived foam cell formation. It has been observed that HYQT and ox-LDL regulate lipophagy through the mTOR/TFEB signalling pathway, rather than the mTOR/4EBP1/P70S6K pathway. Additionally, HYQT is found to regulate cholesterol efflux through the mTORC1/TFEB/ABCA1-SCARB1 signal axis, while taurine plays a significant role in lipophagy.
Collapse
Affiliation(s)
- Yue Li
- Department of Cardiologythe Affiliated Hospital of Liaoning University of Traditional Chinese MedicineShenyangChina
- Liaoning Provincial Key Laboratory of TCM Geriatric Cardio‐Cerebrovascular DiseasesShenyangChina
| | - Jiaxiang Pan
- Department of Cardiologythe Affiliated Hospital of Liaoning University of Traditional Chinese MedicineShenyangChina
- Graduate School of Liaoning University of Traditional Chinese MedicineShenyangChina
| | - J. J. Jiajia Yu
- Postdoctoral Program of Liaoning University of Traditional Chinese MedicineShenyangChina
| | - Xize Wu
- Graduate School of Liaoning University of Traditional Chinese MedicineShenyangChina
- Nantong Hospital of Traditional Chinese MedicineNantong Hospital Affiliated to Nanjing University of Chinese MedicineNantongChina
| | - Guanlin Yang
- Innovation Engineering Technology Center of Traditional Chinese MedicineLiaoning University of Traditional Chinese MedicineShenyangChina
| | - Xue Pan
- Graduate School of Liaoning University of Traditional Chinese MedicineShenyangChina
- Dazhou Vocational College of Chinese MedicineDazhouChina
| | - Guoyuan Sui
- Innovation Engineering Technology Center of Traditional Chinese MedicineLiaoning University of Traditional Chinese MedicineShenyangChina
| | - Mingyang Wang
- College of Animal Science and Veterinary Medicine of Shenyang Agricultural UniversityShenyangChina
| | - Meijia Cheng
- Experimental Center of Traditional Chinese Medicinethe Affiliated Hospital of Liaoning University of Traditional Chinese MedicineShenyangChina
| | - Shu Zhu
- Department of Paediatric Dentistry, School of StomatologyChina Medical UniversityShenyangChina
| | - He Tai
- School of PharmacyLiaoning University of Traditional Chinese MedicineDalianChina
| | - Honghe Xiao
- School of PharmacyLiaoning University of Traditional Chinese MedicineDalianChina
| | - Lili Xu
- Department of Cardiology, 924 Hospital of Joint Logistic Support Force of PLAGuilinChina
| | - Jin Wu
- Innovation Engineering Technology Center of Traditional Chinese MedicineLiaoning University of Traditional Chinese MedicineShenyangChina
| | - Yongju Yang
- Experimental Center of Traditional Chinese Medicinethe Affiliated Hospital of Liaoning University of Traditional Chinese MedicineShenyangChina
| | - Jing Tang
- Department of Cardiologythe Affiliated Hospital of Liaoning University of Traditional Chinese MedicineShenyangChina
| | - Lihong Gong
- Department of Cardiologythe Affiliated Hospital of Liaoning University of Traditional Chinese MedicineShenyangChina
- Liaoning Provincial Key Laboratory of TCM Geriatric Cardio‐Cerebrovascular DiseasesShenyangChina
| | - Lianqun Jia
- Innovation Engineering Technology Center of Traditional Chinese MedicineLiaoning University of Traditional Chinese MedicineShenyangChina
| | - Dongyu Min
- Experimental Center of Traditional Chinese Medicinethe Affiliated Hospital of Liaoning University of Traditional Chinese MedicineShenyangChina
| |
Collapse
|
29
|
Wang L, Hong W, Zhu H, He Q, Yang B, Wang J, Weng Q. Macrophage senescence in health and diseases. Acta Pharm Sin B 2024; 14:1508-1524. [PMID: 38572110 PMCID: PMC10985037 DOI: 10.1016/j.apsb.2024.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/16/2023] [Accepted: 12/06/2023] [Indexed: 04/05/2024] Open
Abstract
Macrophage senescence, manifested by the special form of durable cell cycle arrest and chronic low-grade inflammation like senescence-associated secretory phenotype, has long been considered harmful. Persistent senescence of macrophages may lead to maladaptation, immune dysfunction, and finally the development of age-related diseases, infections, autoimmune diseases, and malignancies. However, it is a ubiquitous, multi-factorial, and dynamic complex phenomenon that also plays roles in remodeled processes, including wound repair and embryogenesis. In this review, we summarize some general molecular changes and several specific biomarkers during macrophage senescence, which may bring new sight to recognize senescent macrophages in different conditions. Also, we take an in-depth look at the functional changes in senescent macrophages, including metabolism, autophagy, polarization, phagocytosis, antigen presentation, and infiltration or recruitment. Furthermore, some degenerations and diseases associated with senescent macrophages as well as the mechanisms or relevant genetic regulations of senescent macrophages are integrated, not only emphasizing the possibility of regulating macrophage senescence to benefit age-associated diseases but also has an implication on the finding of potential targets or drugs clinically.
Collapse
Affiliation(s)
- Longling Wang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
| | - Wenxiang Hong
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hong Zhu
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
| | - Bo Yang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Wang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
- Taizhou Institute of Zhejiang University, Taizhou 318000, China
| | - Qinjie Weng
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
- Taizhou Institute of Zhejiang University, Taizhou 318000, China
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| |
Collapse
|
30
|
Cai M, Li Q, Cao Y, Huang Y, Yao H, Zhao C, Wang J, Zhu H. Quercetin activates autophagy to protect rats ovarian granulosa cells from H 2O 2-induced aging and injury. Eur J Pharmacol 2024; 966:176339. [PMID: 38272342 DOI: 10.1016/j.ejphar.2024.176339] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024]
Abstract
Autophagy is closely related to the aging of various organ systems, including ovaries. Quercetin has a variety of biological activities, including potential regulation of autophagy. However, whether quercetin-regulated autophagy activity affects the process of ovarian aging and injury has not been clarified yet. This study explores whether quercetin can resist H2O2-induced aging and injury of granulosa cells by regulating autophagy and its related molecular mechanisms in vitro experiments. The cell viability, endocrine function, cell aging, and apoptosis were detected to evaluate the effects of quercetin and autophagy regulators like 3-methyladenine and rapamycin. The levels of autophagy markers Atg5, Atg12, Atg16L, Lc3B II/I, and Beclin1 were determined by Western blot to assess the effects of quercetin, 3-methyladenine and rapamycin on autophagy. Our results showed quercetin resisted H2O2-induced granulosa cell aging and injury by activating protective autophagy. The treatment of 3-methyladenine and rapamycin confirmed the protective function of autophagy in H2O2-induced granulosa cells. 3-methyladenine treatment inhibited the expression of autophagy markers Atg5, Atg12, Atg16L, Lc3B II/I, and Beclin1 and abolished the positive effects on cell viability, estradiol secretion, and cell apoptosis activated by quercetin. In conclusion, quercetin activates autophagy by upregulating the expression of autophagy-related proteins to resist H2O2-induced aging and injury, which is crucial for stabilizing the function of granulosa cells under oxidative injury conditions and delaying aging. This study may explain the protective effects of quercetin on ovarian aging and injury from the perspective of regulating autophagy.
