1
|
Hua S, Sun L, Zhang H, Shiu C, Zhang S, Zhu Y, Yan X, Gu P, Huang Z, Jiang W. Yiqi Wenyang decoction protects against the development of atherosclerosis by inhibiting vascular inflammation. PHARMACEUTICAL BIOLOGY 2025; 63:264-274. [PMID: 40254717 PMCID: PMC12010649 DOI: 10.1080/13880209.2025.2492650] [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: 08/30/2024] [Revised: 03/17/2025] [Accepted: 04/08/2025] [Indexed: 04/22/2025]
Abstract
CONTEXT Vascular inflammation is a key process in the pathogenesis of atherosclerosis, which is regulated by NF-κB pathway. Yiqi Wenyang decoction (YQWY), a Traditional Chinese medicine (TCM) formula, has anti-inflammatory properties and may inhibit this pathway, potentially offering anti-atherosclerotic effects. OBJECTIVE The purpose of this study is to investigate the effects of YQWY on atherosclerosis and the underlying mechanism. Materials and methods: ApoE-/- mice were fed a Western diet and administered with YQWY (low or high dose), atorvastatin, or vehicle for 13 weeks. The size of atherosclerotic plaques was assessed using ORO staining. Vascular inflammation was evaluated with IF or IHC staining. The mechanisms and signaling pathways underlying the effect of YQWY on vasculature were studied using transcriptomic analysis and were validated in vitro in endothelial cells and macrophages. RESULTS YQWY attenuated atherosclerotic plaque development which was associated with reduced vascular inflammation as demonstrated by transcriptomic analysis of aorta. This was verified by reduced expression of proinflammatory chemokines, adhesion molecules, and inflammatory cytokines in aortas from YQWY-treated mice at both mRNA and protein levels. Mechanistically, YQWY suppressed NF-κB activation in endothelial cells and, to a lesser extent, macrophages possibly. DISCUSSION AND CONCLUSIONS YQWY protects against vascular inflammation and atherosclerosis by suppressing NF-κB pathway, suggesting the potential of YQWY and its active ingredients as novel anti-atherosclerotic therapeutics.
Collapse
Affiliation(s)
- Shuang Hua
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, China
| | - Lingling Sun
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Han Zhang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chiwen Shiu
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Shujie Zhang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yao Zhu
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xingqun Yan
- Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Gu
- Department of Endocrinology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
- Department of Endocrinology, Jinling Hospital, the First School of Clinical Medicine, Southern Medical University, Nanjing, China
| | - Zhe Huang
- Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Department of Cardiology, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Weimin Jiang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
2
|
Liu Z, Wang H, Dai L, Zeng H, Zhong X. Y-box binding protein 1: A critical target for understanding and treating cardiovascular disease. Cell Signal 2025; 132:111797. [PMID: 40204098 DOI: 10.1016/j.cellsig.2025.111797] [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: 03/12/2025] [Revised: 04/01/2025] [Accepted: 04/06/2025] [Indexed: 04/11/2025]
Abstract
Cardiovascular diseases (CVDs) remain a significant public health burden, characterized by escalating morbidity and mortality rates and demanding novel therapeutic approaches. Cold shock protein Y-box binding protein 1 (YB-1), a highly conserved RNA/DNA-binding protein, has emerged as a pivotal regulator in various pathophysiological processes, including CVDs. YB-1 exerts pleiotropic functions by modulating gene transcription, pre-mRNA splicing, mRNA translation, and stability. The expression and function of YB-1 are intricately regulated by its subcellular localization, post-translational modifications, upstream regulatory signals. YB-1 plays a multifaceted role in CVDs, influencing inflammation, oxidative stress, cell proliferation, apoptosis, phenotypic switching of smooth muscle cells, and mitochondrial dysfunction. However, the regulation of YB-1 expression and function in CVDs is complex and context-dependent, exhibiting divergent effects even in the same disease across different cell types or at disease stages. This review comprehensively explores the structure, regulation, and functional significance of YB-1 in CVDs. We delve into the transcriptional and translational control mechanisms of YB-1, as well as its post-translational modifications. Furthermore, we elucidate the upstream signaling pathways that influence YB-1 expression, with a particular emphasis on non-coding RNAs and specific upstream molecules. Finally, we systematically examine the role of YB-1 in CVDs, summarizing its expression patterns, regulatory mechanisms, and therapeutic potential as a promising target for novel therapeutic interventions. By providing a comprehensive overview of YB-1's involvement in CVDs, this review aims to stimulate further research and facilitate the development of targeted therapies to improve cardiovascular health.
Collapse
Affiliation(s)
- Zixuan Liu
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan 430030, Hubei, China
| | - Hongjie Wang
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan 430030, Hubei, China
| | - Lei Dai
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan 430030, Hubei, China
| | - Hesong Zeng
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan 430030, Hubei, China.
| | - Xiaodan Zhong
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan 430030, Hubei, China.
| |
Collapse
|
3
|
Huang Z, Li A, Huang R, Shi M, Yang R, Wang W, Huang Z, Liu Y, Wu J. Effect of xylitol on low‑density lipoprotein‑stimulated oxidative stress in THP‑1 cells. Mol Med Rep 2025; 32:190. [PMID: 40341459 PMCID: PMC12076051 DOI: 10.3892/mmr.2025.13555] [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: 12/17/2024] [Accepted: 04/03/2025] [Indexed: 05/10/2025] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease caused by oxidative stress and the oxidation of low‑density lipoprotein (LDL). Xylitol, a widely used sugar substitute, has antioxidant potential; however, its effects on LDL‑induced oxidative stress in AS remain unclear. Using western blot, reverse transcription‑quantitative PCR, flow cytometry and so on, the present study investigated the role of xylitol in mitigating oxidative stress induced by high levels of LDL in Tohoku Hospital Pediatrics‑1 (THP‑1) human monocytic cell line), a model for studying AS. Xylitol significantly alleviated high LDL‑induced oxidative stress in THP‑1 cells and decreased reactive oxygen species levels, malondialdehyde content and the expression of NADPH oxidase family enzymes. Concurrently, xylitol enhanced the activity and expression of superoxide dismutase and increased the glutathione levels. Mechanistically, xylitol activated the nuclear factor erythroid 2‑related factor 2 (Nrf2)/heme oxygenase‑1 (HO‑1) axis by increasing the NADPH/NADP+ ratio via the regulation of the pentose phosphate pathway via the Nrf2 transcription factor. This led to a decrease in LDL oxidative modification in THP‑1 cells (Figs. 6,7). Overall, xylitol attenuates high LDL level‑induced oxidative stress in THP‑1 cells by modulating the Nrf2‑mediated pentose phosphate pathway and activating the Nrf2/HO‑1 axis, highlighting its potential for the prevention and treatment of AS.
Collapse
Affiliation(s)
- Zile Huang
- Taikang Medical School, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430072, P.R. China
| | - Anke Li
- Taikang Medical School, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430072, P.R. China
| | - Rui Huang
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Mengmeng Shi
- Taikang Medical School, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430072, P.R. China
| | - Rujing Yang
- Taikang Medical School, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430072, P.R. China
| | - Wenyan Wang
- Taikang Medical School, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430072, P.R. China
| | - Zhen Huang
- Taikang Medical School, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430072, P.R. China
| | - Yanhong Liu
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Junzhu Wu
- Taikang Medical School, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430072, P.R. China
| |
Collapse
|
4
|
Zhong C, Deng K, Lang X, Shan D, Xie Y, Pan W, Yu J. Therapeutic potential of natural flavonoids in atherosclerosis through endothelium-protective mechanisms: An update. Pharmacol Ther 2025; 271:108864. [PMID: 40274196 DOI: 10.1016/j.pharmthera.2025.108864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/27/2025] [Accepted: 04/20/2025] [Indexed: 04/26/2025]
Abstract
Atherosclerosis and its associated cardiovascular complications remain significant global public health challenges, underscoring the urgent need for effective therapeutic strategies. Endothelial cells are critical for maintaining vascular health and homeostasis, and their dysfunction is a key contributor to the initiation and progression of atherosclerosis. Targeting endothelial dysfunction has, therefore, emerged as a promising approach for the prevention and management of atherosclerosis. Among natural products, flavonoids, a diverse class of plant-derived phenolic compounds, have garnered significant attention for their anti-atherosclerotic properties. A growing body of evidence demonstrates that flavonoids can mitigate endothelial dysfunction, highlighting their potential as endothelial dysfunction-targeted therapeutics for atherosclerosis. In this review, we summarize current knowledge on the roles of natural flavonoids in modulating various aspects of endothelial dysfunction and their therapeutic effects on atherosclerosis, focusing on the underlying molecular mechanisms. We also discuss the challenges and future prospects of translating natural flavonoids into clinical applications for cardiovascular medicine. This review aims to provide critical insights to advance the development of novel endothelium-protective pharmacotherapies for atherosclerosis.
Collapse
Affiliation(s)
- Chao Zhong
- Center for Translational Medicine, Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Keke Deng
- Center for Translational Medicine, Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Xiaoya Lang
- Center for Translational Medicine, Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Dan Shan
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Yanfei Xie
- Center for Translational Medicine, Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Wen Pan
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi University of Chinese Medicine, Nanchang 330006, China.
| | - Jun Yu
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
5
|
De Bartolo A, Romeo N, Angelone T, Rocca C. Specialized Pro-Resolving Mediators as Emerging Players in Cardioprotection: From Inflammation Resolution to Therapeutic Potential. Acta Physiol (Oxf) 2025; 241:e70062. [PMID: 40433738 PMCID: PMC12117521 DOI: 10.1111/apha.70062] [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: 02/28/2025] [Revised: 04/18/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025]
Abstract
AIM Timely myocardial reperfusion is essential for restoring blood flow to post-ischemic tissue, thereby reducing cardiac injury and limiting infarct size. However, this process can paradoxically result in additional, irreversible myocardial damage, known as myocardial ischemia-reperfusion injury (MIRI). The goal of this review is to explore the role of specialized pro-resolving mediators (SPMs) in atherosclerosis and MIRI, and to assess the therapeutic potential of targeting inflammation resolution in these cardiovascular conditions. METHODS This review summarizes current preclinical and clinical evidence on the involvement of SPMs in the pathogenesis of atherosclerosis and MIRI, acknowledging that several cellular and molecular aspects of their mechanisms of action remain to be fully elucidated. RESULTS MIRI is a complex phenomenon in which inflammation, initially triggered during ischemia and further amplified upon reperfusion, plays a central role in its pathogenesis. Various cellular and molecular players mediate the initial pro-inflammatory response and the subsequent anti-inflammatory reparative phase following acute myocardial infarction (AMI), contributing both to ischemia- and reperfusion-induced damage as well as to the healing process. SPMs have emerged as key endogenous immunoresolvents with potent anti-inflammatory, antioxidant, and pro-resolving properties that contribute to limit excessive acute inflammation and promote tissue repair. While dysregulated SPM-related signaling has been linked to various cardiovascular diseases (CVD), their precise role in AMI and MIRI remains incompletely understood. CONCLUSION Targeting inflammation resolution may represent a promising therapeutic strategy for mitigating atheroprogression and addressing a complex condition such as MIRI.
Collapse
Affiliation(s)
- Anna De Bartolo
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. And E. S. (DiBEST)University of CalabriaCosenzaItaly
| | - Naomi Romeo
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. And E. S. (DiBEST)University of CalabriaCosenzaItaly
| | - Tommaso Angelone
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. And E. S. (DiBEST)University of CalabriaCosenzaItaly
- National Institute of Cardiovascular Research (INRC)BolognaItaly
| | - Carmine Rocca
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. And E. S. (DiBEST)University of CalabriaCosenzaItaly
- National Institute of Cardiovascular Research (INRC)BolognaItaly
| |
Collapse
|
6
|
Wang SH, Xu XL, Chen W. How Do Organelle-Targeting Nanotherapeutics Treat Inflammatory Diseases? A Comprehensive Review of the Literature. Int J Nanomedicine 2025; 20:7133-7152. [PMID: 40491850 PMCID: PMC12146407 DOI: 10.2147/ijn.s516260] [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: 01/24/2025] [Accepted: 05/20/2025] [Indexed: 06/11/2025] Open
Abstract
Inflammation is a protective response of the body, but when excessive or prolonged, it can contribute to disease progression and tissue damage. Identifying more effective and less toxic drugs for treating both acute and chronic inflammatory diseases is a major challenge. Organelle-targeting strategies, which deliver drugs directly to specific organelles, offer a promising solution by improving treatment efficiency and minimizing toxic effects on healthy cells. However, despite the potential of organelles as therapeutic targets, precise targeting remains challenging. This review systematically summarizes organelle-targeting nanodelivery strategies for major organelles-mitochondria, the endoplasmic reticulum, lysosomes, and the Golgi apparatus-and the research progress in evaluating the potential of these strategies for treating inflammation-related diseases. This study focuses on the applications of these strategies for the treatment of sepsis, inflammatory bowel disease, atherosclerosis, and osteoarthritis. Additionally, this review outlines future directions and key challenges in this field, aiming to provide a scientific reference for the application of organelle-targeting nanotherapeutics for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Si-Hui Wang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People’s Republic of China
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, 310015, People’s Republic of China
| | - Xiao-Ling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, 310015, People’s Republic of China
| | - Wei Chen
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People’s Republic of China
| |
Collapse
|
7
|
Xue K, Li Y, Hu R, Hu X, Guo R, Guo H, Li G. Predictive value of fecal sulfatide and neutrophil-to‑lymphocyte ratio in coronary heart disease. Exp Ther Med 2025; 29:126. [PMID: 40357311 PMCID: PMC12067041 DOI: 10.3892/etm.2025.12876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/20/2025] [Indexed: 05/15/2025] Open
Abstract
The incidence of coronary atherosclerotic heart disease is on the rise, posing a serious threat to public health. Emerging evidence highlights the interplay between systemic inflammation and cardiovascular pathophysiology, suggesting novel diagnostic avenues. The aim of the present study was to evaluate the predictive value of fecal sulfatide and neutrophil-to-lymphocyte ratio (NLR), both individually and in combination, for coronary heart disease (CHD). A total of 523 patients diagnosed with CHD at the Cardiovascular Department of Hebei General Hospital (Shijiazhuang, China) from August 2022 to September 2023 were included in the experimental group, along with 198 healthy controls. The CHD group was further subdivided into stable angina pectoris (n=194), unstable angina pectoris (n=134), and acute myocardial infarction (AMI) groups (n=195). Fecal sulfatide and serum NLR levels were measured in both the experimental and control groups, as well as within each CHD subgroup. Multivariate logistic regression was utilized to assess whether these biomarkers serve as independent risk factors for CHD. The predictive value of fecal sulfatide and serum NLR was evaluated using receiver operating characteristic curves. Fecal sulfatide and serum NLR levels were distinctly higher in the CHD group compared with the control group (2.40±0.48 vs. 1.64±0.39 µmol/l and 2.92 vs. 1.65; P<0.05). Patients with AMI had higher NLR levels than those with stable and unstable angina pectoris (5.55 vs. 2.65 and 2.68; P<0.05). Fecal sulfatide levels were also elevated in patients with AMI (2.50±0.44 µmol/l) compared with patients with stable angina pectoris (2.32±0.48 µmol/l). Both fecal sulfatide (AUC=0.899) and NLR (AUC=0.811) exhibited strong predictive accuracy for CHD. When combined, the predictive value (AUC=0.945) was further improved. Elevated levels of fecal sulfatide and serum NLR in patients with CHD revealed that these biomarkers may serve as valuable adjuncts in the diagnosis of CHD. The combined use of these biomarkers enhances the accuracy and reliability of CHD prediction.
