1
|
Al Fahad MA, Lee HY, Park M, Lee BT. A cardiac extracellular matrix-based bilayer vascular graft with controlled microstructures for the reconstruction of small-diameter blood vessels. Biomaterials 2025; 320:123264. [PMID: 40121829 DOI: 10.1016/j.biomaterials.2025.123264] [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: 12/09/2024] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Despite recent progress, challenges with small-diameter vascular grafts, including mechanical strength, intimal hyperplasia, thrombosis, and poor endothelialization, remain unresolved. The present study reports a novel bilayer vascular graft designed to mimic the anatomical features of small-diameter blood vessels. The electrospun graft consists of a dense micro/nanofibrous inner layer of cardiac extracellular matrix (cECM), polycaprolactone (PCL) loaded with heparin (P-cECM-H), and a super porous and micro-fibrous PCL outer layer. Liquid chromatography-mass spectrometry (LC-MS/MS) proteome analysis of the cECM revealed that it is enriched with several bioactive proteins related to angiogenesis, wound regeneration, cell migration, etc. The porosities of the two layers are tailored according to endothelial and smooth muscle cell biology. The graft exhibited excellent mechanical properties, and the heparinized P-cECM inner layer improved hemocompatibility and anticoagulation efficacy. A significant increase in endothelial cell proliferation was noted in the P-cECM-H group after 7 days compared with the control group (p < 0.05). The bilayer graft maintained 100 % patency after 10 weeks of rat abdominal aorta implantation. Histological evaluation revealed smooth muscle cell infiltration inside the highly porous outer layer and neointima regeneration in the inner layer with a complete endothelial lining. RNA sequencing (RNA-Seq) analysis further confirmed smooth muscle formation and endothelial layer formation. The gene expression data also suggested that the hypoxia-inducible factor-1 (HIF-) and vascular endothelial growth factor (VEGF) signaling pathways are involved in endothelial layer remodeling. These promising results indicate that cECM could be a key material for vascular tissue regeneration.
Collapse
Affiliation(s)
- Md Abdullah Al Fahad
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Hyun-Yong Lee
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan, 31151, Republic of Korea
| | - Myeongki Park
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Byong-Taek Lee
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea; Institute of Tissue Regeneration, Soonchunhyang University, Cheonan, 31151, Republic of Korea.
| |
Collapse
|
2
|
Zheng Q, Lin Y, Zeng L, Chen S, Chen L, Lin X, Zhu J, Lin J, Weng X, Chai D. ITE-mediated AhR activation attenuates atherosclerosis by promoting macrophage M2-like polarization through NF-κB/LCN2 pathway suppression. Life Sci 2025; 375:123715. [PMID: 40389023 DOI: 10.1016/j.lfs.2025.123715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 05/04/2025] [Accepted: 05/10/2025] [Indexed: 05/21/2025]
Abstract
AIMS Atherosclerosis (AS) is a chronic inflammatory disease characterized by lipid accumulation and inflammation. Macrophage phenotypic transformation plays a critical role in AS progression. Aryl hydrocarbon receptor (AhR) has been proved to regulate the phenotype of macrophages. This study investigates the role and molecular mechanism of AhR activation by its endogenous ligand, 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE) attenuates AS. MATERIALS AND METHODS We employed Western blotting to analyze the expression of AhR, NF-κB, and lipocalin-2 (LCN2). Flow cytometry and immunofluorescence staining were used to assess the phenotype of macrophages. Plaque progression was evaluated using pathological staining. Transcriptome sequencing was utilized to explore the potential mechanism by which AhR promotes macrophage phenotypic transformation. CUT&Tag-qPCR and lentivirus infection confirmed that the AhR/NF-κB/LCN2 pathway regulates macrophage polarization. KEY FINDINGS Activation of AhR by ITE reduced plaque area and inhibited lipid deposition. ITE significantly increased the number of M2-like macrophages both in vivo and in vitro. Transcriptome sequencing identified LCN2 as a key target for AhR-mediated macrophage M2-like polarization. Furthermore, AhR activation suppressed the NF-κB/LCN2 pathway. SIGNIFICANCE Our findings reveal that AhR promotes the macrophages to exhibit M2-like characteristics to attenuate AS by inhibiting the NF-κB/LCN2 pathway. These results suggest that AhR may serve as a novel therapeutic target for AS.
Collapse
Affiliation(s)
- Qiaowen Zheng
- Cardiovascular Department, Fuqing City Hospital, Fuzhou 350005, China
| | - Yifei Lin
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Key Laboratory of Metabolic Heart Disease in Fujian Province, Clinical Research Centre of Metabolic Cardiovascular Disease in Fujian Province, Fuzhou 350005, China; Cardiovascular Department, National Regional Medical Center, Binhai Branch of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Lishan Zeng
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Key Laboratory of Metabolic Heart Disease in Fujian Province, Clinical Research Centre of Metabolic Cardiovascular Disease in Fujian Province, Fuzhou 350005, China; Cardiovascular Department, National Regional Medical Center, Binhai Branch of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Shuaijie Chen
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Key Laboratory of Metabolic Heart Disease in Fujian Province, Clinical Research Centre of Metabolic Cardiovascular Disease in Fujian Province, Fuzhou 350005, China; Cardiovascular Department, National Regional Medical Center, Binhai Branch of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Longqing Chen
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Key Laboratory of Metabolic Heart Disease in Fujian Province, Clinical Research Centre of Metabolic Cardiovascular Disease in Fujian Province, Fuzhou 350005, China; Cardiovascular Department, National Regional Medical Center, Binhai Branch of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Xiaoyan Lin
- Echocardiological Department, the First Affiliated Hospital, Fujian Medical University, Fujian Institute of Hypertension, Fuzhou 350005, China
| | - Jiang Zhu
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Key Laboratory of Metabolic Heart Disease in Fujian Province, Clinical Research Centre of Metabolic Cardiovascular Disease in Fujian Province, Fuzhou 350005, China; Cardiovascular Department, National Regional Medical Center, Binhai Branch of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Jinxiu Lin
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Key Laboratory of Metabolic Heart Disease in Fujian Province, Clinical Research Centre of Metabolic Cardiovascular Disease in Fujian Province, Fuzhou 350005, China; Cardiovascular Department, National Regional Medical Center, Binhai Branch of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Xiuzhu Weng
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Key Laboratory of Metabolic Heart Disease in Fujian Province, Clinical Research Centre of Metabolic Cardiovascular Disease in Fujian Province, Fuzhou 350005, China; Cardiovascular Department, National Regional Medical Center, Binhai Branch of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| | - Dajun Chai
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Key Laboratory of Metabolic Heart Disease in Fujian Province, Clinical Research Centre of Metabolic Cardiovascular Disease in Fujian Province, Fuzhou 350005, China; Cardiovascular Department, National Regional Medical Center, Binhai Branch of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| |
Collapse
|
3
|
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] [MESH Headings] [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
|
4
|
Li X, Huang X, Song K, Liu J, Jin Y, Li T, Zhang L, Zhang H. Qingre Sanjie Formula alleviates atherosclerosis by promoting LXR-α/ABCG5/G8-mediated reverse cholesterol transport and bile acid synthesis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156691. [PMID: 40286749 DOI: 10.1016/j.phymed.2025.156691] [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: 01/28/2025] [Revised: 03/11/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Atherosclerosis is the leading cause of cardiovascular disease-related morbidity and mortality. The traditional Chinese medicine Qingre Sanjie Formula (QRSJF), composed of Prunellae Spica, Sargassum, Fritillariae Thunbergii Bulbus, Leonuri Herba, and Forsythiae Fructus, has shown efficacy in treating cardiovascular diseases, although its mechanisms are unclear. PURPOSE This study aimed to explore the protective effects of QRSJF against atherosclerosis and the mechanisms involved. METHODS The composition of QRSJF was analyzed using Ultra Performance Liquid Chromatography Quadrupole Time-of-Flight Mass Spectrometry. An 8-week high-fat diet (HFD)-induced atherosclerosis model was established in ApoE-/- mice. Following model induction, mice received 12 weeks of QRSJF treatment at high- and low doses (3.16 and 1.58 g drug/kg/day, respectively) via oral gavage, while simvastatin (2.6 mg/kg/day) as the positive control. Various techniques, including biochemical assays, vascular ultrasonography, histopathology, untargeted metabolomics, and molecular biology techniques were utilized to evaluate therapeutic effects. The underlying mechanism was investigated in vitro using free fatty acids -induced HepG2 cells. RESULTS Both low- and high-dose QRSJF effectively attenuated dyslipidemia and decreased serum inflammatory cytokine levels in HFD-fed ApoE-/- mice. In addition, QRSJF alleviated atherosclerotic plaque formation, reduced arterial narrowing, and enhanced plaque stability. Plasma and liver metabolomic analyses further identified that ABC (ATP binding cassette) subfamily transporters and bile acid metabolism as key pathways through which QRSJF ameliorates atherosclerosis. QRSJF also alleviated liver lipid accumulation and increased the expression of liver proteins, including scavenger receptor class B type 1, low-density lipoprotein receptor, ABC subfamily A member 1, cholesterol 7α-hydroxylase (CYP7A1), ABC transporter G5/G8 (ABCG5/G8), bile salt output pump, and liver X receptor alpha (LXR-α). In vitro, QRSJF activated LXR-α expression in HepG2 cells, thereby enhancing the expression of the downstream targets, CYP7A1 and ABCG5/8, and reducing free fatty acid-induced lipid accumulation. Notably, the beneficial effects of QRSJF were abrogated by the LXR-α inhibitor GSK2033. CONCLUSION QRSJF improves dyslipidemia and reduces atherosclerotic plaque in ApoE-/- mice by activating the LXR-α/ABCG5/G8 pathway. This facilitates cholesterol transport to the liver and promotes bile acid synthesis and cholesterol excretion into the bile and intestine, thereby exerting anti-atherosclerotic effects.
Collapse
Affiliation(s)
- Xiao Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xianglong Huang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Keyan Song
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jinjie Liu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ya Jin
- Department of Pharmacology, Xinjiang Second Medical College, Karamay 834000, China
| | - Tianxiang Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lishuang Zhang
- Tianjin Binhai New Area Hospital of Traditional Chinese Medicine, Fourth Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300450, China
| | - Han Zhang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
5
|
Bi A, Liu R, Xie M, He B, Yan T, Du Y, Jia Y. Semen Ziziphi Spinosae alleviates cardiomyocyte apoptosis in rats with coronary heart disease via the AMPK/SIRT1/PGC-1α signaling pathway activation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156743. [PMID: 40250033 DOI: 10.1016/j.phymed.2025.156743] [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: 02/01/2025] [Revised: 03/23/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Coronary heart disease (CHD) represents a significant cardiovascular condition, with its occurrence increasing as a result of alterations in lifestyle and dietary habits. Semen Ziziphi Spinosae (SZS) is commonly utilized for the management of disorders associated with the nervous system, including conditions like depression and insomnia. Recent research has revealed its potential therapeutic properties for cardiovascular issues. Nevertheless, there exists a limited amount of research addressing the mechanisms involved. PURPOSE This research seeks to explore the protective effects that SZS has on cardiac tissue, specifically within the framework of CHD. By conducting this investigation, the study aims to uncover the various mechanisms that play a role in these protective effects. This understanding could yield significant insights into how SZS may result in the preservation and enhancement of cardiac health in patients affected by CHD. STUDY DESIGN The study innovatively combines multiple advanced techniques. It first integrates UPLC-Q-TOF/MS analysis and network pharmacology to identify SZS components. In vitro experiments were conducted using H9c2 rat cardiomyocytes, and in vivo experiments used a CHD model in SD rats. Multiple assays were performed for multi - level and multi - dimensional validation. METHODS In the initial stage, the primary components of SZS and their possible mechanisms for combating CHD were examined through UPLC-Q-TOF/MS analysis in conjunction with network pharmacology approaches. For the in vitro investigation, an ischemia-hypoxia model was established utilizing H9c2 rat cardiomyocytes. The CCK-8 assay was used to assess myocardial injury markers. TUNEL staining and Western blot techniques were employed to confirm the impact of SZS treatment on apoptosis in H9c2 cells. The expression levels of proteins associated with the AMPK/SIRT1/PGC-1α signaling pathway were measured using RT-qPCR and Western blotting, and the results were validated with the AMPK inhibitor, compound C. In the in vivo segment, a model of coronary heart disease (CHD) in SD rats was established through the administration of a high-fat emulsion diet combined with pituitrin injections. Cardiac function in the rats was evaluated through electrocardiograms and echocardiograms. Pathological changes in the heart were observed utilizing TTC and H&E staining. Kits were implemented to measure the serum biochemical indicators in the rats.RT - qPCR and Western blotting were employed to measure the expression levels of proteins related to the AMPK/SIRT1/PGC - 1α signaling pathway. RESULTS The study identified 67 in vitro components, 27 blood - absorbed components, and 12 metabolic components of SZS. Network pharmacology analysis suggested the AMPK/SIRT1/PGC - 1α signaling pathway as a key mechanism. In vitro and in vivo experiments showed that SZS increased cell viability, reduced apoptosis, and activated the AMPK/SIRT1/PGC - 1α signaling pathway. Inhibiting AMPK abolished SZS's effects. SZS also improved cardiac function and reduced myocardial damage in rats with CHD. CONCLUSION This study for the first time highlights that Semen Ziziphi Spinosae plays a beneficial role in cardiovascular health by activating the AMPK/SIRT1/PGC-1α signaling pathway and reducing apoptosis in cardiomyocytes. These findings support its potential application in the treatment of CHD and other cardiac conditions.
Collapse
Affiliation(s)
- Anqi Bi
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Rihong Liu
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Min Xie
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Bosai He
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Tingxu Yan
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Yiyang Du
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China.
| | - Ying Jia
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China.
| |
Collapse
|
6
|
Qian Z, Huang Y, Yang N, Fang Z, Zhang Y, Huang Y, Luo M, Ji T, Chen Z, Gao S, Li Y, Yan J, Jiang D, Ruan L, Liu A, Zhang C, Zhang L. miR-34a-5p/MARCHF8/ADAM10 axis in the regulation of vascular endothelial cell dysfunction and senescence. Mech Ageing Dev 2025; 225:112060. [PMID: 40222711 DOI: 10.1016/j.mad.2025.112060] [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: 01/25/2025] [Revised: 04/06/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
Vascular aging is a key driver of age-related cardiovascular and metabolic diseases, with endothelial dysfunction and senescence as a central mechanism. In our recent study, we observed elevated ADAM10 protein levels in senescent endothelial cells, which worsened endothelial dysfunction and senescence. However, the regulatory mechanisms controlling ADAM10 expression are poorly understood. In this study, we show that ADAM10 undergoes post-transcriptional modification in senescent human umbilical vein endothelial cells (HUVECs), with the E3 ubiquitin ligase MARCHF8 predicted to facilitate its ubiquitination-dependent degradation. We also found that MARCHF8 expression was significantly reduced in senescent HUVECs. Knockdown of MARCHF8 in young HUVECs induced endothelial senescence and impaired key endothelial functions, including migration, proliferation, angiogenesis, and nitric oxide production. Conversely, overexpression of MARCHF8 in senescent HUVECs ameliorated senescence-associated dysfunctions. RNA sequencing analysis revealed that MARCHF8 knockdown disrupted pathways linked to cell senescence and atherosclerosis. In vivo, MARCHF8 overexpression in high-fat diet-fed apoE-/- mice reduced plasma interleukin-6 levels and attenuated atherosclerosis progression. Additionally, miR-34a-5p upregulation in senescence inhibited MARCHF8 expression, compromising its protective effects in delaying endothelial senescence. Collectively, these findings reveal a novel miR-34a-5p/MARCHF8/ADAM10 axis in vascular endothelial senescence, positioning MARCHF8 as a potential biomarker and therapeutic target for vascular aging and related diseases.