Collapse
Affiliation(s)
- Minghui Cai
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Qiuyuan Li
- Department of Physiology, Harbin Medical University, Harbin, China; Department of Physiology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yang Cao
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Yujia Huang
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Haixu Yao
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Chen Zhao
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Jiao Wang
- Department of Physiology, Harbin Medical University, Harbin, China.
| | - Hui Zhu
- Department of Physiology, Harbin Medical University, Harbin, China.
| |
Collapse
|
31
|
Zhang C, Huang X, Xie B, Lian D, Chen J, Li W, Lin Y, Cai X, Li J. The multi-protective effect of IL-37-Smad3 against ox-LDL induced dysfunction of endothelial cells. Biomed Pharmacother 2024; 172:116268. [PMID: 38359489 DOI: 10.1016/j.biopha.2024.116268] [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: 09/03/2023] [Revised: 01/30/2024] [Accepted: 02/07/2024] [Indexed: 02/17/2024] Open
Abstract
Atherosclerosis is a lipid-driven inflammatory arterial disease, with one crucial factor is oxidized low-density lipoprotein (ox-LDL), which can induce endothelial dysfunction through endoplasmic reticulum stress (ERS). Interleukin-37 (IL-37) exerts vascular protective functions. This study aims to investigates whether IL-37 can alleviate ERS and autophagy induced by ox-LDL, therely potentialy treating atherosclerosis. We found that ox-LDL enhances the wound healing rate in Rat Coronary Artery Endothelial Cells (RCAECs) and IL-37 reduce the ox-LDL-induced pro-osteogenic response, ERS, and autophagy by binding to Smad3. In RCAECs treated with ox-LDL and recombinant human IL-37, the wound healing rate was mitigated. The expression of osteogenic transcription factors and proteins involved in the ERS pathway was reduced in the group pretreated with IL-37 and ox-LDL. However, these responses were not alleviated when Smads silenced. Electron microscopy revealed that the IL-37/Smad3 complex could suppress endoplasmic reticulum autophagy under ox-LDL stimulation. Thus, IL-37 might treat atherosclerosis through its multi-protective effect by binding Smad3.
Collapse
Affiliation(s)
- Changyi Zhang
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou City, Guangdong province, China
| | - Xiaojun Huang
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou City, Guangdong province, China
| | - Bin Xie
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou City, Guangdong province, China
| | - Danchun Lian
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou City, Guangdong province, China
| | - Jinhao Chen
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou City, Guangdong province, China
| | - Weiwen Li
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou City, Guangdong province, China
| | - Ying Lin
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou City, Guangdong province, China
| | - Xiangna Cai
- Department of Plastic Surgeon, First Affiliated Hospital of Shantou University Medical College, Shantou City, Guangdong province, China.
| | - Jilin Li
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou City, Guangdong province, China.
| |
Collapse
|
32
|
Gu Y, Cui M, Wang W, Zhang J, Wang H, Zheng C, Lei L, Ji M, Chen W, Xu Y, Wang P. Visualization of the Ferroptosis in Atherosclerotic Plaques with Nanoprobe Engineered by Macrophage Cell Membranes. Anal Chem 2024; 96:281-291. [PMID: 38153251 DOI: 10.1021/acs.analchem.3c03999] [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: 12/29/2023]
Abstract
Atherosclerosis (AS) is the root cause of cardiovascular diseases. Ferroptosis is characterized by highly iron-dependent lipid peroxidation and has been reported to play an important role in the pathogenesis of AS. Visualization of the ferroptosis process in atherosclerotic plaques is of great importance for diagnosing and treating AS. In this work, the rationally designed fluorescent probe FAS1 exhibited excellent advantages including large Stokes shift, sensitivity to environmental viscosity, good photostability, and improved water solubility. It also could co-locate with commercial lipid droplets (LDs) probes (BODIPY 493/503) well in RAW264.7 cells treated by the ferroptosis inducer. After self-assembly into nanoparticles and then encapsulation with macrophage membranes, the engineered FAS1@MM NPs could successfully target the atherosclerotic plaques in Western diet-induced apolipoprotein E knockout (ApoE-/-) mice and reveal the association of ferroptosis with AS through fluorescence imaging in vivo. This study may provide additional insights into the roles of ferroptosis in the diagnosis and treatment of AS.
Collapse
Affiliation(s)
- Yinhui Gu
- Department of Nuclear Medicine & Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610044, China
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Mengyuan Cui
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Weizhi Wang
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing 100050, China
| | - Jiaqi Zhang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Huizhe Wang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Cheng Zheng
- Department of Nuclear Medicine & Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610044, China
| | - Lijuan Lei
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing 100050, China
| | - Min Ji
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Wei Chen
- Department of Nuclear Medicine & Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610044, China
| | - Yanni Xu
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing 100050, China
| | - Peng Wang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
33
|
Li F, Li D, Yan X, Zhu F, Tang S, Liu J, Yan J, Chen H. Quercetin Promotes the Repair of Mitochondrial Function in H9c2 Cells Through the miR-92a-3p/Mfn1 Axis. Curr Pharm Biotechnol 2024; 25:1858-1866. [PMID: 38173217 DOI: 10.2174/0113892010266863231030052150] [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: 06/12/2023] [Revised: 09/25/2023] [Accepted: 10/04/2023] [Indexed: 01/05/2024]
Abstract
OBJECTIVE Cardiocerebrovascular disease is a severe threat to human health. Quercetin has a wide range of pharmacological effects such as antitumor and antioxidant. In this study, we aimed to determine how quercetin regulates mitochondrial function in H9c2 cells. METHODS An H9c2 cell oxygen glucose deprivation/reoxygenation (OGD/R) model was constructed. The expression of miR-92a-3p and mitofusin 1 (Mfn1) mRNA in the cells was detected using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Changes in the mitochondrial membrane potential of cells were examined by JC-1 staining. ATP production in the cells was detected using a biochemical assay. Mitochondrial morphological changes were observed using transmission electron microscopy. Detection of miR-92a-3p binding to Mfn1 was done using dual luciferase. Western blotting was used to detect the protein expression of Mfn1 in the cells. RESULTS miR-92a-3p is essential in regulating cell viability, apoptosis, and tumor cell metastasis. OGD/R induced miR-92a-3p expression, decreased mitochondrial membrane potential and mitochondrial ATP production, and increased mitochondrial damage. Mitochondria are the most critical site for ATP production. Continued opening of the mitochondrial permeability transition pore results in an abnormal mitochondrial transmembrane potential. Both quercetin and inhibition of miR-29a-3p were able to downregulate miR-29a-3p levels, increase cell viability, mitochondrial membrane potential, and ATP levels, and improve mitochondrial damage morphology. Furthermore, we found that downregulation of miR-29a-3p upregulated the protein expression of Mfn1 in cells. Additionally, miR-92a-3p was found to bind to Mfn1 in a luciferase assay. miR- 29a-3p overexpression significantly inhibited the protein expression level of Mfn1. Quercetin treatment partially reversed the effects of miR-29a-3p overexpression in H9c2 cells. CONCLUSION Quercetin promoted the recovery of mitochondrial damage in H9c2 cells through the miR-92a-3p/Mfn1 axis.