Collapse
Affiliation(s)
- Kefan Xue
- Division of Cardiology, Institute of Geriatric Diseases, Hebei General Hospital, Shijiazhuang, Hebei 050000, P.R. China
- Graduate School of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Yihan Li
- Division of Cardiology, Institute of Geriatric Diseases, Hebei General Hospital, Shijiazhuang, Hebei 050000, P.R. China
- Graduate School of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Rui Hu
- General Clinical Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Xiao Hu
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, Hebei 050000, P.R. China
| | - Ran Guo
- The Third General Surgery Department, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Hongxia Guo
- Department of Anesthesiology, Hebei General Hospital, Shijiazhuang, Hebei 050000, P.R. China
| | - Gang Li
- Division of Cardiology, Institute of Geriatric Diseases, Hebei General Hospital, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
8
|
Zheng Q, Zheng Y, Junaid M, Zeng M, Liao H, Li Y, Zhao Y, Huang Q, Wang J. Biochar alleviates nanoplastics and bisphenol A mediated immunological, neurological and gut microbial toxicity in channel catfish Ictaluruspunctatus. CHEMOSPHERE 2025; 378:144422. [PMID: 40252530 DOI: 10.1016/j.chemosphere.2025.144422] [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: 06/19/2024] [Revised: 03/20/2025] [Accepted: 04/11/2025] [Indexed: 04/21/2025]
Abstract
Nanoplastics (NPs) and bisphenol A (BPA), exhibit abundant industrial applications, are produced in large volumes and ubiquitously released into the environment, posing a serious threat to ecological and human health. Biochar has been extensively studied for its ability to mitigate the negative effects of contaminants on plants. Therefore, this study aims to investigate whether biochar co-exposure with polystyrene nanoplastics (PS-NPs, size 80 nm) and BPA mitigate their toxic impacts on Ictalurus punctatus and maintain its normal growth. The I. punctatus was exposed individually to PS-NPs (0.5 mg/L) and BPA (0.2 mg/L) as well as co-exposed to PS-NPs + biochar and BPA + biochar for 7 days. Results showed PS-NPs and BPA single exposure caused tissue damage in terms of hepatocyte swelling and gut villi diffusion, and induced oxidative stress. PS-NPs and BPA single exposures led to significant changes in enzymatic activities and genetic expressions of biomarkers related to the immune system, producing inflammatory response. It also led to dysregulation of neurotransmitter enzymes (ACH, ChAT, AChE) and overexpression of neuron genes, resulting in neurotoxicity. Moreover, there was an increase in the diversity and alteration in composition of the gut microbiota (Plesiomonas, Pseudomonas), resulting in dysbiosis of the gut microbiota. However, biochar presence (0.5 g/L) reduced the accumulation of PS-NPs and BPA in fish and contributed to various degrees of mitigation for the toxic impacts of PS-NPs and BPA. Overall, biochar helped to mitigate the negative effects of PS-NPs and BPA on oxidative stress, histopathology, immune system, neurological responses and gut microbiota. This study emphasized the potential of biochar to mitigate the negative impacts of NPs and BPA on aquatic organisms.
Collapse
Affiliation(s)
- Qingzhi Zheng
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Yufeng Zheng
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Muhammad Junaid
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Ming Zeng
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Hongping Liao
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Ye Li
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Yuanqi Zhao
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Qian Huang
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Jun Wang
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; Institute of Eco-Environmental Research, Guangxi Academy of Sciences, Nanning, 530007, China; Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai, 528478, China.
| |
Collapse
|
9
|
Guo Z, Zhao C, Fang Y, Yue X, Wang Q, Qu C, Cui J. Solasodine inhibits the Th2 immune response and airway remodeling in asthmatic mice through the Runx3/NLRP3 pathway. Toxicol Appl Pharmacol 2025; 499:117351. [PMID: 40268171 DOI: 10.1016/j.taap.2025.117351] [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: 12/20/2024] [Revised: 03/21/2025] [Accepted: 04/18/2025] [Indexed: 04/25/2025]
Abstract
PURPOSE To explore the therapeutic effects of Solasodine on Th2 immune responses and airway remodeling, and to assess whether its mechanism involves NLRP3 inflammasome inactivation mediated by Runx3. METHODS We created an asthma model with wild-type and Runx3 knockout mice using ovalbumin (OVA). After oral administration of Solasodine, we assessed inflammatory and Th2 immune responses using HE staining, ELISA, and flow cytometry. Airway remodeling was assessed with Masson's trichrome staining and α-SMA and TGF-β immunohistochemistry. Mucus secretion was analyzed through MUC5AC immunohistochemistry, and expectoration assays. We studied NLRP3 inflammasome activation using immunohistochemistry and Western blot. We used western blotting and flow cytometry to evaluate how Solasodine regulates Runx3 protein levels. RESULTS Solasodine effectively inhibited the inflammatory response in OVA-induced asthmatic mice, evidenced by reducing inflammatory cell infiltration and lower IL-4, IL-5, and IL-13 levels, decreasing airway remodeling and mucus secretion. Solasodine reduced airway hyperresponsiveness, shown by a lower Penh value. Solasodine boosts Runx3 expression and suppresses NLRP3 inflammasome activation in asthmatic mice. We created an asthma model in Runx3 knockout mice and administered Solasodine at a consistent dose. Following OVA induction, Runx3 knockout mice showed greater inflammation, a Th2 immune response, airway remodeling, and mucus secretion than wild-type mice. Solasodine is less effective in Runx3 knockout asthmatic mice than in wild-type mice. CONCLUSION The anti-asthmatic effects of Solasodine are shown through the inhibition in the Th2 immune response, airway remodeling, hyperresponsiveness, and mucus secretion. The effectiveness may be linked to Runx3-mediated the NLRP3 inflammasomes.
Collapse
Affiliation(s)
- Zhen Guo
- Department of Paediatrics, Yantaishan Hospital, Yantai 264003, China
| | - Cuixiang Zhao
- Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, China
| | - Yanni Fang
- Department of Paediatrics, Yantaishan Hospital, Yantai 264003, China
| | - Xiuxuan Yue
- Department of Paediatrics, Yantaishan Hospital, Yantai 264003, China
| | - Qiuxia Wang
- Department of Paediatrics, Yantaishan Hospital, Yantai 264003, China
| | - Changhua Qu
- Department of Clinical Laboratory Medicine, Yantaishan Hospital, Yantai 264003, China.
| | - Jinpeng Cui
- Department of Clinical Laboratory Medicine, Yantaishan Hospital, Yantai 264003, China.
| |
Collapse
|
10
|
Tornifoglio B, Hughes C, Digeronimo F, Guendouz Y, Johnston RD, Lally C. Imaging the microstructure of the arterial wall - ex vivo to in vivo potential. Acta Biomater 2025; 199:18-34. [PMID: 40348073 DOI: 10.1016/j.actbio.2025.05.022] [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: 12/18/2024] [Revised: 05/01/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Microstructural imaging enables researchers to visualise changes in the arterial wall, allowing for (i) a deeper understanding of the role of specific components in arterial mechanics, (ii) the observation of cellular responses, (iii) insights into pathological alterations in tissue microstructure, and/or (iv) advancements in tissue engineering aimed at replicating healthy native tissue. In this prospective review, we present various imaging modalities spanning from ex vivo to in vivo applications within arterial tissue. The pros, cons, and sensitivities of these modalities are highlighted. By consolidating the latest advancements in microstructural imaging of arterial tissue, the authors aim for this paper to serve as a guide for researchers designing experiments at various stages. Furthermore, the integration of non-invasive, non-destructive imaging techniques into studies provides an additional layer of microstructural information, enhancing scientific findings, improving our understanding of disease, and potentially enabling earlier or more effective diagnostic capabilities. STATEMENT OF SIGNIFICANCE: Imaging the specific microstructural components of the arterial wall provides critical insights into vascular biology, mechanics, and pathology. It enables the visualisation of key structural components and their roles in arterial function, supports the analysis of cell-matrix interactions, and reveals microarchitectural changes associated with disease progression. This level of specificity also informs the design of biomimetic materials and scaffolds in tissue engineering, facilitating the replication of native arterial properties. By synthesising recent developments in microstructural imaging techniques, this paper serves as a reference for investigators designing experiments across a range of vascular research applications. Moreover, the incorporation of non-invasive, non-destructive imaging methods offers a means to acquire detailed microstructural data without compromising tissue integrity. This enhances the interpretability and translational potential of findings, deepens our understanding of vascular disease mechanisms, and may ultimately contribute to the development of earlier and more precise diagnostic approaches.
Collapse
Affiliation(s)
- B Tornifoglio
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland.
| | - C Hughes
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - F Digeronimo
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - Y Guendouz
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - R D Johnston
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - C Lally
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Ireland.
| |
Collapse
|
11
|
Chen T, Ma W, Wang X, Ye Q, Hou X, Wang Y, Jiang C, Meng X, Sun Y, Cai J. Insights of immune cell heterogeneity, tumor-initiated subtype transformation, drug resistance, treatment and detecting technologies in glioma microenvironment. J Adv Res 2025; 72:527-554. [PMID: 39097088 DOI: 10.1016/j.jare.2024.07.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/30/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024] Open
Abstract
BACKGROUND With the gradual understanding of glioma development and the immune microenvironment, many immune cells have been discovered. Despite the growing comprehension of immune cell functions and the clinical application of immunotherapy, the precise roles and characteristics of immune cell subtypes, how glioma induces subtype transformation of immune cells and its impact on glioma progression have yet to be understood. AIM OF THE REVIEW In this review, we comprehensively center on the four major immune cells within the glioma microenvironment, particularly neutrophils, macrophages, lymphocytes, myeloid-derived suppressor cells (MDSCs), and other significant immune cells. We discuss (1) immune cell subtype markers, (2) glioma-induced immune cell subtype transformation, (3) the mechanisms of each subtype influencing chemotherapy resistance, (4) therapies targeting immune cells, and (5) immune cell-associated single-cell sequencing. Eventually, we identified the characteristics of immune cell subtypes in glioma, comprehensively summarized the exact mechanism of glioma-induced immune cell subtype transformation, and concluded the progress of single-cell sequencing in exploring immune cell subtypes in glioma. KEY SCIENTIFIC CONCEPTS OF REVIEW In conclusion, we have analyzed the mechanism of chemotherapy resistance detailly, and have discovered prospective immunotherapy targets, excavating the potential of novel immunotherapies approach that synergistically combines radiotherapy, chemotherapy, and surgery, thereby paving the way for improved immunotherapeutic strategies against glioma and enhanced patient outcomes.
Collapse
Affiliation(s)
- Tongzheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenbin Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qile Ye
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xintong Hou
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yiwei Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chuanlu Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; The Six Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangqi Meng
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Ying Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Jinquan Cai
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
12
|
Zeng W, Huang Z, Huang Y, Xiong K, Sheng Y, Lin X, Zhong X, Ye J, Guo Y, Arkin G, Xu J, Fei H, Liu Y. Dual-targeted microbubbles for atherosclerosis therapy: Inducing M1 macrophage apoptosis by inhibiting telomerase activity. Mater Today Bio 2025; 32:101675. [PMID: 40225135 PMCID: PMC11986608 DOI: 10.1016/j.mtbio.2025.101675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 03/05/2025] [Accepted: 03/16/2025] [Indexed: 04/15/2025] Open
Abstract
The progression of atherosclerosis (AS) is closely associated with M1 macrophages. Although the activation of macrophage telomerase during plaque formation has been documented, targeted modulation strategies remain challenging. In this study, we developed a dual-target microbubble-delivery system (Ab-MMB1532) encapsulating BIBR1532, a telomerase inhibitor, for the targeted therapy of AS. This system exhibited remarkable targeting capabilities towards M1 macrophages, with its targeting advantage notably accentuated under high shear forces. Mechanistically, Ab-MMB1532 inhibited telomerase activity by downregulating telomerase reverse transcriptase (TERT) expression, subsequently inducing caspase-3-mediated apoptosis. Integrated multi-omics profiling revealed that the inhibition of the NF-κB pathway served as the central regulatory hub. In vivo studies further confirmed that Ab-MMB1532 effectively targets and accumulates within AS lesions, promoting M1 macrophage apoptosis through the inhibition of the TERT/NF-κB signaling axis, and significantly reducing plaque burden (25.4 % reduction vs. controls, p < 0.001). In summary, our findings suggest a novel approach for telomerase-targeted therapy in AS.
Collapse
Affiliation(s)
- Wei Zeng
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Zhengan Huang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 519041, China
| | - Yalan Huang
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
- Post-doctoral Scientific Research Station of Basic Medicine, Jinan University, Guangzhou, 510632, China
| | - Kaifen Xiong
- Department of Dermatology, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Yuanyuan Sheng
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Xiaoxuan Lin
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Xiaofang Zhong
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Jiayu Ye
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Yanbin Guo
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Gulzira Arkin
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Jinfeng Xu
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Hongwen Fei
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 519041, China
| | - Yingying Liu
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| |
Collapse
|
13
|
Wang R, He Y, Wang Y, Wang J, Ding H. Palmitoylation in cardiovascular diseases: Molecular mechanism and therapeutic potential. IJC HEART & VASCULATURE 2025; 58:101675. [PMID: 40242212 PMCID: PMC12002947 DOI: 10.1016/j.ijcha.2025.101675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025]
Abstract
Cardiovascular disease is one of the leading causes of mortality worldwide, and involves complex pathophysiological mechanisms that encompass various biological processes and molecular pathways. Post-translational modifications of proteins play crucial roles in the occurrence and progression of cardiovascular diseases, among which palmitoylation is particularly important. Various proteins associated with cardiovascular diseases can be palmitoylated to enhance the hydrophobicity of their molecular subdomains. This lipidation can significantly affect some pathophysiological processes, such as metabolism, inflammation by altering protein stability, localization, and signal transduction. In this review, we narratively summarize recent advances in the palmitoylation of proteins related to cardiovascular diseases and discuss its potential as a therapeutic target.