Collapse
Affiliation(s)
- Zonghao Qian
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Yuzhen Huang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Ni Yang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Ziwei Fang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Yucong Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Yi Huang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Mandi Luo
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Tianyi Ji
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Zuoguan Chen
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Beijing 100730, China
| | - Shang Gao
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Beijing 100730, China
| | - Yongjun Li
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Beijing 100730, China
| | - Jinhua Yan
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Dingsheng Jiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Lei Ruan
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Anding Liu
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan 430100, China
| | - Cuntai Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China.
| | - Le Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China.
| |
Collapse
|
7
|
Okunlola FO, Okunlola AR, Adetuyi BO, Soliman MES, Alexiou A, Papadakis M, Fawzy MN, El-Saber Batiha G. Beyond the gut: Unraveling the multifaceted influence of microbiome on cardiovascular health. Clin Nutr ESPEN 2025; 67:71-89. [PMID: 40064239 DOI: 10.1016/j.clnesp.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
Cardiovascular disease is one of the leading causes of death worldwide. Even while receiving adequate pharmacological treatment for their hypertension, people are nonetheless at greater risk for cardiovascular disease. There is growing evidence that the gut microbiota may have major positive and negative effects on blood pressure and illnesses related with it as more study into this topic is conducted. Trimethylamine n-oxide (TMAO) and short-chain fatty acids (SCFA) are two major by-products of the gut microbiota. TMAO is involved in the formation of other coronary artery diseases, including atherosclerosis and hypertension, while SCFAs play an important role in controlling blood pressure. Numerous investigations have confirmed the established link between dietary salt intake and hypertension. Reducing sodium in the diet is linked to lower rates of cardiovascular disease morbidity and mortality as well as lower rates of blood pressure and hypertension. In both human and animal research, high salt diets increase local and systemic tissue inflammation and compromise gut architecture. Given that the gut microbiota constantly interacts with the immune system and is required for the correct maturation of immune cells, it is scientifically conceivable that it mediates the inflammatory response. This review highlights the therapeutic possibilities for focusing on intestinal microbiomes as well as the potential functions of the gut microbiota and its metabolites in the development of hypertension.
Collapse
Affiliation(s)
- Felix Oladele Okunlola
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Abimbola Rafiat Okunlola
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Babatunde Oluwafemi Adetuyi
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Mahmoud E S Soliman
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India; Department of Research & Development, Funogen, Athens, 11741, Greece.
| | - Marios Papadakis
- University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Mohamed N Fawzy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish, 45511, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira, 22511, Egypt.
| |
Collapse
|
8
|
Zhuang C, Cui F, Chen J, He D, Sun T, Wang P. Rbm39 ameliorates metabolic dysfunction-associated steatotic liver disease by regulating Apob and Fabp4. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167815. [PMID: 40147697 DOI: 10.1016/j.bbadis.2025.167815] [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: 11/29/2024] [Revised: 03/12/2025] [Accepted: 03/22/2025] [Indexed: 03/29/2025]
Abstract
Excessive hepatic lipid accumulation is the hallmark of metabolic dysfunction-associated steatotic liver disease (MASLD), yet its underlying mechanisms still not fully understood. In this study, we identified RNA binding motif protein 39 (Rbm39) as a key modulator of hepatic lipid homeostasis during MASLD progression. To establish in vivo MASLD model, mice were fed either a high-fat diet (HFD) or a Gubra-Amylin NASH (GAN) diet. We employed adeno-associated virus to manipulate Rbm39 expression levels to assess its role in MASLD. Transcriptome analysis was conducted to pinpoint the genes targeted by Rbm39. Western blot, RT-PCR, dual-luciferase reporter gene assays, and alternative splicing analysis were utilized to delve into the molecular mechanisms. Our results showed that Rbm39 expression was notably decreased in the livers of MASLD mice. Knockdown of hepatic Rbm39 aggravated HFD-induced hepatic steatosis and GAN diet-induced MASH, along with a notable decrease in serum lipid levels. Conversely, overexpression of Rbm39 attenuated MASLD development and progression. RNA sequencing data analysis indicated that Rbm39 regulated the expression of apolipoprotein B (Apob) and fatty acid-binding protein 4 (Fabp4), both of which are crucial for lipid transport. Mechanistically, Rbm39 enhanced the transcription of Apob by upregulating hepatocyte nuclear factor 4α (Hnf4α), while it suppressed Fabp4 transcription by regulating alternative splicing of hypoxia inducible factor-1α (Hif-1α). These findings highlight the pivotal role of Rbm39 in maintaining hepatic lipid homeostasis and suggest its potential as a therapeutic target for MASLD.
Collapse
Affiliation(s)
- Chunbo Zhuang
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Fangfang Cui
- Department of Gastroenterology, Kaifeng People's Hospital, Kaifeng, Henan 475000, PR China
| | - Jin Chen
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Dezhi He
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Ting Sun
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Pei Wang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China.
| |
Collapse
|
9
|
Cui J, Zhang Y, Zhang W, Li D, Hong Z, Zhao L, Sun J, Chen Y, Zhang N. Research Hotspots and Development Trends on Apolipoprotein B in the Field of Atherosclerosis: A Bibliometric Analysis. Mol Biotechnol 2025; 67:2204-2222. [PMID: 38963531 DOI: 10.1007/s12033-024-01218-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/15/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Cardiovascular diseases caused by atherosclerosis (AS) are the leading causes of disability and death worldwide. Apolipoprotein B (ApoB), the core protein of low-density lipoproteins, is a major contributor to cardiovascular disease-related morbidity and mortality, with apolipoprotein B (ApoB) playing a critical role in its pathogenesis. However, no bibliometric studies on the involvement of ApoB in AS have been published. This study aimed to conduct a comprehensive bibliometric analysis to explore the current and future trends regarding the role of ApoB in AS. METHODS Utilizing the Web of Science Core Collection, a thorough search was conducted for ApoB in AS-related papers related to research on ApoB in the field of AS during 1991-2023. The analysis focused on annual publication trends, leading countries/regions and institutions, influential authors, journal and key journals. CiteSpace and VOSviewer were employed to visualize reference co-citations, and keyword co-occurrences, offering insights into the research landscape and emerging trends. RESULTS This bibliometric analysis employed network diagrams for cluster analysis of a total of 2105 articles and reviews, evidencing a discernible upward trend in annual publication volume. This corpus of research emanates from 76 countries/regions and 2343 organizations, illustrating the widespread international engagement in ApoB-related AS studies. Notably, the United States and the University of California emerge as the most prolific contributors, which underscores their pivotal roles in advancing this research domain. The thematic investigation has increasingly focused on elucidating the mechanistic involvement of ApoB in atherosclerosis, its potential as a diagnostic biomarker, and its implications for therapeutic strategies. CONCLUSION This bibliometric analysis provides the first comprehensive perspective on the evolving promise of ApoB in AS-related research, emphasizing the importance of this molecule in opening up new diagnostic and therapeutic avenues. This study emphasizes the need for continued research and interdisciplinary efforts to strengthen the fight against AS. Furthermore, it emphasizes the critical role of international collaboration and interdisciplinary exploration in leveraging new insights to achieve clinical breakthroughs, thereby addressing the complexities of AS by focusing on ApoB.
Collapse
Affiliation(s)
- Jing Cui
- Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China
- Navy Clinical College, The Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Yan Zhang
- Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Wenhong Zhang
- Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China
- Navy Clinical College, The Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Dongtao Li
- Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Zhibo Hong
- Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Li Zhao
- Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Jiachen Sun
- Department of Dermatology, Peking University Third Hospital, Beijing, China
| | - Yu Chen
- Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China.
- Navy Clinical College, The Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China.
| | - Ningkun Zhang
- Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China.
| |
Collapse
|
10
|
Gao J, Pan H, Guo X, Huang Y, Luo JY. Endothelial Krüppel-like factor 2/4: Regulation and function in cardiovascular diseases. Cell Signal 2025; 130:111699. [PMID: 40023301 DOI: 10.1016/j.cellsig.2025.111699] [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: 12/02/2024] [Revised: 02/09/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
This review presents an overview of the regulation, function, disease-relevance and pharmacological regulation of the critical endothelial transcription factors KLF2/4 in vasculature. The regulatory mechanisms of KLF2/4 expression and activity in vascular endothelium in response to hemodynamic forces and biochemical stimuli are depicted. The functional effects mediated by direct or indirect target genes of KLF2/4 in endothelial cells are systematically summarized. The contributory roles that dysregulated KLF2/4 play in relevant cardiovascular pathologies, such as atherosclerotic vascular lesions, pulmonary arterial hypertension and vascular complications of diabetes were reviewed. Moreover, this review also discusses the pharmacological regulation of KLF2/4 by drugs used in clinics and therapeutic possibility by directly targeting these two transcription factors for treating atherosclerotic cardiovascular diseases. Finally, prospective opinions on the gaps in disclosing novel vascular function mediated by KLF2/4 and future research needs are expressed.
Collapse
Affiliation(s)
- Jing Gao
- Department of Cardiology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, China
| | - Hongjie Pan
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Jiang-Yun Luo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
11
|
Naeini MK, Cecelja M, Freidin MB, Smith IG, Hysi P, Nielsen CS, Williams FMK. Chronic widespread musculoskeletal pain shares a highly heritable latent pathway with atherosclerosis and arterial stiffness. Pain 2025; 166:1425-1435. [PMID: 39620366 PMCID: PMC12067610 DOI: 10.1097/j.pain.0000000000003486] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 05/14/2025]
Abstract
ABSTRACT Chronic widespread pain (CWP) is prevalent and associated with reduced life expectancy. Cardiovascular disease is one possible mechanism for this. The purpose of this study was to examine the association of CWP with arterial stiffness and carotid plaque measured using ultrasound to determine if shared environmental or genetic factors might account for any observed association. Around 3000 participants from the TwinsUK with CWP information and measures of carotid-femoral pulse wave velocity (cfPWV), carotid intima-media thickness (cIMT), and plaque were considered. The relationship between CWP and cfPWV, cIMT, and plaque was determined. UK Biobank data were used to replicate the association. Cholesky decomposition and multivariate pathway twin models were examined. Using a 2-sample Mendelian randomisation approach, the causal association between CWP and coronary artery disease was assessed. TwinsUK participants demonstrated a significant association between CWP and increased cfPWV consistent with arterial stiffening (OR = 1.35, P -value = 0.012), as well as the presence of carotid plaque (OR = 1.45, P -value = 0.8e-5). The twin modelling showed a common latent component and pathway underlying CWP, cfPWV, and carotid plaque, with genetic factors accounting for 68% and 90% of the latent factor variation, respectively. The 2-sample MR revealed a potential causal association between CWP and coronary artery disease. This study found that those with CWP have increased the risk of arterial stiffness and atherosclerosis and suggests that CWP leads to an increased risk of cardiovascular disease through genetic factors.
Collapse
Affiliation(s)
- Maryam Kazemi Naeini
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, United Kingdom
| | - Marina Cecelja
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, United Kingdom
| | - Maxim B. Freidin
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, United Kingdom
- Department of Biology, School of Biological and Behavioural Sciences, Queen Mary University of London, London, United Kingdom
| | - Isabelle Granville Smith
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, United Kingdom
| | - Pirro Hysi
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, United Kingdom
| | - Christopher Sivert Nielsen
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
- Division of Emergencies and Critical Care, Department of Pain Management and Research, Oslo University Hospital, Oslo, Norway
| | - Frances M. K. Williams
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, United Kingdom
| |
Collapse
|
12
|
Yan H, Lv S, Pi H, Yu H, Yin W, Wang Y, Lan Y, Liu W. Inflammation mediates the relationship between cardiometabolic index and vulnerable plaque in patients with acute coronary syndrome. Lipids Health Dis 2025; 24:194. [PMID: 40437601 PMCID: PMC12121154 DOI: 10.1186/s12944-025-02608-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Accepted: 05/14/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND As a novel indicator reflecting metabolic status and visceral adiposity distribution, the cardiometabolic index (CMI) has gained attention in cardiovascular risk stratification. This investigation employed optical coherence tomography (OCT) to examine potential associations between CMI and vulnerable plaque, as well as the role of inflammation. METHODS This study conducted a cross-sectional analysis of 270 acute coronary syndrome (ACS) patients who had OCT imaging evaluation. Patients were categorized based on CMI tertiles, with CMI calculated using the formula [waist (cm)/height (cm)]×[triglycerides (mmol/L)/HDL-C (mmol/L)]. OCT was used to assess plaque events in culprit lesions and plaque components in non-culprit lesions, and inflammatory markers were measured. A mediation analysis framework was implemented to investigate inflammatory pathways in CMI-vulnerable plaque relationships. RESULTS CMI tertiles were linked to vulnerable plaque traits: thin-cap fibroatheromas (TCFA), macrophages (Tertiles1 vs. Tertiles2 vs. Tertiles3, TCFA: 10.0% vs. 20.0% vs. 26.7%, P = 0.016; macrophages: 17.8% vs. 28.9% vs. 36.7%, P = 0.019). Multivariate regression demonstrated CMI elevation independently predicted a higher prevalence of TCFA (OR:1.40, 95%CI: 1.25-2.89, P = 0.003), more macrophage infiltration (OR:1.61, 95% CI:1.09-2.37, P = 0.017), reduced FCT (β:-30.65, 95% CI:-50.72-10.57, P = 0.003), and enlarged maximum lipid arc (β:20.78, 95% CI:6.55-35.01, P = 0.004). Moreover, CMI was positively related to hsCRP, WBC, and neutrophils. Mediation analysis revealed that hsCRP mediated about 17.0% of the association between CMI and minimum FCT [Indirect effect=-5.21, 95% CI=(-12.70, -1.27), P = 0.016]. CONCLUSIONS CMI is a key forecaster of vulnerable plaque in patients with ACS. Systemic inflammation is associated with the relationship between CMI and vulnerable plaque features, suggesting a potential mechanistic link.
Collapse
Affiliation(s)
- Haihao Yan
- Department of Cardiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Sai Lv
- Department of Cardiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Haiyao Pi
- Department of Cardiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Haixu Yu
- Department of Cardiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Weijun Yin
- Department of Cardiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Yaran Wang
- Department of Cardiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Yonghao Lan
- Department of Cardiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Wei Liu
- Department of Cardiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China.
| |
Collapse
|
13
|
Wang J, Zheng W, Shang H, Pan L, Yuan Y, Chen W, Guo C, Li S, Sun X, Guo J, Zhang X. Radial artery intima-media thickening is a sensitive marker of atherosclerosis and coronary artery stenosis, a lesson from a 6-year study of a spontaneous monkey model. Mol Cell Biochem 2025:10.1007/s11010-025-05315-x. [PMID: 40411734 DOI: 10.1007/s11010-025-05315-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 05/15/2025] [Indexed: 05/26/2025]
Abstract
Atherosclerosis is the primary driver of cardiovascular and cerebral vascular diseases globally. Atherosclerotic plaques have been detected in multiple arterial locations, such as the aorta, carotids, and coronaries. However, it remains uncertain if there are variations in susceptibility and association among arteries of different calibers. Utilizing a spontaneous rhesus monkey model of metabolic syndrome (MetS), we assessed the susceptibility of atherosclerosis among the radial artery, femoral artery, and carotid artery and their correlation with coronary heart disease (CHD). The development of atherosclerosis in the three arteries mentioned above was evaluated by Intima-media thickness (IMT) and plaques using echo imaging over 6 years in a cohort of elderly monkeys with metabolic disorders. Coronary artery stenosis was assessed by coronary flow reserve (CFR) simultaneously. The diagnosis was further confirmed by histopathological examination, and RNA sequencing was employed to probe the transcriptional underpinnings of atherosclerotic development. The spontaneous development of atherosclerosis was observed in elderly monkeys, and the incidence of atherosclerosis was increased by three times in the MetS monkeys compared to the age-matched control group. During the 6-year follow-up, there was a notable increase in the IMT across all three arteries, with the radial artery showing the most pronounced thickening. Moreover, only the radial IMT correlated with CFR, suggesting its potential as a non-invasive diagnostic indicator for CHD. Histopathology confirmed the findings by echo imaging and identified different extracellular matrix (ECM) remodeling patterns in the arteries. In addition, transcriptomic analysis revealed that ECM remodeling and inflammation-related pathways were significantly upregulated in radial atherosclerotic samples, multiple inflammatory pathways were upregulated in the femoral lesion samples, and the carotid samples failed to enrich any pathways due to a lack of differentially expressed genes compared to the control samples. Non-human primates, which share extensive genetic and physiological similarities with humans, develop atherosclerosis spontaneously. This provides an invaluable platform for investigating the intricate mechanisms of arterial disease and evaluating potential treatments. Using the monkey model, we identified the radial artery as a sensitive indicator for assessing the occurrence and progression of atherosclerosis and coronary stenosis.
Collapse
Affiliation(s)
- Jue Wang
- Institute of Molecular Medicine and National Biomedical Imaging Center, College of Future Technology, Peking University, Beijing, 100871, China.