Collapse
Affiliation(s)
- Fen Li
- Department of Neurology, Wuhan Third Hospital and Tongren Hospital of Wuhan University, Wuhan, 430056, Hubei, China
| | - Dongsheng Li
- Department of Cardiology, Wuhan Third Hospital and Tongren Hospital of Wuhan University, Wuhan, 430056, Hubei, China
| | - Xisheng Yan
- Department of Cardiology, Wuhan Third Hospital and Tongren Hospital of Wuhan University, Wuhan, 430056, Hubei, China
| | - Fen Zhu
- Department of Cardiology, Wuhan Third Hospital and Tongren Hospital of Wuhan University, Wuhan, 430056, Hubei, China
| | - Shifan Tang
- Department of Cardiology, Wuhan Third Hospital and Tongren Hospital of Wuhan University, Wuhan, 430056, Hubei, China
| | - Jianguang Liu
- Department of Neurology, Wuhan Third Hospital and Tongren Hospital of Wuhan University, Wuhan, 430056, Hubei, China
| | - Jie Yan
- Department of Forensic Science, Changsha, 410013, Hunan, China
| | - Haifeng Chen
- Department of Clinical Medicine, Jianghan University, Wuhan, 430056, Hubei, China
| |
Collapse
|
34
|
Huang J, Xu S, Liu L, Zhang J, Xu J, Zhang L, Zhou X, Huang L, Peng J, Wang J, Gong Z, Chen Y. Targeted treatment of atherosclerosis with protein-polysaccharide nanoemulsion co-loaded with photosensitiser and upconversion nanoparticles. J Drug Target 2023; 31:1111-1127. [PMID: 37962293 DOI: 10.1080/1061186x.2023.2284093] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/09/2023] [Indexed: 11/15/2023]
Abstract
Macrophages are the most abundant cell group in atherosclerosis (AS) lesions and play a vital role in all stages of AS progression. Recent research has shown that reactive oxygen species (ROS) generation from photodynamic therapy (PDT) induces macrophage autophagy to improve abnormal lipid metabolism and inflammatory environment. Especially in macrophage-derived foam cells, which has become a potential strategy for the treatment of AS. In this study, we prepared the conjugate (DB) of dextran (DEX) and bovine serum albumin (BSA). The DB was used as the emulsifier to prepare nanoemulsion loaded with upconversion nanoparticles (UCNPs) and chlorin e6 (Ce6) (UCNPs-Ce6@DB). The DEX modified on the surface of the nanoemulsion can recognise and bind to the scavenger receptor class A (SR-A) highly expressed on macrophages and promote the uptake of macrophage-derived foam cells in AS plates through SR-A-mediated endocytosis. In addition, UCNPs-Ce6@DB-mediated PDT enhanced ROS generation and induced autophagy in macrophage-derived foam cells, enhanced the expression of ABCA1, a protein closely related to cholesterol efflux, and inhibited the secretion of pro-inflammatory cytokines. Ultimately, UCNPs-Ce6@DB was shown to inhibit plaque formation in mouse models of AS. In conclusion, UCNPs-Ce6@DB offers a promising treatment for AS.
Collapse
Affiliation(s)
- Jing Huang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Shan Xu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Lina Liu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Jiyuan Zhang
- School of Pharmacy, Fudan University, Shanghai, China
| | - Jinzhuan Xu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Lili Zhang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Xiang Zhou
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Lei Huang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Jianqing Peng
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Jianing Wang
- Department of Pharmacy, The Affiliated Jiangning Hospital with Nanjing Medical University, Jiangsu, Nanjing, China
| | - Zipeng Gong
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Yi Chen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| |
Collapse
|
35
|
Xu YH, Xu JB, Chen LL, Su W, Zhu Q, Tong GL. Protective mechanisms of quercetin in neonatal rat brain injury induced by hypoxic-ischemic brain damage (HIBD). Food Sci Nutr 2023; 11:7649-7663. [PMID: 38107093 PMCID: PMC10724619 DOI: 10.1002/fsn3.3684] [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: 07/13/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 12/19/2023] Open
Abstract
Neonatal hypoxic-ischemic brain damage (HIBD) is a leading cause of infant mortality worldwide. This study explored whether quercetin (Que) exerts neuroprotective effects in a rat model of HIBD. A total of 36 seven-day-old Sprague-Dawley rats were divided into control, Que, HI, and HI + Que groups. The Rice method was used to establish HIBD in HI and HI + Que rats, which were treated with hypoxia (oxygen concentration of 8%) for 2 h after ligation of the left common carotid artery. The rats in the HI + Que group were intraperitoneally injected with Que (30 mg/kg) 1 h before hypoxia, and the rats in the Que group were only injected with the same amount of Que. Brain tissues were harvested 24 h postoperation and assessed by hematoxylin and eosin staining, 2,3,5-triphenyltetrazolium chloride staining, and terminal deoxynucleotidyl transferase dUTP nick-end labeling assay; relative gene and protein levels were evaluated by RT-qPCR, IHC, or western blot (WB) assay. Brain tissue morphologies were characterized by transmission electron microscopy (TEM); LC3B protein levels were assessed by immunofluorescence staining. Escape latencies and platform crossing times were significantly improved (p < .05) in HI + Que groups; infarct volume significantly decreased (p < .001), whereas the numbers of autophagic bodies and apoptotic cells increased and decreased, respectively. Meanwhile, NLRX1, ATG7, and Beclin1 expressions were significantly upregulated, and mTOR and TIM23 expressions, LC3B protein level, and LC 3II/LC 3I ratio were significantly downregulated. Que exerted neuroprotective effects in a rat model of HIBD by regulating NLRX1 and autophagy.