Collapse
Affiliation(s)
- Rongli Wang
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, PR China
| | - Yi He
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, PR China
| | - Yan Wang
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, PR China
- Genetic Diagnosis Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuan 430030, PR China
| | - Jing Wang
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, PR China
| | - Hu Ding
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, PR China
- Genetic Diagnosis Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuan 430030, PR China
| |
Collapse
|
14
|
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
|
15
|
Wang X, Chen L, Wei J, Zheng H, Zhou N, Xu X, Deng X, Liu T, Zou Y. The immune system in cardiovascular diseases: from basic mechanisms to therapeutic implications. Signal Transduct Target Ther 2025; 10:166. [PMID: 40404619 PMCID: PMC12098830 DOI: 10.1038/s41392-025-02220-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/22/2024] [Accepted: 03/20/2025] [Indexed: 05/24/2025] Open
Abstract
Immune system plays a crucial role in the physiological and pathological regulation of the cardiovascular system. The exploration history and milestones of immune system in cardiovascular diseases (CVDs) have evolved from the initial discovery of chronic inflammation in atherosclerosis to large-scale clinical studies confirming the importance of anti-inflammatory therapy in treating CVDs. This progress has been facilitated by advancements in various technological approaches, including multi-omics analysis (single-cell sequencing, spatial transcriptome et al.) and significant improvements in immunotherapy techniques such as chimeric antigen receptor (CAR)-T cell therapy. Both innate and adaptive immunity holds a pivotal role in CVDs, involving Toll-like receptor (TLR) signaling pathway, nucleotide-binding oligomerization domain-containing proteins 1 and 2 (NOD1/2) signaling pathway, inflammasome signaling pathway, RNA and DNA sensing signaling pathway, as well as antibody-mediated and complement-dependent systems. Meanwhile, immune responses are simultaneously regulated by multi-level regulations in CVDs, including epigenetics (DNA, RNA, protein) and other key signaling pathways in CVDs, interactions among immune cells, and interactions between immune and cardiac or vascular cells. Remarkably, based on the progress in basic research on immune responses in the cardiovascular system, significant advancements have also been made in pre-clinical and clinical studies of immunotherapy. This review provides an overview of the role of immune system in the cardiovascular system, providing in-depth insights into the physiological and pathological regulation of immune responses in various CVDs, highlighting the impact of multi-level regulation of immune responses in CVDs. Finally, we also discuss pre-clinical and clinical strategies targeting the immune system and translational implications in CVDs.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Liming Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianming Wei
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Hao Zheng
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Ning Zhou
- Department of Cardiovascular Medicine, Anzhen Hospital Affiliated to Capital Medical University, Beijing, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Deng
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tao Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China.
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Jiangsu, Nanjing, China.
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
- Institutes of Advanced Medical Sciences and Huaihe Hospital, Henan University, Kaifeng, Henan, China.
| |
Collapse
|
16
|
Ahari RK, Moohebati M, Tajfard M, Esmaily H, Ferns G, Ghayour-Mobarhan M, Saffar Soflaei S. Introducing Two Novel Indices for Evaluating Coronary Artery Stenosis: Athero-Inflammatory Index and Athero-Inflammatory Glucose Index. J Clin Lab Anal 2025:e70050. [PMID: 40401782 DOI: 10.1002/jcla.70050] [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: 10/26/2024] [Revised: 04/16/2025] [Accepted: 04/24/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND We investigated the association of two novel indices, the athero-inflammatory (AI) index and athero-inflammatory glucose (AIG) index, with coronary artery stenosis (CAS). METHODS In this case-control study, the cases were grouped as angiography (+) and angiography (-) according to the angiographic results. The control group comprised subjects who attended clinics for routine check-ups or pre-employment medical assessments. The AI index and AIG index were compared between the groups using ANOVA. Binary logistic regression (LR) was performed to find the association of the indices with angiography (+). Receiver operating characteristic (ROC) curve analysis was used to establish the cut-off values in differentiating angiography (+) from angiography (-) and healthy subjects. p < 0.05 were considered statistically significant. RESULTS Among a total of 2326 participants (761 angiography (+), 406 angiography (-), and 1159 controls), the AI index and AIG index were significantly different between the groups (p < 0.001). In LR analysis, after adjustment for potential confounders, the AI index and AIG index were independently associated with angiography (+). ROC curve analysis showed that the AI index (AUC: 0.895; 95% CI: 0.880, 0.908; p < 0.0001) and AIG index (AUC: 0.918; 95% CI: 0.905, 0.930; p < 0.0001) performed better diagnostic performance in differentiating angiography (+) from healthy subjects. CONCLUSION AI index demonstrated higher AUC compared to other biomarkers in differentiating angiography (+) from angiography (-) and healthy subjects. If it combines with fasting glucose (AIG index), it is a promising indicator for the identification of the CAS particularly from a healthy population, with a promising AUC.
Collapse
Affiliation(s)
- Rana Kolahi Ahari
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Moohebati
- Department of Cardiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Tajfard
- Department of Health Education and Health Promotion, Faculty of Health, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Habibollah Esmaily
- Social Determinants of Health Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon Ferns
- Division of Medical Education, Brighton and Sussex Medical School, Brighton, UK
| | - Majid Ghayour-Mobarhan
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sara Saffar Soflaei
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
17
|
Wang J, Yao J, Wang Z. Identification of shared mechanisms between Alzheimer's disease and atherosclerosis by integrated bioinformatics analysis. Eur J Med Res 2025; 30:408. [PMID: 40405226 PMCID: PMC12096694 DOI: 10.1186/s40001-025-02642-z] [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: 11/08/2024] [Accepted: 04/27/2025] [Indexed: 05/24/2025] Open
Abstract
Alzheimer's disease (AD) and atherosclerosis (AS) are two interacting diseases mostly affecting aged adults. AD is characterized by the deposition of neuritic plaques mainly consisting of Aβ, and AS is defined by the formation of atheromatous plaque along the vascular wall. The shared mechanisms underlying the pathogenesis of AD and AS remain unclear. Here we applied several bioinformatic analyses of bulk sequencing data sets of AD brain tissues and atherosclerotic plaques to seek relevant genes between AD and AS. WIPF3, was identified as the most affected gene in both diseases using weighted gene co-expression network analysis, machine-learning-based Lasso Cox regression analysis and random forest analysis. Furthermore, immune cell infiltration analysis of AS data sets and cell portion of single-cell RNA sequencing data from AD patients revealed an essential role of inflammation in the co-occurrence of AD and AS. Taken together, WIPF3 deficiency and inflammation may simultaneously mediate both AD and AS and could be potential targets for the prevention and therapy of these two closely related diseases.
Collapse
Affiliation(s)
- Jukun Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jing Yao
- Cerebrovascular Disease Department, Neurological Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Zhe Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
18
|
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
|
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
|
Lin C, Li X, Wu Y, Wang Y, Song W, Yan F, Sun L. Ultrasound Molecular Imaging of Blood Vessel Walls and Vulnerable Plaques via CXCR4-Targeted Nanoscale GVs. Int J Nanomedicine 2025; 20:6205-6220. [PMID: 40395655 PMCID: PMC12091238 DOI: 10.2147/ijn.s504265] [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: 11/02/2024] [Accepted: 04/06/2025] [Indexed: 05/22/2025] Open
Abstract
Purpose C-X-C chemokine receptor 4 (CXCR4) mediates the inflammatory response of atherosclerotic vulnerable plaques (ASVP) and is a potential biomarker of atherosclerotic vulnerable plaques. The purpose of this study was to use the imaging ability of a new type of ultrasound contrast agent, nanoscale biosynthetic gas vesicles (GVs), on the vascular wall and to combine the specific ligand of CXCR4 to construct a targeted molecular probe to achieve early identification of atherosclerotic vulnerable plaques and guide clinical treatment decisions. Materials and Methods Compared three contrast agents: GVs, the micro-contrast agent SonoVue, and polyethylene glycol (PEG)-modified GVs in the carotid artery. The expression of CXCR4 in atherosclerotic plaques was demonstrated using flow cytometry and immunofluorescence experiments. Cell adhesion and in vivo ultrasound imaging experiments demonstrated their ability to target the nanoscale biosynthetic gas vesicles. The safety of GVs, PEG-GVs, and CXCR4-GVs was tested the CCk8 test, H&E staining, and serum detection. Results Strong CXCR4 expression was observed in plaques, whereas little expression was observed in normal vessels. GVs can produce stable contrast signals on the carotid artery walls of rats, whereas PEG-GVs can produce more lasting contrast signals on the carotid artery wall of rats. CXCR4-GVs exhibited excellent binding capability to ox-LDL-induced RAW264.7 cells. Animal experiments showed that compared with Con-GVs, CXCR4-GVs injected plaque imaging signal was stronger and more durable. In vitro scanning of vulnerable plaques in rats injected with fluorescent vesicles demonstrated that CXCR4-GVs oozed through the neovasculars within vulnerable plaques and aggregated in vulnerable plaques. Through the CCK8 test, H&E staining, and serum detection, the safety of CXCR4-GVs was confirmed. Conclusion CXCR4-GVs were constructed as targeted molecular probes, which can be proven to have good targeting properties to vulnerable atherosclerotic plaques.
Collapse
Affiliation(s)
- Chen Lin
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
- Department of Ultrasound Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
| | - Xiaoying Li
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Yingnan Wu
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Yuanyuan Wang
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People’s Republic of China
| | - Weijian Song
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Fei Yan
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People’s Republic of China
| | - Litao Sun
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| |
Collapse
|
21
|
Wang Y, Dou W, Qian X, Chen H, Zhang Y, Yang L, Wu Y, Xu X. Advancements in the study of short-chain fatty acids and their therapeutic effects on atherosclerosis. Life Sci 2025; 369:123528. [PMID: 40049368 DOI: 10.1016/j.lfs.2025.123528] [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: 12/01/2024] [Revised: 02/15/2025] [Accepted: 03/02/2025] [Indexed: 03/09/2025]
Abstract
Atherosclerosis (AS) remains a leading cause of cardiovascular disease and mortality globally. This chronic condition is characterized by inflammation, lipid accumulation, and the deposition of cellular components within arterial walls. Emerging evidence has highlighted the multifaceted therapeutic potential of short-chain fatty acids (SCFAs) in mitigating AS progression. SCFAs have demonstrated anti-inflammatory properties and the ability to regulate immune responses, metabolic pathways, vascular integrity, and intestinal barrier function in animal models of AS. Consequently, SCFAs have garnered significant attention as a promising approach for the prevention and treatment of AS. However, further clinical trials and studies are necessary to fully elucidate the underlying mechanisms and effects of SCFAs. Additionally, different types of SCFAs may exert distinct impacts, necessitating more in-depth investigation into their specific roles and mechanisms. This review provides an overview of the diverse cellular mechanisms contributing to AS formation, as well as a discussion of the significance of SCFAs in AS pathogenesis and their multifaceted therapeutic potential. Nonetheless, additional research is warranted to comprehensively understand and harness the potential of various SCFAs in the context of AS.
Collapse
Affiliation(s)
- Yongsen Wang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China; Department of Hepatobiliary Pancreatic and Splcnic Surgery, Luzhou People's Hospital, Luzhou, Sichuan 646000, PR China; Department of Vascular and Breast Surgery, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan 621000, PR China
| | - Wei Dou
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Xin Qian
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Hao Chen
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Yi Zhang
- Department of Vascular and Breast Surgery, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan 621000, PR China
| | - Liu Yang
- Department of Hepatobiliary Pancreatic and Splcnic Surgery, Luzhou People's Hospital, Luzhou, Sichuan 646000, PR China
| | - Ya Wu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Xiongfei Xu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China.
| |
Collapse
|
22
|
Antonello J, Roy P. Damage-Associated Molecular Patterns (DAMPs) In Vascular Diseases. J Biol Chem 2025:110241. [PMID: 40381697 DOI: 10.1016/j.jbc.2025.110241] [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: 03/10/2025] [Revised: 05/02/2025] [Accepted: 05/07/2025] [Indexed: 05/20/2025] Open
Abstract
Research into the role of chronic sterile inflammation (i.e. a prolonged inflammatory state not caused by an infectious agent), in vascular disease progression has continued to grow over the last few decades. DAMPs have a critical role in this research due to their ability to link stress-causing cardiovascular risk factors to inflammatory phenotypes seen in vascular disease. In this mini-review, we will briefly summarize the DAMPs and receptor signaling pathways that have been extensively studied in the context of vascular disease, including TLRs, RAGE, cGAS-STING, and the NLRP3 inflammasome. In particular, we will discuss how these pathways can promote the release of pro-inflammatory cytokines and chemokines as well as vascular remodeling. Next, we will summarize the results of studies which have linked the various pro-inflammatory effects of DAMPs with the phenotypes in the context of vascular diseases including atherosclerosis, fibrosis, aneurysm, ischemia, and hypertension. Finally, we will discuss some pre-clinical and clinical trials that have targeted DAMPs, their receptors, or the products of their signaling pathways, and discuss the outlook and future directions for the field at large.
Collapse
Affiliation(s)
| | - Partha Roy
- Bioengineering, University of Pittsburgh; Pathology, University of Pittsburgh.
| |
Collapse
|
23
|
Geng Y, Song M, Huang B, Lin R, Wu S, Lin A. Safranal ameliorates atherosclerosis progression partly via repressing PI3K/Akt and NF-κB signaling pathways in ApoE (-/-) mice. J Nat Med 2025:10.1007/s11418-025-01913-7. [PMID: 40366560 DOI: 10.1007/s11418-025-01913-7] [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: 01/01/2025] [Accepted: 05/03/2025] [Indexed: 05/15/2025]
Abstract
Atherosclerosis (AS) remains the main cause of vascular diseases. This study reveals the effects of safranal and underlying mechanisms in RAW264.7 macrophages under AS context, which is hoped to facilitate its clinical application. Safranal reduced AS progression in ApoE (-/-) mice, and it also increased the serum level of HDL-C and decreased the levels of TG, TC, and LDL-C as well as ALT and AST. Besides, safranal repressed the pathophysiological processes of OS (downregulated levels of ROS and MDA and upregulated biosynthesis of GSH), ERS (decreased protein levels of activating transcription factor 6, X-Box Binding Protein 1, and glucose-regulated protein, 78 kDa), and inflammation (downregulated serum levels of TNF-α, IL-1β, and IL-6) in vivo. Mechanistically, safranal repressed PI3K/Akt and NF-κB signaling pathways in vivo. On the cellular level, safranal treatment relieved the uptake of ox-LDL, and decreased contents of TG, TC, and LDL-C while increasing HDL-C level in ox-LDL-treated RAW264.7 macrophages. It also reduced the molecular indexes of pathophysiological processes (OS, ESR, and release of inflammatory mediators) in ox-LDL-exposed RAW264.7 macrophages. Notably, safranal treatment also impaired PI3K/Akt and NF-κB signaling pathways in ox-LDL-exposed RAW264.7 macrophages. Additionally, the PI3K agonist 740Y-P notably reversed the in vitro inhibitory effects of safranal on lipid deposition, productions of TC and TNF-α, and protein levels of molecules of PI3K/Akt and NF-κB signaling pathways. Safranal exerts anti-AS effects via repressing OS, ERS, and inflammation in ApoE (-/-) mice, and it also negatively modulates PI3K/Akt and NF-κB signaling pathways in RAW264.7 macrophages.