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China.
| | - Wen Zheng
- Institute of Molecular Medicine and National Biomedical Imaging Center, College of Future Technology, Peking University, Beijing, 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | - Haibao Shang
- Laboratory Animal Center, Peking University, Beijing, 100871, China
| | - Lin Pan
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Ye Yuan
- Institute of Molecular Medicine and National Biomedical Imaging Center, College of Future Technology, Peking University, Beijing, 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | - Wenli Chen
- Institute of Molecular Medicine and National Biomedical Imaging Center, College of Future Technology, Peking University, Beijing, 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | - Chunguang Guo
- Institute of Molecular Medicine and National Biomedical Imaging Center, College of Future Technology, Peking University, Beijing, 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | - Shihan Li
- Institute of Molecular Medicine and National Biomedical Imaging Center, College of Future Technology, Peking University, Beijing, 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | - Xueting Sun
- Institute of Molecular Medicine and National Biomedical Imaging Center, College of Future Technology, Peking University, Beijing, 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | - Jing Guo
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Xiuqin Zhang
- Institute of Molecular Medicine and National Biomedical Imaging Center, College of Future Technology, Peking University, Beijing, 100871, China.
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China.
| |
Collapse
|
14
|
Mohanta SK, Heron C, Klaus-Bergmann A, Horstmann H, Brakenhielm E, Giannarelli C, Habenicht AJR, Gerhardt H, Weber C. Metabolic and Immune Crosstalk in Cardiovascular Disease. Circ Res 2025; 136:1433-1453. [PMID: 40403115 DOI: 10.1161/circresaha.125.325496] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
Cardiovascular diseases including atherosclerosis and heart failure, arise from the intricate interplay of metabolic, immune, and neural dysregulation within vascular and cardiac tissues: This review focuses on integrating recent advances in metabolic and immune crosstalk of the cardiac vasculature that affects cardiometabolic health and disease progression. Coronary and lymphatic endothelial cells regulate cardiac metabolism, and their dysfunction is linked to cardiovascular diseases. Lymphatics maintain tissue homeostasis, including clearing metabolic waste, lipids, and immune cells, and their maladaptation in metabolic diseases worsens outcomes. Altered vascular endothelial metabolism in heart failure drives immune-mediated inflammation, fibrosis, and adverse cardiac remodeling. Concurrently, artery tertiary lymphoid organs formed in the adventitia of advanced atherosclerotic arteries, serve as pivotal neuroimmune hubs, coordinating local immunity through T and B cell activation and neurovascular signaling via artery-brain circuits. T cells within plaques and artery tertiary lymphoid organs undergo clonal expansion as a result of peripheral tolerance breakdown, with proinflammatory CD4+ and CD8+ subsets amplifying atherosclerosis, effects further shaped by systemic immune activation. Therapeutic strategies targeting endothelial cell metabolism, lymphatic dysfunction, neuroimmune crosstalk, and T cell plasticity hold promise for integrated cardiovascular disease management.
Collapse
Affiliation(s)
- Sarajo K Mohanta
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU), LMU University Hospital, Munich, Germany (S.K.M., A.J.R.H., C.W.)
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany (S.K.M., A.J.R.H., C.W.)
| | - Coraline Heron
- UnivRouen Normandie, INSERM EnVI, UMR 1096, Rouen, France (C.H., E.B.)
| | - Alexandra Klaus-Bergmann
- Integrative Vascular Biology Laboratory, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (A.K.-B., H.G.)
- German Centre for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany (A.K.-B., H.G.)
| | - Hauke Horstmann
- Cardiology and Angiology, Medical Center (H.H.), University of Freiburg, Freiburg, Germany
- Faculty of Medicine (H.H.), University of Freiburg, Freiburg, Germany
- Department of Medicine, Division of Cardiology (H.H., C.G.), NYU Grossman School of Medicine, New York, NY
| | - Ebba Brakenhielm
- UnivRouen Normandie, INSERM EnVI, UMR 1096, Rouen, France (C.H., E.B.)
| | - Chiara Giannarelli
- Department of Medicine, Division of Cardiology (H.H., C.G.), NYU Grossman School of Medicine, New York, NY
- Department of Pathology (C.G.), NYU Grossman School of Medicine, New York, NY
| | - Andreas J R Habenicht
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU), LMU University Hospital, Munich, Germany (S.K.M., A.J.R.H., C.W.)
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany (S.K.M., A.J.R.H., C.W.)
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China (A.J.R.H.)
| | - Holger Gerhardt
- Integrative Vascular Biology Laboratory, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (A.K.-B., H.G.)
- German Centre for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany (A.K.-B., H.G.)
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU), LMU University Hospital, Munich, Germany (S.K.M., A.J.R.H., C.W.)
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany (S.K.M., A.J.R.H., C.W.)
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands (C.W.)
| |
Collapse
|
15
|
Wu J, Zhang L, Zhao Z, Liu Y, Li Z, Feng X, Zhang L, Yao X, Du J, Chen L, Zhou Z. Advancing T-cell immunotherapy for cellular senescence and disease: Mechanisms, challenges, and clinical prospects. Ageing Res Rev 2025; 109:102783. [PMID: 40412763 DOI: 10.1016/j.arr.2025.102783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 05/12/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
Cellular senescence is a complex biological process with a dual role in tissue homeostasis and aging-related pathologies. Accumulation of senescent cells promotes chronic inflammation, tissue dysfunction, age-related diseases, and tumor suppression. Recent advancements in immunotherapy have positioned T cell-based approaches as precision tools for the targeted clearance of senescent cells, offering a novel avenue for anti-aging interventions. This review explores the molecular mechanisms underlying cellular senescence, focusing on its immunogenic features and interactions with T cells, including T-cell activation, antigen recognition, modulation of tumor microenvironment (TME), and immune evasion strategies. Innovations such as chimeric antigen receptor (CAR)-T cells, immune checkpoint therapies, and SASP-neutralizing approaches are highlighted as breakthrough strategies for enhancing senescent cell eradication. The integration of multi-omics and artificial intelligence is further catalyzing the development of personalized therapies to amplify immune surveillance and tissue rejuvenation. Clinically, T cell-based interventions hold promise for mitigating age-related pathologies and extending healthspan, yet challenges remain in optimizing target specificity, countering immunosuppressive niches, and overcoming immune senescence in aging populations. This review synthesizes current advances and challenges, highlighting the potential of T cell immunotherapy as a cornerstone of anti-aging medicine and emphasizing the need for interdisciplinary innovation to translate preclinical findings into transformative therapies for aging and age-related diseases.
Collapse
Affiliation(s)
- Jizhun Wu
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Lu Zhang
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Zihan Zhao
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yuping Liu
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Zhengxing Li
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xiaohang Feng
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Lin Zhang
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xiang Yao
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jun Du
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Liang Chen
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China.
| | - Zhuolong Zhou
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China; Biomedical Sciences, College of Medicine and Veterinary Medicine, Edinburgh Medical School, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
16
|
Steffensen LB, Kavan S, Jensen PS, Pedersen MK, Bøttger SM, Larsen MJ, Dembic M, Bergman O, Matic L, Hedin U, Andersen LVB, Lindholt JS, Houlind KC, Riber LP, Thomassen M, Rasmussen LM. Mutational landscape of atherosclerotic plaques reveals large clonal cell populations. JCI Insight 2025; 10:e188281. [PMID: 40198128 DOI: 10.1172/jci.insight.188281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 04/04/2025] [Indexed: 04/10/2025] Open
Abstract
The notion of clonal cell populations in human atherosclerosis has been suggested but not demonstrated. Somatic mutations are used to define cellular clones in tumors. Here, we characterized the mutational landscape of human carotid plaques through whole-exome sequencing to explore the presence of clonal cell populations. Somatic mutations were identified in 12 of 13 investigated plaques, while no mutations were detected in 11 non-atherosclerotic arteries. Mutated clones often constituted over 10% of the sample cell population, with genes related to the contractile apparatus enriched for mutations. In carriers of clonal hematopoiesis of indeterminate potential (CHIP), hematopoietic clones had infiltrated the plaque tissue and constituted substantial fractions of the plaque cell population alongside locally expanded clones. Our findings establish somatic mutations as a common feature of human atherosclerosis and demonstrate the existence of mutated clones expanding locally, as well as CHIP clones invading from the circulation. While our data do not support plaque monoclonality, we observed a pattern suggesting the coexistence of multiple mutated clones of considerable size spanning different regions of plaques. Mutated clones are likely to be relevant to disease development, and somatic mutations will serve as a convenient tool to uncover novel pathological processes of atherosclerosis in future studies.
Collapse
Affiliation(s)
- Lasse Bach Steffensen
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Centre for Individualized Medicine in Arterial Diseases (CIMA)
| | - Stephanie Kavan
- Centre for Individualized Medicine in Arterial Diseases (CIMA)
- Department of Clinical Biochemistry and Pharmacology, and
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Pia Søndergaard Jensen
- Centre for Individualized Medicine in Arterial Diseases (CIMA)
- Department of Clinical Biochemistry and Pharmacology, and
| | - Matilde Kvist Pedersen
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Centre for Individualized Medicine in Arterial Diseases (CIMA)
| | - Steffen Møller Bøttger
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Clinical Genome Center, Department of Clinical Research
| | - Martin Jakob Larsen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Clinical Genome Center, Department of Clinical Research
- Department of Clinical Research, and
| | - Maja Dembic
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Clinical Genome Center, Department of Clinical Research
- Department of Clinical Research, and
- Department of Mathematics and Computer Science (IMADA), University of Southern Denmark, Odense, Denmark
| | - Otto Bergman
- Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Ljubica Matic
- Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Lars van Brakel Andersen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Clinical Genome Center, Department of Clinical Research
| | - Jes Sanddal Lindholt
- Centre for Individualized Medicine in Arterial Diseases (CIMA)
- Department of Cardiothoracic Surgery, Odense University Hospital, Odense, Denmark
| | | | - Lars Peter Riber
- Department of Cardiothoracic Surgery, Odense University Hospital, Odense, Denmark
| | - Mads Thomassen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Clinical Genome Center, Department of Clinical Research
| | - Lars Melholt Rasmussen
- Centre for Individualized Medicine in Arterial Diseases (CIMA)
- Department of Clinical Biochemistry and Pharmacology, and
| |
Collapse
|
17
|
Bu R, Zhao W, Liang R. Downregulation of ATP8B2 in atherosclerosis exacerbates foam cell-like pathological changes via impairing lysosomal membrane fusion. Mol Biol Rep 2025; 52:485. [PMID: 40402302 DOI: 10.1007/s11033-025-10565-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 05/01/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Atherosclerosis, a major cause of global mortality, involves the transformation of macrophages into foam cells, which is a key pathological process. This study aims to elucidate the molecular mechanisms that contribute to foam cell formation and the progression of atherosclerosis. METHODS AND RESULTS We performed a comprehensive bioinformatics analysis of transcriptome data to identify differentially expressed genes (DEGs) associated with atherosclerosis. Using the human acute monocytic leukemia cell line THP-1, we established in vitro models of macrophages and foam cells to simulate the atherosclerotic microenvironment. Functional studies were conducted using siRNA-mediated knockdown, real-time PCR, Western blotting, and immunofluorescence imaging. Our results showed that ATP8B2 was significantly down-regulated in atherosclerotic foam cells. The downregulation of ATP8B2 led to impaired lysosomal membrane fusion, evidenced by an increase in CD63-positive compartments without a change in CD63 protein levels. Additionally, under starvation conditions, there was a significant accumulation of autophagosomes, indicating a defect in the autophagy-lysosomal pathway. CONCLUSIONS This study, for the first time, demonstrates that the downregulation of ATP8B2 exacerbates atherosclerosis by disrupting lysosomal membrane fusion, leading to lipid accumulation and foam cell formation. These findings provide novel insights into the pathogenesis of atherosclerosis and suggest that ATP8B2 could be a potential therapeutic target for the prevention or treatment of this disease.
Collapse
Affiliation(s)
- Rui Bu
- The Fourth Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, China
| | - Weihao Zhao
- Heilongjiang Red Cross Sengong General Hospital, Harbin City, Heilongjiang Province, China
| | - Rui Liang
- The Fourth Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, China.
| |
Collapse
|
18
|
Han GM, Liu JQ, Dai ZQ, Jin WL, Cai QL, Kong DM, Zhu LN. Multifunctional cerium-based nanozymes as moonlighting protein mimics for atherosclerosis diagnosis and therapy. Chem Sci 2025; 16:8772-8782. [PMID: 40271043 PMCID: PMC12013363 DOI: 10.1039/d5sc01014d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 04/10/2025] [Indexed: 04/25/2025] Open
Abstract
Moonlighting proteins are multifunctional proteins widely present in organisms, playing crucial roles in various physiological activities. Drawing inspiration from the moonlight proteins, we developed a cerium (Ce)-based nanozyme CF, featuring multiple enzymatic activities along with robust cargo-loading and transport capabilities. The CF was synthesized through a one-step assembly between Ce3+ and a phosphorylated amino acid derivative, achieving high biostability through a simple heat treatment. The nanozyme possesses both superoxide dismutase (SOD) and catalase (CAT) activities, enabling scavenging of reactive oxygen species (ROS) and modulation of inflammation by inhibiting NF-κB pathway activation. Besides its enzymatic activities, CF can also serve as a versatile nanocarrier for various cargoes through one-pot co-assembly. Herein, the CF-based nanoassembly loaded with a near infrared fluorescent dye was demonstrated to work well for the diagnosis of atherosclerotic plaques. The nanoassembly co-assembled with probucol exhibited superior ROS-scavenging and anti-inflammatory effects compared to either CF nanozyme or probucol, attributed to the synergy of the nanozyme and the drug, thus facilitating a highly efficient treatment of atherosclerosis. This work introduces a novel Ce-based nanozyme with multifunctional properties, providing a promising approach to endow nanozymes with moonlighting protein-like characteristics, thereby enhancing their functional capabilities and broadening their application potential in various fields.
Collapse
Affiliation(s)
- Gui-Mei Han
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center of Analytical Science, College of Chemistry, Nankai University Tianjin 300071 P. R. China
- College of Chemistry and Chemical Engineering, Qilu Normal University Jinan 250200 P. R. China
| | - Jing-Qi Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center of Analytical Science, College of Chemistry, Nankai University Tianjin 300071 P. R. China
| | - Zhi-Qi Dai
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center of Analytical Science, College of Chemistry, Nankai University Tianjin 300071 P. R. China
| | - Wei-Liang Jin
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center of Analytical Science, College of Chemistry, Nankai University Tianjin 300071 P. R. China
- Department of Chemistry, School of Science, Tianjin University Tianjin 300354 P. R. China
| | - Qi-Liang Cai
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology Tianjin 300211 P. R. China
| | - De-Ming Kong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center of Analytical Science, College of Chemistry, Nankai University Tianjin 300071 P. R. China
| | - Li-Na Zhu
- Department of Chemistry, School of Science, Tianjin University Tianjin 300354 P. R. China
| |
Collapse
|
19
|
Ma C, Hua Y, Yang S, Zhao Y, Zhang W, Miao Y, Zhang J, Feng B, Zheng G, Li L, Liu Z, Zhang H, Zhu M, Gao X, Fan G. Wogonin Attenuates Atherosclerosis via KLF11-Mediated Suppression of PPARα-YAP1-Driven Glycolysis and Enhancement of ABCA1/G1-Mediated Cholesterol Efflux. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500610. [PMID: 40397286 DOI: 10.1002/advs.202500610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/04/2025] [Indexed: 05/22/2025]
Abstract
Atherosclerosis, a chronic inflammatory disorder and leading cause of cardiovascular disease, is characterized by macrophage-derived inflammation and foam cell formation. Emerging evidence suggests that metabolic reprogramming of macrophages represents a promising therapeutic approach for atherosclerosis management. In this study, the therapeutic potential of wogonin, a bioactive flavonoid isolated from Scutellaria baicalensis, in modulating macrophage metabolism and attenuating atherogenesis is investigated. Wogonin reduces lesion size and plaque vulnerability, accompanied by a reduction in foam cell formation and inflammation. Mechanistically, wogonin reprogrammes macrophage metabolism from glycolysis to fatty acid oxidation (FAO) by activating the PPARα-CPT1α pathway and acts as a mitochondrial protector by activating PPARα. Wogonin also promotes the KLF11 expression and KLF11 knockout exacerbated atherosclerosis and abolishes the inhibitory effect of wogonin on glycolysis and atherosclerosis. KLF11 forms a transcriptional complex with PPARα and YAP1, serving both as a brake on PPARα-YAP1-mediated glycolysis and a transcriptional activator of ABCA1/G1. Collectively, wogonin reprograms macrophage metabolism from glycolysis to FAO through activation of the PPARα-KLF11-YAP1 pathway, thereby reducing inflammation and foam cell formation, ultimately attenuating atherogenesis.