Collapse
Affiliation(s)
- Yan-Hong Xu
- Anhui Provincial Children's Hospital Hefei China
| | - Jin-Bo Xu
- Anhui Provincial Children's Hospital Hefei China
| | - Lu-Lu Chen
- Anhui Provincial Children's Hospital Hefei China
| | - Wei Su
- Anhui Provincial Children's Hospital Hefei China
| | - Qing Zhu
- Anhui Provincial Children's Hospital Hefei China
| | | |
Collapse
|
36
|
Huang Q, Zhou H, Yu S. Long non‑coding RNA PEG13 regulates endothelial cell senescence through the microRNA‑195/IRS1 axis. Exp Ther Med 2023; 26:584. [PMID: 38023368 PMCID: PMC10665998 DOI: 10.3892/etm.2023.12283] [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: 05/04/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by endothelial dysfunction and plaque formation. The present study aimed to elucidate the pathological role of the long non-coding RNA (lncRNA) paternally expressed 13 (PEG13) in the onset and progression of atherosclerosis. Specifically, its effects on human umbilical vein endothelial cell (HUVEC) proliferation, angiogenesis, senescence and senescence-associated secretory phenotype (SASP)-related factors were investigated using cell proliferation, cellular angiogenesis, β-galactosidase staining, reverse transcription-quantitative PCR and enzyme-linked immunosorbent assays. The results showed that oxidized low-density lipoprotein (ox-LDL) inhibited lncRNA PEG13 expression and HUVEC viability in a dose-dependent manner and PEG13 overexpression partially reversed these effects. Additionally, PEG13 overexpression ameliorated the ox-LDL-induced impairment of angiogenesis, cellular senescence and SASP. Furthermore, lncRNA PEG13 directly targeted microRNA (miR/miRNA)-195-5p, suppressing the ox-LDL-induced upregulation of the miRNA. The gene coding for insulin receptor substrate 1 (IRS1), an activator of the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway, was confirmed as a direct target of miR-195. PEG13 overexpression attenuated the ox-LDL-induced inhibition of IRS1 expression and PI3K/AKT signaling and its protective effects on HUVEC viability, angiogenesis and senescence were partially reversed by small interfering RNAs targeting IRS1. The present study demonstrated that lncRNA PEG13 attenuates ox-LDL-induced senescence in HUVECs by modulating the miR-195/IRS1/PI3K/AKT signaling pathway, suggesting a potential therapeutic target for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Qin Huang
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi 330006, P.R. China
| | - Haiwen Zhou
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi 330006, P.R. China
| | - Songping Yu
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
37
|
Qin X, Xie Z, Chen X, Wang X, Ma L. Network pharmacology and molecular docking-based study on exploring the potential mechanism of Lycium barbarum L: In the treatment of atherosclerosis. Medicine (Baltimore) 2023; 102:e35734. [PMID: 37932998 PMCID: PMC10627648 DOI: 10.1097/md.0000000000035734] [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: 03/04/2023] [Accepted: 09/29/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND Goji berries (Lycium barbarum L) are herbal medicine that have a long history of use and multiple pharmacological activities. In this study, we investigated the potential therapeutic effects of Goji berries on atherosclerosis (AS) using network pharmacology and molecular docking. METHODS The active compounds of Goji berries were identified using the Traditional Chinese Medicine Systems Pharmacology platform, as well as the literature and the targets of each active compound were obtained using the Swiss Target Prediction database. The AS-related targets were collected from the GeneCards and OMIM databases to obtain the common targets of Goji berries and AS. The drug-compound-target-disease network and protein-protein interaction network were constructed using the Cytoscape software to obtain the core target proteins of Goji berries related to AS. Gene ontology analysis of the core targets and Kyoto encyclopedia of genes and genomes pathway enrichment analysis were performed by Metascape. The target-chemical correlations were verified using AutoDock molecular docking. RESULTS After analysis, 44 active compounds within Goji berries were obtained that exhibit associations with AS. Among these, the proteins exhibiting the highest degrees of interaction within the compound-targeted protein protein-protein interaction network were AKT1, SRC, MAPK3, MAPK1, RELA, and STAT3. The gene ontology-biology process analysis showed that compound-targeted proteins were mainly involved in regulating small molecule metabolic process, cellular response to chemical stress, reactive oxygen species metabolic process, and regulation of inflammatory response. Kyoto encyclopedia of genes and genomes pathway mainly included lipid and AS in which AKT1, SRC, MAPK3, and MAPK1 were involved. Advanced glycation end-product-receptor for advanced glycation end-product signaling pathway in diabetic complications, Chagas disease, and pancreatic disease. Molecular docking assessment showed that fucosterol is bound to AKT1, MAPK3, and SRC. CONCLUSION This study demonstrates that network pharmacology and molecular docking analyses contribute to a better understanding of Goji berries active compounds and targets as potential therapeutic drugs for treating AS.
Collapse
Affiliation(s)
- Xinchen Qin
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Zikai Xie
- Independent Researcher, Zhuhai, China
| | - Xi Chen
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
- Guangdong Second Provincial General Hospital, Postdoctoral Research Station of Basic Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Xiaoxuan Wang
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Lijuan Ma
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| |
Collapse
|
38
|
Zhang N, Ye T, Lu X, Li ZH, Li L. Radix Scrophulariae Extracts Exert Effect on Hyperthyroidism via MST1/Hippo Signaling Pathway. Chin J Integr Med 2023; 29:998-1006. [PMID: 37661231 DOI: 10.1007/s11655-023-3744-7] [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] [Accepted: 04/15/2023] [Indexed: 09/05/2023]
Abstract
OBJECTIVE To explore the mechanism of Radix Scrophulariae (RS) extracts in the treatment of hyperthyroidism rats by regulating proliferation, apoptosis, and autophagy of thyroid cell through the mammalian sterile 20-like kinase 1 (MST1)/Hippo pathway. METHODS Twenty-four rats were randomly divided into 4 groups according to a random number table: control, model group, RS, and RS+Hippo inhibitor (XMU-MP-1) groups (n=6 per group). Rats were gavaged with levothyroxine sodium tablet suspension (LST, 8 μ g/kg) for 21 days except for the control group. Afterwards, rats in the RS group were gavaged with RS extracts at the dose of 1,350 mg/kg, and rats in the RS+XMU-MP-1 group were gavaged with 1,350 mg/kg RS extracts and 1 mg/kg XMU-MP-1. After 15 days of administration, thyroid gland was taken for gross observation, and histopathological changes were observed by hematoxylin-eosin staining. The structure of Golgi secretory vesicles in thyroid tissues was observed by transmission electron microscopy. The expression of thyrotropin receptor (TSH-R) was observed by immunohistochemistry. Terminal-deoxynucleoitidyl transferase mediated nick end labeling assay was used to detect cell apoptosis in thyroid tissues. Real-time quantity primer