1
|
Mo Y, Ge Y, Wang D, Wang J, Zhang R, Hu Y, Qin X, Hu Y, Lu S, Liu Y, Zhang WS. Comprehensive analysis of single-cell and bulk transcriptome unravels immune landscape of atherosclerosis and develops a S100 family based-diagnostic model. Comput Biol Chem 2025; 117:108436. [PMID: 40163962 DOI: 10.1016/j.compbiolchem.2025.108436] [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: 12/22/2024] [Revised: 03/05/2025] [Accepted: 03/17/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND The S100 family of calcium-binding proteins (S100s) had been tightly related to the biological processes of various cardiovascular diseases. This study aims to investigate the expression of S100s in Atherosclerosis (AS) and explore their potential as diagnostic biomarkers and therapeutic targets. METHODS We analyzed multiple sequencing datasets from the GEO database to compare the expression profiles of S100s in AS tissues versus normal samples. Employing unsupervised clustering techniques, AS subtypes were discerned based on the intricate variations in S100-related gene expression profiles. Subsequent analyses delved into immune cell infiltration and GSVA pathway enrichment, shedding light on the nuanced immune landscape characterizing diverse AS subtypes. Machine learning techniques were employed to develop a diagnostic model for AS. Single-cell RNA analysis was utilized to investigate the cellular distribution of S100 hub genes in AS. RESULTS Unsupervised clustering analysis identified two distinct AS subtypes (C1 and C2), characterized by specific S100 gene expression patterns. The RF-based diagnostic model exhibited the highest efficacy (AUC=0.881), and the top five genes (S100A4, S100A10, S100A11, S100A13, S100Z) were used to construct a diagnostic nomogram. CONCLUSION This study systematically elucidates the roles of S100s in AS, offering insights into molecular subtyping, immune characteristics, and diagnostic model construction. The findings provide valuable implications for the precise treatment and prognosis assessment of AS and pave the way for further research into related mechanisms.
Collapse
Affiliation(s)
- Yanfei Mo
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China; Department of Cardiology, Pukou Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China; Jiangsu Medical College, Yancheng, Jiangsu, China
| | - Yaoqi Ge
- Department of General Practice, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Dan Wang
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jizheng Wang
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rihua Zhang
- Department of the Core Facility, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yifang Hu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoxuan Qin
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yanyan Hu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shan Lu
- Maternity and Child Dept, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Yun Liu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Wen-Song Zhang
- Department of the Core Facility, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
2
|
Li L, Lai L, Qiu D, Ding Y, Yu M, Zhang T, Wang Z, Wang S. P2Y 6 receptor: A promising therapeutic target for atherosclerosis. Eur J Pharmacol 2025; 998:177513. [PMID: 40097133 DOI: 10.1016/j.ejphar.2025.177513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/18/2025] [Accepted: 03/14/2025] [Indexed: 03/19/2025]
Abstract
Atherosclerosis is induced by lipid accumulation, inflammation, and endothelial dysfunction, and is the leading cause of death from cardiovascular disease worldwide. The P2Y6 receptor can be activated by the extracellular release of UDP. The evidence from the last decade has highlighted its critical therapeutic effect in atherosclerosis, yet with unclear mechanisms. This review introduced the P2Y6 receptor in atherosclerosis, and its mechanisms of atherosclerosis-promoting in macrophages, endothelial cells, and vascular smooth muscle cells. Finally, we discussed the development and potential of P2Y6 receptor antagonists in treating atherosclerosis.
Collapse
Affiliation(s)
- Lixia Li
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Liting Lai
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Dan Qiu
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yang Ding
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Meiling Yu
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Tingyu Zhang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zongbao Wang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Shuzhi Wang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
3
|
Athauda-Arachchi PM, Kaththiriarachchi L, Salgado W, De Silva S, Salgado T, Farooq M, Jibran MJ, Godakanda Arachchi Y, Premanath S, Senaratne SL, Samarakoon SP, Rathnayake H, Shajahan M. Cross-sectional study of lipoprotein(a) and the severity of coronary artery disease, cerebrovascular disease, and peripheral vascular disease in a group of South Asian patients. Cardiovasc Endocrinol Metab 2025; 14:e00327. [PMID: 40160971 PMCID: PMC11952828 DOI: 10.1097/xce.0000000000000327] [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: 01/09/2025] [Accepted: 02/27/2025] [Indexed: 04/02/2025]
Abstract
Background Atherosclerotic cardiovascular diseases (ASCVD), including coronary artery disease (CAD), cerebrovascular disease (CVD), and peripheral vascular disease (PVD), remain the leading cause of death globally. South Asians exhibit a higher incidence of cardiovascular diseases than other ethnicities, attributed to a range of genetic, environmental, and lifestyle factors. Lipoprotein(a) [Lp(a)] with a unique apolipoprotein(a) component, has emerged as a marker of atherosclerosis and ASCVD risk, with evidence to promote arterial plaque formation and thrombogenesis. Objective The aim of this study was to explore the associations between Lp(a) levels and the severity of CAD, CVD, and PVD in a group of South Asian patients. Methods Following ethical approval, 60 consecutive patients who underwent coronary angiography for any indication were reviewed. There were 51 eligible participants who were evaluated for Lp(a) level, Synergy Between Percutaneous Coronary Intervention with Taxus and Cardiac Surgery (SYNTAX) score, and severity of CVD and PVD. The SYNTAX-I score was calculated using two-observer consensus on coronary angiograms. Assessment of CVD was by ultrasound/Doppler, and PVD by estimating ankle-brachial index using Doppler. The multisite arterial disease score 2 (MADS2) and SYNTAX score tertiles were used to group the patients. Statistical analysis was performed using the SPSS software. Results and discussion In this group, we identified a statistically significant difference with higher Lp(a) levels being associated with more severe coronary disease (SYNTAX tertile 2,3). Despite a numerical trend, statistical significance was not confirmed for Lp(a) levels in relation to MADS2-CVD or MADS2-PVD scores. A larger study may be required to assess these aspects.
Collapse
|
4
|
Ke S, Cao X, Lu X, Xu J, Zhang CY, Xu L, Jiang X. Atheroprotective roles of exercise-regulated microRNAs. Atherosclerosis 2025; 405:119229. [PMID: 40367725 DOI: 10.1016/j.atherosclerosis.2025.119229] [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: 09/11/2024] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 05/16/2025]
Abstract
Cardiovascular disease remains the leading cause of death worldwide, with atherosclerosis (AS) serving as a critical underlying pathological process and major risk factor. Regular physical exercise is widely recognized as an effective strategy to reduce the risks and severity of AS, yet the precise molecular mechanisms through which exercise exerts its protective effects are still not fully understood. MicroRNAs (miRNAs), key regulators of gene expression, play integral roles in the progression of AS by influencing vascular function, lipid metabolism, and inflammation. The exercise-induced improvement of AS is a complex process, with miRNAs playing essential roles not only within cells and tissues but also circulating stably in the bloodstream as novel signaling molecules. These circulating miRNAs mediate communication between organs and tissues, acting as potential biomarkers that could provide deeper, systemic insights into the metabolic benefits of exercise. In this review, we explore recent advancements in our understanding of how exercise affects both intracellular and circulating miRNAs. We emphasize how exercise-regulated miRNAs contribute to endothelial function, promote lipid metabolism across various metabolic organs, and reduce monocyte-mediated systemic inflammation, while also addressing their role in alleviating frailty. Circulating miRNAs, which dynamically reflect tissue-specific responses to exercise, hold great promise as diagnostic and prognostic biomarkers for AS. Moreover, we discuss the challenges and future directions in this field, aiming to uncover how exercise-induced miRNA modulation could offer innovative therapeutic strategies for the prevention and treatment of AS.