Collapse
Affiliation(s)
- Chuanrui Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300381, China
| | - Yunqing Hua
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300381, China
| | - Shu Yang
- Department of Geriatrics, Peking University Shenzhen Hospital, Shenzhen, China, Shenzhen, Guangdong, 518000, China
| | - Yun Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Wei Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Yaodong Miao
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300250, P. R. China
| | - Jing Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Boxuan Feng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Guobin Zheng
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Lan Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
| | - Zhihao Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
| | - Han Zhang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
| | - Mingjun Zhu
- Department of Cardiovascular Diseases, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, China
| | - Xiumei Gao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 300193, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300381, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 300193, China
| |
Collapse
|
20
|
Zhang Q, Ma S, Kang X, Liu Y, Ma F, Yu F, Luo X, Li G, Hao Y, Zhang H, Liu B, Jiang Y. A dual-targeting bio-liposomes nanodrug repair endothelial cell dysfunction and restore macrophage cholesterol flow homeostasis to treat early atherosclerosis. J Nanobiotechnology 2025; 23:365. [PMID: 40394654 PMCID: PMC12090647 DOI: 10.1186/s12951-025-03436-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 05/01/2025] [Indexed: 05/22/2025] Open
Abstract
Hyperhomocysteinemia (HHy) can lead to vascular endothelial cell dysfunction, progressive inflammation and lipid metabolism disorder, which finally result in the onset and development of atherosclerosis, a major contributor to cardiovascular diseases. Given the complexity of pathological process, treatments based on a single target often showed limited therapeutic efficacy against AS. Thus, developing nanodrug for enhanced multi-targets therapy is promising. In this study, we constructed a dual-targeting nanodrug (HA-ML@ES NPs) co-loaded with Shikonin (SKN) and Evolocumab (Evol). In vitro results showed that HA-ML@ES NPs could simultaneously target dysfunctional endothelial cell and inflammatory macrophage through the interaction between HA and CD44. In vivo assay indicated that HA-ML@ES NPs with long circulation and plaque accumulation efficiently attenuate endothelial cell dysfunction by inhibiting glycolysis and restore cholesterol flow homeostasis in macrophage by reprogramming macrophage phenotype, which finally attenuated the development of atherosclerosis. Collectively, these results present a highly promising dual-cell therapeutic approach based on HA-ML@ES NPs for the management of early atherosclerosis.
Collapse
Grants
- 2023AAC005035 The Natural Science Foundation of Ningxia Hui Autonomous Region
- XJKF240301, XJKF240304, XJKF230125 and XJKF240326 Open Competition Mechanism to Select the Best Candidates for Key Research Projects of Ningxia Medical University
- XJKF240301, XJKF240304, XJKF230125 and XJKF240326 Open Competition Mechanism to Select the Best Candidates for Key Research Projects of Ningxia Medical University
- XJKF240301, XJKF240304, XJKF230125 and XJKF240326 Open Competition Mechanism to Select the Best Candidates for Key Research Projects of Ningxia Medical University
- XJKF240301, XJKF240304, XJKF230125 and XJKF240326 Open Competition Mechanism to Select the Best Candidates for Key Research Projects of Ningxia Medical University
- 2023BEG02074, 2022BFH02013, 2022BEG02054 Key Projects of the Key R&D Program of the Ning Xia Hui Autonomous Region
- U21A20343 National Natural Science Foundation of China
- 2024ZD0531200 Noncommunicable Chronic Diseases-National Science and Technology Major Project
- Key Projects of the Key R&D Program of the Ning Xia Hui Autonomous Region
Collapse
Affiliation(s)
- Qi Zhang
- School of Inspection, Ningxia Medical University, Yinchuan, 750004, China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
| | - Shengchao Ma
- School of Inspection, Ningxia Medical University, Yinchuan, 750004, China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
| | - Xue Kang
- Department of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Yi Liu
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Fei Ma
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Feifei Yu
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Xiaolan Luo
- School of Inspection, Ningxia Medical University, Yinchuan, 750004, China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
| | - Guizhong Li
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Yinju Hao
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
- Medical Experimental Center, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
| | - Huiping Zhang
- Medical Experimental Center, General Hospital of Ningxia Medical University, Yinchuan, 750004, China.
- Department of Medical Genetics, Hunan Provincial Maternal and Child Health Hospital, Changsha, 410008, China.
| | - Bin Liu
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China.
- College of Biology, Hunan University, Changsha, 410082, China.
| | - Yideng Jiang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China.
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
21
|
Zhao M, Pang S, Gao Y, Li T, Jiang H. Effect of Chitooligosaccharides on TLR2/NF-κB Signaling in LPS-Stimulated RAW 264.7 Macrophages. Molecules 2025; 30:2226. [PMID: 40430398 PMCID: PMC12114287 DOI: 10.3390/molecules30102226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 05/13/2025] [Accepted: 05/17/2025] [Indexed: 05/29/2025] Open
Abstract
Chitooligosaccharides (COSs), degraded products of chitosan or chitin, are attracting growing interest owing to their low degree of polymerization (DP), high solubility, and prominent anti-inflammatory activity. However, the correlation between their structure and anti-inflammatory activities still needs to be explored. In this study, we use LPS-stimulated RAW 264.7 macrophages as an inflammatory model to systematically evaluate COS1-7 for their effects on inflammatory mediators and NF-κB signaling pathways. The results of Griess assay, ELISA, and real-time quantitative PCR show that COSs can inhibit the expression of NO, iNOS, and pro-inflammatory cytokines (IL-6, TNF-α, MCP-1 and IL-1β), thereby attenuating inflammatory signaling. Notably, chitohexaose (COS6) exhibits the most significant anti-inflammatory effect, reducing the mRNA levels of LPS-induced iNOS, IL-6, and IL-1β and the production of IL-6 and TNF-α by more than 50%. Transcriptome, western blotting, and real-time quantitative PCR analysis reveal that COSs can inhibit the activation of the NF-κB signal pathway by down-regulating TLR2 levels. Additionally, molecular docking confirms that COSs retard TLR2/4 dimerization and LPS recognition by TLR4, affecting downstream signaling cascades. In summary, this study provides a valuable insight into the potential anti-inflammatory mechanism of COSs and highlights the possible applications in human health promotion by modulating receptor-mediated signaling pathways.
Collapse
Affiliation(s)
- Mengting Zhao
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China; (M.Z.); (S.P.); (Y.G.); (T.L.)
| | - Shurong Pang
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China; (M.Z.); (S.P.); (Y.G.); (T.L.)
| | - Yiqing Gao
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China; (M.Z.); (S.P.); (Y.G.); (T.L.)
| | - Ting Li
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China; (M.Z.); (S.P.); (Y.G.); (T.L.)
| | - Hongrui Jiang
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China; (M.Z.); (S.P.); (Y.G.); (T.L.)
- Key Laboratory of Deep Processing and Safety Control for Specialty Agricultural Products in Guangxi Universities, Education Department of Guangxi, Nanning 530004, China
| |
Collapse
|
22
|
Cui Y, Wu Y, Shi P, Ni Y, Zeng H, Zhang Z, Zhao C, Sun W, Yi Q. Mitigating microplastic-induced organ Damage: Mechanistic insights from the microplastic-macrophage axes. Redox Biol 2025; 84:103688. [PMID: 40412021 DOI: 10.1016/j.redox.2025.103688] [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: 01/17/2025] [Revised: 05/10/2025] [Accepted: 05/17/2025] [Indexed: 05/27/2025] Open
Abstract
We live in a world increasingly dominated by plastic, leading to the generation of microplastic particles that pose significant global health concerns. Microplastics can enter the body via ingestion, inhalation, and direct contact, accumulating in various tissues and potentially causing harm. Despite this, the specific cellular mechanisms and signaling pathways involved remain poorly understood. Macrophages are essential in absorbing, distributing, and eliminating microplastics, playing a key role in the body's defense mechanisms. Recent evidence highlights oxidative stress signaling as a key pathway in microplastic-induced macrophage dysfunction. The accumulation of microplastics generates reactive oxygen species (ROS), disrupting normal macrophage functions and exacerbating inflammation and organ damage. This review serves as the first comprehensive examination of the interplay between microplastics, macrophages, and oxidative stress. It discusses how oxidative stress mediates macrophage responses to microplastics and explores the interactions with gut microbiota. Additionally, it reviews the organ damage resulting from alterations in macrophage function mediated by microplastics and offers a novel perspective on the defense, assessment, and treatment of microplastic-induced harm from the viewpoint of macrophages.
Collapse
Affiliation(s)
- Yinxing Cui
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China; Department of General Surgery, Dongguan Huangjiang Hospital, Dongguan, 523061, Guangdong, China
| | - Yuqi Wu
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China
| | - Pan Shi
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China
| | - Yan Ni
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China
| | - Huaying Zeng
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China
| | - Zhao Zhang
- Department of General Surgery, Dongguan Huangjiang Hospital, Dongguan, 523061, Guangdong, China
| | - Chunling Zhao
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China.
| | - Weichao Sun
- Department of Orthopedics, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, 518035, China.
| | - Qian Yi
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China.
| |
Collapse
|
23
|
Chen M, Wang P, Li Y, Liu X. Inhibitory mechanism study of soyasaponins on thrombin: extraction, purification, and functional property analysis. Food Funct 2025; 16:4149-4160. [PMID: 40314673 DOI: 10.1039/d5fo00522a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
This study aimed to investigate the inhibitory effects of soyasaponins extracted from soybean meal on thrombin, thereby providing scientific evidence for the application of soybean meal byproducts in functional foods and cardiovascular disease prevention. Using a combination of resin column chromatography and preparative liquid chromatography, six extracts were isolated and purified. Among these, four extracts (SE3, SE4, SE5, SE6) exhibited significant thrombin inhibitory activity. Enzyme activity assays revealed that SE6 showed the strongest inhibitory effect, with an IC50 of 1.36 mg mL-1. Component analysis indicated that SE3 contained a soyasaponins content as high as 91.2%, while SE4, SE5, and SE6 were mixtures of soyasaponins and isoflavones. Molecular docking analysis demonstrated that soyasaponins, such as Soyasapogenol E, had the highest binding affinity to thrombin. Moreover, the interactions of SE3, SE4, SE5, and SE6 with thrombin induced structural changes, reducing or eliminating the α-helical structures. These interactions were accompanied by fluorescence quenching effects, further elucidating the inhibition mechanism. The findings confirm that soyasaponins exhibit thrombin inhibitory activity. Additionally, a novel hypothesis was proposed, suggesting a potential synergistic effect between saponins and isoflavones. This synergy highlights the potential of soyasaponins and their complexes in the development of functional foods for cardiovascular health.
Collapse
Affiliation(s)
- Mengqian Chen
- China Food Flavor and Nutrition Health Innovation Center, Beijing Technology and Business University; National Soybean Processing Industry Technology Innovation Center, Beijing Technology and Business University, Beijing 100048, China.
| | - Pengcheng Wang
- China Food Flavor and Nutrition Health Innovation Center, Beijing Technology and Business University; National Soybean Processing Industry Technology Innovation Center, Beijing Technology and Business University, Beijing 100048, China.
| | - You Li
- China Food Flavor and Nutrition Health Innovation Center, Beijing Technology and Business University; National Soybean Processing Industry Technology Innovation Center, Beijing Technology and Business University, Beijing 100048, China.
| | - Xinqi Liu
- China Food Flavor and Nutrition Health Innovation Center, Beijing Technology and Business University; National Soybean Processing Industry Technology Innovation Center, Beijing Technology and Business University, Beijing 100048, China.
- Jiangsu Protec Biotechnology Co., Ltd, China
| |
Collapse
|
24
|
Zhang B, Li Q, Wang W, Tian M, Xu D, Xie Y. PFOS and Its Substitute OBS Cause Endothelial Dysfunction to Promote Atherogenesis in ApoE -/- Mice. ENVIRONMENT & HEALTH (WASHINGTON, D.C.) 2025; 3:526-538. [PMID: 40400551 PMCID: PMC12090012 DOI: 10.1021/envhealth.4c00206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 01/24/2025] [Accepted: 01/30/2025] [Indexed: 05/23/2025]
Abstract
Perfluorooctanesulfonate (PFOS), an emerging contaminant with widespread concern, has been associated with the pathogenesis of atherosclerosis (AS). As a substitute for PFOS, sodium p-perfluorous nonenoxybenzenesulfonate (OBS) is extensively utilized in various applications and detected in human blood. However, its potential health risk in AS remain unclear. In this study, we investigated the comparative impacts of PFOS and OBS on endothelial dysfunction and atherogenesis. In the in vivo study, Apolipoprotein E knockout (ApoE-/-) mice were exposed to 0.4 or 4 mg/L PFOS/OBS for 12 weeks. We found that dyslipidemia developed more rapidly in the OBS-exposed mice than in the PFOS-exposed mice. PFOS exhibited a higher enrichment capacity in both blood and aortic tissues than OBS. Remarkably, OBS induced a more pronounced inflammatory response and caused a more significant disruption of the endothelial barrier in the aorta of ApoE-/- mice compared to PFOS. In vitro experiments showed that OBS, at the same exposure concentrations and durations as PFOS (0.1-20 μmol/L, 48 h), more effectively inhibited cell viability of human umbilical vein endothelial cells (HUVECs), caused higher levels of lactate dehydrogenase (LDH) release, and enhanced cell adhesion between HUVECs and monocytes. Both PFOS and OBS were found to activate the NF-κB signaling pathway and upregulate the expression of inflammatory factors. Notably, the use of OBS, but not PFOS, was shown to disrupt cell junctions and increase endothelial permeability by activating the MAPK/ERK signaling pathway. Our findings suggest that OBS may lead to endothelial dysfunction and have a greater impact on AS compared to PFOS, presenting significant health risks in cardiovascular diseases.
Collapse
Affiliation(s)
- Boxiang Zhang
- Institute
of Environmental Systems Biology, Environment Science and Engineering
College, Dalian Maritime University, Linghai Road 1, Dalian 116026, China
| | - Qing Li
- Institute
of Environmental Systems Biology, Environment Science and Engineering
College, Dalian Maritime University, Linghai Road 1, Dalian 116026, China
| | - Wensheng Wang
- Institute
of Environmental Systems Biology, Environment Science and Engineering
College, Dalian Maritime University, Linghai Road 1, Dalian 116026, China
| | - Mingming Tian
- Institute
of Environmental Systems Biology, Environment Science and Engineering
College, Dalian Maritime University, Linghai Road 1, Dalian 116026, China
| | - Dan Xu
- Institute
of Environmental Systems Biology, Environment Science and Engineering
College, Dalian Maritime University, Linghai Road 1, Dalian 116026, China
| | - Ying Xie
- The
Second Affiliated Hospital of Dalian Medical University, No. 467 Zhongshan Road, Dalian 116023, China
| |
Collapse
|
25
|
Shi P, Tang B, Xie W, Li K, Guo D, Li Y, Yao Y, Cheng X, Xu C, Wang QK. LncRNA-induced lysosomal localization of NHE1 promotes increased lysosomal pH in macrophages leading to atherosclerosis. J Biol Chem 2025:110246. [PMID: 40383150 DOI: 10.1016/j.jbc.2025.110246] [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: 01/31/2025] [Revised: 04/30/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025] Open
Abstract
ANRIL, also referred to as CDKN2B-AS1, is a lncRNA gene implicated in the pathogenesis of multiple human diseases including atherosclerotic coronary artery disease, however, definitive in vivo evidence is lacking and the underlying molecular mechanism is largely unknown. In this study, we show that ANRIL overexpression causes atherosclerosis in vivo as transgenic mouse overexpression of full-length ANRIL (NR_003529) increases inflammation and aggravates atherosclerosis under ApoE-/- background (ApoE-/-ANRIL mice). Mechanistically, ANRIL reduces the expression of miR-181b-5p, which leads to increased TMEM106B expression. TMEM106B is significantly up-regulated in atherosclerotic lesions of both human CAD patients and ApoE-/-ANRIL mice. TMEM106B interacts and co-localizes with Na+-H+ exchanger NHE1, which results in mis-localization of NHE1 from cell membranes to lysosomal membranes, leading to increased lysosomal pH in macrophages. Large truncation and point mutation analyses define the critical amino acids for TMEM106B-NHE1 interaction and lysosomal pH regulation as F115 and F117 on TMEM106B and I537, C538, and G539 on NHE1. Topological analysis suggests that both N-terminus and C-terminus of NHE1 are located inside lysosomal lumen, and NHE1 is an important new proton efflux channel involved in raising lysosomal pH. A short TMEM106B peptide (YGRKKRRQRRR-L111A112V113F114F115L116F117) disrupting the TMEM106B-NHE1 interaction normalized lysosomal pH in macrophages with ANRIL overexpression. Our data demonstrate that ANRIL promotes atherosclerosis in vivo and identify the ANRIL/miR-181b-5p/TMEM106B-NHE1/lysosomal pH axis as the underlying molecular pathogenic mechanism for the chromosome 9p21.3 genetic locus for coronary artery disease.