chain reaction and Western blot were used to detect the expressions of MST1, p-large tumor suppressor gene 1 (LATS1), p-Yes1 associated transcriptional regulator (YAP), proliferating cell nuclear antigen (PCNA), G1/S-specific cyclin-D1 (Cyclin D1), B-cell lymphoma-2 (Bcl-2), Caspase-3, microtubule-associated proeins light chain 3 II/I (LC3-II/I), and recombinant human autophagy related 5 (ATG5). Thyroxine (T4) level was detected by enzyme-linked immunosorbent assay. RESULTS The thyroid volume of rats in the model group was significantly increased compared to the normal control group (P<0.01), and pathological changes such as uneven size of follicular epithelial cells, disorderly arrangement, and irregular morphology occurred. The secretion of small vesicles by Golgi apparatus was reduced, and the expressions of receptor protein TSH-R and T4 were significantly increased (P<0.01), while the expressions of MST1, p-LATS1, p-YAP, Caspase-3, LC3-II/I, and ATG5 were significantly decreased (P<0.01). The expressions of Bcl-2, PCNA, and cyclin D1 were significantly increased (P<0.01). Compared with the model group, RS extracts reduced the volume of thyroid gland, improved pathological condition of the thyroid gland, promoted secretion of the secretory vesicles with double-layer membrane structure in thyroid Golgi, significantly inhibited the expression of TSH-R and T4 levels (P<0.01), upregulated MST1, p-LATS1, p-YAP, Caspase-3, LC3-II/I, and ATG5 expressions (P<0.01), and downregulated Bcl-2, PCNA, and Cyclin D1 expressions (P<0.01). XMU-MP-1 inhibited the intervention effects of RS extracts (P<0.01). CONCLUSION RS extracts could inhibit proliferation and promote apoptosis and autophagy in thyroid tissues through MST1/Hippo pathway for treating hyperthyroidism.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Pharmacy, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 50001, China
| | - Tao Ye
- Department of Chinese Medicine Rehabilitation, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 50001, China
| | - Xu Lu
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Zi-Hui Li
- College of Life and Health, Dalian University, Dalian, Liaoning Province, 116622, China
| | - Ling Li
- Department of Pharmacy, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 50001, China.
| |
Collapse
|
39
|
Zhou Y, Qian C, Tang Y, Song M, Zhang T, Dong G, Zheng W, Yang C, Zhong C, Wang A, Zhao Y, Lu Y. Advance in the pharmacological effects of quercetin in modulating oxidative stress and inflammation related disorders. Phytother Res 2023; 37:4999-5016. [PMID: 37491826 DOI: 10.1002/ptr.7966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/27/2023] [Accepted: 07/07/2023] [Indexed: 07/27/2023]
Abstract
Numerous pharmacological effects of quercetin have been illustrated, including antiinflammation, antioxidation, and anticancer properties. In recent years, the antioxidant activity of quercetin has been extensively reported, in particular, its impacts on glutathione, enzyme activity, signaling transduction pathways, and reactive oxygen species (ROS). Quercetin has also been demonstrated to exert a striking antiinflammatory effect mainly by inhibiting the production of cytokines, reducing the expression of cyclooxygenase and lipoxygenase, and preserving the integrity of mast cells. By regulating oxidative stress and inflammation, which are regarded as two critical processes involved in the defense and regular physiological operation of biological systems, quercetin has been validated to be effective in treating a variety of disorders. Symptoms of these reactions have been linked to degenerative processes and metabolic disorders, including metabolic syndrome, cardiovascular, neurodegeneration, cancer, and nonalcoholic fatty liver disease. Despite that evidence demonstrates that antioxidants are employed to prevent excessive oxidative and inflammatory processes, there are still concerns regarding the expense, accessibility, and side effects of agents. Notably, natural products, especially those derived from plants, are widely accessible, affordable, and generally safe. In this review, the antioxidant and antiinflammatory abilities of the active ingredient quercetin and its application in oxidative stress-related disorders have been outlined in detail.
Collapse
Affiliation(s)
- Yueke Zhou
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Cheng Qian
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu Tang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Mengyao Song
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Teng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Guanglu Dong
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weiwei Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chunmei Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chongjin Zhong
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Aiyun Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Zhao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
40
|
Li Q, Chai Y, Li W, Guan L, Fan Y, Chen Y. Mechanism of Simiao Decoction in the treatment of atherosclerosis based on network pharmacology prediction and molecular docking. Medicine (Baltimore) 2023; 102:e35109. [PMID: 37682164 PMCID: PMC10489409 DOI: 10.1097/md.0000000000035109] [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: 03/17/2023] [Accepted: 08/16/2023] [Indexed: 09/09/2023] Open
Abstract
To explore the molecular mechanism of Simiao Decoction (SMD) intervening atherosclerosis (AS). The main components and potential mechanisms of SMD remain unknown. This study aims to initially clarify the potential mechanism of SMD in the treatment of AS based on network pharmacology and molecular docking techniques. The principal components and corresponding protein targets of SMD were searched on Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform and the compound-target network was constructed by Cytoscape3.9.1. AS targets were searched on DrugBank, OMIM, and TTD databases. The intersection of compound target and disease target was obtained and the coincidence target was imported into STRING database to construct a protein-protein interaction network. We further performed Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis on the targets. The molecular docking method was used to verify the interaction between core components of SMD and targets. We created the active compounds-targets network and the active compounds-AS-targets network based on the network database containing Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform, DrugBank, OMIM, and TTD. We discovered that the therapy of AS with SMD involves 3 key substances-quercetin, kaempferol, and luteolin-as well as 5 crucial targets-ALB, AKT1, TNF, IL6, and TP53. The Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis revealed that the shared targets involved a number of signaling pathways, including the advanced glycosylation end product-receptor of AGE signaling pathway in diabetic complications, Hepatitis B, Lipid and atherosclerosis, Chemical Carcinogenesis-Receptor Activation, and Pathways in Cancer. The molecular docking demonstrated that the binding energies of quercetin, kaempferol, and luteolin with 5 important targets were favorable. This study reveals the active ingredients and potential molecular mechanism of SMD in the treatment of AS, and provides a reference for subsequent basic research.