Collapse
Affiliation(s)
- Shuo Ke
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Xiaoqin Cao
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Xiaohui Lu
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Jinhong Xu
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Chen-Yu Zhang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China; Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, 210023, China
| | - Li Xu
- Clinical Trial Institution, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Number 321 Zhongshan Road, Nanjing, Jiangsu, 210008, China.
| | - Xiaohong Jiang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China; Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, 210023, China.
| |
Collapse
|
5
|
Lou Y, Chen H, Fei S, Chen X, Guo L, Pan Q. Role of denosumab in lipid metabolism disorders: clinical significance and potential mechanisms. Arch Osteoporos 2025; 20:68. [PMID: 40418391 DOI: 10.1007/s11657-025-01546-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 04/11/2025] [Indexed: 05/27/2025]
Abstract
PURPOSE Lipid metabolism disorders, characterized by abnormal blood lipid levels, are central to the pathogenesis of obesity, nonalcoholic fatty liver disease (NAFLD), and atherosclerosis. These conditions increase the risk of type 2 diabetes, cardiovascular diseases (CVD), and stroke, highlighting the need for novel therapeutic approaches. Emerging evidence suggests a complex interplay between bone and lipid metabolism, with RANKL playing a key role. This review explores the potential of denosumab, a RANKL-targeting monoclonal antibody, in modulating lipid metabolism and its broader metabolic implications. METHODS We conducted a comprehensive literature review to analyze the molecular mechanisms by which denosumab influences lipid metabolism, with a focus on the OPG/RANKL/RANK signaling pathway. Additionally, we examined the roles of immune modulation, bone marrow adipose tissue, and gut microbiota in metabolic diseases. RESULTS Denosumab, primarily known for its anti-resorptive effects in osteoporosis, may also exert beneficial effects on lipid metabolism. Preclinical and clinical studies suggest its potential in ameliorating obesity, NAFLD, and atherosclerosis. The OPG/RANKL/RANK axis appears to mediate crosstalk between bone and metabolic pathways, while immune regulation and gut microbiota may further contribute to these effects. CONCLUSION Denosumab shows promise as a therapeutic agent for lipid metabolism disorders, though long-term metabolic effects remain unclear. Further research is needed to validate its efficacy and elucidate underlying mechanisms, which could pave the way for novel treatments targeting metabolic diseases.
Collapse
Affiliation(s)
- Yuan Lou
- Department of Endocrinology Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100000, China
- Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, Beijing, 100000, China
| | - Huan Chen
- Department of Endocrinology Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100000, China
- Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, Beijing, 100000, China
| | - Sijia Fei
- Department of Endocrinology Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100000, China
| | - Xinda Chen
- Department of Endocrinology, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Beijing, 100000, China
| | - Lixin Guo
- Department of Endocrinology Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100000, China.
- Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, Beijing, 100000, China.
| | - Qi Pan
- Department of Endocrinology Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100000, China.
- Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, Beijing, 100000, China.
| |
Collapse
|
6
|
Xu R, Wang Z, Dong J, Yu M, Zhou Y. Lipoprotein(a) and panvascular disease. Lipids Health Dis 2025; 24:186. [PMID: 40413492 DOI: 10.1186/s12944-025-02600-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 05/08/2025] [Indexed: 05/27/2025] Open
Abstract
Panvascular disease (PVD) is an emerging clinical concept that encompasses a spectrum of atherosclerotic conditions involving multiple major vascular beds, including the coronary, cerebral, peripheral, and valvular arteries. Although not formally recognized as a nosological entity, in this review, PVD is adopted as a conceptual framework to reflect the systemic nature of atherosclerosis affecting vascular territories supplying the heart, brain, and peripheral circulation. This perspective enables a more integrated understanding of disease processes across organ systems that are often studied in isolation. Lipoprotein(a) [Lp(a)] is a genetically regulated, low-density lipoprotein (LDL)-like particle that has garnered increasing attention as an independent pathogenic risk factor for PVD. Accumulating evidence from epidemiological, genetic, and mechanistic studies has confirmed the multifaceted role of Lp(a) in promoting atherogenesis, vascular calcification, inflammation, and thrombogenesis across multiple vascular beds. Elevated Lp(a) levels are associated with increased cardiovascular and cerebrovascular event risk, even after controlling for traditional risk factors. This review systematically outlines the structure, genetic determinants, and pathogenic mechanisms of Lp(a), and synthesizes current clinical evidence regarding its role in various PVD subtypes. The interactions between Lp(a) and traditional cardiovascular risk factors such as hypercholesterolemia, diabetes, and hypertension are explored in depth, highlighting their synergistic contributions to vascular injury and disease progression. Furthermore, sex-based differences in Lp(a)-associated risk, response to therapy, and biological behavior are discussed, providing insights into personalized cardiovascular risk stratification. In addition, the review summarizes current and emerging therapeutic strategies targeting Lp(a), including niacin, antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), and gene-editing technologies. These advances offer promising new avenues for reducing residual cardiovascular risk attributable to elevated Lp(a). In conclusion, viewing Lp(a)-associated pathology through the lens of PVD provides a comprehensive and unifying approach to understanding its systemic impact. This framework supports the development of integrated risk assessment tools and multi-targeted interventions, ultimately aiming to improve outcomes for patients with complex, multisite vascular involvement.
Collapse
Affiliation(s)
- Ruiyan Xu
- Department of Clinical Medicine, Queen Mary School of Nanchang University, Nanchang, 330031, China
| | - Zhenwei Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Jiayu Dong
- Department of Clinical Medicine, Queen Mary School of Nanchang University, Nanchang, 330031, China
| | - Miao Yu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330008, China.
| | - Yue Zhou
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330008, China.
| |
Collapse
|
7
|
Gualtero DF, Buitrago DM, Pinzón-García AD, Cely Veloza WF, Figueroa-Ariza LT, Torres-Morales S, Rodriguez-Navarrete JD, Jimenez VJ, Lafaurie GI. Biopolymers of Polycaprolactone Loaded with Caffeic Acid and Trametes versicolor Extract Induced Proliferation in Human Coronary Artery Endothelial Cells and Inhibited Platelet Activity. Int J Mol Sci 2025; 26:4949. [PMID: 40430091 PMCID: PMC12112702 DOI: 10.3390/ijms26104949] [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: 09/17/2024] [Revised: 03/27/2025] [Accepted: 03/27/2025] [Indexed: 05/29/2025] Open
Abstract
In atherosclerosis, the proliferation and migration of endothelial and smooth muscle cells (SMCs) and platelet activation alter endothelial function. Naturally occurring substances, such as caffeic acid (CA) and Trametes versicolor extract (TvE), have medicinal properties and are traditionally used for their antiproliferative, antioxidant, and anti-inflammatory effects. Electrospun 5% and 8% polycaprolactone-loaded CA or TvE was developed as a delivery system. Cytocompatibility was evaluated using human coronary artery endothelial cells (HCAECs), coronary artery SMCs (CASMCs), and platelets. Three types of systems (µF-CA, µF-TvE, and µF-CA/TvE) were developed and microscopically characterized. Analysis with scanning electron microscopy showed multidirectional fibers with diameters of 2-4.5 μm. The µF systems were hydrophobic and low cellular adhesion. The viability of CASMCs decreased with microfibers of 8% PCL and high CA concentration. However, the viability of CASMCs and HCAECs improved with 5% PCL and low CA concentration. Treatment with µF-TvE and µF-CA/TvE increased cell viability. HCAEC proliferation was affected by µF-CA, but incorporating TvE improved it. Platelet viability was unaffected by any µF system, but µF-CA and µF-CA/TvE inhibited the activation and adhesion of platelets. The results suggest that microfibers loaded with CA and TvE play a dual role in modifying HCAEC proliferation and blocking human platelet activation and adhesion. These findings have the potential to mitigate the atherosclerotic process.
Collapse
Affiliation(s)
- Diego Fernando Gualtero
- Unidad de Investigación Básica Oral—UIBO, Vicerrectoría de Investigación, Facultad de Odontología, Universidad El Bosque, Bogotá 111321, Colombia; (D.M.B.); (A.D.P.-G.); (L.T.F.-A.)
| | - Diana Marcela Buitrago
- Unidad de Investigación Básica Oral—UIBO, Vicerrectoría de Investigación, Facultad de Odontología, Universidad El Bosque, Bogotá 111321, Colombia; (D.M.B.); (A.D.P.-G.); (L.T.F.-A.)
| | - Ana Delia Pinzón-García
- Unidad de Investigación Básica Oral—UIBO, Vicerrectoría de Investigación, Facultad de Odontología, Universidad El Bosque, Bogotá 111321, Colombia; (D.M.B.); (A.D.P.-G.); (L.T.F.-A.)
| | - Willy Fernando Cely Veloza
- Unidad de Investigación Básica Oral—UIBO, Vicerrectoría de Investigación, Facultad de Odontología, Universidad El Bosque, Bogotá 111321, Colombia; (D.M.B.); (A.D.P.-G.); (L.T.F.-A.)
| | - Leydy Tatiana Figueroa-Ariza
- Unidad de Investigación Básica Oral—UIBO, Vicerrectoría de Investigación, Facultad de Odontología, Universidad El Bosque, Bogotá 111321, Colombia; (D.M.B.); (A.D.P.-G.); (L.T.F.-A.)
| | - Santiago Torres-Morales
- Facultad de Odontología—UBSIFO, Universidad El Bosque, Bogotá 111321, Colombia; (S.T.-M.); (J.D.R.-N.); (V.J.J.)
| | | | - Victor Junior Jimenez
- Facultad de Odontología—UBSIFO, Universidad El Bosque, Bogotá 111321, Colombia; (S.T.-M.); (J.D.R.-N.); (V.J.J.)
| | - Gloria Inés Lafaurie
- Unidad de Investigación Básica Oral—UIBO, Vicerrectoría de Investigación, Facultad de Odontología, Universidad El Bosque, Bogotá 111321, Colombia; (D.M.B.); (A.D.P.-G.); (L.T.F.-A.)