Collapse
Affiliation(s)
- Pengcheng Shi
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Bo Tang
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Wen Xie
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Ke Li
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Di Guo
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Yining Li
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Yufeng Yao
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College
| | - Chengqi Xu
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Qing K Wang
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology; Maternal and Child Health Hospital of Hubei Province, Women and Children's Hospital of Hubei Province, Huazhong University of Science and Technology, Wuhan, P. R. China.
| |
Collapse
|
26
|
Wang M, Li B, Nie S, Meng X, Wang G, Yang M, Dang W, He K, Sun T, Xu P, Yang X, Ye K. Asparagine endopeptidase cleaves apolipoprotein A1 and accelerates pathogenesis of atherosclerosis. J Clin Invest 2025; 135:e185128. [PMID: 40371638 PMCID: PMC12077905 DOI: 10.1172/jci185128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 03/12/2025] [Indexed: 05/16/2025] Open
Abstract
Atherosclerosis is a slowly progressing inflammatory disease characterized with cholesterol disorder and intimal plaques. Asparagine endopeptidase (AEP) is an endolysosomal protease that is activated under acidic conditions and is elevated substantially in both plasma and plaques of patients with atherosclerosis. However, how AEP accelerates atherosclerosis development remains incompletely understood, especially from the view of cholesterol metabolism. This project aims to reveal the crucial substrate of AEP during atherosclerosis plaque formation and to lay the foundation for developing novel therapeutic agents for Atherosclerosis. Here, we show that AEP is augmented in the atherosclerosis plaques obtained from patients and proteolytically cuts apolipoprotein A1 (APOA1) and impairs cholesterol efflux and high-density lipoprotein (HDL) formation, facilitating atherosclerosis pathologies. AEP is activated in the liver and aorta of apolipoprotein E-null (APOE-null) mice, and deletion of AEP from APOE-/- mice attenuates atherosclerosis. APOA1, an essential lipoprotein in HDL for cholesterol efflux, is cleaved by AEP at N208 residue in the liver and atherosclerotic macrophages of APOE-/- mice. Blockade of APOA1 cleavage by AEP via N208A mutation or its specific inhibitor, #11a, substantially diminishes atherosclerosis in both APOE-/- and LDLR-/- mice. Hence, our findings support that AEP disrupts cholesterol metabolism and accelerates the development of atherosclerosis.
Collapse
Affiliation(s)
- Mengmeng Wang
- Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Bowei Li
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology (SUAT), University of Chinese Academy of Science, Shenzhen, Guangdong, China
| | - Shuke Nie
- Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Xin Meng
- Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Guangxing Wang
- Department of Laboratory Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Menghan Yang
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology (SUAT), University of Chinese Academy of Science, Shenzhen, Guangdong, China
| | - Wenxin Dang
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology (SUAT), University of Chinese Academy of Science, Shenzhen, Guangdong, China
| | - Kangning He
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, International Campus, Zhejiang University, Haining, China
| | - Tucheng Sun
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ping Xu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Discipline of Health Toxicology (2020–2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Keqiang Ye
- Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology (SUAT), University of Chinese Academy of Science, Shenzhen, Guangdong, China
| |
Collapse
|
27
|
Peluzzo AM, St Paul A, Corbett CB, Kelemen SE, Fossati S, Liu X, Autieri MV. IL-19 Is a Novel Lymphangiocrine Factor Inducing Lymphangiogenesis and Lymphatic Junctional Regulation. Arterioscler Thromb Vasc Biol 2025. [PMID: 40371466 DOI: 10.1161/atvbaha.125.322669] [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: 02/20/2025] [Accepted: 05/01/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND The lymphatic system functions by removing fluid, macromolecules, and immune cells to maintain tissue homeostasis. The structural organization of junctional protein complexes is vital to lymphatic function where initial lymphatics have permeable button junctions and collecting lymphatics have relatively impermeable zipper junctions. During inflammation, this junctional morphology appears to reverse, contributing to overall lymphatic malfunction. Little is known about the effects of immunomodulatory cytokines on lymphatic vessel formation and function during inflammation. The purpose of this study is to test the hypothesis that IL (interleukin)-19 promotes lymphangiogenesis and proper lymphatic function during inflammation. METHODS We used cultured human dermal lymphatic endothelial cells to determine IL-19 expression and its effects on lymphangiogenesis assays. Immunocytochemistry and electric cell-substrate impedance sensing determined effects on junctional morphology as it relates to permeability in vitro. RNA sequencing determined the effects of IL-19 on gene expression. Il19-/-Ldlr-/- double knockout mice were used to determine IL-19 effects on lymphatic function and lymphatic vessel visualization in vivo. RESULTS Endogenous IL-19 expression is induced by exogenous IL-19 and VEGF (vascular endothelial growth factor) C stimulation. IL-19 is lymphangiogenic, increasing human dermal lymphatic endothelial cell migration, network formation, and proliferation. IL-19 induces expression of transcription factors and permeability-associated genes. IL-19 induces rapid VE-cadherin (vascular endothelial cadherin) phosphorylation, increases permeability of human dermal lymphatic endothelial cell monolayers, and mitigates oxidized low-density lipoprotein-associated decrease in human dermal lymphatic endothelial cell permeability. In vivo, Il19-/-Ldlr-/- double knockout mice on a high-fat diet have impaired lymphatic drainage, decreased lymphatic branch points, and increased percentage of zippered junctions compared with control mice. CONCLUSIONS Taken together, these data show that IL-19 has potent effects on lymphatic vessel formation and function in vitro and that IL-19 regulates lymphatic drainage in vivo. IL-19 may represent an immunomodulatory cytokine with therapeutic potential for improving impaired lymphatic function consequent to inflammation.
Collapse
Affiliation(s)
- Amanda M Peluzzo
- Lemole Center for Integrated Lymphatics and Vascular Research (A.M.P., A.S.P., C.B.C., S.E.K., X.L., M.V.A.)
| | - Amanda St Paul
- Lemole Center for Integrated Lymphatics and Vascular Research (A.M.P., A.S.P., C.B.C., S.E.K., X.L., M.V.A.)
| | - Cali B Corbett
- Lemole Center for Integrated Lymphatics and Vascular Research (A.M.P., A.S.P., C.B.C., S.E.K., X.L., M.V.A.)
| | - Sheri E Kelemen
- Lemole Center for Integrated Lymphatics and Vascular Research (A.M.P., A.S.P., C.B.C., S.E.K., X.L., M.V.A.)
| | - Silvia Fossati
- Alzheimer's Center at Temple, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (S.F.)
| | - Xiaolei Liu
- Lemole Center for Integrated Lymphatics and Vascular Research (A.M.P., A.S.P., C.B.C., S.E.K., X.L., M.V.A.)
| | - Michael V Autieri
- Lemole Center for Integrated Lymphatics and Vascular Research (A.M.P., A.S.P., C.B.C., S.E.K., X.L., M.V.A.)
| |
Collapse
|
28
|
Liu W, Wang YR, Wu H, Cui W, Xu X. The role of myeloperoxidase in the pathogenesis of stroke. Brain Res 2025; 1861:149705. [PMID: 40379076 DOI: 10.1016/j.brainres.2025.149705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 05/01/2025] [Accepted: 05/11/2025] [Indexed: 05/19/2025]
Abstract
Stroke is the leading cause of mortality and morbidity worldwide, significantly impacting human welfare and overall health. Myeloperoxidase (MPO), a heme peroxidase secreted by neutrophils, plays a crucial role in the body's defense mechanisms, exhibiting pro-inflammatory and pro-oxidative properties. Additionally, MPO compromises the structural integrity and functional capacity of blood vessels, potentially leading to the formation and dislodgement of atherosclerotic plaques, vascular stenosis, thrombosis, and ultimately contributing to stroke occurrence. Following a stroke, a significant influx of neutrophils infiltrates the cerebral tissue, leading to an excessive release of MPO-derived oxidants and the subsequent promotion of various inflammatory mediators, thereby exacerbating cerebral tissue damage. Numerous studies have consistently demonstrated the pivotal role of MPO in the pathogenesis and progression of stroke, establishing it as a reliable prognostic indicator. Exploring the association between MPO and stroke enhances our understanding of the pathological mechanisms underlying stroke and aids in the development of therapeutic interventions. This review provides a comprehensive analysis of the molecular structure and cellular localization of MPO, elucidating its critical role in mediating vascular injury, the formation of Neutrophil Extracellular Traps (NETs), oxidative stress, neuroinflammation, disruption of the blood-brain barrier (BBB), and neuronal apoptosis during stroke pathogenesis. Additionally, we discuss recent advancements in MPO-targeted drugs and Traditional Chinese Medicine compounds as potential therapeutic strategies for stroke treatment.
Collapse
Affiliation(s)
- Wei Liu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Yi-Ran Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Hongyun Wu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China; Department of Neurology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China.
| | - Wenqiang Cui
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China; Department of Neurology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China.
| | - Xiangqing Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China; Department of Neurology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China.
| |
Collapse
|
29
|
Cao P, Yang Y, Zhang N, Wang B, Gong Z. Inflammasomes: novel therapeutic targets for metabolic syndrome? Front Endocrinol (Lausanne) 2025; 16:1569579. [PMID: 40433411 PMCID: PMC12106043 DOI: 10.3389/fendo.2025.1569579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 04/18/2025] [Indexed: 05/29/2025] Open
Abstract
Chronic inflammation is a hallmark for Metabolic Syndrome (MetS). It is also one of the most important risk factors for insulin resistance and metabolic disorders. Inflammasomes, which are intracellular multiprotein complexes within the innate immune system, regulate the production and maturation of pro-inflammatory cytokines including interleukin-1β (IL-1β) and IL-18 upon sensing pathogens or danger signals in the cytosol. A growing body of evidence indicates that inflammasomes play a pivotal role in the pathophysiology and progression of metabolic diseases, as deficiency in the key component of inflammasomes protects mice from high fat diet induced obesity and insulin resistance. Thus, in this review, we will summarize the role of inflammasomes in MetS and how to treat MetS by targeting inflammasomes. This may provide novel insights and therapeutic targets for treating metabolic disorders.
Collapse
Affiliation(s)
- Pengyu Cao
- The Second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Yulin Yang
- The Second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Ningning Zhang
- The Second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Bojian Wang
- School of Nursing, Jilin University, Changchun, Jilin, China
| | - Zhenwei Gong
- Division of Endocrinology, Department of Pediatrics, Children’s Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
30
|
Kubryń N, Fijałkowski Ł, Nowaczyk J, Jamil A, Nowaczyk A. PROTAC Technology as a New Tool for Modern Pharmacotherapy. Molecules 2025; 30:2123. [PMID: 40430296 PMCID: PMC12114078 DOI: 10.3390/molecules30102123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/05/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
The publication focuses on the innovative applications of PROTAC (proteolysis-targeting chimera) technology in modern pharmacotherapy, with particular emphasis on cancer treatment. PROTACs represent an advanced therapeutic strategy that enables selective protein degradation, opening new possibilities in drug design. This technology shows potential in the treatment of cancers, viral infections (such as HIV and COVID-19), and chronic diseases including atherosclerosis, Alzheimer's disease, atopic dermatitis, and Huntington's disease. Promising results from clinical studies on the compound ARV-471 confirm the effectiveness of this approach. New types of PROTACs, like TF-PROTAC and PhosphoTAC, are designed to enhance the effectiveness, stability, and absorption of treatment drugs. The conclusions of the review highlight the broad therapeutic potential of PROTACs in various diseases and their relevance for the future of therapies, particularly in oncology.
Collapse
Affiliation(s)
- Natalia Kubryń
- Department of Organic Chemistry, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 2 dr. A. Jurasza St., 85-094 Bydgoszcz, Poland; (N.K.); (A.N.)
| | - Łukasz Fijałkowski
- Department of Organic Chemistry, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 2 dr. A. Jurasza St., 85-094 Bydgoszcz, Poland; (N.K.); (A.N.)
| | - Jacek Nowaczyk
- Department of Physical Chemistry and Physicochemistry of Polymers, Faculty of Chemistry, Nicolaus Copernicus University, 7 Gagarina St., 87-100 Toruń, Poland;
| | - Amer Jamil
- Department of Biochemistry, University of Agriculture, Faisalabad 38040, Pakistan;
| | - Alicja Nowaczyk
- Department of Organic Chemistry, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 2 dr. A. Jurasza St., 85-094 Bydgoszcz, Poland; (N.K.); (A.N.)
| |
Collapse
|
31
|
Dai Y, Yang L, Cao G, Mo L, Yang C, Zhu Y, Guo Y, Hong Y, Xu H, Lu S, Du S, He J. Combination therapy and drug co-delivery systems for atherosclerosis. J Control Release 2025; 381:113543. [PMID: 39986476 DOI: 10.1016/j.jconrel.2025.02.039] [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: 11/13/2024] [Revised: 01/25/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of plaque within the arteries. Despite advances in therapeutic strategies including anti-inflammatory, antioxidant, and lipid metabolism modulation treatments over the past two decades, the treatment of atherosclerosis remains challenging, as arterial damage is the result of interconnected pathological factors. Therefore, current monotherapies often fail to address the complex nature of this disease, leading to insufficient therapeutic outcomes. This review addressed this paucity of effective treatment options by comprehensively exploring the potential for combination therapies and advanced drug co-delivery systems for the treatment of atherosclerosis. We investigated the pathological features of and risk factors for atherosclerosis, underscoring the importance of drug combination therapies for the treatment of atherosclerotic diseases. We discuss herein mathematical models for quantifying the efficacy of the combination therapies and provide a systematic summary of drug combinations for the treatment of atherosclerosis. We also provide a detailed review of the latest advances in nanoparticle-based drug co-delivery systems for the treatment of atherosclerosis, focusing on the design of carriers with high biocompatibility and efficacy. By exploring the possibilities and challenges inherent to this approach, we aim to highlight cutting-edge technologies that can foster the development of innovative strategies, optimize drug co-administration, improve treatment outcomes, and reduce the burden of atherosclerosis-related morbidity and mortality on the healthcare system.
Collapse
Affiliation(s)
- Yingxuan Dai
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Li Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Guosheng Cao
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China
| | - Liqing Mo
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Can Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yuxi Zhu
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210, USA; Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, OH 44106, USA
| | - Yujie Guo
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yi Hong
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Hanlin Xu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Shan Lu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China.
| | - Shi Du
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210, USA; Division of Pharmaceutics and Pharmacology, College of Pharmacy, Ohio State University, Columbus, OH 43210, USA.
| | - Jianhua He
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China.
| |
Collapse
|
32
|
Kou H, Wang J, Yannie PJ, Huang D, Korzun WJ, Kakiyama G, Ghosh SS, Yang H. Enhanced Cholesterol Efflux and Atherosclerosis Regression via CEH Gene Delivery Using Galactose-Functionalized Dendrimeric Nanoparticles. ACS Pharmacol Transl Sci 2025; 8:1359-1365. [PMID: 40370995 PMCID: PMC12070320 DOI: 10.1021/acsptsci.5c00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/29/2025] [Accepted: 04/02/2025] [Indexed: 05/16/2025]
Abstract
Cholesteryl ester hydrolase (CEH) is a critical enzyme in cholesterol ester hydrolysis, influencing cholesterol metabolism and efflux. This study demonstrates that CEH overexpression promotes free cholesterol efflux from macrophages, thereby reducing the lipid burden in existing atherosclerotic plaques. To enable targeted delivery, galactose-functionalized polyamidoamine (PAMAM) dendrimeric nanoparticles were utilized as nanocarriers for hepatic delivery of the CEH expression vector. The therapeutic potential of CEH plasmid-loaded dendrimeric nanoparticles was evaluated in Ldlr-/- mice. Results showed a significant reduction in total lesion area (21%) and aortic arch lesion area (23%) compared to baseline. Lesion component analysis revealed marked decreases in total cholesterol (36%), free cholesterol (35%), and cholesterol esters (44%). Collectively, these results support CEH overexpression as an effective strategy to enhance cholesterol efflux and mitigate lipid accumulation in atherosclerotic plaques. Moreover, galactose-functionalized PAMAM dendrimeric nanoparticles demonstrate strong potential as a targeted hepatic gene delivery system for therapeutic intervention in atherosclerosis.