Collapse
Affiliation(s)
- Qian Li
- Guizhou University of Traditional Chinese Medicine, Guiyang City, China
| | - Yihui Chai
- Guizhou University of Traditional Chinese Medicine, Guiyang City, China
| | - Wen Li
- Guizhou University of Traditional Chinese Medicine, Guiyang City, China
| | - Liancheng Guan
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang City, China
| | - Yizi Fan
- Chongqing High-tech Zone People’s Hospital, Chongqing City, China
| | - Yunzhi Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang City, China
| |
Collapse
|
41
|
Ji L, Song T, Ge C, Wu Q, Ma L, Chen X, Chen T, Chen Q, Chen Z, Chen W. Identification of bioactive compounds and potential mechanisms of scutellariae radix-coptidis rhizoma in the treatment of atherosclerosis by integrating network pharmacology and experimental validation. Biomed Pharmacother 2023; 165:115210. [PMID: 37499457 DOI: 10.1016/j.biopha.2023.115210] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023] Open
Abstract
OBJECTIVE This study aims at investigating the potential targets and functional mechanisms of Scutellariae Radix-Coptidis Rhizoma (QLYD) against atherosclerosis (AS) through network pharmacology, molecular docking, bioinformatic analysis and experimental validation. METHODS The compositions of QLYD were collected from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) and literature, where the main active components of QLYD and corresponding targets were identified. The potential therapeutic targets of AS were excavated using the OMIM database, DrugBank database, DisGeNET database, CTD database and GEO datasets. The protein-protein interaction (PPI) network of common targets was constructed and visualized by Cytoscape 3.7.2 software. Gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) analysis were performed to analyze the function of core targets in the PPI network. Molecular docking was carried out using AutoDockTools, AutoDock Vina, and PyMOL software to verify the correlation between the main components of QLYD and the core targets. Mouse AS model was established and the results of network pharmacology were verified by in vivo experiments. RESULTS Totally 49 active components and 225 corresponding targets of QLYD were obtained, where 68 common targets were identified by intersecting with AS-related targets. Five hub genes including IL6, VEGFA, AKT1, TNF, and IL1B were screened from the PPI network. GO functional analysis reported that these targets had associations mainly with cellular response to oxidative stress, regulation of inflammatory response, epithelial cell apoptotic process, and blood coagulation. KEGG pathway analysis demonstrated that these targets were correlated to AGE-RAGE signaling pathway in diabetic complications, TNF signaling pathway, IL-17 signaling pathway, MAPK signaling pathway, and NF-kappa B signaling pathway. Results of molecular docking indicated good binding affinity of QLYD to FOS, AKT1, and TNF. Animal experiments showed that QLYD could inhibit inflammation, improve blood lipid levels and reduce plaque area in AS mice to prevent and treat AS. CONCLUSION QLYD may exert anti-inflammatory and anti-oxidative stress effects through multi-component, multi-target and multi-pathway to treat AS.
Collapse
Affiliation(s)
- Lingyun Ji
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - Ting Song
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250011, China
| | - Chunlei Ge
- Department of Respiratory Medicine, Linyi Tradition Chinese Medical Hospital, Linyi, Shandong Province 276600, China
| | - Qiaolan Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - Lanying Ma
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - Xiubao Chen
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250011, China
| | - Ting Chen
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - Qian Chen
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - Zetao Chen
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250011, China; Subject of Integrated Chinese and Western Medicine,Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China.
| | - Weida Chen
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250011, China.
| |
Collapse
|
42
|
Sun N, Yang N, Zhou J, He Y, Wang J, Liang Y, Dai R, Bai J, Chen Z. Yiqi Qingre Xiaozheng formula protects against diabetic nephropathy by restoring autophagy in mice. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2023; 10:310-320. [DOI: 10.1016/j.jtcms.2023.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
|
43
|
Fan W, Lan S, Yang Y, Liang J. Network pharmacology prediction and molecular docking-based strategy to discover the potential pharmacological mechanism of Huang-Qi-Gui-Zhi-Wu-Wu decoction against deep vein thrombosis. J Orthop Surg Res 2023; 18:475. [PMID: 37391801 DOI: 10.1186/s13018-023-03948-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 06/22/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND Huangqi Guizhi Wuwu decoction (HQGZWWD) has been used to treat and prevent deep vein thrombosis (DVT) in China. However, its potential mechanisms of action remain unclear. This study aimed to utilize network pharmacology and molecular docking technology to elucidate the molecular mechanisms of action of HQGZWWD in DVT. METHODS We identified the main chemical components of HQGZWWD by reviewing the literature and using a Traditional Chinese Medicine Systems Pharmacology (TCMSP) database. We used GeneCards and Online Mendelian Inheritance in Man databases to identify the targets of DVT. Herb-disease-gene-target networks using Cytascape 3.8.2 software; a protein-protein interaction (PPI) network was constructed by combining drug and disease targets on the STRING platform. Additionally, we conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. Finally, molecular docking verification of active components and core protein targets was conducted. RESULTS A total of 64 potential targets related to DVT were identified in HQGZWWD, with 41 active components; quercetin, kaempferol, and beta-sitosterol were the most effective compounds. The PPI network analysis revealed that AKT1, IL1B, and IL6 were the most abundant proteins with the highest degree. GO analysis indicated that DVT treatment with HQGZWWD could involve the response to inorganic substances, positive regulation of phosphorylation, plasma membrane protein complexes, and signaling receptor regulator activity. KEGG analysis revealed that the signaling pathways included pathways in cancer, lipid and atherosclerosis, fluid shear stress and atherosclerosis, and the phosphatidylinositol 3-kinases/protein kinase B(PI3K-Akt) and mitogen-activated protein kinase (MAPK) signaling pathways. The molecular docking results indicated that quercetin, kaempferol, and beta-sitosterol exhibited strong binding affinities for AKT1, IL1B, and IL6. CONCLUSION Our study suggests that AKT1, IL1B, and IL6 are promising targets for treating DVT with HQGZWWD. The active components of HQGZWWD likely responsible for its effectiveness against DVT are quercetin, kaempferol, and beta-sitosterol, they may inhibit platelet activation and endothelial cell apoptosis by regulating the PI3K/Akt and MAPK signaling pathways, slowing the progression of DVT.
Collapse
Affiliation(s)
- Wei Fan
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Provincial Laboratory of Orthopaedic Engineering, Luzhou, China
| | - Shuangli Lan
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yunkang Yang
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
- Sichuan Provincial Laboratory of Orthopaedic Engineering, Luzhou, China.
| | - Jie Liang
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
- Sichuan Provincial Laboratory of Orthopaedic Engineering, Luzhou, China.