| |
Collapse
|
8
|
Dai Y, Yang L, Cao G, Mo L, Yang C, Zhu Y, Guo Y, Hong Y, Xu H, Lu S, Du S, He J. Combination therapy and drug co-delivery systems for atherosclerosis. J Control Release 2025; 381:113543. [PMID: 39986476 DOI: 10.1016/j.jconrel.2025.02.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/25/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of plaque within the arteries. Despite advances in therapeutic strategies including anti-inflammatory, antioxidant, and lipid metabolism modulation treatments over the past two decades, the treatment of atherosclerosis remains challenging, as arterial damage is the result of interconnected pathological factors. Therefore, current monotherapies often fail to address the complex nature of this disease, leading to insufficient therapeutic outcomes. This review addressed this paucity of effective treatment options by comprehensively exploring the potential for combination therapies and advanced drug co-delivery systems for the treatment of atherosclerosis. We investigated the pathological features of and risk factors for atherosclerosis, underscoring the importance of drug combination therapies for the treatment of atherosclerotic diseases. We discuss herein mathematical models for quantifying the efficacy of the combination therapies and provide a systematic summary of drug combinations for the treatment of atherosclerosis. We also provide a detailed review of the latest advances in nanoparticle-based drug co-delivery systems for the treatment of atherosclerosis, focusing on the design of carriers with high biocompatibility and efficacy. By exploring the possibilities and challenges inherent to this approach, we aim to highlight cutting-edge technologies that can foster the development of innovative strategies, optimize drug co-administration, improve treatment outcomes, and reduce the burden of atherosclerosis-related morbidity and mortality on the healthcare system.
Collapse
Affiliation(s)
- Yingxuan Dai
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Li Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Guosheng Cao
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China
| | - Liqing Mo
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Can Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yuxi Zhu
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210, USA; Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, OH 44106, USA
| | - Yujie Guo
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yi Hong
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Hanlin Xu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Shan Lu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China.
| | - Shi Du
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210, USA; Division of Pharmaceutics and Pharmacology, College of Pharmacy, Ohio State University, Columbus, OH 43210, USA.
| | - Jianhua He
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China.
| |
Collapse
|
9
|
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:S1742-7061(25)00346-0. [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] [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
|
10
|
Yilmaz R, Toprak K, Karagoz A, Yontar OC, Ucar M, Kokcu HI, Ozturk B, Kaya E, Yilmaz M, Öz E. Prognostic Value of Non-Traditional Lipid Indices for In-Hospital Mortality in Patients with Acute Coronary Syndromes. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:846. [PMID: 40428804 PMCID: PMC12113601 DOI: 10.3390/medicina61050846] [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: 04/13/2025] [Accepted: 05/02/2025] [Indexed: 05/29/2025]
Abstract
Background and Objectives: Acute coronary syndrome (ACS) is a life-threatening cardiovascular condition with high mortality rates, necessitating accurate and early risk assessment to optimize patient outcomes. While traditional lipid markers, such as low-density lipoprotein-cholesterol (LDL-C) and high-density lipoprotein-cholesterol (HDL-C), are widely used, non-traditional lipid indices, including the lipoprotein combined index (LCI), atherogenic index of plasma (AIP), atherogenic index (AI), Castelli risk indices (CRI-I, CRI-II), and atherogenic combined index (ACI) may offer additional prognostic insights by reflecting the underlying atherogenic and inflammatory processes. This study aimed to assess the prognostic value of these non-traditional lipid indices, along with traditional lipid and biochemical markers, for in-hospital mortality in ACS patients. Materials and Methods: This retrospective observational study analyzed data from ACS patients admitted to the coronary care unit (CCU) between January 2019 and September 2024. A cohort of 920 patients was divided into survivor (n = 823, 89.46%) and non-survivor (n = 97, 10.54%) groups based on in-hospital mortality outcomes. Demographic, hematological, biochemical, and lipid profile data, including traditional and non-traditional lipid indices, were collected. Separate logistic regression models were developed for each index, adjusting for demographic and clinical variables in order to assess the independent predictive power of each non-traditional lipid index. Results: Significant differences were observed between survivor and non-survivor groups in terms of age, c-reactive protein (CRP), white blood cell count (WBC), hemoglobin (HGB), and creatinine levels (all p-values < 0.05). While traditional lipid markers, such as LDL-C and HDL-C, showed limited predictive value, non-traditional lipid indices demonstrated stronger associations. The highest Exp (Beta) values were observed for the CRI-II, AI, and CRI-I. An ROC analysis further confirmed that the CRI-II, AI, and CRI-I had the highest AUC values, with pairwise comparisons underscoring the CRI-II's superior accuracy. These findings suggest that non-traditional lipid indices predict atherogenic risk better than traditional markers alone. Conclusions: Non-traditional lipid indices, particularly the CRI-I and II, AI, LCI, ACI, and AIP, were found to be significantly associated with in-hospital mortality in ACS patients. These indices may provide additional prognostic value beyond traditional lipid parameters; however, further prospective studies are needed to confirm their clinical utility. These results underscore the importance of integrating non-traditional lipid indices into routine risk assessments to improve mortality predictions and inform targeted interventions in high-risk ACS patients.
Collapse
Affiliation(s)
- Rustem Yilmaz
- Department of Cardiology, Faculty of Medicine, Samsun University, Samsun 33805, Turkey; (A.K.); (O.C.Y.); (M.U.); (H.I.K.); (B.O.); (E.K.); (M.Y.)
| | - Kenan Toprak
- Department of Cardiology, Faculty of Medicine, Harran University, Şanlıurfa 63050, Turkey;
| | - Ahmet Karagoz
- Department of Cardiology, Faculty of Medicine, Samsun University, Samsun 33805, Turkey; (A.K.); (O.C.Y.); (M.U.); (H.I.K.); (B.O.); (E.K.); (M.Y.)
| | - Osman Can Yontar
- Department of Cardiology, Faculty of Medicine, Samsun University, Samsun 33805, Turkey; (A.K.); (O.C.Y.); (M.U.); (H.I.K.); (B.O.); (E.K.); (M.Y.)
| | - Melisa Ucar
- Department of Cardiology, Faculty of Medicine, Samsun University, Samsun 33805, Turkey; (A.K.); (O.C.Y.); (M.U.); (H.I.K.); (B.O.); (E.K.); (M.Y.)
| | - Halil Ibrahim Kokcu
- Department of Cardiology, Faculty of Medicine, Samsun University, Samsun 33805, Turkey; (A.K.); (O.C.Y.); (M.U.); (H.I.K.); (B.O.); (E.K.); (M.Y.)
| | - Berkant Ozturk
- Department of Cardiology, Faculty of Medicine, Samsun University, Samsun 33805, Turkey; (A.K.); (O.C.Y.); (M.U.); (H.I.K.); (B.O.); (E.K.); (M.Y.)
| | - Enes Kaya
- Department of Cardiology, Faculty of Medicine, Samsun University, Samsun 33805, Turkey; (A.K.); (O.C.Y.); (M.U.); (H.I.K.); (B.O.); (E.K.); (M.Y.)
| | - Mustafa Yilmaz
- Department of Cardiology, Faculty of Medicine, Samsun University, Samsun 33805, Turkey; (A.K.); (O.C.Y.); (M.U.); (H.I.K.); (B.O.); (E.K.); (M.Y.)
| | - Ersoy Öz
- Department of Statistics, Yildiz Technical University, Istanbul 34220, Turkey
| |
Collapse
|
11
|
Satapathy T, Minj A, Verma M. Impact of NSAIDs corticosteroids DMARDs biologics and their comparisons with natural products in C-reactive proteins (CRP) linked cardiovascular disorders. Inflammopharmacology 2025:10.1007/s10787-025-01767-1. [PMID: 40319427 DOI: 10.1007/s10787-025-01767-1] [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: 10/01/2024] [Accepted: 01/20/2025] [Indexed: 05/07/2025]
Abstract
An important part of the pathophysiology of atherosclerosis is the involvement of inflammatory processes, which mediate various stages of the formation of atheroma, from the first leukocyte recruitment to the final rupture of the unstable atherosclerotic plaque. Acute phase reactant C-reactive protein (CRP), which represents varying degrees of inflammation, has been identified as a separate risk factor for several cardiovascular diseases (CVD), particularly unstable coronary syndrome. We hypothesize that CRP is a direct cause of CVD in addition to being an inflammatory marker. Therefore, therapies aimed at lowering CRP should be beneficial for both primary and secondary CVD prevention. It has been demonstrated that the use of many drugs, particularly statins, alters CRP levels while also lowering cardiovascular events. The use of inflammatory biomarkers aids in the discovery of CVDs and tracks the assessment, prognosis, and administration of treatment. An acute-phase protein called C-reactive protein (CRP) is created in response to pro-inflammatory cytokines. CRP is a key modulator of atherosclerosis and a biomarker of the inflammatory response. It is also regarded as a CVD risk factor since it actively promotes the growth of atherosclerotic plaque, instability, and consequent clot. Patients with intermediate risk for cardiovascular diseases have been using the plasma concentration of hsCRP (high sensitivity CRP) as a biomarker for disease prognosis since 2010.