Collapse
Affiliation(s)
- Yining Geng
- Department of Acupuncture, Hainan General Hospital, Hainan Medical University Hainan Hospital, No. 19, Xiuhua Road, Xiuying District, Haikou, 570300, China.
| | - Manping Song
- Department of Acupuncture, Hainan General Hospital, Hainan Medical University Hainan Hospital, No. 19, Xiuhua Road, Xiuying District, Haikou, 570300, China
| | - Bing Huang
- Department of Acupuncture, Hainan General Hospital, Hainan Medical University Hainan Hospital, No. 19, Xiuhua Road, Xiuying District, Haikou, 570300, China
| | - Ru Lin
- Department of Acupuncture, Hainan General Hospital, Hainan Medical University Hainan Hospital, No. 19, Xiuhua Road, Xiuying District, Haikou, 570300, China
| | - Shiwen Wu
- Department of Traditional Chinese Medicine and Western Medicine, Hainan General Hospital Branch-Tunchang Hospital, Tunchang, China
| | - An Lin
- Department of Traditional Chinese Medicine and Western Medicine, Hainan General Hospital Branch-Tunchang Hospital, Tunchang, China
| |
Collapse
|
24
|
Bai Y, Wu J, Jian W. Trained immunity in diabetes: emerging targets for cardiovascular complications. Front Endocrinol (Lausanne) 2025; 16:1533620. [PMID: 40438395 PMCID: PMC12116311 DOI: 10.3389/fendo.2025.1533620] [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: 11/24/2024] [Accepted: 04/24/2025] [Indexed: 06/01/2025] Open
Abstract
Diabetes is a metabolic disorder primarily characterized by persistent hyperglycemia. Diabetes-induced inflammation significantly compromises cardiovascular health, greatly increasing the risk of atherosclerosis. The increasing prevalence of harmful lifestyle habits and overconsumption has contributed substantially to the global rise in diabetes-related cardiovascular diseases, creating a significant economic and healthcare burden. Although current therapeutic strategies focus on blood glucose control and metabolic regulation, clinical observations show that diabetic patients still face persistent residual risk of AS even after achieving metabolic stability. Recent studies suggest that this phenomenon is linked to diabetes-induced trained immunity. Diabetes can induce trained immunity in bone marrow progenitor cells and myeloid cells, thus promoting the long-term development of AS. This article first introduces the concept and molecular mechanisms of trained immunity, with particular emphasis on metabolic and epigenetic reprogramming, which plays a crucial role in sustaining chronic inflammation during trained immunity. Next, it summarizes the involvement of trained immunity in diabetes and its contribution to AS, outlining the cell types that can be trained in AS. Finally, it discusses the connection between diabetes-induced trained immunity and AS, as well as the potential of targeting trained immunity as an intervention strategy. Understanding the molecular mechanisms of trained immunity and their impact on disease progression may provide innovative strategies to address the persistent clinical challenges in managing diabetes and its complications.
Collapse
Affiliation(s)
- Yanan Bai
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Jianglan Wu
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Weixiong Jian
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
- Diagnostics of Traditional Chinese Medicine, National Key Discipline, Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
25
|
Coșarcă MC, Șincaru SV, Horváth E, Sala DT, Lazăr NA, Szanto LA, Harpa MM, Carașcă C, Ráduly G, Bândea P, Mureșan VA. The Role of Admission Glucose and Inflammatory Markers in Histopathological Features of Atherosclerotic Plaques in Carotid and Femoro-Popliteal Arteries. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:879. [PMID: 40428837 PMCID: PMC12113057 DOI: 10.3390/medicina61050879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 05/03/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025]
Abstract
Background and Objectives: Atherosclerosis is a chronic inflammatory disease significantly contributing to cardiovascular morbidity and mortality. This study primarily aims to evaluate the role of baseline blood glucose levels and inflammatory markers in the histopathological features of atherosclerotic plaques in the carotid and femoro-popliteal arteries. Materials and Methods: In this retrospective, observational, and monocentric study, 165 patients diagnosed with infrainguinal peripheral arterial disease or carotid artery disease hospitalized in the Vascular Surgery Clinic, between January 2019 and December 2023, were included. From the electronic database of the hospital, we documented demographic data, cardiovascular comorbidities, including hypertension, atrial fibrillation, ischemic heart disease, and chronic heart failure, as well as chronic kidney disease, diabetes, and prevalent risk factors such as active smoking, dyslipidemia, and obesity. Additionally, we recorded the arterial site from which the atherosclerotic plaque was obtained, along with laboratory data obtained at the time of admission prior to the surgery. The patients were divided into "Carotid Artery" and "Femoro-Popliteal Axis" based on anatomical location. Results: A greater prevalence of male patients (p = 0.008) and dyslipidemia (p = 0.002) was found in the group with atherosclerotic plaques from the femoro-popliteal axis. Laboratory data also showed increased lymphocyte (p = 0.020) and PLT (p = 0.028) levels in this group. There was no significant difference in the types of atherosclerotic plaques between the two patient groups. However, those in the Carotid Artery group showed a higher rate of antiaggregant treatment and a reduced incidence of dual therapy (p < 0.001). The Spearman correlation analysis revealed a positive correlation between baseline glucose levels and NLR (r = 0.402, p < 0.001), MLR (r = 0.217, p = 0.005), PLR (r = 0.306, p < 0.001), and LGI (r = 0.693, p < 0.001). Furthermore, the predictive roles of glucose, NLR, MLR, and LGI were assessed through multivariate analysis. Consequently, elevated baseline values of the parameters above were associated with unstable atherosclerotic plaques, independent of demo-graphic data, standard cardiovascular risk factors, site of artery harvest, and chronic vascular treatments at the time of admission (for all p < 0.05). Conclusions: This study highlights the significant relationships between glucose levels and various inflammatory markers in patients with different histopathological diagnoses of atherosclerotic plaques. Additionally, elevated glycemic and systemic inflammation biomarkers were associated with unstable atherosclerotic plaque, independent of demographic data, comorbidities, cardiovascular risk factors, anatomical artery harvest, and vascular chronic medication at the time of admission.
Collapse
Affiliation(s)
- Mircea Cătălin Coșarcă
- Department of Anatomy, ‘George Emil Palade’ University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania;
- Clinic of Vascular Surgery, Mures County Emergency Hospital (SCJU Târgu-Mureș), 540136 Targu Mures, Romania
| | - Suzana Vasilica Șincaru
- Clinic of Vascular Surgery, Mures County Emergency Hospital (SCJU Târgu-Mureș), 540136 Targu Mures, Romania
| | - Emőke Horváth
- Department of Morphopatology, ‘George Emil Palade’ University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
| | - Daniela Tatiana Sala
- Department of General Surgery, ‘George Emil Palade’ University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
| | - Nicolae Alexandru Lazăr
- Clinic of Vascular Surgery, Mures County Emergency Hospital (SCJU Târgu-Mureș), 540136 Targu Mures, Romania
| | - Ludovic Alexandru Szanto
- Clinic of Vascular Surgery, Mures County Emergency Hospital (SCJU Târgu-Mureș), 540136 Targu Mures, Romania
| | - Marius Mihai Harpa
- Department of Surgery Number IV, ‘George Emil Palade’ University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
| | - Cosmin Carașcă
- Department of Forensic Medicine, ‘George Emil Palade’ University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania;
| | - Gergő Ráduly
- Department of Anatomy, ‘George Emil Palade’ University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania;
| | - Paula Bândea
- Clinic of Vascular Surgery, Mures County Emergency Hospital (SCJU Târgu-Mureș), 540136 Targu Mures, Romania
| | - Vasile Adrian Mureșan
- Clinic of Vascular Surgery, Mures County Emergency Hospital (SCJU Târgu-Mureș), 540136 Targu Mures, Romania
- Department of Vascular Surgery, ‘George Emil Palade’ University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
| |
Collapse
|
26
|
Ding S, Choi SH, Miller YI. Amyloid β-Induced Inflammarafts in Alzheimer's Disease. Int J Mol Sci 2025; 26:4592. [PMID: 40429737 PMCID: PMC12111532 DOI: 10.3390/ijms26104592] [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: 04/20/2025] [Revised: 05/07/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
The formation of amyloid beta (Aβ) plaques is a central process in the development of Alzheimer's disease (AD). Although its causative role or the effectiveness of therapeutic targeting is still debated, the key involvement of Aβ in the pathogenesis of neuroinflammation and neurodegeneration in AD is broadly accepted. In this review, we emphasize the role of lipid rafts, both in APP cleavage producing Aβ in neurons and in mediating Aβ inflammatory signaling in microglia. We introduce the term inflammarafts to characterize the Aβ-driven formation of enlarged, cholesterol-rich lipid rafts in activated microglia, which support protein-protein and lipid-protein interactions of inflammatory receptors. Examples reviewed include toll-like receptors (TLR2, TLR4), scavenger receptors (CD36, RAGE), and TREM2. The downstream pathways lead to the production of cytokines and reactive oxygen species, intensifying neuroinflammation and resulting in neuronal injury and cognitive decline. We further summarize emerging therapeutic strategies and emphasize the utility of apolipoprotein A-I binding protein (AIBP) in selective targeting of inflammarafts and attenuation of microglia-driven inflammation. Unlike the targeting of a single inflammatory receptor or a secretase, selective disruption of inflammarafts and preservation of physiological lipid rafts offer a novel approach to targeting multiple components and processes that contribute to neuroinflammation in AD.
Collapse
Affiliation(s)
| | | | - Yury I. Miller
- Department of Medicine, University of California, San Diego, CA 92093, USA; (S.D.); (S.-H.C.)
| |
Collapse
|
27
|
Owczarek AJ, Ochman A, Chudek A, Mossakowska M, Puzianowska-Kuźnicka M, Kujawska-Danecka H, Zdrojewski T, Więcek A, Chudek J, Olszanecka-Glinianowicz M. Pentraxin-3 and C-reactive protein plasma levels predict survival in older adults with or without metabolic syndrome - results of the PolSenior2 substudy. Immun Ageing 2025; 22:16. [PMID: 40340865 PMCID: PMC12060356 DOI: 10.1186/s12979-025-00509-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 04/17/2025] [Indexed: 05/10/2025]
Abstract
OBJECTIVE There are no published data on the associations between plasma concentration of pentraxin-3 (PTX-3) - a marker of vascular inflammation and mortality in older subjects with or without metabolic syndrome (MS). Therefore, we aimed to compare the prognostic significance of increased PTX-3 and CRP levels on overall survival in subjects aged 60 and older with and without MS. MATERIALS AND METHODS Study participants (N = 3534) were categorized according to the presence or absence of MS and then each of these groups was stratified into 3 subgroups based on concentrations of CRP (≤ 3 mg/dL and > 3 mg/dL) and PTX-3 (< and ≥ the sex-specific cut-off values, based on the ROC curve analysis with the Youden index): double-negative inflammatory markers (low CRP and PTX-3 plasma concentrations); single-positive inflammatory marker (increased CRP or PTX-3 plasma concentrations) and double-positive inflammatory markers subgroup (increased CRP and PTX-3 plasma concentrations). During the 4.19-year follow-up, 678 (19.2% of the entire cohort) individuals died including 401 men (22.9%) and 277 women (15.5% ). RESULTS The optimal cut-off for PTX-3 plasma concentration associated with an increased risk of death was 2.07 ng/mL for men and 2.23 ng/mL for women. The death rates were increased for single-positive and were highest in double-positive subgroups both for men and women, with or without MS. Kaplan-Meier analysis showed no effect of MS on survival in men and women in subgroups within specific inflammatory marker categories. Of note, the inflammatory markers class effect on survival was already significant in the single-positive subgroups (34% and 44% higher risk for death for men and women), and even more pronounced for the double-positive subgroup (more than two and almost three times higher risk of death for men and women, respectively). In the entire study group, a weak correlation was found between plasma concentrations of PTX-3 and hs-CRP (ρ = 0.11, p < 0.001) and slightly higher in undernourished subjects with hs-CRP > 3 mg/dL (ρ = 0.28, p < 0.001). CONCLUSION Our study suggests that in the age-advanced Caucasian population, the inflammatory status with increased plasma levels of both PTX-3 and CRP is associated with a higher risk of all-cause mortality, regardless of the occurrence of MS. However, due to the retrospective study design, these results require confirmation in prospective studies with an analysis of the underlying causes of death.
Collapse
Affiliation(s)
- Aleksander J Owczarek
- Health Promotion and Obesity Management Unit, Department of Pathophysiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland.
| | - Anna Ochman
- Department of Internal Medicine and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Anna Chudek
- Health Promotion and Obesity Management Unit, Department of Pathophysiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Małgorzata Mossakowska
- Study on Aging and Longevity, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Monika Puzianowska-Kuźnicka
- Department of Human Epigenetics, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
- Department of Geriatrics and Gerontology, Medical Centre of Postgraduate Education, Warsaw, Poland
| | - Hanna Kujawska-Danecka
- Division of Preventive Medicine and Education, Medical University of Gdansk, Gdansk, Poland
| | - Tomasz Zdrojewski
- Division of Preventive Medicine and Education, Medical University of Gdansk, Gdansk, Poland
| | - Andrzej Więcek
- Department of Nephrology, Transplantation and Internal Medicine, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Jerzy Chudek
- Department of Internal Medicine and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Magdalena Olszanecka-Glinianowicz
- Health Promotion and Obesity Management Unit, Department of Pathophysiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| |
Collapse
|
28
|
Li Y, Wang YB, Zhu M, Du XY, Hou YY, Wu BB, Sun YX. Increased Hepatorenal Index Is Associated with the Risk of Developing Stroke in Patients with Nonalcoholic Fatty Liver Disease. Curr Med Sci 2025:10.1007/s11596-025-00051-1. [PMID: 40332736 DOI: 10.1007/s11596-025-00051-1] [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: 08/10/2024] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 05/08/2025]
Abstract
OBJECTIVE The aim of this study was to examine the relationship between the sonographic hepatorenal index and stroke risk in patients with nonalcoholic fatty liver disease (NAFLD). METHODS From December 2023 to July 2024, 72 NAFLD patients with stroke, 53 stroke-free NAFLD patients, and 54 healthy controls were enrolled in our study. The hepatorenal index was calculated as the ratio of the echo intensity of the liver to that of the renal cortex. The mean brightness values for one region of interest within the right hepatic lobe and the other size-matched region at the same depth of field within the right kidney were obtained with two-dimensional ultrasound and a 1- to 6-MHz convex array probe. Laboratory tests were performed with a Cobas 8000 automatic biochemical analyzer. Univariate and multivariate analyses were adopted to analyze the risk factors for stroke in NAFLD patients. RESULTS NAFLD patients had a greater hepatorenal index than healthy controls did (P < 0.05). Additionally, NAFLD patients with stroke had an even greater hepatorenal index than did those with stroke-free NAFLD (P < 0.001). Multivariate regression analysis further revealed that the hepatorenal index was independently associated with stroke risk in NAFLD patients (β = 8.897, P < 0.001) after controlling for age, body mass index, systolic blood pressure, and serum glucose, total cholesterol, alanine transaminase, and creatinine concentrations. Receiver operating characteristic curve analysis revealed a sensitivity of 62.5% and specificity of 95.3% for the hepatorenal index, with a cutoff value of 1.255 and an area under the curve of 0.80. CONCLUSION The increased sonographic hepatorenal index could be an independent predictor of stroke development in patients with NAFLD.