| |
Collapse
|
44
|
Wu X, Pan J, Yu JJ, Kang J, Hou S, Cheng M, Xu L, Gong L, Li Y. DiDang decoction improves mitochondrial function and lipid metabolism via the HIF-1 signaling pathway to treat atherosclerosis and hyperlipidemia. JOURNAL OF ETHNOPHARMACOLOGY 2023; 308:116289. [PMID: 36822344 DOI: 10.1016/j.jep.2023.116289] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/28/2023] [Accepted: 02/15/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE DiDang Decoction (DDD) is a traditional classical prescription that has been used to treat atherosclerosis (AS) and hyperlipidemia (HLP) in China. Nevertheless, the underlying mechanism of DDD remains unclear. AIM OF THE STUDY To validate the mechanism of DDD in AS and HLP based on network pharmacology and in vitro experiments. MATERIALS AND METHODS The chemical components of DDD were obtained from the Traditional Chinese Medicine System Pharmacology Database and Analysis Platform (TCMSP) database and literature mining, and the disease targets of AS and HLP were obtained from the Gencards, OMIM, and DisGeNET databases. The intersection genes were imported into the STRING database to construct protein-protein interaction (PPI) network, and the DAVID database was used for gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Combined with the results of KEGG pathway analysis, the HIF-1 signaling pathway was selected for further in vitro experiments. RESULTS The results showed that network pharmacology predicted 112 targets related to DDD treatment of AS and HLP, and the top 10 related pathways are: Lipid and atherosclerosis, AGE-RAGE signaling pathway in diabetic complications, Chemical carcinogenesis - receptor activation, Pathways in cancer, Proteoglycans in cancer, Fluid shear stress and atherosclerosis, HIF-1 signaling pathway, Alcoholic liver disease, PPAR signaling pathway, and Coronavirus disease-COVID-19. In vitro experiments showed that DDD effectively reduced lipid accumulation in FFA-treated L02 cells; DDD attenuated mitochondrial damage and reduced ROS content; DDD inhibited ferroptosis and apoptosis; DDD up-regulated the expression of HIF-1α, Glutathione Peroxidase 4(GPX4), and Bcl2 proteins, and down-regulated expression of Bax protein. CONCLUSION DDD exerts therapeutic effects on AS and HLP through multiple targets and pathways, and improves mitochondrial function, reduces ROS content, inhibits ferroptosis and apoptosis by activating the HIF-1 signaling pathway, which provides reliable theoretical and experimental support for DDD treatment of AS and HLP.
Collapse
Affiliation(s)
- Xize Wu
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110032, China.
| | - Jiaxiang Pan
- The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110032, China.
| | - Jj Jiajia Yu
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110032, China.
| | - Jian Kang
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110032, China.
| | - Siyi Hou
- The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110032, China; Liaoning Provincial Key Laboratory of TCM Geriatric Cardio-Cerebrovascular Diseases, Shenyang, 110032, China.
| | - Meijia Cheng
- The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110032, China.
| | - Lili Xu
- Department of Cardiology, 924 Hospital of Joint Logistic Support Force of PLA, Guilin, 541002, China.
| | - Lihong Gong
- The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110032, China; Liaoning Provincial Key Laboratory of TCM Geriatric Cardio-Cerebrovascular Diseases, Shenyang, 110032, China.
| | - Yue Li
- The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110032, China; Liaoning Provincial Key Laboratory of TCM Geriatric Cardio-Cerebrovascular Diseases, Shenyang, 110032, China.
| |
Collapse
|
45
|
Xuan X, Zhang J, Fan J, Zhang S. Research progress of Traditional Chinese Medicine (TCM) in targeting inflammation and lipid metabolism disorder for arteriosclerosis intervention: A review. Medicine (Baltimore) 2023; 102:e33748. [PMID: 37144986 PMCID: PMC10158879 DOI: 10.1097/md.0000000000033748] [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: 03/23/2023] [Accepted: 04/21/2023] [Indexed: 05/06/2023] Open
Abstract
Atherosclerosis (AS) is a chronic disease caused by inflammation and lipid deposition. Immune cells are extensively activated in the lesions, producing excessive pro-inflammatory cytokines, which accompany the entire pathological process of AS. In addition, the accumulation of lipid-mediated lipoproteins under the arterial intima is a crucial event in the development of AS, leading to vascular inflammation. Improving lipid metabolism disorders and inhibiting inflammatory reactions are the primary treatment methods currently used in medical practice to delay AS progression. With the development of traditional Chinese medicine (TCM), more mechanisms of action of the monomer of TCM, Chinese patent medicine, and compound prescription have been studied and explored. Research has shown that some Chinese medicines can participate in treating AS by targeting and improving lipid metabolism disorders and inhibiting inflammatory reactions. This review explores the research on Chinese herbal monomers, compound Chinese medicines, and formulae that improve lipid metabolism disorders and inhibit inflammatory reactions to provide new supplements for treating AS.
Collapse
Affiliation(s)
- Xiaoyu Xuan
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jingyi Zhang
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jilin Fan
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shiliang Zhang
- Department of Cardiology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
46
|
Yin Y, Tan M, Han L, Zhang L, Zhang Y, Zhang J, Pan W, Bai J, Jiang T, Li H. The hippo kinases MST1/2 in cardiovascular and metabolic diseases: A promising therapeutic target option for pharmacotherapy. Acta Pharm Sin B 2023; 13:1956-1975. [PMID: 37250161 PMCID: PMC10213817 DOI: 10.1016/j.apsb.2023.01.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/09/2022] [Accepted: 11/18/2022] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular diseases (CVDs) and metabolic disorders are major components of noncommunicable diseases, causing an enormous health and economic burden worldwide. There are common risk factors and developmental mechanisms among them, indicating the far-reaching significance in exploring the corresponding therapeutic targets. MST1/2 kinases are well-established proapoptotic effectors that also bidirectionally regulate autophagic activity. Recent studies have demonstrated that MST1/2 influence the outcome of cardiovascular and metabolic diseases by regulating immune inflammation. In addition, drug development against them is in full swing. In this review, we mainly describe the roles and mechanisms of MST1/2 in apoptosis and autophagy in cardiovascular and metabolic events as well as emphasis on the existing evidence for their involvement in immune inflammation. Moreover, we summarize the latest progress of pharmacotherapy targeting MST1/2 and propose a new mode of drug combination therapy, which may be beneficial to seek more effective strategies to prevent and treat CVDs and metabolic disorders.
Collapse
Affiliation(s)
- Yunfei Yin
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Mingyue Tan
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Lianhua Han
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Lei Zhang
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yue Zhang
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jun Zhang
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Wanqian Pan
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jiaxiang Bai
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Department of Orthopedics, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Tingbo Jiang
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Hongxia Li
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
47
|
Sang M, Huang Y, Wang L, Chen L, Nawsherwan, Li G, Wang Y, Yu X, Dai C, Zheng J. An "AND" Molecular Logic Gate as a Super-Enhancers for De Novo Designing Activatable Probe and Its Application in Atherosclerosis Imaging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207066. [PMID: 36808894 PMCID: PMC10131802 DOI: 10.1002/advs.202207066] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/31/2023] [Indexed: 09/30/2023]
Abstract
Developing activatable fluorescent probes with superlative fluorescence enhancement factor (F/F0 ) to improve the signal-to-noise (S/N) ratio is still an urgent issue. "AND" molecular logic gates are emerging as a useful tool for enhanced probes selectivity and accuracy. Here, an "AND" logic gate is developed as super-enhancers for designing activatable probes with huge F/F0 and S/N ratio. It utilizes lipid-droplets (LDs) as controllable background input and sets the target analyte as variable input. The fluorescence is tremendously quenching due to double locking, thus an extreme F/F0 ratio of target analyte is obtained. Importantly, this probe can transfer to LDs after a response occurs. The target analyte can be directly visualized through the spatial location without a control group. Accordingly, a peroxynitrite (ONOO- ) activatable probe (CNP2-B) is de novo designed. The F/F0 of CNP2-B achieves 2600 after reacting with ONOO- . Furthermore, CNP2-B can transfer from mitochondria to lipid droplets after being activated. The higher selectivity and S/N ratio of CNP2-B are obtained than commercial probe 3'-(p-hydroxyphenyl) fluorescein (HPFin vitro and in vivo. Therefore, the atherosclerotic plaques at mouse models are delineated clearly after administration with in situ CNP2-B probe gel. Such input controllable "AND" logic gate is envisioned to execute more imaging tasks.