Collapse
Affiliation(s)
- Trilochan Satapathy
- Department of Pharmacology, Columbia Institute of Pharmacy, Vill-Tekari, Near Vidhansabha, Raipur, CG, 493111, India
| | - Anjali Minj
- Department of Pharmacology, Columbia Institute of Pharmacy, Vill-Tekari, Near Vidhansabha, Raipur, CG, 493111, India.
| | - Mansi Verma
- Department of Pharmacology, Columbia Institute of Pharmacy, Vill-Tekari, Near Vidhansabha, Raipur, CG, 493111, India
| |
Collapse
|
12
|
Wang T, Wang X, Ren W, Sun Z, Zhang Y, Wu N, Diao H. Cardiomyocyte proliferation: Advances and insights in macrophage-targeted therapy for myocardial injury. Genes Dis 2025; 12:101332. [PMID: 39935606 PMCID: PMC11810708 DOI: 10.1016/j.gendis.2024.101332] [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: 08/17/2023] [Revised: 02/18/2024] [Accepted: 03/20/2024] [Indexed: 02/13/2025] Open
Abstract
In the mammalian heart, cardiomyocytes undergo a transient window of proliferation that leads to regenerative impairment, limiting cardiomyocyte proliferation and myocardial repair capacity. Cardiac developmental patterns exacerbate the progression of heart disease characterized by myocardial cell loss, ultimately leading to cardiac dysfunction and heart failure. Myocardial infarction causes the death of partial cardiomyocytes, which triggers an immune response to remove debris and restore tissue integrity. Interestingly, when transient myocardial injury triggers irreversible loss of cardiomyocytes, the subsequent macrophages responsible for proliferation and regeneration have a unique immune phenotype that promotes the formation of pre-existing new cardiomyocytes. During mammalian regeneration, mononuclear-derived macrophages and self-renewing resident cardiac macrophages provide multiple cytokines and molecular signals that create a regenerative environment and cellular plasticity capacity in postnatal cardiomyocytes, a pivotal strategy for achieving myocardial repair. Consistent with other human tissues, cardiac macrophages originating from the embryonic endothelium produce a hierarchy of contributions to monocyte recruitment and fate specification. In this review, we discuss the novel functions of macrophages in triggering cardiac regeneration and repair after myocardial infarction and provide recent advances and prospective insights into the phenotypic transformation and heterogeneous features involving cardiac macrophages. In conclusion, macrophages contribute critically to regeneration, repair, and remodeling, and are challenging targets for cardiovascular therapeutic interventions.
Collapse
Affiliation(s)
- Tao Wang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Xueyao Wang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Weibin Ren
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Zeyu Sun
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Yanhui Zhang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Nanping Wu
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Hongyan Diao
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
13
|
Miki S, Takashima M, Suzuki JI. Anti?atherosclerotic effect of aged garlic extract: Mode of action and therapeutic benefits (Review). Exp Ther Med 2025; 29:104. [PMID: 40171135 PMCID: PMC11959349 DOI: 10.3892/etm.2025.12854] [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: 09/25/2024] [Accepted: 02/06/2025] [Indexed: 04/03/2025] Open
Abstract
Atherosclerosis, a chronic inflammatory disease characterized by plaque buildup within the arteries that obstructs blood flow and significantly increases the morbidity and mortality rates associated with cardiovascular diseases caused by impaired blood flow due to vascular stenosis or occlusion, such as angina and myocardial infarction. The development of atherosclerosis involves a complex interplay of endothelial dysfunction, accumulation of oxidized low-density lipoprotein and macrophage-driven inflammation. The risk factors for atherosclerosis include chronic inflammation, hyperlipidemia and hypertension. Effective management of these risk factors can prevent and delay the onset and progression of atherosclerosis. Garlic and its processed preparations have previously been utilized to mitigate cardiovascular risk factors and continue to be used in traditional medicine in several countries. Among these preparations, aged garlic extract (AGE) has been shown to improve atherosclerosis in clinical trials and animal studies. AGE contains various compounds with potential anti-atherosclerotic properties, such as S-1-propenylcysteine, S-allylcysteine and other sulfur-containing constituents, which may help prevent the development and progression of atherosclerosis. The present manuscript reviewed and discussed the anti-atherogenic effect of AGE and its constituents by highlighting their mode of action and potential benefits for prevention and therapy in the management of atherosclerosis.
Collapse
Affiliation(s)
- Satomi Miki
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., Hiroshima 739-1195, Japan
| | - Miyuki Takashima
- Drug Discovery Laboratory, Wananga Pharmaceutical Co., Ltd., Hiroshima 739-1195, Japan
| | - Jun-Ichiro Suzuki
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., Hiroshima 739-1195, Japan
| |
Collapse
|
14
|
Li P, Wang F, Yue A, Xuan Y, Huang Y, Xu J, Weng J, Li Y, Sun K. LncRNA uc003pxg.1 Interacts With miR-339-5p Promote Vascular Endothelial Cell Proliferation, Migration and Angiogenesis. Korean Circ J 2025; 55:440-455. [PMID: 39733458 PMCID: PMC12088996 DOI: 10.4070/kcj.2024.0153] [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: 05/06/2024] [Revised: 08/29/2024] [Accepted: 10/09/2024] [Indexed: 12/31/2024] Open
Abstract
BACKGROUND AND OBJECTIVES This study aimed to investigate the roles of lncRNA uc003pxg.1 and miR-339-5p in regulating the occurrence and development of coronary heart disease. METHODS First, the expression levels of uc003pxg.1 and miR-339-5p were verified in peripheral blood mononuclear cells of clinical samples. Then, the target gene was identified using high-throughput sequencing combined with bioinformatics. Human umbilical vein endothelial cells (HUVECs) were transfected with si-uc003pxg.1, miR-339-5p mimic and miR-339-5p inhibitor, and the expression of related genes was detected by reverse transcription-quantitative polymerase chain reaction and western blotting. EdU, CCK-8, Cell scratch and Transwell assays were used to analyze the effects of uc003pxg.1 and miR-339-5p on cell proliferation and migration. RESULTS The expression of uc003pxg.1 and miR-339-5p was negatively correlated in clinical samples and HUVECs. The si-uc003pxg.1 and miR-339-5p mimic decreased the proliferation and migration of HUVECs and decreased the expression of transforming growth factor (TGF)-β1 and α-smooth muscle actin (SMA). The protein expression levels of TGF-β1, α-SMA, CD31, collagen I, collagen III and endoglin were decreased, and angiogenesis was weakened. The miR-339-5p inhibitor had the opposite effect. CONCLUSIONS Our study revealed that upregulation of uc003pxg.1 and downregulation of miR-339-5p in vitro promote cell proliferation, cell migration and angiogenesis and upregulate the expression of TGF-β1, α-SMA, CD31, collagen I, collagen III and endoglin, which may lead to the development of vascular atherosclerosis.
Collapse
Affiliation(s)
- Ping Li
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, P. R. China
- Department of Central Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, P. R. China
| | - Feng Wang
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, P. R. China
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, P. R. China
| | - Anna Yue
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, P. R. China
| | - Yanling Xuan
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Ying Huang
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, P. R. China
- Department of Central Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, P. R. China
| | - Jingyi Xu
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, P. R. China
- Department of Central Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, P. R. China
| | - Jiayi Weng
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, P. R. China
| | - Yuan Li
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, P. R. China.
| | - Kangyun Sun
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, P. R. China.
| |
Collapse
|
15
|
Fei X, Song C, Cui J, Li Y, Lei X, Tang H. The role of deubiquitinases in cardiovascular diseases: mechanisms and therapeutic implications. Front Cardiovasc Med 2025; 12:1582049. [PMID: 40376148 PMCID: PMC12078317 DOI: 10.3389/fcvm.2025.1582049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Accepted: 04/22/2025] [Indexed: 05/18/2025] Open
Abstract
Cardiovascular diseases (CVDs) have become the leading cause of death globally, surpassing infectious diseases and other chronic illnesses. The incidence and mortality rates of CVDs are rising worldwide, posing a key challenge in public health. The ubiquitination system is a vast and complex. It is an important post-translational modification that plays a crucial role in various cellular processes. Deubiquitination is catalyzed by deubiquitinases (DUBs), which remove ubiquitin (Ub) from ubiquitinated proteins, thereby reversing the ubiquitination process. DUBs play an important role in many biological processes, such as DNA repair, cell metabolism, differentiation, epigenetic regulation, and protein stability control. They also participate in the regulation of many signaling pathways associated with the development and progression of CVDs. In this review, we primarily focus on the role of DUBs in various key pathological mechanisms of atherosclerosis (AS), such as foam cell formation, vascular remodeling (VR), endothelial-to-mesenchymal transition (End-MT), and clonal hematopoiesis (CH). In the heart, we summarize the involvement of DUBs in diseases and pathological processes, including heart failure (HF), myocardial infarction (MI), myocardial hypertrophy (MH) and ischemia/reperfusion (I/R) injury. Additionally, we also explore the diabetic cardiomyopathy (DCM) and the use of doxorubicin-induced cardiotoxicity in clinical settings. A comprehensive understanding of deubiquitination may provide new insights for the treatment and drug design of CVDs.