Collapse
Affiliation(s)
- Yang Li
- Department of Medical Ultrasound, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China
| | - Yi-Bin Wang
- Department of Medical Ultrasound, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Min Zhu
- Department of Medical Ultrasound, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China
| | - Xiao-Ying Du
- Department of Medical Ultrasound, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China
| | - Ying-Ying Hou
- Department of Medical Ultrasound, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China
| | - Ban-Ban Wu
- Department of Clinical and Research, Shenzhen Mindray Bio-Medical Electronics Company Limited, Shenzhen, 518000, China
| | - Yi-Xue Sun
- Department of Medical Ultrasound, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China
| |
Collapse
|
29
|
Meng Q, Huang Y, Long X, Liu L, Tang Y, He J, Luo Y. Scutellarin mitigates high glucose-induced pyroptosis in diabetic atherosclerosis: Role of Nrf2-FBXL2-mediated NLRP3 degradation. Endocr J 2025; 72:495-507. [PMID: 40010729 PMCID: PMC12086276 DOI: 10.1507/endocrj.ej24-0505] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/30/2024] [Indexed: 02/28/2025] Open
Abstract
This study investigated the role of scutellarin (Scu) and Nrf2 in diabetic atherosclerosis, focusing on their effects on FBXL2 and NLRP3 ubiquitination. Human umbilical vein endothelial cells were treated with high glucose (HG) to model diabetic atherosclerosis in vitro. Cell viability, cytotoxicity, pyroptosis, and inflammatory cytokine levels were assessed, and gene interactions were examined by dual-luciferase reporter assays. Ubiquitination and protein levels were analyzed through immunoprecipitation and western blotting. The results revealed that HG treatment decreased Nrf2 and FBXL2 levels and enhanced NLRP3-mediated pyroptosis. However, Scu treatment increased Nrf2 expression, improved cell viability, and inhibited pyroptosis. Nrf2 knockdown downregulated FBXL2 and reversed the protective effects of Scu. Additionally, FBXL2 promoted the ubiquitination-mediated degradation of NLRP3 and suppressed pyroptosis. The activation of NLRP3 reversed the protective effects of Scu on diabetic atherosclerosis. These findings suggest that Scu alleviated diabetic atherosclerosis by increasing Nrf2 and FBXL2 expression, promoting NLRP3 ubiquitination-mediated degradation, and suppressing pyroptosis.
Collapse
Affiliation(s)
- Qingxin Meng
- School of Medicine, Changsha Social Work College, Changsha, Hunan Province 410004, China
| | - Yongpan Huang
- School of Medicine, Changsha Social Work College, Changsha, Hunan Province 410004, China
| | - Xian Long
- School of Medicine, Changsha Social Work College, Changsha, Hunan Province 410004, China
| | - Lijing Liu
- School of Medicine, Changsha Social Work College, Changsha, Hunan Province 410004, China
| | - Yani Tang
- School of Medicine, Changsha Social Work College, Changsha, Hunan Province 410004, China
| | - Jingjing He
- School of Medicine, Changsha Social Work College, Changsha, Hunan Province 410004, China
| | - Yayuan Luo
- Department of Neurology, Hunan Aerospace Hospital, Changsha, Hunan Province 410205, China
| |
Collapse
|
30
|
Chen Q, Lai H, Chen Y, Peng Z, Wu S, Liu D. Characterization of circRNA expression profiles and functional roles in a mouse model of liver injury induced by OSA. Sci Rep 2025; 15:15615. [PMID: 40320447 PMCID: PMC12050294 DOI: 10.1038/s41598-025-99612-6] [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: 01/12/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025] Open
Abstract
Despite mounting evidence linking circular RNAs (circRNAs) to various diseases, their specific role in liver damage triggered by obstructive sleep apnea (OSA) remains ambiguous. This study investigates alterations in circRNA expression patterns in a mouse model subjected to chronic intermittent hypoxia (CIH), aiming to elucidate the pathways that lead to liver damage associated with OSA. We established the CIH model and conducted circRNA microarray analysis on liver samples from both CIH and control groups. The findings were substantiated via qRT-PCR. Furthermore, a comprehensive circRNA-miRNA-mRNA (ceRNA) network was developed, followed by the analysis of GO and KEGG pathways to further elucidate the underlying biological processes. We identified 259 differentially expressed circRNAs, comprising 86 that were upregulated and 173 that were downregulated in CIH mice. The ceRNA analysis suggested that these circRNAs may modulate gene expression by sequestering miRNAs. Our findings highlight potential therapeutic targets for liver pathologies associated with OSA.
Collapse
Affiliation(s)
- Qingshi Chen
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.
| | - Huiting Lai
- Department of Interventional Therapy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yuwei Chen
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Zhuli Peng
- Department of Interventional Therapy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Siying Wu
- Department of Interventional Therapy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Dexin Liu
- Department of Interventional Therapy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.
| |
Collapse
|
31
|
López-González ÁA, Ramírez-Manent JI, Martínez-Almoyna Rifá E, Paublini Oliveira H, Martorell Sánchez C, Tárraga López PJ. Association between sociodemographic variables, lifestyle factors, and stress with lipoprotein ratio values. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2025:500803. [PMID: 40318947 DOI: 10.1016/j.arteri.2025.500803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/30/2025] [Accepted: 04/08/2025] [Indexed: 05/07/2025]
Abstract
INTRODUCTION Atherosclerosis is a multifactorial process underlying major cardiovascular diseases (CVDs). Among the associated risk factors, lipoprotein ratios have been identified as key indicators of atherogenic risk. However, the influence of sociodemographic variables, lifestyle factors, and stress levels on lipoprotein ratios remains underexplored. OBJECTIVE To analyze the relationship between sociodemographic variables, healthy lifestyle habits, and stress levels with lipoprotein ratios and atherogenic risk in a large cohort of Spanish workers. MATERIALS AND METHODS A cross-sectional study was conducted in 24,244 Spanish workers. The association between age, gender, socioeconomic status, tobacco and alcohol consumption, physical activity, adherence to the Mediterranean diet, and stress levels with atherogenic risk scales, including atherogenic dyslipidemia (AD) and atherogenic índices, was evaluated. RESULTS A significant association was found between all analyzed variables and lipoprotein ratios. The strongest associations were observed with age and gender, followed by occupational status, tobacco and alcohol consumption, physical activity, diet, and stress levels. Men exhibited a higher atherogenic risk compared to women (OR 2.36; 95% CI 2.18-2.55). The risk increased significantly with age, peaking in the 50-69 age group (OR 5.00; 95% CI 3.70-6.31). Manual workers had a higher prevalence of atherogenic dyslipidemia compared to non-manual workers (OR 1.35; 95% CI 1.27-1.41). Furthermore, smoking (OR 1.84; 95% CI 1.71-1.97) and alcohol consumption (OR 1.45; 95% CI 1.36-1.55), physical inactivity (OR 1.90; 95% CI 1.76-2.05), and low adherence to the Mediterranean diet (OR 1.72; 95% CI 1.60-1.84) were associated with higher atherogenic index values. CONCLUSIONS The presence of an adverse lipoprotein profile is strongly influenced by sociodemographic factors, lifestyle habits, and stress. Identifying these determinants may enable the implementation of preventive strategies aimed at reducing atherogenic risk and improving cardiovascular health in working populations.
Collapse
Affiliation(s)
- Ángel Arturo López-González
- Grupo ADEMA-Salud, Instituto Universitario de Ciencias de la Salud (IUNICS) de Baleares, Palma de Mallorca, España; Facultad de Odontología, Escuela Universitaria ADEMA-UIB, Palma de Mallorca, España; Servicio de Salud de las Islas Baleares, España
| | - José Ignacio Ramírez-Manent
- Grupo ADEMA-Salud, Instituto Universitario de Ciencias de la Salud (IUNICS) de Baleares, Palma de Mallorca, España; Servicio de Salud de las Islas Baleares, España; Facultad de Medicina, Universidad de las Islas Baleares, Palma de Mallorca, España
| | - Emilio Martínez-Almoyna Rifá
- Grupo ADEMA-Salud, Instituto Universitario de Ciencias de la Salud (IUNICS) de Baleares, Palma de Mallorca, España; Facultad de Odontología, Escuela Universitaria ADEMA-UIB, Palma de Mallorca, España
| | - Hernán Paublini Oliveira
- Grupo ADEMA-Salud, Instituto Universitario de Ciencias de la Salud (IUNICS) de Baleares, Palma de Mallorca, España; Facultad de Odontología, Escuela Universitaria ADEMA-UIB, Palma de Mallorca, España
| | - Cristina Martorell Sánchez
- Grupo ADEMA-Salud, Instituto Universitario de Ciencias de la Salud (IUNICS) de Baleares, Palma de Mallorca, España; Facultad de Odontología, Escuela Universitaria ADEMA-UIB, Palma de Mallorca, España
| | | |
Collapse
|
32
|
Khandelwal P, Fish JD, Gorfinkel L, Guilcher GMT, Howell J, Nishitani M, Nuechterlein B, Obeng E, Shah AJ, Cuvelier GDE, Rotz S, Williams KM, Duncan CN. Pediatric Transplant and Cellular Therapy Consortium RESILIENT Conference on Pediatric Chronic Graft-versus-Host Disease Survivorship after Hematopoietic Cell Transplantation: Part III. Long-Term Impact of Chronic Graft-versus-Host Disease on Endocrinologic, Cardiovascular, and Metabolic Outcomes in Survivors of Pediatric Hematopoietic Cell Transplantation. Transplant Cell Ther 2025; 31:297.e1-297.e15. [PMID: 39952365 PMCID: PMC12082646 DOI: 10.1016/j.jtct.2025.01.891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/07/2025] [Accepted: 01/27/2025] [Indexed: 02/17/2025]
Abstract
Chronic graft-versus-host disease (cGVHD) has a profound impact on the endocrinologic and cardiovascular health of survivors of transplantation performed in childhood. The impact of cGVHD is long-lasting and contributes to morbidity and early mortality through multiple mechanisms. Organs and tissues may be direct targets of alloreactive donor-derived immune cells. Corticosteroids and other cGVHD-directed therapies influence hormonal actions, alter bone metabolism, and negatively impact cardiometabolic health. Pediatric survivors are particularly vulnerable to the endocrinologic and cardiovascular effects of cGVHD as it develops during periods of intense growth and development, although little is known about the direct contribution to late effects. The Research and Education Toward Solutions for Late Effects to Innovate, Excel, and Nurture after cGVHD (RESILIENT after cGVHD) effort brought together content experts to determine the state of the science, develop clinical recommendations, and propose a research agenda in endocrine, cardiovascular, and metabolic cGVHD survivorship, which are detailed in this report.
Collapse
Affiliation(s)
| | - Jonathan D Fish
- Steven and Alexandra Cohen Children's Medical Center, Queens, New York
| | | | | | - Jonathan Howell
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | | | - Brandon Nuechterlein
- University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado
| | - Esther Obeng
- Saint Jude Children's Research Hospital Memphis, Tennessee
| | - Ami J Shah
- Stanford University School of Medicine, Stanford, California
| | | | | | | | | |
Collapse
|
33
|
Rikken SAOF, Gibson CM, Bahit MC, Duffy D, Chi G, Korjian S, Mohammadnia N, White H, Anschuetz G, Kingwell BA, Ophuis TO, Nicolau JC, Lopes RD, Lewis BS, El Messaoudi S, Vinereanu D, Ten Berg JM, Goodman SG, Bode C, Steg PG, Libby P, Bainey KR, van 't Hof AWJ, Ridker PM, Mahaffey KW, Nicholls SJ, Mehran R, Harrington RA, Cornel JH, AEGIS-II Committees and Investigators. Impact of Apolipoprotein A-I Infusions on Cardiovascular Events Post-MI by Neutrophil-Lymphocyte Ratio and LDL-Cholesterol Levels. JACC. ADVANCES 2025; 4:101727. [PMID: 40288083 PMCID: PMC12059331 DOI: 10.1016/j.jacadv.2025.101727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND The AEGIS-II (ApoA-I Event Reducing in Ischemic Syndromes-II; NCT03473223) trial evaluated CSL112, a human plasma-derived apolipoprotein A-I therapy, for reducing cardiovascular events after acute myocardial infarction (AMI). Given CSL112's potential anti-inflammatory properties, we conducted an exploratory post hoc analysis to determine if its efficacy is influenced by baseline neutrophil-lymphocyte ratio (NLR), a marker of systemic inflammation, and low-density lipoprotein cholesterol (LDL-C). OBJECTIVES The purpose of this study was to investigate the association of baseline NLR and cardiovascular events and explore whether NLR and LDL-C modify CSL112's efficacy in post-AMI patients. METHODS A total of 18,219 participants with AMI, multivessel coronary artery disease, and additional cardiovascular risk factors were randomized to 4 weekly infusions of 6 g CSL112 or placebo. The primary endpoint was a composite of cardiovascular death, myocardial infarction, or stroke (major adverse cardiovascular events [MACE]). Cox proportional hazards models evaluated risk by dichotomized baseline NLR (>median vs ≤median). Treatment interactions with NLR and LDL-C (≥100 vs <100 mg/dL) were assessed. RESULTS Among 15,966 participants, those with baseline NLR >median (>3.3) had a significantly greater risk of MACE at 90 days (HR: 1.40; 95% CI: 1.21-1.63), persisting at 180 and 365 days. CSL112 reduced MACE at 90 days among participants with elevated NLR and LDL-C ≥100 mg/dL (HR: 0.63; 95% CI: 0.42-0.93), with sustained benefits at 180 and 365 days. Significant interactions were observed between treatment and NLR (Pinteraction = 0.010) and among treatment, NLR, and LDL-C at 180 days (Pinteraction = 0.029). CONCLUSIONS Baseline elevated NLR predicts MACE in post-AMI patients, and CSL112 showed an associated reduction in MACE in patients with elevated NLR and LDL-C ≥100 mg/dL.