Collapse
Affiliation(s)
- Huari Kou
- Linda
and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Jing Wang
- Department
of Internal Medicine, Virginia Commonwealth
University Medical Center, Richmond, Virginia 23298, United States
| | - Paul J. Yannie
- Department
of Safety and Risk Management, Virginia
Commonwealth University, Richmond, Virginia 23298, United States
| | - Da Huang
- Linda
and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
- College
of Biological Science and Engineering, Fuzhou
University, Fuzhou, Fujian 350108, China
| | - William J. Korzun
- Department
of Clinical Laboratory Sciences, Virginia
Commonwealth University, Richmond, Virginia 23298, United States
| | - Genta Kakiyama
- Department
of Internal Medicine, Virginia Commonwealth
University Medical Center, Richmond, Virginia 23298, United States
- Research
Services, Central Virginia VA Health Care System, Richmond, Virginia 23249, United States
| | - Siddhartha S. Ghosh
- Department
of Internal Medicine, Virginia Commonwealth
University Medical Center, Richmond, Virginia 23298, United States
| | - Hu Yang
- Linda
and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| |
Collapse
|
33
|
Gao C, Zhu J, Wu F, Cui Z, Fang M, Zhu Z, He B. Simulation and experimental study on processing behavior of coronary artery calcified tissue removal. Sci Rep 2025; 15:16116. [PMID: 40341621 PMCID: PMC12062317 DOI: 10.1038/s41598-025-01236-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 05/05/2025] [Indexed: 05/10/2025] Open
Abstract
Coronary artery atherosclerosis is a prevalent cardiovascular disease and a leading cause of major adverse cardiovascular events (MACE). Rotational atherectomy (RA) is an effective interventional technique for treating severe calcified stenosis. However, excessive forces, heat, and debris are prone to lead to serious surgical complications, such as slow flow/no-reflow and blood clots. To mitigate excessive force and heat generation during RA, a novel high-performance cutting tool was designed and fabricated for coronary artery calcified tissue removal. An RA simulation model was developed to simulate the procedure. The results showed that the forces, temperatures, and debris size remained within predefined safety thresholds. Using the 1.5 mm tool as an illustration, the peak cutting force was 1.062 N, and the peak temperature rise reached 1.170 °C. Debris distribution exhibited a normal pattern, with 90% of particles measuring below 14 μm. The experimental results closely matched the simulation values, showcasing errors under 10% and affirming the simulation model's precision. This research provides theoretical support for the study of mechanisms and contributes to optimizing the effectiveness of RA.
Collapse
Affiliation(s)
- Chuhang Gao
- School of Mechanical Engineering and Automation, Fuzhou University, Fuzhou, 350108, China
- Fujian Engineering Research Center of Joint Intelligent Medical Engineering, Fuzhou, 350108, China
| | - Jialiang Zhu
- School of Mechanical Engineering and Automation, Fuzhou University, Fuzhou, 350108, China
- Fujian Engineering Research Center of Joint Intelligent Medical Engineering, Fuzhou, 350108, China
| | - Fan Wu
- School of Mechanical Engineering and Automation, Fuzhou University, Fuzhou, 350108, China
- Fujian Engineering Research Center of Joint Intelligent Medical Engineering, Fuzhou, 350108, China
| | - Ziyu Cui
- School of Mechanical Engineering and Automation, Fuzhou University, Fuzhou, 350108, China
- Fujian Engineering Research Center of Joint Intelligent Medical Engineering, Fuzhou, 350108, China
| | - Mingcheng Fang
- Fujian Provincial Hospital, Fuzhou, 350001, China.
- Department of Cardiology, Fujian Provincial Hospital, Fuzhou, 350001, China.
- Fuzhou University Town, No. 2 Wulongjiang North Avenue, Fuzhou City, Fuzhou, Fujian Province, China.
| | - Zhaoju Zhu
- School of Mechanical Engineering and Automation, Fuzhou University, Fuzhou, 350108, China.
- Fujian Engineering Research Center of Joint Intelligent Medical Engineering, Fuzhou, 350108, China.
| | - Bingwei He
- School of Mechanical Engineering and Automation, Fuzhou University, Fuzhou, 350108, China
- Fujian Engineering Research Center of Joint Intelligent Medical Engineering, Fuzhou, 350108, China
| |
Collapse
|
34
|
Kong DZ, Zhang XZ, Lu Y, Zhou YY, Pan YH, Zhao XQ, Wu XY, Li WW, Ye H. Jian-pi Qu-tan Hua-yu Decoction improves oxidative stress-induced inflammation in endothelial cells in atherosclerotic ApoE-/- mice through the NOX1-ROS-ERK1/2 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156815. [PMID: 40398180 DOI: 10.1016/j.phymed.2025.156815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 04/20/2025] [Accepted: 04/26/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Atherosclerosis (AS) is a key mechanism in cardiovascular diseases and a major target for interventions. Jian-pi Qu-tan Hua-yu Decoction (JPQTHYD), a herbal formula, has been shown to alleviate AS. OBJECTIVE This study aimed to evaluate the therapeutic effect of JPQTHYD on AS and explore its molecular mechanisms. MATERIALS AND METHODS In vivo, we established a mouse model through a high-fat diet for 16 weeks combined with a 4-week exhaustive swimming experiment. The body weight and food intake of the mice were measured every 4 weeks. At the end of the 16 weeks, the moisture content of the mice's feces was measured, the morphology of the thoracic aorta and myocardium was observed using HE staining, and lipid deposition in the aorta and myocardium was assessed using Oil Red O staining. The ultrastructure of myocardial tissue was observed via transmission electron microscopy. Levels of TC, TG, and LDL-C were measured using an automatic biochemical analyzer. ELISA was used to detect the levels of ROS, IL-6, IL-10, TNF-α, hs-CRP, VCAM-1, and ICAM-1. In vitro, we induced HUVECs injury using 700 nM of Ang II. Cell viability was assessed using the CCK-8 assay, while ROS levels were measured by a ROS detection kit. NOX1 gene suppression was achieved using a NOX1 inhibitor. Protein expression levels of NOX1, ERK1/2, P-ERK1/2, VCAM-1, and ICAM-1 were measured by Western blot both in vivo and in vitro. RESULTS 1. JPQTHYD improved fecal water content and exercise capacity in ApoE-/- mice. 2. JPQTHYD reduced TC, TG, LDL-C levels, decreased arterial intimal thickness, and inhibited atherosclerotic plaque formation. 3. JPQTHYD decreased proinflammatory factors and adhesion molecules by inhibiting the NOX1-ROS-ERK1/2 pathway. 4. In vitro, JPQTHYD suppressed endothelial inflammation by reducing NOX1-ROS-ERK1/2 signaling. CONCLUSION JPQTHYD reduced blood lipids, inhibited oxidative stress-induced inflammation, and alleviated AS in ApoE-/- mice, likely through the NOX1-ROS-ERK1/2 pathway. This study offers a novel investigation into the mechanisms and regulatory pathways through which traditional Chinese medicine contributes to the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- De Zhao Kong
- Peking University First Hospital, Beijing, PR China; Peking University Health Science Center, Beijing, PR China; Peking University First Hospital Taiyuan Branch (Taiyuan Central Hospital of Shanxi Medical University), PR China.
| | - Xue Zhi Zhang
- Peking University First Hospital, Beijing, PR China.
| | - Yi Lu
- Tiantai Hospital of Traditional Chinese Medicine, Taizhou, PR China.
| | - Yuan Yuan Zhou
- Liaoning University of Traditional Chinese Medicine, Shenyang, PR China.
| | - Yi Hui Pan
- Liaoning University of Traditional Chinese Medicine, Shenyang, PR China.
| | - Xin Qi Zhao
- Liaoning University of Traditional Chinese Medicine, Shenyang, PR China.
| | - Xin Yu Wu
- Liaoning University of Traditional Chinese Medicine, Shenyang, PR China.
| | - Wei Wei Li
- Peking University First Hospital, Beijing, PR China.
| | - Hui Ye
- Peking University First Hospital, Beijing, PR China.
| |
Collapse
|
35
|
Zhang H, Zhang X, Yun Z, Chen Y, Cang S, Shao Y, Jia E, Chen R. Loss of diurnal oscillatory rhythms in gut microbiota correlates with progression of atherosclerosis. Food Funct 2025; 16:3423-3438. [PMID: 40201963 DOI: 10.1039/d4fo05227g] [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: 04/10/2025]
Abstract
Circadian rhythms in gut microbiota composition are crucial for metabolic function and disease progression, yet the diurnal oscillation patterns of gut microbiota in atherosclerotic cardiovascular disease (ASCVD) and their role in disease progression remain unknown. Here, we investigated gut bacterial dynamics in Apoe-/- mice over 24 hours and elucidated dynamic changes in fecal microbiota composition and function among C57BL/6 and Apoe-/- mice with standard chow diet or high-fat/high-cholesterol diet under ad libitum conditions. Compared with C57BL/6 mice, Apoe-/- mice exhibited significant differences in fecal microbial composition. Rhythmicity analysis revealed that the temporal dynamics of fecal microbiota composition and function in Apoe-/- mice differed significantly from those in C57BL/6 mice, particularly in B. coccoides-dominated oscillatory modules. Functional annotation showed that rhythmic B. coccoides strains inhibited ASCVD progression by enhancing intestinal and endothelial barrier functions. These findings demonstrate that diurnal oscillations in gut microbiota are closely associated with ASCVD progression and provide new insights for microbiota-targeted precision therapies.
Collapse
Affiliation(s)
- He Zhang
- School of Life Sciences, Xuzhou Medical University, 221004, Xuzhou, China.
| | - Xiaohan Zhang
- School of Life Sciences, Xuzhou Medical University, 221004, Xuzhou, China.
| | - Zihan Yun
- School of Life Sciences, Xuzhou Medical University, 221004, Xuzhou, China.
| | - Yang Chen
- School of Life Sciences, Xuzhou Medical University, 221004, Xuzhou, China.
| | - Suhua Cang
- School of Life Sciences, Xuzhou Medical University, 221004, Xuzhou, China.
| | - Yating Shao
- School of Life Sciences, Xuzhou Medical University, 221004, Xuzhou, China.
| | - Erteng Jia
- Thoracic Surgery Laboratory, the First College of Clinical Medicine, Xuzhou Medical University, 221004, Xuzhou, China.
| | - Renjin Chen
- School of Life Sciences, Xuzhou Medical University, 221004, Xuzhou, China.
| |
Collapse
|
36
|
Cao Y, Tang K, Feng J, Wang W, Jin H, Zheng Z, Lin B, Ding Y, Yang S. Knowledge, attitudes, and practice toward atherosclerosis and ultrasound screening in adults in Shanghai. Sci Rep 2025; 15:15480. [PMID: 40319079 PMCID: PMC12049462 DOI: 10.1038/s41598-025-00561-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 04/29/2025] [Indexed: 05/07/2025] Open
Abstract
This study investigates the knowledge, attitudes, and practices (KAP) regarding atherosclerosis ultrasound screening among adults in the general population of Shanghai. A cross-sectional study was conducted from April 5 to May 30, 2024, at the Department of Ultrasound Medicine, Shanghai Eighth People's Hospital, involving the general public. An investigator-designed questionnaire collected demographic information and KAP scores. Path analysis was performed to assess the relationships between overall KAP scores, with subgroup analysis for participants with atherosclerotic cardiovascular disease (CVD). A total of 617 valid questionnaires were analyzed, revealing a high reliability coefficient of 0.949. Mean KAP scores were as follows: knowledge 9.73 ± 5.80 (/24, 40.54%), attitude 56.38 ± 10.94 (/65, 86.74%), and practice 21.66 ± 5.54 (/35, 61.89%), indicating poor knowledge but positive attitudes and proactive practices. Knowledge significantly influenced both attitude (β = 0.354, P = 0.028) and practice (β = 0.357, P = 0.021). In participants with CVD, mean scores were 11.14 ± 4.10 for knowledge, 59.69 ± 11.94 for attitude, and 23.17 ± 5.32 for practice. In the general Shanghai population, knowledge about atherosclerosis ultrasound screening is poor, yet attitudes are positive, and practices are proactive. Targeted educational interventions are necessary to enhance knowledge and attitudes, thus improving screening practices.
Collapse
Affiliation(s)
- Yan Cao
- Department of Ultrasound, Shanghai Eighth People's Hospital, Shanghai, 200235, China
| | - Keqiang Tang
- Department of Intensive Care Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital, Shanghai, China
| | - Jiangbo Feng
- Department of Ultrasound, Shanghai Eighth People's Hospital, Shanghai, 200235, China
| | - Wenwen Wang
- Department of Ultrasound, Shanghai Eighth People's Hospital, Shanghai, 200235, China
| | - Hua Jin
- Department of Ultrasound, Shanghai Kongjiang Hospital, Shanghai, 200093, Postcode, China
| | - Ziwei Zheng
- Department of Ultrasound, Shanghai Eighth People's Hospital, Shanghai, 200235, China
| | - Bo Lin
- Department of Ultrasound, Shanghai Eighth People's Hospital, Shanghai, 200235, China
| | - Yueyou Ding
- Department of Cardiology, Shanghai Eighth People's Hospital, Shanghai, 200235, China.
| | - Shaoling Yang
- Department of Ultrasound, Shanghai Eighth People's Hospital, Shanghai, 200235, China.
| |
Collapse
|
37
|
Zhou Y, Guo Y. Circ-PSMB1 knockdown inhibits the pyroptosis of ox-LDL treated human aortic cells via the miR-624-3p/ASC axis. J Cardiothorac Surg 2025; 20:226. [PMID: 40317040 PMCID: PMC12048961 DOI: 10.1186/s13019-025-03457-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: 12/17/2024] [Accepted: 04/21/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND Atherosclerosis (AS) is a cardiovascular disease that is caused by a variety of factors, including hypertension, diabetes, hyperlipidaemia and smoking. Circular RNAs (circRNAs) have been reported to participate in the progression of AS. Here, we investigated the mechanism by which circ-proteasome 20 S subunit beta 1 (PSMB1) participates in AS. METHODS HAECs were stimulated with oxidized low-density lipoprotein (ox-LDL) to establish a model of AS in vitro. Cell viability was investigated with MTT assays. Western blotting and qRT‒PCR were used to measure relative protein and mRNA expression. Cell pyroptosis was analysed by flow cytometry. Lactate dehydrogenase (LDH) levels were measured with a commercial kit. RESULTS We found that circ-PSMB1 and apoptosis-associated speck-like protein containing a CARD (ASC) were overexpressed and miR-624-3p was expressed at low levels in HAECs treated with ox-LDL. Circ-PSMB1 silencing enhanced cell viability and decreased pyroptosis, as shown by the downregulation of IL-1β and IL-18 mRNA expression as well as NOD-like receptor thermal protein domain associated protein 3 (NLRP3) and GasderminD-N (GSDMD-N) protein expression. In addition, the miR-624-3p inhibitor neutralized the effects of si-circ-PSMB1, and ASC overexpression neutralized the effects of the miR-624-3p mimic in ox-LDL-treated HAECs. CONCLUSION This research demonstrated that circ-PSMB1 might participate in AS development through regulating the pyroptosis of HAECs via the miR-624-3p/ASC axis.