Collapse
Affiliation(s)
- Xiangyu Fei
- School of Pharmacy, Hengyang Medical College, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, China
- Department of Cardiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Myocardial Injury in Hunan Province, The First Affiliated Hospital, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Chao Song
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, China
- Department of Cardiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Myocardial Injury in Hunan Province, The First Affiliated Hospital, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Jian Cui
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, China
- Department of Cardiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Myocardial Injury in Hunan Province, The First Affiliated Hospital, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Yuqing Li
- School of Pharmacy, Hengyang Medical College, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, China
- Department of Cardiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Myocardial Injury in Hunan Province, The First Affiliated Hospital, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Xiaoyong Lei
- School of Pharmacy, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Huifang Tang
- School of Pharmacy, Hengyang Medical College, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, China
- Department of Cardiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Myocardial Injury in Hunan Province, The First Affiliated Hospital, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| |
Collapse
|
16
|
He Y, Wang J, Chen C, Wang R, Ma X, Ma R, Sun Y, Wang L, Ding H. Comprehensive profiling of tsRNAs in acute coronary syndrome: expression patterns, clinical correlations, and functional insights. Hum Genet 2025; 144:575-590. [PMID: 40232417 PMCID: PMC12033100 DOI: 10.1007/s00439-025-02742-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/28/2025] [Indexed: 04/16/2025]
Abstract
Transfer RNA-derived small RNAs (tsRNAs) have emerged as potential biomarkers of various human diseases. However, the clinical utility and biological functions of tsRNA in acute coronary syndrome (ACS) remain poorly understood. To investigate this, we performed high-throughput small RNA sequencing on peripheral blood monocyte cells (PBMCs) from 24 ACS patients and 12 healthy controls. Our analysis revealed distinct and characteristic expression patterns of tsRNAs in response to ACS, highlighting their potential as disease signatures in human PBMCs. Differentially expressed tsRNAs were validated using RT-qPCR in two independent case-control sets. Among these, tRF-Gly-GCC-06 was significantly upregulated in volunteers with unstable angina (UA) and acute myocardial infarction (AMI) (p < 0.05) and showed a statistically significant positive correlation with the Gensini score (r = 0.353, p < 0.001). Moreover, this tsRNA was independently associated with an increased risk of ACS after adjusting for conventional cardiovascular risk factors (odds ratio (OR) = 1.58, 95% confidence interval (CI): 1.37-1.83, p < 0.001). A series of functional studies showed that tRF-Gly-GCC-06 significantly facilitated macrophage proliferation and migration and modulated inflammation-related gene expression in vitro. This study identified a novel functional gene associated with ACS, tRF-Gly-GCC-06, as a potential clinical biomarker and therapeutic target.
Collapse
Affiliation(s)
- Yi He
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan, 430030, P.R. China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, P.R. China
| | - Jing Wang
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan, 430030, P.R. China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, P.R. China
| | - Chen Chen
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan, 430030, P.R. China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, P.R. China
| | - Rongli Wang
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan, 430030, P.R. China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, P.R. China
| | - Xiaozhu Ma
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan, 430030, P.R. China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, P.R. China
| | - Ruiying Ma
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan, 430030, P.R. China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, P.R. China
| | - Yang Sun
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan, 430030, P.R. China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, P.R. China
| | - Luyun Wang
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan, 430030, P.R. China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, P.R. China.
- Genetic Diagnosis Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, P.R. China.
| | - Hu Ding
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan, 430030, P.R. China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, P.R. China.
- Genetic Diagnosis Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, P.R. China.
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, P.R. China.
| |
Collapse
|
17
|
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
|
18
|
Wang J, An H, Tao N. Association of non-insulin-based insulin resistance indices, mean platelet volume and prostate cancer: a cross-sectional study. BMC Cancer 2025; 25:795. [PMID: 40295970 PMCID: PMC12039131 DOI: 10.1186/s12885-025-13839-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 02/28/2025] [Indexed: 04/30/2025] Open
Abstract
PURPOSE Insulin resistance and prostate cancer (PCa) association results remain controversial. However, few studies have compared the role of various non-insulin-based insulin resistance (NI-IR) indices and mean platelet volume (MPV) in PCa. METHODS We conducted a cross-sectional study, the case group included 354 patients with PCa, and the control group included 1,498 non-PCa participants. We performed inverse probability weighting to reduce the impact of differences in baseline information between the case and control groups on results. Weighted logistic regression analysis for assessing the relationship between NI-IR indices and PCa risk. Fitting 4-point restricted cubic spline (RCS) plots to show the trend of NI-IR indices with PCa risk. The interaction between insulin resistance and platelet volume based on generalized additive model (GAM) to reveal the impact of the interaction between insulin resistance and cardiovascular risk on PCa. In the end, we performed three sensitivity analyses to verify the stability of results. RESULTS Weighted logistic regression analysis revealed that all NI-IR indices were associated with PCa. When NI-IR indices were evaluated as continuous variables, in the all variables adjusted model (model 3), the adjusted OR of ZJU index was 1.337 (95%CI: 1.296-1.379), the adjusted OR of TyG index was 5.300 (95%CI:4.208-6.675), the adjusted OR of TG/HDL-c was 1.431 (95%CI:1.335-1.534), and the adjusted OR of METS-IR was 1.129 (95%CI:1.110-1.149). When NI-IR indices were analyzed as categorical variables, also in model 3, using Q1 as reference, the adjusted OR of ZJU index in Q5 was 15.592 (95%CI:10.809-22.492), the adjusted OR of TyG index in Q5 was 7.306 (95%CI:5.182-10.301), the adjusted OR of TG/HDL-c in Q5 was 4.790 (95%CI:3.459-6.632), and the adjusted OR of METS-IR in Q5 was 9.844 (95%CI:6.862-14.121). RCS displayed that PCa risk tended to increase as the ZJU index, TyG index, TG/HDL-c, and METS-IR increased. The interaction test based on the GAM indicated that the value of the interaction between TG/HDL-c and MPV on the PCa risk was χ2 = 6.924(P = 0.009). With the increase in TG/HDL-c and the decrease in MPV, the PCa risk progressively increases. The sensitivity analysis further confirmed the robustness of the results. CONCLUSIONS NI-IR indices were associated with an increased PCa risk. The interaction between MPV and insulin resistance may further contribute to the PCa risk.
Collapse
Affiliation(s)
- Jinru Wang
- College of Public Health, Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Hengqing An
- Department of Urology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, China.
| | - Ning Tao
- College of Public Health, Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, China.
| |
Collapse
|
19
|
Duan C, Du Y, Chen J, Shi S, Zhang X, Hu Y. Dynamic and Static Effects of the Systemic Inflammatory Response Index on All-Cause Mortality in Individuals With Atherosclerotic Cardiovascular Disease: Evidence From National Health and Nutrition Examination Survey. Mediators Inflamm 2025; 2025:5343213. [PMID: 40270516 PMCID: PMC12017944 DOI: 10.1155/mi/5343213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 03/12/2025] [Indexed: 04/25/2025] Open
Abstract
Objective: This research focuses on analyzing the link between the systemic inflammatory response index (SIRI) and all-cause mortality in individuals with atherosclerotic cardiovascular disease (ASCVD) . Methods: This research analyzed data from 4693 patients using nine cycles of the National Health and Nutrition Examination Survey (NHANES). The connection between SIRI and mortality was determined by employing survey-weighted Cox models, with hazard ratios (HRs) and 95% confidence intervals (CIs) being computed. Kaplan-Meier method illustrated survival differences across SIRI levels. Sensitivity analyses involved restricted cubic splines (RCS), stratified analysis, and E-value calculations. Landmark analysis assessed survival differences at multiple follow-up intervals, while time-dependent receiver operating characteristic curves evaluated SIRI's prognostic value. Mediation analysis identified potential intermediaries impacting the SIRI-mortality relationship. Results: Over 406,564 person-months, 1933 deaths occurred. Adjusted Cox models discovered that higher SIRI was connected with elevated overall mortality [HR 1.192, (95% CI 1.131-1.256), p < 0.001]. Higher SIRI consistently showed lower survival probabilities. RCS and stratified analysis confirmed the robustness of these findings. Survival probability at different follow-up periods was considerably lower in those with higher SIRI. Additionally, SIRI demonstrated a prognostic value of 0.66 for all-cause mortality at 1 year and 3 years, and 0.65 at 5 years. Notably, serum uric acid (6.2%) partially mediated the connection between SIRI and mortality from all causes. Conclusion: In ASCVD patients, SIRI was robustly correlated with all-cause mortality, partially mediated by serum uric acid.