Collapse
Affiliation(s)
- Sem A O F Rikken
- Department of Cardiology, St. Antonius Hospital, Nieuwegein, the Netherlands; Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands; Baim Institute for Clinical Research, Boston, Massachusetts, USA
| | - C Michael Gibson
- Baim Institute for Clinical Research, Boston, Massachusetts, USA; Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.
| | - M Cecilia Bahit
- Baim Institute for Clinical Research, Boston, Massachusetts, USA; INECO Neurociencias Rosario, Rosario, Argentina
| | | | - Gerald Chi
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Serge Korjian
- Baim Institute for Clinical Research, Boston, Massachusetts, USA; Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Harvey White
- Baim Institute for Clinical Research, Boston, Massachusetts, USA; Health New Zealand Te Toka Tumai Auckland City Hospital, Auckland, New Zealand
| | | | | | | | - Jose C Nicolau
- Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Renato D Lopes
- Duke Clinical Research Institute, Duke Health, Durham, North Carolina, USA; Brazilian Clinical Research Institute, Sao Paulo, SP, Brazil
| | - Basil S Lewis
- Baim Institute for Clinical Research, Boston, Massachusetts, USA; Lady Davis Carmel Medical Center, Haifa, Israel; The Technion-Israel Institute of Technology, Haifa, Israel
| | - Saloua El Messaoudi
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Dragos Vinereanu
- University of Medicine and Pharmacy Carol Davila, University and Emergency Hospital, Bucharest, Romania
| | - Jurriën M Ten Berg
- Department of Cardiology, St. Antonius Hospital, Nieuwegein, the Netherlands; Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands; Department of Cardiology, University Medical Center Maastricht, Maastricht, the Netherlands
| | - Shaun G Goodman
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada; St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; Peter Munk Cardiac Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Christoph Bode
- Heart Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - P Gabriel Steg
- Baim Institute for Clinical Research, Boston, Massachusetts, USA; Université Paris-Cité, INSERM U_1148, FACT and AP-HP, Hôpital Bichat, Paris, France
| | - Peter Libby
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kevin R Bainey
- Baim Institute for Clinical Research, Boston, Massachusetts, USA; Walter Mackenzie Health Sciences Centre, University of Alberta Hospital, Edmonton, Alberta, Canada
| | - Arnoud W J van 't Hof
- Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands; University of Medicine and Pharmacy Carol Davila, University and Emergency Hospital, Bucharest, Romania; Department of Cardiology, Zuyderland Medical Center, Heerlen, The Netherlands
| | - Paul M Ridker
- Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Kenneth W Mahaffey
- Stanford Center for Clinical Research, Department of Medicine, Stanford University School of Medicine, Palo Alto, California, USA
| | - Stephen J Nicholls
- Baim Institute for Clinical Research, Boston, Massachusetts, USA; Victorian Heart Institute, Monash University, Melbourne, Victoria, Australia
| | - Roxana Mehran
- Icahn School of Medicine at Mount Sinai, Zena and Michael A. Wiener Cardiovascular Institute, New York, New York, USA
| | | | - Jan H Cornel
- Baim Institute for Clinical Research, Boston, Massachusetts, USA; Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands; Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands
| | | |
Collapse
|
34
|
Pei J, Chen S, Ke Q, Pang A, Niu M, Li N, Li J, Wang Z, Wu H, Nie P. Immune response to polystyrene microplastics: Regulation of inflammatory response via the ROS-driven NF-κB pathway in zebrafish (Danio rerio). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2025; 282:107308. [PMID: 40058300 DOI: 10.1016/j.aquatox.2025.107308] [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: 01/11/2025] [Revised: 02/16/2025] [Accepted: 03/02/2025] [Indexed: 04/05/2025]
Abstract
There is increasing apprehension regarding the rising prevalence of microplastics (MPs) in aquatic ecosystems. Although MPs cause toxicological effect on fish via diverse pathways, the precise immunotoxicological mechanism is yet to be fully understood. Utilizing zebrafish in early developmental stages and zebrafish embryonic fibroblast (ZF4) as models, this study delved into the immune response elicited by polystyrene MPs (PS-MPs). It was observed that larvae predominantly accumulate 3 μm PS-MPs in their intestines through ingestion, leading to notable changes in locomotor behavior and histopathological alterations. Further investigation revealed that short-term exposure to PS-MPs triggers oxidative stress (OS) and inflammation in zebrafish. This is evidenced by the upregulation of OS and inflammation-related genes, increased levels of reactive oxygen species (ROS), malonaldehyde (MDA), and inflammatory cytokines, altered activities of antioxidant enzymes, along with induced recruitment of leukocyte in larvae. Cellular assays confirmed that PS-MPs elevate intracellular ROS in ZF4 cells and enhance the nuclear translocation of NF-κB P65. Notably, the activation of NF-κB and the upsurge in inflammatory cytokines can be mitigated by inhibiting ROS. This research highlights the significance of the ROS-triggered NF-κB signaling cascade in PS-MPs-mediated inflammation within zebrafish, illuminating the possible processes that underlie the innate immune system of fish toxicity caused by MPs.
Collapse
Affiliation(s)
- Jincheng Pei
- Hubei Key Laboratory of Multi-media Pollution Cooperative Control in Yangtze Basin, School of Environmental Science & Engineering, Huazhong University of Science and Technology (HUST), Wuhan, Hubei Province 430074, PR China
| | - Shannan Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province 430072, PR China
| | - Qingxia Ke
- Yangxin County Fishery Service Center, Huangshi, Hubei Province 435200, PR China
| | - Anning Pang
- State Key Laboratory of Freshwater Ecology and Biotechnology and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province 430072, PR China
| | - Mengmeng Niu
- State Key Laboratory of Freshwater Ecology and Biotechnology and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province 430072, PR China
| | - Nan Li
- State Key Laboratory of Freshwater Ecology and Biotechnology and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province 430072, PR China
| | - Jiayi Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266109, PR China
| | - Zhi Wang
- Yangxin County Fishery Service Center, Huangshi, Hubei Province 435200, PR China
| | - Hongjuan Wu
- Hubei Key Laboratory of Multi-media Pollution Cooperative Control in Yangtze Basin, School of Environmental Science & Engineering, Huazhong University of Science and Technology (HUST), Wuhan, Hubei Province 430074, PR China.
| | - Pin Nie
- State Key Laboratory of Freshwater Ecology and Biotechnology and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province 430072, PR China; School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266109, PR China.
| |
Collapse
|
35
|
Abavisani M, Tafti P, Khoshroo N, Ebadpour N, Khoshrou A, Kesharwani P, Sahebkar A. The heart of the matter: How gut microbiota-targeted interventions influence cardiovascular diseases. Pathol Res Pract 2025; 269:155931. [PMID: 40174272 DOI: 10.1016/j.prp.2025.155931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 03/10/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
The human body is habitat to a wide spectrum of microbial populations known as microbiota, which play an important role in overall health. The considerable research has mostly focused on the gut microbiota due to its potential to impact numerous physiological functions and its correlation with a variety of disorders, such as cardiovascular diseases (CVDs). Imbalances in the gut microbiota, known as dysbiosis, have been linked to the development and progression of CVDs through various processes, including the generation of metabolites like trimethylamine-N-oxide and short-chain fatty acids. Studies have also looked at the idea of using therapeutic interventions, like changing your diet, taking probiotics or prebiotics, or even fecal microbiota transplantation (FMT), to change the gut microbiota's make-up and how it works in order to prevent or treat CVDs. Exploring the cause-and-effect connection between the gut microbiota and CVDs offers a hopeful path for creating innovative microbiome-centered strategies to prevent and cure CVDs. This review presents an in-depth review of the correlation between the gut microbiota and CVDs, as well as potential therapeutic approaches for manipulating the gut microbiota to enhance cardiovascular health.
Collapse
Affiliation(s)
- Mohammad Abavisani
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Pourya Tafti
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niloofar Khoshroo
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Ebadpour
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Khoshrou
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pardesh, India; University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
36
|
Wang H, Ding K, He J, Wang J. Tetrahydropalmatine promotes macrophage autophagy by inhibiting the AMPK/mTOR pathway to attenuate atherosclerosis. Histol Histopathol 2025; 40:697-710. [PMID: 39359170 DOI: 10.14670/hh-18-809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
BACKGROUND Atherosclerosis (AS) is a chronic progressive arterial disease that is associated with macrophage autophagy and AMP-activated protein kinase (AMPK)/mechanistic target of the rapamycin (mTOR) pathway. Tetrahydropalmatine (THP) can activate AMPK-dependent autophagy. We aim to study the mechanism of macrophage autophagy mediated by THP in the treatment of AS via the AMPK/mTOR pathway. METHODS High-fat diet apolipoprotein E-deficient mice and ox-LDL-induced RAW264.7 cells were used to mimic the AS model, then THP was administered. Cell viability was detected by MTT. Pathological aorta lesions were detected using Hematoxylin and Eosin, Masson, and oil red staining. Lipid metabolism indices and inflammatory factors were measured using ELISA. A transmission electron microscope was used to observe autophagosomes. Autophagy and AMPK/mTOR pathway protein expression was detected by immunofluorescence and Western blot. The AMPK inhibitor 9-β-d-Arabinofuranosyl Adenine (Ara-A) was used to validate the effect of THP. The mRNA expression of Beclin-1 and MCP-1 was detected by q-PCR. RESULTS THP administration regulated lipid metabolism by lowering total cholesterol, triacylglycerol, low-density lipoprotein, and high-density lipoprotein levels, and suppressed aortic damage. THP suppressed aortic damage and regulated lipid metabolism by altering serum lipid levels. THP reduced inflammation and macrophage CD68 expression. Twenty μg/mL THP reduced cell viability. THP decreased cholesterol uptake and increased efflux, promoting autophagy. THP increased autophagosome number, LC3B expression, and autophagy markers p-AMPK/AMPK and LC3-II/LC3-I. THP also decreased p-mTOR/mTOR and P62. THP increased Beclin-1 mRNA expression and decreased MCP-1 mRNA expression. Ara-A reversed THP's effects. CONCLUSION THP promotes macrophage autophagy by inhibiting the AMPK/mTOR pathway to attenuate AS.
Collapse
Affiliation(s)
- Hui Wang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ke Ding
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Jiaqi He
- Traditional Chinese Medicine Dispensary, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jiahong Wang
- Traditional Chinese Medicine Dispensary, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
37
|
Xing Y, Lin X. Challenges and advances in the management of inflammation in atherosclerosis. J Adv Res 2025; 71:317-335. [PMID: 38909884 DOI: 10.1016/j.jare.2024.06.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 06/25/2024] Open
Abstract
INTRODUCTION Atherosclerosis, traditionally considered a lipid-related disease, is now understood as a chronic inflammatory condition with significant global health implications. OBJECTIVES This review aims to delve into the complex interactions among immune cells, cytokines, and the inflammatory cascade in atherosclerosis, shedding light on how these elements influence both the initiation and progression of the disease. METHODS This review draws on recent clinical research to elucidate the roles of key immune cells, macrophages, T cells, endothelial cells, and clonal hematopoiesis in atherosclerosis development. It focuses on how these cells and process contribute to disease initiation and progression, particularly through inflammation-driven processes that lead to plaque formation and stabilization. Macrophages ingest oxidized low-density lipoprotein (oxLDL), which partially converts to high-density lipoprotein (HDL) or accumulates as lipid droplets, forming foam cells crucial for plaque stability. Additionally, macrophages exhibit diverse phenotypes within plaques, with pro-inflammatory types predominating and others specializing in debris clearance at rupture sites. The involvement of CD4+ T and CD8+ T cells in these processes promotes inflammatory macrophage states, suppresses vascular smooth muscle cell proliferation, and enhances plaque instability. RESULTS The nuanced roles of macrophages, T cells, and the related immune cells within the atherosclerotic microenvironment are explored, revealing insights into the cellular and molecular pathways that fuel inflammation. This review also addresses recent advancements in imaging and biomarker technology that enhance our understanding of disease progression. Moreover, it points out the limitations of current treatment and highlights the potential of emerging anti-inflammatory strategies, including clinical trials for agents such as p38MAPK, tumor necrosis factor α (TNF-α), and IL-1β, their preliminary outcomes, and the promising effects of canakinumab, colchicine, and IL-6R antagonists. CONCLUSION This review explores cutting-edge anti-inflammatory interventions, their potential efficacy in preventing and alleviating atherosclerosis, and the role of nanotechnology in delivering drugs more effectively and safely.
Collapse
Affiliation(s)
- Yiming Xing
- Cardiology Department, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, 230022, China
| | - Xianhe Lin
- Cardiology Department, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, 230022, China.
| |
Collapse
|
38
|
Baltramonaityte V, Karhunen V, Felix JF, Penninx BWJH, Cecil CAM, Fairchild G, Milaneschi Y, Walton E. Biological pathways underlying the relationship between childhood maltreatment and Multimorbidity: A two-step, multivariable Mendelian randomisation study. Brain Behav Immun 2025; 126:59-69. [PMID: 39900145 DOI: 10.1016/j.bbi.2025.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/29/2025] [Accepted: 01/31/2025] [Indexed: 02/05/2025] Open
Abstract
Childhood maltreatment has been associated with multimorbidity of depression, coronary artery disease and type 2 diabetes. However, the biological mechanisms underlying this association remain unclear. We employed two-step and multivariable Mendelian randomisation (MR) to understand the role of three potential biological mediating mechanisms - inflammation (92 proteins), metabolic processes (54 markers), and cortisol - in the link between childhood maltreatment liability and multimorbidity. Using summary statistics from large-scale genome-wide association studies of European ancestry for childhood maltreatment (N = 185,414) and multimorbidity (Neffective = 156,717), we tested for the presence of an indirect effect via each mediator individually. We found a potential role of metabolic pathways. Up to 11% of the effect of childhood maltreatment on multimorbidity was mediated by triglycerides (indirect effect [95% CI]: 0.018 [0.009-0.027]), 8% by glycated haemoglobin (indirect effect: 0.013 [0.003-0.023]), and up to 7% by high-density lipoprotein cholesterol (indirect effect: 0.011 [0.005-0.017]). We did not find evidence for mediation via any inflammatory protein or cortisol. Our findings shed light on the biological mechanisms linking childhood maltreatment liability to multimorbidity, highlighting the role of metabolic pathways. Future studies may explore underlying pathways via non-biological mediators (e.g., lifestyle factors) or via multiple mediators simultaneously.