Collapse
Affiliation(s)
- Yupu Zhou
- Department of Vascular Surgery, Jiangjin Centre Hospital, No. 725, Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing, Chongqing, 402260, China
| | - Yongchuan Guo
- Department of Vascular Surgery, Jiangjin Centre Hospital, No. 725, Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing, Chongqing, 402260, China.
| |
Collapse
|
38
|
Lai Z, Kong D, Li Q, Wang Y, Li K, Duan X, Shao J, Xie Y, Chen J, Zhang T, Feng Y, Deng H, Wang J, Wang C, Shu K, Zhao H, Du H, Jia C, Dai H, Xie L, Liu J, Luo X, Wang L, Xu L, Zhu Z, Lei X, Wang Y, Yang Y, Liu Y, Liang Y, Yang Y, Xie J, Liu B, Deng Z, Liu X. Single-cell spatial transcriptomics of tertiary lymphoid organ-like structures in human atherosclerotic plaques. NATURE CARDIOVASCULAR RESEARCH 2025; 4:547-566. [PMID: 40295810 DOI: 10.1038/s44161-025-00639-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/20/2025] [Indexed: 04/30/2025]
Abstract
Tertiary lymphoid organs have been identified in the arterial adventitia in both mouse models of atherosclerosis and patients with atherosclerosis, yet their role in the disease remains insufficiently explored. Here we present a spatially resolved single-cell transcriptome atlas of human atherosclerotic plaques, identifying 14 distinct cell types and providing evidence of plaque tertiary lymphoid organs (PTLOs). The development of PTLOs was associated with the expression of lymphangiogenic chemokine genes and the adhesion molecule gene in fibroblast-like smooth muscle cells. PTLOs harbor abundant B cells with expanded and diversified B cell receptors, suggesting substantial immune involvement. We also observed that B cells may be exchanged between PTLOs and perivascular adipose tissues. The presence of PTLO-like structures correlates with cerebrovascular events, which may be mediated by PTLO-derived IgG antibodies enhancing macrophage functional activity. Our findings suggest the existence and characteristics of PTLOs in human atherosclerosis, elucidating their cellular functions and clinical implications and offering avenues for understanding, diagnosing and treating this condition.
Collapse
Affiliation(s)
- Zhichao Lai
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Deqiang Kong
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | - Yue Wang
- BGI Research, Beijing, China
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Kang Li
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaohan Duan
- BGI Research, Beijing, China
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiang Shao
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yiyun Xie
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Junye Chen
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Tianjing Zhang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yuyao Feng
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | - Jiaxian Wang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Chaonan Wang
- Department of Hemangiomas and Vascular Malformations, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Keqiang Shu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Hongmei Zhao
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hanze Du
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Translation Medicine Centre, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Congwei Jia
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Huanyu Dai
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Lizhi Xie
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | | | - Lin Wang
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Leyin Xu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhan Zhu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiangling Lei
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yuru Wang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yixuan Yang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yanan Liu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | | | - Jun Xie
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China
| | - Bao Liu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| | | | - Xin Liu
- BGI Research, Beijing, China.
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
39
|
He Y, Wang J, Chen C, Wang R, Ma X, Ma R, Sun Y, Wang L, Ding H. Comprehensive profiling of tsRNAs in acute coronary syndrome: expression patterns, clinical correlations, and functional insights. Hum Genet 2025; 144:575-590. [PMID: 40232417 PMCID: PMC12033100 DOI: 10.1007/s00439-025-02742-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/28/2025] [Indexed: 04/16/2025]
Abstract
Transfer RNA-derived small RNAs (tsRNAs) have emerged as potential biomarkers of various human diseases. However, the clinical utility and biological functions of tsRNA in acute coronary syndrome (ACS) remain poorly understood. To investigate this, we performed high-throughput small RNA sequencing on peripheral blood monocyte cells (PBMCs) from 24 ACS patients and 12 healthy controls. Our analysis revealed distinct and characteristic expression patterns of tsRNAs in response to ACS, highlighting their potential as disease signatures in human PBMCs. Differentially expressed tsRNAs were validated using RT-qPCR in two independent case-control sets. Among these, tRF-Gly-GCC-06 was significantly upregulated in volunteers with unstable angina (UA) and acute myocardial infarction (AMI) (p < 0.05) and showed a statistically significant positive correlation with the Gensini score (r = 0.353, p < 0.001). Moreover, this tsRNA was independently associated with an increased risk of ACS after adjusting for conventional cardiovascular risk factors (odds ratio (OR) = 1.58, 95% confidence interval (CI): 1.37-1.83, p < 0.001). A series of functional studies showed that tRF-Gly-GCC-06 significantly facilitated macrophage proliferation and migration and modulated inflammation-related gene expression in vitro. This study identified a novel functional gene associated with ACS, tRF-Gly-GCC-06, as a potential clinical biomarker and therapeutic target.
Collapse
Affiliation(s)
- Yi He
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan, 430030, P.R. China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, P.R. China
| | - Jing Wang
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan, 430030, P.R. China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, P.R. China
| | - Chen Chen
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan, 430030, P.R. China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, P.R. China
| | - Rongli Wang
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan, 430030, P.R. China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, P.R. China
| | - Xiaozhu Ma
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan, 430030, P.R. China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, P.R. China
| | - Ruiying Ma
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan, 430030, P.R. China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, P.R. China
| | - Yang Sun
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan, 430030, P.R. China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, P.R. China
| | - Luyun Wang
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan, 430030, P.R. China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, P.R. China.
- Genetic Diagnosis Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, P.R. China.
| | - Hu Ding
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan, 430030, P.R. China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, P.R. China.
- Genetic Diagnosis Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, P.R. China.
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, P.R. China.
| |
Collapse
|
40
|
Song C, Yang J, Gu Z. Latest developments of microphysiological systems (MPS) in aging-related and geriatric diseases research: A review. Ageing Res Rev 2025; 107:102728. [PMID: 40058462 DOI: 10.1016/j.arr.2025.102728] [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: 12/02/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Aging is a gradual and irreversible process accompanied by the decline in tissue function and a significantly increased risk of various aging-related and geriatric diseases. Especially in the paradoxical context of accelerated global aging and the widespread emergence of pandemics, aging-related and geriatric diseases have become leading causes of individual mortality and disability, drawing increasing attention from researchers and investors alike. Despite the utility of current in vitro systems and in vivo animal models for studying aging, these approaches are limited by insurmountable inherent constraints. In response, microphysiological systems (MPS), leveraging advances in tissue engineering and microfluidics, have emerged as highly promising platforms. MPS are capable of replicating key features of the tissue microenvironment within microfabricated devices, offering biomimetic tissue culture conditions that enhance the in vitro simulation of intact or precise human body structure and function. This capability improves the predictability of clinical trial outcomes while reducing time and cost. In this review, we focus on recent advancements in MPS used to study age-related and geriatric diseases, with particular emphasis on the application of organoids and organ-on-a-chip technologies in understanding cardiovascular diseases, cerebrovascular diseases, neurodegenerative diseases, fibrotic diseases, locomotor and sensory degenerative disorders, and rare diseases. And we aim to provide readers with critical guidelines and an overview of examples for modeling age-related and geriatric diseases using MPS, exploring mechanisms, treatments, drug screening, and other subsequent applications, from a physiopathological perspective, emphasizing the characteristic of age-related and geriatric diseases and their established correlations with the aging process. We also discuss the limitations of current models and propose future directions for MPS in aging research, highlighting the potential of interdisciplinary approaches to address unresolved challenges in the field.
Collapse
Affiliation(s)
- Chao Song
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Jiachen Yang
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Zhongze Gu
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing, China.
| |
Collapse
|
41
|
Sun Q, Jing M, Xi H, Ren W, Zhu H, Wang Y, Liu Q, Zhou J. Different plaque types and its association with the volume and attenuation of pericoronary adipose tissue as assessed by coronary computed tomography angiography. Clin Radiol 2025; 84:106814. [PMID: 40069975 DOI: 10.1016/j.crad.2025.106814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 11/15/2024] [Accepted: 01/09/2025] [Indexed: 04/20/2025]
Abstract
AIM To explore the relationship between different plaque types and pericoronary adipose tissue (PCAT) volume and attenuation values in patients with stable coronary artery disease (CAD) based on coronary computed tomographic angiography (CCTA). MATERIALS AND METHODS Three hundred twenty one patients with stable CAD who underwent CCTA from May 2022 to March 2023 were enrolled. Using semi-automatic software, PCAT volumes and CT attenuation values were measured around the plaque and in the segment and proximal coronary artery where the plaque was located. To compare whether there was a statistical difference in PCAT volume and attenuation values among different plaque types in the periplaque, the segment, and proximal coronary artery in which the plaque was positioned. RESULTS In total, 552 lesions were included, with 299 calcified plaques (CPs), 174 noncalcified plaques (NCPs), and 79 mixed plaques (MPs). There were excellent agreements between the two radiologists regarding the measured PCAT volumes and attenuation values (all interclass correlation coefficients values > 0.80). The periplaque PCAT volume was larger in CPs and MPs than in NCPs (291.98[213.25,381.03] mm3 vs. 261.00[173.25,377.85] mm3 vs. 206.54[139.72,284.07] mm3, P < 0.05), and the PCAT attenuation values around the plaque and the segment in which the plaque was positioned were higher in NCPs and MPs compared with CPs (-73.00[-79.00,-68.00] HU vs. -76.00[-79.00,-71.00] HU vs. -85.00[-92.00,-80.00] HU, -81.72 ± 0.70 HU vs. -80.73 ± 1.03 HU vs. -84.31 ± 0.49 HU; P < 0.05). CONCLUSION PCAT volume and attenuation values differed significantly among different plaque types, and the differences are particularly significant in measurements around the plaque.
Collapse
Affiliation(s)
- Q Sun
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - M Jing
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - H Xi
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - W Ren
- GE Healthcare, Computed Tomography Research Center, Beijing, China
| | - H Zhu
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Y Wang
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Q Liu
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - J Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China.
| |
Collapse
|
42
|
He J, Dai Y, Xu F, Huang X, Gao Y, Liu L, Zhang W, Liu J. High-density lipoprotein-based nanoplatforms for macrophage-targeted diagnosis and therapy of atherosclerosis. Int J Biol Macromol 2025; 306:140826. [PMID: 40010459 DOI: 10.1016/j.ijbiomac.2025.140826] [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: 11/01/2024] [Revised: 01/19/2025] [Accepted: 02/07/2025] [Indexed: 02/28/2025]
Abstract
Atherosclerosis, the primary cause of cardiovascular disease, which has the highest mortality worldwide, is a chronic inflammatory disease mainly induced by excessive lipid accumulation in plaque macrophages. Lipid-laden macrophages are crucial at all stages of atherosclerotic lesion progression and are, thus, regarded as popular therapeutic targets for atherosclerosis. High-density lipoprotein (HDL), an endogenous particle with excellent atherosclerotic plaque-homing properties, is considered a potential therapeutic agent for treating atherosclerosis. Based on the excellent properties of HDL, reconstituted HDL (rHDL), with physiological functions similar to those of its natural counterparts, have been successfully prepared as therapeutics and are also recognized as a potential nanoplatform for delivering drugs or contrast agents to atherosclerotic plaques owing to their high biocompatibility, amphiphilic structure, and macrophage-targeting capability. In this review, we focus on the (a) important role of macrophages in atherosclerotic lesions, (b) biological properties of rHDL as a delivery nanoplatform in atherosclerotic diseases, and (c) multiple applications of rHDL in the diagnosis and treatment of atherosclerosis. We systematically summarize the novel applications of rHDL with unique advantages in atherosclerosis, aiming to provide specific insights and inspire additional innovative research in this field.
Collapse
Affiliation(s)
- Jianhua He
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China; School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yingxuan Dai
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Fengfei Xu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China
| | - Xinya Huang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China
| | - Yu Gao
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China
| | - Lisha Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China.
| | - Jianping Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China.
| |
Collapse
|
43
|
He X, Li Z, Li S, Zhang X, Liu D, Han X, He H, Chen J, Dong X, Long W, Lu H, Ye T, Meng F, Liao H, Yang Z, Lu L, Ni S. Huoxue Tongluo tablet enhances atherosclerosis efferocytosis by promoting the differentiation of Trem2 + macrophages via PPARγ signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156579. [PMID: 40068297 DOI: 10.1016/j.phymed.2025.156579] [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: 09/23/2024] [Revised: 02/07/2025] [Accepted: 02/24/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Atherosclerosis (AS) serves as the primary pathological basis for various cardiovascular and cerebrovascular diseases. Impaired efferocytosis by macrophages within AS plaques exacerbates lipid metabolism disorders and inflammatory responses. Huoxue Tongluo Tablet (HXTL), a traditional Chinese medicine formula, has shown efficacy in treating AS and modulating macrophage function. However, its underlying mechanisms remain unclear. It is hypothesized that HXTL ameliorates AS by enhancing macrophage efferocytosis. PURPOSE To assess the efficacy and mechanisms of HXTL in treating AS at the single-cell level. METHODS Ultra-high-performance liquid chromatography-mass spectrometry (UPLC-MS/MS) was used to analyze the constituents of HXTL. HXTL was administered to ApoE⁻/⁻ mice maintained on a high-fat diet. The progression of AS was evaluated by measuring atherosclerotic plaque area, necrotic core formation, collagen depletion, lipid accumulation, lipid profiles, pro-inflammatory mediators, and oxidative stress markers. Transcriptomic analysis was performed to explore the mechanisms underlying the therapeutic effects of HXTL on AS. Efferocytosis-related marker expression was evaluated using immunohistochemistry and quantitative PCR (qPCR), and the efferocytosis index was determined by the co-localization of apoptotic cells and macrophages. Efferocytosis inhibition was induced using Cytochalasin D. Single-cell sequencing was utilized to investigate alterations in Trem2⁺ macrophages following HXTL treatment. Trem2 expression was accessed by immunohistochemistry and qPCR, while flow cytometry and immunofluorescence staining confirmed the changes in Trem2⁺ macrophages. Bioinformatic analyses were conducted to investigate the mechanism through which HXTL enhances efferocytosis by regulating Trem2⁺ macrophage subsets. Western blotting and qPCR were used to assess the expression levels of PPARγ signaling, and the regulatory role of PPARγ signaling in macrophage subpopulation generation and efferocytosis function was accessed using GW9662. RESULTS UPLC-MS/MS analysis identified 99 major components in HXTL. In vivo, medium and high doses of HXTL significantly reduced atherosclerotic plaque area, improved lipid profiles, decreased pro-inflammatory mediators and reactive oxygen species (ROS), and enhanced the efferocytosis function. Inhibition of efferocytosis reversed these beneficial effects. Single-cell sequencing and in vivo validation revealed that HXTL upregulated Trem2⁺ macrophages and efferocytosis-related genes. Bioinformatics and in vivo experiments demonstrated that HXTL activated PPARγ signaling, and inhibition of PPARγ signaling negated the pro-efferocytosis effects and the upregulation of Trem2⁺ macrophage upregulation induced by HXTL. CONCLUSIONS HXTL activates the PPARγ pathway, upregulates Trem2⁺ macrophages, and enhances macrophage efferocytosis, thereby ameliorating AS. This study is the first to demonstrate the regulatory effects of HXTL on macrophage subpopulations and its pro-efferocytosis activity.
Collapse
Affiliation(s)
- Xingling He
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Geriatrics Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangdong Clinical Research Institute of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Ziru Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Geriatrics Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangdong Clinical Research Institute of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Sijing Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Geriatrics Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangdong Clinical Research Institute of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xiaojiao Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Geriatrics Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangdong Clinical Research Institute of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Donghua Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Geriatrics Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangdong Clinical Research Institute of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xiaowei Han
- Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Huan He
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Geriatrics Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangdong Clinical Research Institute of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jiahui Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Geriatrics Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangdong Clinical Research Institute of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xiaoming Dong
- Geriatrics Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Wenjie Long
- Geriatrics Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Huan Lu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Taochun Ye
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Fanhang Meng
- Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Huili Liao
- Geriatrics Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Zhongqi Yang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Geriatrics Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangdong Clinical Research Institute of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Lu Lu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Geriatrics Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangdong Clinical Research Institute of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Shihao Ni
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Geriatrics Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangdong Clinical Research Institute of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| |
Collapse
|
44
|
Kou H, Qi L, Huang D, Wu J, Shi H, Yang H. Scalable Fabrication of High-Payload Dendrimer-Based Nanoparticles for Targeted Atherosclerosis Therapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:24953-24962. [PMID: 40237537 DOI: 10.1021/acsami.5c00816] [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: 04/18/2025]
Abstract
Nanoparticle-based therapeutics hold promise for the treatment of atherosclerosis, but challenges such as low drug-loading capacity and a lack of scalable, controllable production hinder their clinical translation. Flash nanoprecipitation, a continuous synthesis method, offers a potential solution for scalable and reproducible nanoparticle production. In this study, we employed a custom-designed multi-inlet vortex mixer to perform cross-linking reaction-enabled flash nanoprecipitation, facilitating controlled and scalable synthesis of cross-linked polyamidoamine (PAMAM) dendrimer nanoparticles. Notably, this approach allows simultaneous nanoparticle cross-linking and drug loading in a single step. The mannose moiety enabled specific targeting of macrophages via mannose receptors, enhancing the localization of the nanoparticles to atherosclerotic plaques. Atorvastatin calcium, a widely used clinical drug for atherosclerosis treatment, was selected as the model drug. This approach achieved both high production rates and high drug-loading capacities, with an output flow rate of 9.6 L/h and a nanoparticle concentration of approximately 0.4 g/L. The optimized formulation exhibited a drug-loading capacity of 37% and an encapsulation efficiency of 76%. In vitro and in vivo experiments demonstrated effective macrophage and plaque targeting, leading to significant therapeutic benefits. Treatment with these nanoparticles resulted in approximately 40% inhibition of aortic root plaque progression compared to the free drug-treated group. This scalable and efficient nanoparticle platform is a promising strategy for improving atherosclerosis treatment.