Collapse
Affiliation(s)
- Chenglin Duan
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing 100053, China
- Graduate School of Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yihang Du
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing 100053, China
| | - Jiafan Chen
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing 100053, China
| | - Shuqing Shi
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing 100053, China
| | - Xiaohan Zhang
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing 100053, China
| | - Yuanhui Hu
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing 100053, China
- Graduate School of Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
20
|
Mijajlović MD, Bornstein NM, Aleksić V. Secondary stroke prevention beyond antiplatelets: The role of colchicine and GLP-1RA - an ounce of prevention is worth a pound of cure. Ther Adv Neurol Disord 2025; 18:17562864251326769. [PMID: 40291758 PMCID: PMC12033550 DOI: 10.1177/17562864251326769] [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: 11/04/2024] [Accepted: 02/21/2025] [Indexed: 04/30/2025] Open
Abstract
Stroke remains a major global health concern, ranking as the second most common cause of death and the third leading cause of disability worldwide. Despite advances in therapy and management, ischemic stroke patients continue to face high risks of recurrence, cardiovascular events, and mortality. Effective secondary stroke prevention is critical, encompassing antithrombotic therapy, management of vascular risk factors such as hypertension, dyslipidemia, and diabetes mellitus, and conducting healthy lifestyle. Approximately 80% of strokes are ischemic, with a significant proportion attributable to large-artery atherosclerosis of the extra- and intracranial arteries, particularly in the internal carotid artery. Atherothrombotic strokes, linked to plaque rupture and thrombus formation, present a notably high risk of recurrence. Inflammatory and immune mechanisms play pivotal roles in both the initiation and progression of atherosclerosis and stroke. Colchicine, an anti-inflammatory agent, has shown potential in managing cardiovascular disease, though its effects on stroke reduction and prevention have been inconsistent across studies. Its possible protective role against stroke is attributed to its anti-inflammatory actions, which include disrupting microtubule dynamics, inhibiting immune cell movement, and lowering inflammatory markers like L-Selectin and E-Selectin, while also suppressing interleukin release. Glucagon-like peptide-1 receptor agonists (GLP-1RA) agents have emerged as effective therapies for type 2 diabetes with notable cardiovascular benefits. These agents enhance glucose control while also providing protective effects against atherosclerosis and stroke. GLP-1RA drugs work by mimicking the effects of GLP-1, a peptide that regulates insulin release and glucose metabolism. They also exhibit anti-inflammatory properties, potentially reducing stroke risk through mechanisms such as improved endothelial function and reduced plaque formation. Clinical trials have indicated that GLP-1RA agents can significantly lower the incidence of nonfatal strokes and major adverse events. This narrative review underscores the importance of targeting inflammation to reduce the risk of recurrent stroke, emphasizing recent studies on colchicine and GLP-1RA. It consolidates evidence regarding the efficacy of these agents in secondary stroke prevention; however, future studies are needed to further explore their mechanisms and roles in comprehensive stroke management strategies.
Collapse
Affiliation(s)
- Milija D. Mijajlović
- Neurology Clinic, University Clinical Center of Serbia, Dr Subotica Street 6, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Natan M. Bornstein
- Brain Division, Shaare Zedek Medical Center, Jerusalem, Israel
- Shaare Zedek Medical Center, Jerusalem, Israel
| | - Vuk Aleksić
- Department of Neurosurgery, Clinical Hospital Center Zemun, Belgrade, Serbia
- Faculty of Medicine Focˇa, University of East Sarajevo, East Sarajevo, Bosnia and Herzegovina
| |
Collapse
|
21
|
Hu X, Zhao Z, An Q, Li Y, Wang B. Association of independent dietary antioxidant intake, and CDAI level with risks of all-cause and cardiovascular-cause death among population with cardiovascular disease. BMC Public Health 2025; 25:1327. [PMID: 40205537 PMCID: PMC11980126 DOI: 10.1186/s12889-025-22481-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Antioxidants have been investigated for their potential to prevent cardiovascular disease (CVD); however, their association with mortality risk in patients with CVD has not been thoroughly evaluated. The aim of this study was to assess the relationship between individual antioxidants and the composite dietary antioxidant index (CDAI) with the risk of death in patients with CVD. METHODS This study included 1,395 participants with CVD from the National Health and Nutrition Examination Survey (2001-2010). Cox proportional hazards models were employed to estimate the hazard ratios (HRs) and 95% confidence intervals (CIs) for individual antioxidants (including vitamins A, vitamins C, vitamins E, selenium, carotenoids and zinc) and CDAI levels in relation to all-cause mortality and cardiovascular death. Additionally, restricted cubic splines (RCS) were utilized to further investigate potential nonlinear relationships. RESULTS Individual antioxidants, including vitamin C and vitamin E, were inversely associated with both all-cause and CVD-cause mortality in patients with CVD. As for CDAI, compared to participants in the first tertile(T1) of CDAI, the fully adjusted HR for all-cause mortality in the third tertile (T3) was 0.62 (95% CI: 0.46, 0.85). For CVD mortality, individuals with T3 of CDAI also exhibited a significantly reduced risk, with an HR of 0. 58 (95% CI: 0. 35,0.97). RCS analysis revealed a linear relationship between CDAI and all-cause mortality, while a non-linear, inverted L-shaped relationship was observed for CVD mortality. CONCLUSION Higher levels of dietary antioxidants are associated with a reduced risk of both all-cause and cardiovascular-cause mortality in patients with CVD. These findings suggest that increasing antioxidant intake may serve as a potential strategy for improving outcomes in this population.
Collapse
Affiliation(s)
- Xintao Hu
- Department of Vascular Surgery, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfuqian Street, Erqi District, Zhengzhou, Henan, China
| | - Zhao Zhao
- Department of Vascular Surgery, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfuqian Street, Erqi District, Zhengzhou, Henan, China
| | - Qian An
- Department of Vascular Surgery, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfuqian Street, Erqi District, Zhengzhou, Henan, China
| | - Yang Li
- Department of Vascular Surgery, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfuqian Street, Erqi District, Zhengzhou, Henan, China
| | - Bing Wang
- Department of Vascular Surgery, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfuqian Street, Erqi District, Zhengzhou, Henan, China.
| |
Collapse
|
22
|
Tian J, Dong Y, Xu Z, Ke J, Xu H. Association between triglyceride glucose-body mass index and 365-day mortality in patients with critical coronary heart disease. Front Endocrinol (Lausanne) 2025; 16:1513898. [PMID: 40255500 PMCID: PMC12006011 DOI: 10.3389/fendo.2025.1513898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 03/17/2025] [Indexed: 04/22/2025] Open
Abstract
Objectives The aim of this study was to analyze the association between TyG-BMI and 365-day mortality in critically ill patients with CHD. Methods Patient data were extracted from the MIMIC-IV database. All patients were categorized into 3 groups based on TyG-BMI index: Low TyG-BMI index group, Medium TyG-BMI index group, and High TyG-BMI index group. Outcomes included primary and secondary outcomes, with the primary outcome being 365-day mortality and the secondary outcomes being hospital survival, intensive care unit (ICU) survival, and 28-day, 90-day, and 180-day mortality. The Kaplan-Meier survival curves were used to compare the outcomes of the three groups. The relationship between TyG-BMI index and 365-day mortality was assessed using multivariate Cox proportional risk regression models and restricted cubic spline curves (RCS). Results 889 critically ill patients with CHD were analyzed. Among them, 600 (67.50%) were male patients with a mean age of 68.37 years and 289 (32.50%) were female patients with a mean age of 73.91 years. Patients with a medium TyG-BMI index had the best 365-day prognostic outcome and the highest survival rate compared with patients in the Low and High TyG-BMI index groups [201 (67.68%) vs. 166 (56.08%), 188 (63.51%); P=0.013]. After fully adjusted modeling analysis, the hazard ratio (HR) for 365-day mortality was found to be 0.71 (95% CI 0.54-0.93, P=0.012) for the Medium TyG-BMI index group. Meanwhile, RCS analysis showed an L-shaped relationship between TyG-BMI index and 365-day mortality. Conclusions The TyG-BMI index is significantly associated with 365-day mortality in patients with severe CHD.