Collapse
Affiliation(s)
| | - Ville Karhunen
- MRC Biostatistics Unit, University of Cambridge, United Kingdom; Research Unit of Population Health, University of Oulu, Oulu, Finland; Research Unit of Mathematical Sciences, University of Oulu, Oulu, Finland
| | - Janine F Felix
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Amsterdam Public Health, Mental Health Program, Amsterdam, the Netherlands; Amsterdam Neuroscience, Complex Trait Genetics, Amsterdam, the Netherlands
| | - Charlotte A M Cecil
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Graeme Fairchild
- Department of Psychology, University of Bath, Bath, United Kingdom
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Amsterdam Public Health, Mental Health Program, Amsterdam, the Netherlands; Amsterdam Neuroscience, Complex Trait Genetics, Amsterdam, the Netherlands
| | - Esther Walton
- Department of Psychology, University of Bath, Bath, United Kingdom.
| |
Collapse
|
39
|
Hu S, Tang S, Liu D, Xia R, Wang X. Walnut oil as a dietary intervention for atherosclerosis: Efficacy and mechanistic pathways. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159607. [PMID: 40064415 DOI: 10.1016/j.bbalip.2025.159607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 02/15/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND AND AIMS Walnut oil (WO) and peanut oil (PO) are common vegetable oils rich in unsaturated fatty acids, known to alleviate atherosclerosis (AS) and reduce the risk of cardiovascular diseases (CVD). WO contains a higher proportion of polyunsaturated fatty acids (PUFAs) compared to PO. This study aimed to explore the influence of WO on AS and elucidate its potential mechanisms, providing a theoretical basis for enhancing the application of WO in functional foods and pharmaceuticals. METHODS AS was established in rats using a high-fat diet and vitamin D3 injections. Rats with AS were administered WO or PO via gavage at a dose of 1.2 g/kg for 4 weeks. Serum lipid levels and arterial injury were assessed, and transcriptomic and metabolomic analyses of the rat vasculature were performed. RESULTS Both WO and PO significantly lowered serum lipid levels and the atherogenic index (AI) in rats, reducing arterial wall injury and plaque formation. WO exhibited a more pronounced effect, particularly in decreasing serum levels of TG, TC, HDLC, and LDL-C. Transcriptomic analysis indicated that fatty acid, amino acid metabolism were crucial in AS development due to a high-fat diet. Metabolomic analysis indicated significant changes in the metabolism of arginine, proline, cysteine, methionine, glycine, serine and threonine in rats treated with WO. CONCLUSION WO and PO help alleviate AS by regulating lipid metabolism and influencing pivotal metabolic pathways like TCA cycle and cysteine-methionine metabolism. The more significant impact of WO indicates its potential as a dietary supplement for preventing and treating AS.
Collapse
Affiliation(s)
- Shujuan Hu
- School of Education and Physical Education, Yangtze University, Jingzhou, Hubei 434023, China
| | - Si Tang
- Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
| | - Dang Liu
- Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
| | - Ruohan Xia
- Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
| | - Xianwang Wang
- Department of Biochemistry and Molecular Biology, Center for Molecular Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China; Shannan Maternal and Child Health Hospital, Shannan, Xizang 856100, China.
| |
Collapse
|
40
|
Pan J, Huang J, Chen Y, Jiang N, Guo Y, Zhang J, Zhou S, Pu H, Deng Q, Hu B, Zhou Q. Genetic Evidence of Causal Effect between C1q/TNF-Related Protein-1 and Atherosclerosis: a Bidirectional and Multivariate Mendelian Randomization Study. J Atheroscler Thromb 2025; 32:630-648. [PMID: 39617479 PMCID: PMC12055506 DOI: 10.5551/jat.65313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/03/2024] [Indexed: 05/02/2025] Open
Abstract
AIMS To investigate the causal relationship between C1q/TNF-related protein-1 (CTRP1) and atherosclerosis across various vascular sites, informed by studies connecting CTRP1 to coronary artery disease. METHODS Summary statistics of CTRP1 from the available genome-wide association studies and atherosclerosis in classic vascular sites (including cerebral, coronary, and other arteries) from the FinnGen biobank were extracted for a primary MR analysis, and the analysis was replicated using Ischemic Stroke cohort (large artery atherosclerosis) for validation. The inverse variance-weighted method was used for primary assessment. Sensitivity analysis was performed by Cochrane's Q test and leave-one-out analysis. Potential pleiotropic effects were assessed by MR-Egger intercept and MR-PRESSO global test. Additionally, multivariable MR (MVMR) analysis was performed to investigate the independent effect of CTRP1 on atherosclerosis after removing confounding factors. RESULTS Reliable causal evidence was found for CTRP1 involvement in three atherosclerosis endpoints: causal effects of CTRP1 on cerebral atherosclerosis (OR=1.31, CI:1.04-1.66; FDR_P=0.0222)], coronary atherosclerosis (OR=1.13, CI: 1.08-1.19; FDR_P=2.86e-07), and atherosclerosis at other sites (OR=1.06, CI:1.02-1.11; FDR_P=0.0125). The validation cohort further confirmed its causal effect on large-artery atherosclerosis (OR=1.10, CI:1.03-1.18; FDR_P=0.0115). The reverse MR analysis did not support the causal effect of atherosclerosis on CTRP1. Moreover, the MVMR analysis, adjusting for confounders (CTRP3, CTRP5, and CTRP9A), highlighted a significant independent causal effect of CTRP1 remaining on atherosclerosis. CONCLUSION CTRP1 may represent a promising target for preventing and treating systemic atherosclerosis.
Collapse
Affiliation(s)
- Juhong Pan
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jia Huang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yueying Chen
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, China
| | - Nan Jiang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuxin Guo
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ji Zhang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shiyuan Zhou
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, China
| | - Huan Pu
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing Deng
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bo Hu
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing Zhou
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
41
|
Kang CM, Zhao JJ, Xie XX, Yu KW, Lai BC, Wang YX, Li TT, Ke PF, Huang XZ. Unveiling the role of GATA4 in endothelial cell senescence and atherosclerosis development. Atherosclerosis 2025; 404:119183. [PMID: 40209341 DOI: 10.1016/j.atherosclerosis.2025.119183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 03/17/2025] [Accepted: 04/02/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND AND AIMS Cellular senescence is intimately linked to atherosclerosis development and progression. However, the mechanism is not well known. GATA4 is a classical regulator in human fibroblast senescence. This study aimed to determine the role of GATA4 in endothelial cell (EC) senescence and atherosclerosis development and the mechanisms by which it acts. METHODS Senescence ECs were induced using H2O2 by isolating human primary umbilical vein ECs from umbilical veins. The level of GATA4 was examined in endothelial progenitor cells (EPCs), ECs of arterial tissue from older individuals (>65 years), and aged mice (>24 months). Adeno-associated virus with EC-selective Tie1 promoter, an EC-specific gene transduction system, was used to explore the role of GATA4 in EC senescence and atherosclerosis development in ApoE-/- mice. RT-qPCR, Western blot, ChIP-PCR, and ELISA were conducted to further explore the mechanism of GATA4 in EC senescence and atherosclerosis development. RESULTS GATA4 protein levels are elevated in EC senescence induced by H2O2 and EPCs in older individuals. Additionally, GATA4 protein levels are increased in the ECs of arterial tissue from older individuals and aged mice and are strongly correlated with the progression of atherosclerosis plaques. Knockdown of GATA4 decreased EC senescence, dysfunction, and monocyte adhesion. Mechanistically, we found that GATA4 activates NFκB2 transcription and induces senescence-associated secretory phenotype (SASP) expression (IL-6, IL-8, CXCL1, CXCL3, ICAM-1). In vivo experiments on ApoE-/- mice demonstrated that GATA4 overexpression in ECs contributes to higher SASP expression, vascular senescence, atherosclerotic plaque formation, and impaired cardiac function. CONCLUSIONS Taken together, our findings indicate that elevated EC GATA4 levels contribute to the progression of atherosclerosis through the GATA4-NFκB2-SASP pathway, suggesting potential therapeutic targets for atherosclerosis-related diseases.
Collapse
Affiliation(s)
- Chun-Min Kang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, China; Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Jing-Jing Zhao
- Department of Laboratory Medicine, Nanfang Hospital Affiliated to Southern Medical University, Guangdong, 510515, China
| | - Xi-Xi Xie
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, China; Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Ke-Wei Yu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, China; Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Bai-Cong Lai
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Yun-Xiu Wang
- Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Ting Ting Li
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, China; Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Pei-Feng Ke
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, China; Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Xian-Zhang Huang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, China; Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong, 510120, 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
|
Zhang C, Zhang L, Chen L, Zheng Y, Zhang S, Guo S, Hu S. Mitigating effects of hydroxysafflor yellow a on atherosclerotic inflammatory responses based on flavonoid macromolecule compound: Inhibition of Piezo1-YAP/JNK protein pathway. Int J Biol Macromol 2025; 309:142961. [PMID: 40220830 DOI: 10.1016/j.ijbiomac.2025.142961] [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: 03/09/2025] [Revised: 03/27/2025] [Accepted: 04/06/2025] [Indexed: 04/14/2025]
Abstract
Atherosclerosis (AS) is an important cardiovascular disease caused by inflammation. The inhibitory effect of HysA on the Piezo1-YAP/JNK signaling pathway in a mouse model of AS was studied to clarify its anti-inflammatory effects. The study evaluated HysA's ability to bind Piezo1 through molecular docking analysis. C57BL/6 mice were used to establish AS model, and cell treatment and animal experiments were carried out. Inflammatory markers and lipid accumulation were evaluated using quantitative real-time PCR (qRT-PCR), Westernblot, enzyme-linked immunosorbent assay (ELISA), immunofluorescence, and oil red O staining techniques. Weight changes in mice were recorded to monitor disease progression. The molecular docking results show that HysA has a good binding affinity with Piezo1. In a mouse model of AS, treatment with HysA significantly reduced levels of inflammatory cytokines and inhibited the activation of YAP and JNK signaling pathways. The HysA treatment group showed lower oil red O staining levels, indicating that it effectively mitigated lipid deposition. Therefore, HysA significantly alleviates the inflammatory response of atherosclerosis by inhibiting the Piezo1-YAP/JNK signaling pathway, suggesting its potential application in the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Chunxiao Zhang
- Traditional Chinese Medicine Department, Shaoxing Hospital of Traditional Chinese Medicine, Shaoxing City 312000, Zhejiang Province, China
| | - Lei Zhang
- Internal Medicine Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan City 250011, Shandong Province, China
| | - Lanyong Chen
- Clinical College, Zhejiang Chinese Medical University, Hangzhou City 310000, Zhejiang Province, China
| | - Yuhui Zheng
- Traditional Chinese Medicine Department, Shaoxing Hospital of Traditional Chinese Medicine, Shaoxing City 312000, Zhejiang Province, China
| | - Shengyang Zhang
- Traditional Chinese Medicine Department, Shaoxing Hospital of Traditional Chinese Medicine, Shaoxing City 312000, Zhejiang Province, China
| | - Shuren Guo
- Clinical College, Zhejiang Chinese Medical University, Hangzhou City 310000, Zhejiang Province, China
| | - Songfeng Hu
- Traditional Chinese Medicine Department, Shaoxing Hospital of Traditional Chinese Medicine, Shaoxing City 312000, Zhejiang Province, China.
| |
Collapse
|
44
|
Yang X, Wu W, Huang W, Fang J, Chen Y, Chen X, Lin X, He Y. Exosomes derived from baicalin‑pretreated mesenchymal stem cells mitigate atherosclerosis by regulating the SIRT1/NF‑κB signaling pathway. Mol Med Rep 2025; 31:126. [PMID: 40084693 PMCID: PMC11924171 DOI: 10.3892/mmr.2025.13491] [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: 10/15/2024] [Accepted: 02/13/2025] [Indexed: 03/16/2025] Open
Abstract
Atherosclerosis (AS) is a disease with high global incidence and mortality rates. Currently, the treatment of AS in clinical practice carries a high risk of adverse effects and toxic side effects. The pretreatment of mesenchymal stem cells (MSCs) with drugs may enhance the bioactivity of MSC‑derived exosomes (MSC‑exos), which could be a promising candidate for inhibiting the progression of AS. The aim of the present study was to investigate the ability of exos derived from baicalin‑preconditioned MSCs (Ba‑exos) to exhibit an inhibitory effect on AS progression and to explore the potential molecular mechanisms. Exos were isolated from untreated MSCs and MSCs pretreated with Ba, and were characterized using transmission electron microscopy, nanoparticle tracking analysis and western blotting. Subsequently, Cell Counting Kit‑8 and Transwell assays, reverse transcription‑quantitative PCR, immunofluorescence, western blotting and ELISA were used to evaluate the effects of Ba‑exos on AS, and the possible molecular mechanisms. Oil Red O and Masson staining were used to assess AS pathological tissue in a high‑fat diet‑induced mouse model of AS. Notably, MSC‑exos and Ba‑exos were successfully isolated. Compared with MSC‑exos, Ba‑exos demonstrated superior inhibitory effects on the viability and migration, and the levels of inflammatory factors in oxidized low‑density lipoprotein (ox‑LDL)‑induced vascular smooth muscle cells (VSMCs). Additionally, compared with MSC‑exos, Ba‑exos significantly inhibited NF‑κB activation by upregulating sirtuin 1 (SIRT1), thereby suppressing inflammation in ox‑LDL‑induced VSMCs to a greater extent. In mice with high‑fat diet‑induced AS, Ba‑exos exhibited the ability to inhibit AS plaque formation and to alleviate AS progression by reducing the levels of inflammatory factors compared with MSC‑exos; however, the difference was not significant. In conclusion, Ba‑exos may serve as a potential strategy for treating AS by regulating the SIRT1/NF‑κB signaling pathway to suppress inflammation.