Collapse
Affiliation(s)
- Huari Kou
- Linda and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Lin Qi
- Linda and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Da Huang
- Linda and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Jiandong Wu
- Linda and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Honglan Shi
- Department of Chemistry, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Hu Yang
- Linda and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| |
Collapse
|
45
|
Liu MN, Liu ZH, Leng RX, Strijdom H, Weng JP, Xu SW. Revisiting the role of GDF15 in atherosclerosis in mouse and human. Acta Pharmacol Sin 2025:10.1038/s41401-025-01561-3. [PMID: 40307459 DOI: 10.1038/s41401-025-01561-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025]
Abstract
Growth differentiation factor 15 (GDF15) is a key regulator of food intake and energy metabolism. GDF15 mimetic drugs for the treatment of metabolic syndrome and obesity are under clinical development. While GDF15 presents a promising target for weight management, its potential cardiovascular actions remain elusive. In this study we investigated the role of GDF15 in macrophage function and atherosclerosis pathogenesis and whether GDF15 acts both as a biomarker and mediator of atherosclerosis severity. ApoE-/- mice were fed a high-cholesterol diet (HCD, 1.25% cholesterol) for 6, 12 or 18 weeks to establish atherosclerotic models. We showed that serum levels of GDF15 were elevated in ApoE-/- mice with atheroprogression; increased serum levels of GDF15 were also observed in patients with coronary artery disease. Enlightened by this finding, we established atherosclerotic model in Gdf15-/- mice by injecting with AAV8-PCSK9D377Y virus and feeding HCD for 12 or 16 weeks. We showed that global Gdf15 knockout, whether in male or female mice, did not alter plaque size in en face aorta, lesion in aortic sinus, size of necrotic core or plaque composition. In macrophage-derived foam cells isolated from atherosclerotic mice, neither Gdf15 deletion nor the treatment with recombinant GDF15 protein (1, 10, 100 ng/mL) affected lipid deposition or macrophage polarization. To translate this finding into a clinically relevant scenario, we performed Mendelian randomization (MR) analysis, and found no significant causal association between circulating GDF15 levels and the incidence of cardiovascular diseases. Furthermore, MR studies suggest that genetic associations between GDF15 and factors such as BMI, ApoB, LDL and HDL were not significant in plasma data from the UK Biobank and the deCODE cohort. In summary, this study demonstrates that global Gdf15 deficiency does not affect the development of atherosclerosis in male or female mice despite the positive association between circulating GDF15 levels and disease progression in mice and human. Thus, GDF15 in circulation is a potential biomarker, but not a causal mediator, of atherosclerosis. Long-term cardiovascular safety of GDF15-targeted therapies warrants further investigation.
Collapse
Affiliation(s)
- Mo-Nan Liu
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Zheng-Hong Liu
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Rui-Xue Leng
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Hans Strijdom
- Centre for Cardio-metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Jian-Ping Weng
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
- Anhui Provincial Key Laboratory of Metabolic Health and Panvascular Diseases, Hefei, 230001, China
- Institute of Endocrine and Metabolic Diseases, University of Science and Technology of China, Hefei, 230001, China
| | - Suo-Wen Xu
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
- Anhui Provincial Key Laboratory of Metabolic Health and Panvascular Diseases, Hefei, 230001, China.
- Institute of Endocrine and Metabolic Diseases, University of Science and Technology of China, Hefei, 230001, China.
| |
Collapse
|
46
|
Wang L, Zhang Y, Yue J, Zhou R. The Role of Ubiquitination on Macrophages in Cardiovascular Diseases and Targeted Treatment. Int J Mol Sci 2025; 26:4260. [PMID: 40362498 PMCID: PMC12072125 DOI: 10.3390/ijms26094260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality worldwide, with macrophage dysfunction playing a central role in its pathogenesis. Ubiquitination, a critical post-translational modification, regulates diverse macrophage functions, including lipoprotein metabolism, inflammation, oxidative stress, mitophagy, autophagy, efferocytosis, and programmed cell death (pyroptosis, necroptosis, ferroptosis, and apoptosis). This review highlights the regulatory roles of ubiquitination in macrophage-driven CVD progression, focusing on its effects on cholesterol metabolism, inflammation, activation, polarization, and the survival of macrophages. Targeting ubiquitination pathways has therapeutic potential by enhancing macrophage autophagy, reducing inflammation, and improving plaque stability. However, challenges, such as off-target effects, ubiquitination crosstalk, and macrophage heterogeneity, must be addressed. By integrating advances in ubiquitination biology, therapeutic strategies can be developed to mitigate CVD and other macrophage-driven inflammatory diseases. This review underscores the potential of ubiquitination-targeting therapies for mitigating CVD and highlights the key areas for further investigation.
Collapse
Affiliation(s)
- Li Wang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (L.W.); (Y.Z.); (J.Y.)
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yan Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (L.W.); (Y.Z.); (J.Y.)
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jianming Yue
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (L.W.); (Y.Z.); (J.Y.)
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ronghua Zhou
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (L.W.); (Y.Z.); (J.Y.)
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
47
|
Fu Y, Alam N, Hua Y, Zhang H, Liu E, Li W. Chronic Alcohol Exposure Modulates Atherosclerotic Plaque Vulnerability in ApoE Knockout Mice. Mol Nutr Food Res 2025:e70021. [PMID: 40296557 DOI: 10.1002/mnfr.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 02/12/2025] [Accepted: 02/24/2025] [Indexed: 04/30/2025]
Abstract
AIM This study aimed to examine the effect of chronic alcohol consumption on the development and progression of atherosclerosis in apolipoprotein E-knockout (ApoE-/-) mice. METHOD Male ApoE-/- mice, aged 8 weeks, were randomly assigned to four groups: control, model, low-dose alcohol, and high-dose alcohol. The mice were fed a normal chow or high-fat diet for 14 weeks, with water provided alongside 1% (v/v) or 5% (v/v) alcohol solutions. RESULTS Mice in the 1% (v/v) alcohol group showed a significant increase in subcutaneous and epididymal fat compared to those in the model group. However, basal metabolic markers remained unchanged in the 5% (v/v) alcohol group, although water intake was significantly lower. Histological analyses of the experimental groups exposed to alcohol revealed no protective effects against atherosclerosis in the aortic tree or plaque deposition in the aortic root. Additionally, no significant changes were observed in the macrophages and smooth muscle cells within the aortic root plaques. Nevertheless, low-dose alcohol exposure protected plaque vulnerability compared with the model group, whereas high-dose alcohol had no effect. Transcriptomic analysis of aortic tissue further indicated that alcohol consumption reduced the expression of genes related to lipid transport and metabolic abnormalities.
Collapse
Affiliation(s)
- Yu Fu
- Clinical Nutrition Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Naqash Alam
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Yuxin Hua
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Huifeng Zhang
- Clinical Nutrition Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Enqi Liu
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Weimin Li
- Clinical Nutrition Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
48
|
Zhao P, Yuan K, Tang Z, Li Y, Yu Y, Gao W, Zhang Y, Wang J, Li X, Tie Y. Investigation of hub-shared genes and regulatory mechanisms of rheumatoid arthritis and atherosclerosis. Clin Rheumatol 2025:10.1007/s10067-025-07423-x. [PMID: 40289029 DOI: 10.1007/s10067-025-07423-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025]
Abstract
OBJECTIVE Study found that patients with rheumatoid arthritis (RA) are more likely to develop atherosclerosis than normal adults. However, their shared mechanisms of action still remain unclear. This study aimed to identify the shared genes between the two diseases and uncover their regulatory mechanisms. METHOD The RA- and atherosclerosis-related microarray datasets were downloaded from public databases. Gene set enrichment, differential expression, and weighted gene co-expression network analyses were performed to identify the shared genes between the two diseases. Functional enrichment analysis and protein-protein interaction networks for shared genes were performed. Through further expression validation using validation datasets, hub-shared genes were identified. The diagnostic values of hub-shared genes and their related transcription factors (TFs), small-molecule drugs, and immune cells were analyzed. RESULTS A total of 82 shared genes, which were significantly involved in nine pathways, including the peroxisome proliferator-activated receptor signaling pathway and Th1 and Th2 cell differentiation, were identified. Two hub-shared genes, CD52 and TNFRSF17, were screened out using validation. CD52 and TNFRSF17 showed high diagnostic performance for both diseases. CD52 and TNFRSF17 could interact with multiple proteins, including TNFSF13, and are regulated by several TFs, including NFKB1 and MEF2A. Moreover, significant correlations were observed between hub-shared genes and the infiltration of several immune cells in the two diseases, such as between gamma delta T cells and TNFRSF17, as well as between neutrophils and CD52. CONCLUSION Two hub-shared genes, CD52 and TNFRSF17, may be key regulators in the development of RA and atherosclerosis. Key Points • Study found that patients with RA are more likely to develop atherosclerosis. The shared mechanisms of action between the two diseases are unclear. We used bioinformatics methods to investigate shared genes and explore the mechanisms. • Our results indicated that the two hub genes, CD52 and TNFRSF17, may be key regulators in the development of atherosclerosis in RA. Further research of these two genes may reveal the mechanism that RA patients are more likely to suffer from atherosclerosis.
Collapse
Affiliation(s)
- Pei Zhao
- Hebei Key Laboratory of Molecular Medicine, Shijiazhuang, 050051, China.
- Hebei Clinical Research Center for Laboratory Medicine, Shijiazhuang, 050051, China.
- Department of Laboratory Medicine, Hebei General Hospital, Shijiazhuang, 050051, China.
| | - Kexin Yuan
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Zhipeng Tang
- Hebei Key Laboratory of Molecular Medicine, Shijiazhuang, 050051, China
- Hebei Clinical Research Center for Laboratory Medicine, Shijiazhuang, 050051, China
- Department of Laboratory Medicine, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Yonghui Li
- Hebei Center for Disease Control and Prevention, Shijiazhuang, 050000, China
| | - Yueqing Yu
- Hebei Key Laboratory of Molecular Medicine, Shijiazhuang, 050051, China
- Hebei Clinical Research Center for Laboratory Medicine, Shijiazhuang, 050051, China
- Department of Laboratory Medicine, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Wei Gao
- Hebei Key Laboratory of Molecular Medicine, Shijiazhuang, 050051, China
- Hebei Clinical Research Center for Laboratory Medicine, Shijiazhuang, 050051, China
- Department of Laboratory Medicine, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Yu Zhang
- Hebei Key Laboratory of Molecular Medicine, Shijiazhuang, 050051, China
- Hebei Clinical Research Center for Laboratory Medicine, Shijiazhuang, 050051, China
- Department of Laboratory Medicine, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Jie Wang
- Hebei Key Laboratory of Molecular Medicine, Shijiazhuang, 050051, China
- Hebei Clinical Research Center for Laboratory Medicine, Shijiazhuang, 050051, China
- Department of Laboratory Medicine, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Xinxin Li
- Hebei Key Laboratory of Molecular Medicine, Shijiazhuang, 050051, China
- Hebei Clinical Research Center for Laboratory Medicine, Shijiazhuang, 050051, China
- Department of Laboratory Medicine, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Yanqing Tie
- Hebei Key Laboratory of Molecular Medicine, Shijiazhuang, 050051, China.
- Hebei Clinical Research Center for Laboratory Medicine, Shijiazhuang, 050051, China.
- Department of Laboratory Medicine, Hebei General Hospital, Shijiazhuang, 050051, China.
| |
Collapse
|
49
|
Zhang S, Qian L, Li S, Liu Z. Association between soluble suppression of tumorigenicity 2 and risk and severity of coronary artery disease: a case control study. BMC Cardiovasc Disord 2025; 25:334. [PMID: 40295953 PMCID: PMC12038991 DOI: 10.1186/s12872-025-04787-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Accepted: 04/21/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND To investigate the differential expression of soluble suppression of tumorigenicity 2 (sST2) in patients with coronary artery disease (CAD) and healthy controls, and the correlation between sST2 and the severity of coronary artery atherosclerosis. METHODS A total of 911 CAD patients were selected as the CAD group, and 322 healthy people were selected as the control group. We measured serum sST2 level by chemiluminescence immunoassay, and applied the Gensini scoring system to quantify the severity of coronary artery atherosclerosis. We utilized Mann-Whitney U test to assess the difference of sST2 level between the two groups, and adopted Spearman correlation test to evaluate the correlation between sST2 level and Gensini score and inflammatory indexes. RESULTS Compared with the control group, the expression level of sST2 in CAD group was significantly increased [29.20 (20.67, 46.34) vs. 19.69 (15.97, 25.02), P < 0.001]. Logistic regression showed that sST2 expression could increase CAD risk (OR = 1.099, 95%CI: 1.080 ~ 1.119, P < 0.001). Analysis of variance revealed that the sST2 expression level increased gradually in unstable angina pectoris group (UA), non-ST elevation myocardial infarction group (NSTEMI) and ST elevation myocardial infarction group (STEMI) [UA: 23.05 (17.54, 30.75), NSTEMI: 30.71 (21.31, 42.97), STEMI: 51.05 (32.85, 80.04), P < 0.001]. Spearman correlation analysis demonstrated significantly positive associations between sST2 expression level and Gensini score (r = 0.137, P < 0.001), and systemic inflammatory indexes MHR (r = 0.188, P < 0.001), NLR (r = 0.469, P < 0.001), PLR (r = 0.285, P < 0.001) and MLR (r = 0.368, P < 0.001), but negatively correlated with AFR (r=-0.135, P < 0.001). By receiver operating characteristic (ROC) curve analysis, the sST2 expression level had excellent predictive effect in STEMI with the area under the curve (AUC) value of 0.926 (95%CI: 0.903-0.948, P < 0.001) and sensitivity and specificity of 72.3% and 99.7% respectively, superior to NSTEMI with an AUC of 0.760 (95%CI: 0.719-0.802, P < 0.001) and UA with an AUC of 0.616 (95%CI: 0.576-0.656, P < 0.001). CONCLUSIONS sST2 could not only serve as a biomarker for the clinical auxiliary diagnosis of CAD, but also act as a potential indicator for disease progression or risk stratification. Dynamic monitoring of sST2 levels might assist in evaluating treatment efficacy.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, No.99 Huaihai West Road, Xuzhou, 221002, Jiangsu, China
| | - Lu Qian
- Xuzhou Blood Center, Xuzhou, 221002, Jiangsu, China
| | - Shibao Li
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, No.99 Huaihai West Road, Xuzhou, 221002, Jiangsu, China
| | - Zhijian Liu
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, No.99 Huaihai West Road, Xuzhou, 221002, Jiangsu, China.
| |
Collapse
|
50
|
Ogger PP, Murray PJ. Dissecting inflammation in the immunemetabolomic era. Cell Mol Life Sci 2025; 82:182. [PMID: 40293552 PMCID: PMC12037969 DOI: 10.1007/s00018-025-05715-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 04/30/2025]
Abstract
The role of immune metabolism, specific metabolites and cell-intrinsic and -extrinsic metabolic states across the time course of an inflammatory response are emerging knowledge. Targeted and untargeted metabolomic analysis is essential to understand how immune cells adapt their metabolic program throughout an immune response. In addition, metabolomic analysis can aid to identify pathophysiological patterns in inflammatory disease. Here, we discuss new metabolomic findings within the transition from inflammation to resolution, focusing on three key programs of immunity: Efferocytosis, IL-10 signaling and trained immunity. Particularly the tryptophan-derived metabolite kynurenine was identified as essential for efferocytosis and inflammation resolution as well as a potential biomarker in diverse inflammatory conditions. In summary, metabolomic analysis and integration with transcriptomic and proteomic data, high resolution imaging and spatial information is key to unravel metabolic drivers and dependencies during inflammation and progression to tissue-repair.
Collapse
Affiliation(s)
- Patricia P Ogger
- Immunoregulation Research Group, Max Planck Institute of Biochemistry, Martinsried, 82152, Germany
| | - Peter J Murray
- Immunoregulation Research Group, Max Planck Institute of Biochemistry, Martinsried, 82152, Germany.
| |
Collapse
|