Collapse
Affiliation(s)
| | | | | | | | - Hongyang Xu
- Department of Critical Care Medicine, The Affiliated Wuxi People’s Hospital of
Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| |
Collapse
|
23
|
Li M, Hu X, Hu X, Gao F, Cui Y, Wei X, Qin Y, An X, Zhao Y, Gao Y. Inhibition of PHB1/PHB2 suppresses atherosclerotic plaque formation by interrupting PI3K/AKT/mTOR signaling. PLoS One 2025; 20:e0320509. [PMID: 40168274 PMCID: PMC11960914 DOI: 10.1371/journal.pone.0320509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/19/2025] [Indexed: 04/03/2025] Open
Abstract
Prohibitin 1 (PHB1) and prohibitin 2 (PHB2) are highly conserved proteins belonging to the stomatin-prohibitin flotillin-HflC/K (SPFH) protein superfamily. They are ubiquitously expressed and implicated in the regulation of cell proliferation, migration, and survival. However, the expression and biological functions of PHB1/PHB2 in atherosclerosis (AS) remain unclear. In the present study, an enzyme-linked immunosorbent assay was used to detect PHB1/PHB2 expression in the serum of patients with hyperlipidemia. The potential effect and mechanism of PHB1/PHB2 in apolipoprotein E-deficient (ApoE-/-) mice were also investigated. shRNA-PHB1 and shRNA-PHB2 lentiviruses were engineered and tail vein-injected into ApoE-/- mice fed a high-fat diet. IL-8, a proatherogenic cytokine, was used as an inducer in vitro. The effects of a PHB1/PHB2 knockdown on vascular smooth muscle cell (VSMC) proliferation, migration, and autophagy and endothelial cell (EC) adhesion were evaluated using methyl thiazolyl tetrazolium (MTT), Transwell migration, Boyden chamber, and monocyte adhesion assays, as well as transmission electron microscopy. Compared with the healthy subjects, PHB1/PHB2 expression was elevated in the serum of patients with hyperlipidemia. Animal experiments showed that downregulation of PHBs reduced the area of atherosclerotic lesions, and the expression of cyclinD1, MMP9, and LC3. In addition, in vitro experiments showed that downregulating PHB1/PHB2 expression under inflammatory stimulation reduced the adhesion, proliferation, migration, and autophagy of ECs and VSMCs by inhibiting the PI3K/Akt/mTOR pathway activation. Collectively, our findings showed that PHBs are activly associated with AS progression.
Collapse
Affiliation(s)
- Mei Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xiaoyan Hu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xinxin Hu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Fuhua Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Ying Cui
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
- Molecular Medicine Laboratory, Dalian Medical University, Dalian, China
| | - Xiaoqing Wei
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
- Molecular Medicine Laboratory, Dalian Medical University, Dalian, China
| | - Yuanhua Qin
- Department of Parasite, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaohua An
- Clinical Laboratory, Dalian Central Hospital, Dalian, China
| | - Ying Zhao
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
- Molecular Medicine Laboratory, Dalian Medical University, Dalian, China
| | - Ying Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
- Molecular Medicine Laboratory, Dalian Medical University, Dalian, China
| |
Collapse
|
24
|
Yin W, Wang J, Li L, Zheng H, Xu S. NAT10 Modulates Atherosclerosis Progression Mediated by Macrophage Polarization Through Regulating ac4C Modification of TLR9. J Cardiovasc Transl Res 2025; 18:247-256. [PMID: 39699778 DOI: 10.1007/s12265-024-10579-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024]
Abstract
Atherosclerosis (AS) is an inflammatory disease affected by macrophage polarization. N4-acetylcytosine (ac4C) modification mediated by N-acetyltransferase 10 (NAT10). In this study, we aimed to elucidate the role of ac4C modification mediated macrophage polarization in AS through in vivo and in vitro experiments. The ac4C level was measured using dot blot. Macrophage polarization was assessed by quantitative real-time PCR and flow cytometry. Underlying mechanism was analyzed by methylated RNA Immunoprecipitation (MeRIP), RIP and dual luciferase report. Results showed that the NAT10 expression and ac4C level were increased in patients with AS. Additionally, NAT10 knockdown promoted M1 to M2 polarization and suppressed TLR9 ac4C level. TLR9 overexpression reversed macrophage polarization regulated by NAT10 knockdown. Furthermore, M1 polarization and atherosclerosis in vivo was inhibited by NAT10 knockdown. In conclusion, we demonstrated that NAT10 regualted AS progression mediated by macrophage polarization through regulating ac4C modification of TLR9 and provided a new theoretical basis.
Collapse
Affiliation(s)
- Wei Yin
- Department of Cardiology, Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, No.1 Lijiang Road, High-Tech Zone, Suzhou, 215153, China
| | - Jie Wang
- Department of Cardiology, Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, No.1 Lijiang Road, High-Tech Zone, Suzhou, 215153, China
| | - Lingling Li
- Department of Cardiology, Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, No.1 Lijiang Road, High-Tech Zone, Suzhou, 215153, China
| | - Hongyun Zheng
- Department of Cardiology, Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, No.1 Lijiang Road, High-Tech Zone, Suzhou, 215153, China
| | - Shengkai Xu
- Department of Cardiology, Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, No.1 Lijiang Road, High-Tech Zone, Suzhou, 215153, China.
| |
Collapse
|
25
|
Kelesidis T, Fotooh Abadi L, Ruedisueli I, D'Costa ZU, Middlekauff HR. Atherogenic Effects of Acute Electronic Cigarette Compared With Tobacco Cigarette Smoking in People Living With HIV: A Randomized Crossover Trial. J Am Heart Assoc 2025; 14:e038258. [PMID: 40105098 DOI: 10.1161/jaha.124.038258] [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: 08/12/2024] [Accepted: 02/18/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND People living with HIV (PLWH) are disproportionately affected by tobacco-related health disparities. Concurrent recreational drug use is also more prevalent. Switching to electronic cigarettes (ECs) has been proposed as a harm reduction strategy. However, it remains unproven whether ECs are less atherogenic than tobacco cigarettes (TCs). METHODS AND RESULTS PLWH who smoke TCs and met eligibility criteria were invited to enroll in our acute crossover trial (NCT04568395) comparing the effects of using an EC, a TC, and a straw control on different days on monocyte transendothelial migration and monocyte-derived foam cell formation in our novel ex vivo atherogenesis assay. Twenty-eight PLWH (aged 41.8±9.8 years; 27 men and 1 woman, 18 with a positive urine toxicology screen) completed all 3 sessions. The acute rise in plasma nicotine was similar after acute TC and EC use (7.32±0.86 ng/mL versus 6.30±0.92 ng/mL, P=0.69). The monocyte transendothelial migration after acutely smoking a TC increased by a mean of 0.65-fold difference compared with the straw control (P<0.01). The monocyte transendothelial migration after using an EC was not significantly increased compared with the straw control. The monocyte-derived foam cell formation after acutely smoking a TC was increased by a mean of 0.65-fold difference compared with the straw control (P<0.001). The monocyte-derived foam cell formation after acutely using an EC was not significantly increased compared with the straw control. These findings were present in PLWH with and without concurrent recreational drug use. CONCLUSION These data suggest smaller proatherogenic effects following EC versus TC use in PLWH and justify a larger study looking at cardiovascular risks of ECs in PLWH who smoke, including those who use recreational drugs, populations disproportionately affected by tobacco-related health disparities.
Collapse
Affiliation(s)
- Theodoros Kelesidis
- Department of Medicine, Division of Infectious Disease University of Texas-Southwestern Dallas TX USA
| | - Leila Fotooh Abadi
- Department of Medicine, Division of Infectious Disease University of Texas-Southwestern Dallas TX USA
| | - Isabelle Ruedisueli
- Department of Medicine, Division of Cardiology UCLA David Geffen School of Medicine Los Angeles CA USA
| | - Zoee U D'Costa
- Department of Medicine, Division of Cardiology UCLA David Geffen School of Medicine Los Angeles CA USA
| | - Holly R Middlekauff
- Department of Medicine, Division of Cardiology UCLA David Geffen School of Medicine Los Angeles CA USA
| |
Collapse
|
26
|
Liu Y, Wang R, Zhou J, Lyu Q, Zhao X, Yang X, Chen K, Gao Z, Li X. Myricetin alleviates high-fat diet-induced atherosclerosis in ApoE -/- mice by regulating bile acid metabolism involved in gut microbiota remodeling. Food Funct 2025; 16:2737-2749. [PMID: 40059779 DOI: 10.1039/d5fo00374a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
Abstract
Atherosclerosis poses a significant threat to global health. This study aimed to investigate the effects of myricetin (MYR) on high-fat diet (HFD)-induced atherosclerosis in ApoE-/- mice. Our findings demonstrated that MYR treatment significantly reduced the formation of atherosclerotic plaques, particularly at a high dose of 100 mg kg-1 day-1. Additionally, MYR markedly attenuated lipid metabolism disorders in ApoE-/- mice by decreasing body weight, improving serum lipid profiles, and reducing lipid deposition. Analysis of 16S rRNA sequencing revealed that MYR treatment enhanced the abundance of probiotic g_Lachnospiraceae_NK4A136, while it reduced that of obesity-associated genera, including Rikenellaceae_RC9_gut_group and Alistipes. Metabolomic analysis and RT-qPCR tests indicated that MYR upregulated hepatic bile acid biosynthesis, evidenced by increased total bile acid levels and enhanced expression of key enzymes CYP7A1 and CYP8B1, particularly through the classical biosynthetic pathway. Spearman's correlation analysis revealed strong associations between the regulated bile acids and these aforementioned bacteria. Therefore, our results demonstrated that MYR exerts an anti-atherosclerotic effect by modulating the gut-liver axis.