Collapse
Affiliation(s)
- Xiaochun Yang
- The First Clinical College of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Wei Wu
- The First Clinical College of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Weitian Huang
- Department of Rehabilitation, Guangdong Work Injury Rehabilitation Hospital, Guangzhou, Guangdong 510000, P.R. China
| | - Junfeng Fang
- Department of Emergency, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510000, P.R. China
| | - Yunli Chen
- Department of Rehabilitation, Guangdong Work Injury Rehabilitation Hospital, Guangzhou, Guangdong 510000, P.R. China
| | - Xiaoyan Chen
- Department of Rehabilitation, Guangdong Work Injury Rehabilitation Hospital, Guangzhou, Guangdong 510000, P.R. China
| | - Xiaolan Lin
- Department of Rehabilitation, Guangdong Work Injury Rehabilitation Hospital, Guangzhou, Guangdong 510000, P.R. China
| | - Yanbin He
- Department of Rehabilitation, Guangdong Work Injury Rehabilitation Hospital, Guangzhou, Guangdong 510000, P.R. China
| |
Collapse
|
45
|
Peng P, Ding S, Liang M, Zheng W, Kang Y, Liu W, Shi H, Gao C. A self-sacrificing anti-inflammatory coating promotes simultaneous cardiovascular repair and reendothelialization of implanted devices. Bioact Mater 2025; 47:502-512. [PMID: 40026826 PMCID: PMC11872464 DOI: 10.1016/j.bioactmat.2025.01.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/27/2025] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
During interventional surgeries of implantable cardiovascular devices in addressing cardiovascular diseases (CVD), the inevitable tissue damage will trigger host inflammation and vascular lumen injury, leading to delayed re-endothelization and intimal hyperplasia. Endowing cardiovascular implants with anti-inflammatory and endothelialization functions is conducive to the target site, offering significant tissue repair and regeneration benefits. Herein, inspired by the snake's molting process, a ShedWise device was developed by using the poly(propylene fumarate) polyurethane (PPFU) as the foundational material, which was clicked with hyperbranched polylysine (HBPL) and followed by conjugation with pro-endothelial functional Arg-Glu-Asp-Val peptide (REDV), and finally coated with a "self-sacrificing" layer having reactive oxygen species (ROS)-scavenging ability and degradability. During the acute inflammation in the initial stage of implantation, the ROS-responsive hyperbranched poly(acrylate-capped thioketone-containing ethylene glycol (HBPAK) coating effectively modulated the level of environmental inflammation and resisted initial protein adsorption, showcasing robust tissue protection. As the coating gradually "sacrificed", the exposed hyperbranched HBPL-REDV layer recruited specifically endothelial cells and promoted surface endothelialization. In a rat vascular injury model, the ShedWise demonstrated remarkable efficiency in reducing vascular restenosis, protecting the injured tissue, and fostering re-endothelization of the target site. This innovative design will introduce a novel strategy for surface engineering of cardiovascular implants and other medical devices.
Collapse
Affiliation(s)
- Pai Peng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Shili Ding
- Department of Hand Surgery, First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, 310003, China
| | - Min Liang
- Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China
| | - Weiwei Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Yongyuan Kang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Wenxing Liu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Haifei Shi
- Department of Hand Surgery, First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, 310003, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
- Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China
- The State Key Laboratory of Transvascular Implantation Devices, Zhejiang University, Hangzhou, 310009, China
| |
Collapse
|
46
|
Wu J, Li W, Tang Y, Wu C, Li W. miR-205-3p inhibits porphyromonas gingivalis lipopolysaccharide-induced human umbilical vein endothelial cells inflammation and apoptosis by targeting PRMT5. Arch Oral Biol 2025; 175:106276. [PMID: 40319839 DOI: 10.1016/j.archoralbio.2025.106276] [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: 02/11/2025] [Revised: 04/12/2025] [Accepted: 04/27/2025] [Indexed: 05/07/2025]
Abstract
OBJECTIVE This study aimed to investigate the regulatory mechanism of miR-205-3p in Porphyromonas gingivalis lipopolysaccharide (P.g-LPS)-induced atherosclerosis. DESIGN In an in vitro setting, human umbilical vein endothelial cells (HUVECs) were exposed to P.g-LPS to simulate the vascular endothelial damage induced by periodontitis. Subsequently, ELISA and flow cytometry were employed to assess the inflammatory response and apoptotic status of these cells.To quantify the expression levels of protein arginine methyltransferase 5 (PRMT5), BCL2-associated X protein (Bax), B-cell lymphoma 2 (Bcl-2), P65 and miR-205-3p within the HUVECs, Western Blot and qPCR were respectively utilized. Moreover, small interfering RNA (siRNA) targeting PRMT5 and miR-205-3p were applied to monitor the changes in PRMT5 expression. Bioinformatics analysis was carried out to predict the potential binding sites between miR-205-3p and PRMT5. Finally, the interaction between miR-205-3p and PRMT5 was validated through the dual-luciferase reporter assay. RESULTS The results indicate that P.g-LPS intervention exacerbates damage to HUVECs and increases the expression of PRMT5. Silencing PRMT5 reduces cell inflammation and apoptosis. After stimulation with P.g-LPS, the level of miR-205-3p decreases, and its overexpression alleviates inflammation and apoptosis in the cells. Bioinformatics analysis and dual luciferase reporter assays confirm that PRMT5 is a target of miR-205-3p, and the overexpression of PRMT5 can reverse the protective effects of miR-205-3p. CONCLUSION miR-205-3p can mitigate vascular endothelial injury by decreasing PRMT5 expression, providing new insights for potential treatments.
Collapse
Affiliation(s)
- Jinsheng Wu
- Stomatology College of Jiamusi University, Jiamusi 154000, China
| | - Weiyi Li
- Stomatology College of Jiamusi University, Jiamusi 154000, China
| | - Ying Tang
- Stomatology College of Jiamusi University, Jiamusi 154000, China
| | - Chang Wu
- Stomatology College of Jiamusi University, Jiamusi 154000, China
| | - Weishan Li
- Stomatology College of Jiamusi University, Jiamusi 154000, China; Department of Periodontal and Mucosal Diseases, Jiamusi University Stomatology Hospital, Jiamusi 154000, China.
| |
Collapse
|
47
|
Tang S, Lu C, Meng Z, Ye Z, Qin Y, Na N, Xian S, Huang F, Zeng Z. USP22 enhances atherosclerotic plaque stability and macrophage efferocytosis by stabilizing PPARγ. Commun Biol 2025; 8:678. [PMID: 40301680 PMCID: PMC12041205 DOI: 10.1038/s42003-025-08116-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 04/22/2025] [Indexed: 05/01/2025] Open
Abstract
Atherosclerosis is a chronic inflammatory disease that strongly threatens human health, and macrophages play a pivotal role in its pathogenesis. Ubiquitin-specific peptidase 22 (USP22) is involved in the regulation of macrophage inflammation. However, its role in the atherosclerotic microenvironment remains unclear. In this study, we found that USP22 overexpression in macrophages alleviated atherosclerosis progression in ApoE-/- mice. In vitro, USP22 silencing enhanced macrophage inflammation and foam cell formation, and macrophage efferocytosis was significantly impaired. Mechanistically, USP22 bound to peroxisome proliferator-activated receptor γ (PPARγ) and inhibited its ubiquitination, thereby stabilizing PPARγ and promoting efferocytosis. In addition, intraperitoneal injection of the USP22 inhibitor USP22i-S02 exacerbated atherosclerosis in ApoE-/- mice. In summary, these findings indicate that USP22 may be a potential therapeutic target for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Senhu Tang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, Guangxi, China
| | - Chuanghong Lu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, Guangxi, China
| | - Zhongyuan Meng
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, Guangxi, China
| | - Zihua Ye
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, Guangxi, China
| | - Yue Qin
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, Guangxi, China
| | - Na Na
- Department of Neuroscience, Scripps Research Institute, San Diego, CA, USA
| | - Shenglin Xian
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, Guangxi, China
| | - Feng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, Guangxi, China.
| | - Zhiyu Zeng
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, Guangxi, China.
| |
Collapse
|
48
|
Otaegui J, Sultan D, Heo GS, Liu Y. Positron Emission Tomography Imaging of the Adaptive Immune System in Cardiovascular Diseases. CHEMICAL & BIOMEDICAL IMAGING 2025; 3:209-224. [PMID: 40313531 PMCID: PMC12042138 DOI: 10.1021/cbmi.4c00117] [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: 12/31/2024] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 05/03/2025]
Abstract
Cardiovascular diseases are the leading cause of death around the globe. In recent years, a crucial role of the immune system has been acknowledged in cardiac disease progression, opening the door for immunomodulatory therapies. To this ongoing change of paradigm, positron emission tomography (PET) imaging of the immune system has become a remarkable tool to reveal immune cell trafficking and monitor disease progression and treatment response. Currently, PET imaging of the immune system in cardiovascular disease mainly focuses on the innate immune system such as macrophages, while the immune cells of the adaptive immune system including B and T cells are less studied. This can be ascribed to the lack of radiotracers specifically binding to B and T cell biomarkers compatible with PET imaging within the cardiovascular system. In this review, we summarize current knowledge about the role of the adaptive immune system (e.g., B and T cells) in major cardiovascular diseases and introduce key biomarkers for specific targeting of these immune cells and their subpopulations. Finally, we present available radiotracers for these biomarkers and propose a pathway for developing probes or optimizing those already used in other fields (e.g., oncology) to make them compatible with the cardiovascular system.
Collapse
Affiliation(s)
- Jaume
Ramon Otaegui
- Mallinckrodt Institute of
Radiology, Washington University, St. Louis, Missouri 63110, United States
| | - Deborah Sultan
- Mallinckrodt Institute of
Radiology, Washington University, St. Louis, Missouri 63110, United States
| | - Gyu Seong Heo
- Mallinckrodt Institute of
Radiology, Washington University, St. Louis, Missouri 63110, United States
| | - Yongjian Liu
- Mallinckrodt Institute of
Radiology, Washington University, St. Louis, Missouri 63110, United States
| |
Collapse
|
49
|
Wang SY, Chen YS, Jin BY, Bilal A. The cGAS-STING pathway in atherosclerosis. Front Cardiovasc Med 2025; 12:1550930. [PMID: 40351606 PMCID: PMC12062000 DOI: 10.3389/fcvm.2025.1550930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 04/09/2025] [Indexed: 05/14/2025] Open
Abstract
Atherosclerosis (AS), a chronic inflammatory disease, remains a leading contributor to cardiovascular morbidity and mortality. Recent studies highlight the critical role of the cGAS-STING pathway-a key innate immune signaling cascade-in driving AS progression. This pathway is activated by cytoplasmic DNA from damaged cells, thereby triggering inflammation and accelerating plaque formation. While risk factors such as aging, obesity, smoking, hypertension, and diabetes are known to exacerbate AS, emerging evidence suggests that these factors may also enhance cGAS-STING pathway, which amplifies inflammatory responses. Targeting this pathway offers a promising therapeutic strategy to reduce the burden of cardiovascular diseases (CVD). In this review, we summarize the mechanisms of the cGAS-STING pathway, explore its role in AS, and evaluate potential inhibitors as future therapeutic candidates. By integrating current knowledge, we aim to provide insights for developing novel treatments to mitigate AS and CVD burden.
Collapse
Affiliation(s)
- Si-yu Wang
- Department of Cardiology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
- The First Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Yu-shan Chen
- Department of Cardiology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
- Heart Center/National Regional (Traditional Chinese Medicine) Cardiovascular Diagnosis and Treatment Center, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Bo-yuan Jin
- Department of Cardiology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
- The First Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Ahmad Bilal
- Department of Cardiology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
- The First Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
50
|
Marchiori GN, Paqualini ME, Flores D, Triquell MF, Moreira-Espinoza MJ, Eynard AR, Valentich MA, Defagó MD. Serum and dietary fatty acids and their relationship to vascular inflammation and carotid intima-media thickness: implications for cardiovascular risk in patients with arterial hypertension. Acta Cardiol 2025:1-8. [PMID: 40276965 DOI: 10.1080/00015385.2025.2493978] [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: 01/06/2023] [Revised: 03/24/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Pathogenic mechanisms involved in arterial hypertension (HT) promote the atherosclerosis development. Fatty acids (FAs) could modulate these processes. We investigated the association between dietary and serum FAs and early atherosclerosis development and their correlation with serum vascular inflammation markers in HT. METHODS 96 patients were included. Dietary information was collected through a validated food frequency questionnaire. Serum FAs were detected by gas chromatography. High-sensitivity C-reactive protein (hs-CRP), interleukin 6 (IL-6) and fibrinogen serum were determined by immunoturbidimetry, enzyme immunoassay and Clauss method, respectively. Carotid intima media-thickness (cIMT) was determined by ultrasound. Multivariate linear regression models were applied. RESULTS Patients showed an adverse health profile, with high caloric intake (3113 ± 1477 kcal/day), LDL cholesterol (126.0 ± 35.1 mg/dL), and overweight (BMI 31.1 ± 5.5 kg/t2), and also an inflam-matory process, characterised by higher hs-CRP (3.9 ± 2.7 mg/L) and fibrinogen (433.4 ± 84.6 mg/dL) concentrations. We found significant inverse correlations between dietary monounsaturated FAs (MUFAs), serum omega-3 polyunsaturated FAs (n-3 PUFAs), and hs-CRP (β = -3.59, [95% CI -6.87 to -0.32], p = 0.03 and β = -0.70, [95% CI -1.42-0.01], p = 0.05, respectively), significant positive associations between dietary MUFAs, serum saturated FAs (SFAs) and cIMT (β = 81.96, [95% CI 33.42-130.50], p = 0.002 and β = 36.71, [95% CI 6.34-67.08], p = 0.02, respectively), and significant inverse correlations between serum omega-6 polyunsaturated FAs (n-6 PUFAs) and cIMT (β = -91.44, [95% CI -168.01 to -14.88], p = 0.02 and β = -41.01, [95% CI -70.73-11.28], p = 0.01). CONCLUSIONS Serum n-3 PUFAs modulate hs-CRP levels, while n-6 PUFAs suggest antiatherosclerotic properties in HT. The ambiguous impact of MUFAs requires comprehensive research.
Collapse
Affiliation(s)
- Georgina N Marchiori
- Centro de Investigaciones en Nutrición Humana (CenINH), Escuela de Nutrición, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Instituto de Investigaciones en Ciencias de la Salud (INICSA), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María E Paqualini
- Instituto de Investigaciones en Ciencias de la Salud (INICSA), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
- Instituto de Biología Celular, Cátedra de Biología Celular, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - David Flores
- Hospital Nacional de Clínicas, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba
| | - María F Triquell
- Instituto de Investigaciones en Ciencias de la Salud (INICSA), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
- Instituto de Biología Celular, Cátedra de Biología Celular, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María J Moreira-Espinoza
- Instituto de Biología Celular, Cátedra de Biología Celular, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Aldo R Eynard
- Instituto de Investigaciones en Ciencias de la Salud (INICSA), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
- Instituto de Biología Celular, Cátedra de Biología Celular, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mirta A Valentich
- Instituto de Investigaciones en Ciencias de la Salud (INICSA), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
- Instituto de Biología Celular, Cátedra de Biología Celular, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María D Defagó
- Centro de Investigaciones en Nutrición Humana (CenINH), Escuela de Nutrición, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Instituto de Investigaciones en Ciencias de la Salud (INICSA), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|