Collapse
Affiliation(s)
- Mangmang Sang
- Institute of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityNo. 2999 Jinshan Road, Huli DistrictXiamen361006China
| | - Yibo Huang
- Institute of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityNo. 2999 Jinshan Road, Huli DistrictXiamen361006China
| | - Lu Wang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese MedicineNanjing University of Chinese MedicineNo. 157, Daming Road, Qinhuai DistrictNanjing210000China
| | - Lei Chen
- School of PharmacyGannan Medical UniversityNo. 1 Medical College Road, Zhanggong DistrictGanzhou341000China
| | - Nawsherwan
- Institute of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityNo. 2999 Jinshan Road, Huli DistrictXiamen361006China
| | - Gang Li
- Institute of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityNo. 2999 Jinshan Road, Huli DistrictXiamen361006China
| | - Yan Wang
- Institute of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityNo. 2999 Jinshan Road, Huli DistrictXiamen361006China
| | - Xiu Yu
- Shenzhen Key Laboratory of Respiratory DiseasesShenzhen People's HospitalSouthern University of Science and Technology3046 Shennan East Road, Luohu DistrictShenzhen518055China
| | - Cuilian Dai
- Institute of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityNo. 2999 Jinshan Road, Huli DistrictXiamen361006China
| | - Jinrong Zheng
- Institute of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityNo. 2999 Jinshan Road, Huli DistrictXiamen361006China
| |
Collapse
|
48
|
Shao Y, Wang Y, Sun L, Zhou S, Xu J, Xing D. MST1: A future novel target for cardiac diseases. Int J Biol Macromol 2023; 239:124296. [PMID: 37011743 DOI: 10.1016/j.ijbiomac.2023.124296] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023]
Abstract
Major heart diseases pose a serious threat to human health. Finding early diagnostic markers and key therapeutic targets is an urgent scientific problem in this field. Mammalian sterile 20-like kinase 1 (MST1) is a protein kinase, and the occurrence of many heart diseases is related to the continuous activation of the MST1 gene. With the deepening of the research, the potential role of MST1 in promoting the development of heart disease has become more apparent. Therefore, to better understand the role of MST1 in the pathogenesis of heart disease, this work systematically summarizes the role of MST1 in the pathogenesis of heart disease, gives a comprehensive overview of its possible strategies in the diagnosis and treatment of heart disease, and analyzes its potential significance as a marker for the diagnosis and treatment of heart disease.
Collapse
Affiliation(s)
- Yingchun Shao
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Yanhong Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Li Sun
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Sha Zhou
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Jiazhen Xu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
49
|
Quercetin alleviates atherosclerosis by suppressing oxidized LDL-induced senescence in plaque macrophage via inhibiting the p38MAPK/p16 pathway. J Nutr Biochem 2023; 116:109314. [PMID: 36924853 DOI: 10.1016/j.jnutbio.2023.109314] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 12/10/2022] [Accepted: 02/28/2023] [Indexed: 03/15/2023]
Abstract
Quercetin is a widely known and biologically active phytochemical and exerts therapeutic effects against atherosclerosis. The removal of senescent plaque macrophages effectively slows the progression of atherosclerosis and decreases the plaque burden. Still, whether quercetin alleviates atherosclerosis by inhibiting the senescence of plaque macrophages, including the potential mechanisms, remains unclear. ApoE-/- mice were fed with a normal chow diet or high-fat diet (HFD) supplemented or not with quercetin (100 mg/kg of body weight) for 16 weeks. An accumulation of senescent macrophages was observed in the plaque-rich aortic tissues from the mice with HFD, but quercetin supplementation effectively reduced the amount of senescent plaque macrophage, inhibited the secretion of key senescence-associated secretory phenotype (SASP) factors, and alleviated atherosclerosis by inhibiting p38MAPK phosphorylation and p16 expression. In vitro, SB203580 (a specific inhibitor of p38 MAPK) significantly inhibited oxidized low-density lipoprotein (ox-LDL)-induced senescence in mouse RAW264.7 macrophages, as evidenced by decreased senescence-associated markers (SA-β-gal staining positive cells and p16 expression). Furthermore, quercetin not only effectively reversed ox-LDL-induced senescence in RAW264.7 cells but also decreased the mRNA levels of several key SASP factors by suppressing p38 MAPK phosphorylation and p16 expression. The p38 MAPK agonist asiatic acid reversed the effects of quercetin. In conclusion, these findings indicate that quercetin suppresses ox-LDL-induced senescence in plaque macrophage and attenuates atherosclerosis by inhibiting the p38 MAPK/p16 pathway. This study elucidates the mechanisms of quercetin against atherosclerosis and supports quercetin as a nutraceutical for the management of atherosclerosis.
Collapse
|
50
|
Liu H, He H, Tian Y, Cui J, Wang S, Wang H. Cyclophilin A accelerates SiO 2-induced macrophage foaming. Cell Signal 2023; 103:110562. [PMID: 36535629 DOI: 10.1016/j.cellsig.2022.110562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/06/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Silicosis is a common occupational disease characterized by lung inflammation, fibrosis and pulmonary dysfunction caused by long-term inhalation of free SiO2. Cell foaming and the change of CyPA have been observed in SiO2-induced macrophages, but the specific mechanism of CyPA in SiO2-induced foam cells remains poorly understood. The purpose of this study is to explore the mechanism of CyPA in SiO2-induced macrophage foaming and its effect on silicosis. We found that overexpression of CyPA promoted the macrophage foaming and the expression of COL I and α-SMA, while silencing CyPA inhibites the macrophage foaming and the expression of COL I and α-SMA. After blocking the expression of CD36 on the basis of overexpression CyPA, we found it inhibites the macrophage foaming. In conclusion, CyPA can affect the foaming of macrophages and may participate in silicosis fibrosis.
Collapse
Affiliation(s)
- Heliang Liu
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China; Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Hailan He
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Ying Tian
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Jie Cui
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Shuang Wang
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Hongli Wang
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China.
| |
Collapse
|