Collapse
Affiliation(s)
- Yilong Liu
- Zhejiang Key Laboratory of Horticultural Crop Quality Improvement, Zhejiang University, Hangzhou 310058, China.
| | - Ruoqi Wang
- Zhejiang Key Laboratory of Horticultural Crop Quality Improvement, Zhejiang University, Hangzhou 310058, China.
| | - Jinren Zhou
- Department of Vascular Surgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China.
| | - Qiang Lyu
- School of Pharmacy, Zhejiang Chinese Medical University, 548, Binwen Road, Hangzhou 310053, China
| | - Xiaoyong Zhao
- Zhejiang Key Laboratory of Horticultural Crop Quality Improvement, Zhejiang University, Hangzhou 310058, China.
| | - Xiaochun Yang
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Shandong (Linyi) Institute of Modern Agriculture, Zhejiang University, Linyi 276000, China
| | - Kunsong Chen
- Zhejiang Key Laboratory of Horticultural Crop Quality Improvement, Zhejiang University, Hangzhou 310058, China.
| | - Zhiwei Gao
- Department of Vascular Surgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China.
| | - Xian Li
- Zhejiang Key Laboratory of Horticultural Crop Quality Improvement, Zhejiang University, Hangzhou 310058, China.
- Shandong (Linyi) Institute of Modern Agriculture, Zhejiang University, Linyi 276000, China
| |
Collapse
|
27
|
LIU BQ, YANG C, WEI HY, YU ZX. Global, regional, and national burden of ischemic heart disease attributable to metabolic risks: a systematic analysis of Global Burden of Disease 2021. J Geriatr Cardiol 2025; 22:361-380. [PMID: 40351395 PMCID: PMC12059566 DOI: 10.26599/1671-5411.2025.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025] Open
Abstract
Background Ischemic heart disease (IHD) represents the most significant disease burden among all cardiovascular diseases (CVDs). The increasing prevalence of metabolic risks in the 21st century has a profound impact on the disease burden associated with IHD. We analyzed the global, regional, and national burdens of IHD attributable to metabolic risks from 1990 to 2021. Methods The data were taken from Global Burden of Disease (GBD) study 2021. Deaths, disability-adjusted life years (DALYs), the average annual percent change (AAPC), age-standardized death rates per 100,000 persons (ASDR) and age-standardized rate per 100,000 persons (ASR) of DALYs ranging from 1990 to 2021, were extracted and stratified according to region, nationality, socio-demographic index (SDI), sex, and age. Additionally, the global future trends were predicted using Nordpred prediction model. Results Compared to 1990, in 2021, the number of death and DALYs from metabolic risk-attributed IHD increased globally by 67.35% and 59.91%, respectively; whereas ASDR and ASR of DALYs showed a decreasing trend and the most severe impact was observed in male and elderly populations. In addition, the burden of disease showed an inverted V-shaped relationship with SDI from 1990 to 2021. AAPC showed a significant increase in developing countries and a decrease in developed countries. We also analyzed the effects of different risk factors including metabolic risk factors on IHD in different SDI regions and genders. The prediction of future disease burden showed that the number of death and DALYs will keep rising, while ASDR and ASR of DALYs will maintain a certain downward trend. Conclusions The results of this study highlighted the need for screening and intervention for metabolic risk factors in specific regions and populations, this should call for increased collaboration between developing and developed countries to reduce the burden of disease and improve the prognosis of patients with IHD.
Collapse
Affiliation(s)
- Bo-Qing LIU
- Department of Clinical Medicine, Xiangya Medical School, Central South University, Changsha, China
| | - Chang YANG
- Department of Clinical Medicine, Xiangya Medical School, Central South University, Changsha, China
| | - Heng-Yang WEI
- Department of Clinical Medicine, Xiangya Medical School, Central South University, Changsha, China
| | - Zai-Xin YU
- Department of Cardiology and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, China
| |
Collapse
|
28
|
Do H, Kwon OC, Ha JW, Chung J, Park YB, Huh JH, Lee SW. Remnant Cholesterol Levels at Diagnosis May Predict Acute Coronary Syndrome Occurrence During Follow-Up in Patients with Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. J Clin Med 2025; 14:2260. [PMID: 40217710 PMCID: PMC11989813 DOI: 10.3390/jcm14072260] [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: 03/04/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: Previous studies have revealed the predictive potential of remnant cholesterol (RC) for acute coronary syndrome (ACS) occurrence in the general population. However, whether this association applies to patients with antineutrophil cytoplasmic antibody-associated vasculitis (AAV), in which a lipid paradox exists, remains unclear. We investigated whether RC levels at diagnosis could predict ACS occurrence during follow-up in patients with AAV. Methods: This study included 139 patients with AAV. ACS was defined as ST-elevation myocardial infarction (STEMI), non-STEMI, or unstable angina occurring after AAV diagnosis. RC levels were calculated as (total cholesterol)-(low-density lipoprotein cholesterol)-(high-density lipoprotein cholesterol). Patients were categorised into three groups by RC tertiles: highest (≥26.2 mg/dL), middle (19.1-26.1 mg/dL), and lowest (≤19.0 mg/dL) tertile groups. Results: The median age of the 139 patients (male, 31.7%) was 58.0 years. During follow-up, six, two, and one patients were diagnosed with ACS in the highest, middle, and lowest tertile groups, respectively. Patients in the highest tertile group exhibited a significantly lower ACS-free survival rate than those in the lowest tertile (p = 0.030). In the multivariable Cox hazards model, male sex (hazard ratio [HR] 9.054, 95% confidence interval [CI] 1.786-45.910), Birmingham vasculitis activity score (HR 1.147, 95% CI 1.033-1.274), and the highest tertile of RC levels (HR 10.818, 95% CI 1.867-62.689) were significantly and independently associated with ACS occurrence during follow-up in patients with AAV. Conclusions: Our findings indicate that RC levels at diagnosis may predict ACS occurrence during follow-up in patients with AAV, regardless of the traditional cardiovascular risk factors.
Collapse
Affiliation(s)
- Hyunsue Do
- Division of Rheumatology, Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon-si 24341, Republic of Korea;
| | - Oh Chan Kwon
- Division of Rheumatology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea;
| | - Jang Woo Ha
- Division of Rheumatology, Department of Internal Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin 16995, Republic of Korea;
| | - Jihye Chung
- Division of Rheumatology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.C.); (Y.-B.P.)
| | - Yong-Beom Park
- Division of Rheumatology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.C.); (Y.-B.P.)
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Ji Hye Huh
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea
| | - Sang-Won Lee
- Division of Rheumatology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.C.); (Y.-B.P.)
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
29
|
Wu TW, Chou CL, Liu CC, Chen CF, Cheng CF, Wang LY. Lipid profiles and their association with incident carotid atherosclerosis: A community-based prospective study in Taiwan. Nutr Metab Cardiovasc Dis 2025:104023. [PMID: 40194899 DOI: 10.1016/j.numecd.2025.104023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/04/2025] [Accepted: 03/15/2025] [Indexed: 04/09/2025]
Abstract
BACKGROUND AND AIMS Dyslipidemia, characterized by abnormal blood lipid levels, contributes to atherosclerosis, a condition involving arterial plaque buildup and cardiovascular events. While LDL-C and LDL-to-HDL-C ratios are established atherosclerosis predictors, the role of non-HDL-C is less explored. METHODS AND RESULTS A cohort of 1062 participants without carotid plaque at baseline was analyzed over a 4.0-year follow-up. Age-specific incidence rates were calculated, and baseline characteristics of those who developed plaques were compared using logistic regression and area under the ROC curve (AUROC) analysis to evaluate predictive models. Carotid plaques developed in 284 participants (87 males, 197 females). Incidence rates increased with age, reaching 41.2 % in females and 60.0 % in males aged 70-74 years. Participants with plaques were older (58.2 vs. 55.4 years, p < 0.0001), had higher BMI, blood pressure, and lipid markers, and were more likely to be male, hypertensive, or hyperlipidemic. Logistic regression identified age (OR 1.26 per 5 years), BMI (OR 1.23 per 5 kg/m2), LDL-C (OR 1.07 per 10 mg/dL), and LDL-to-HDL-C ratio (OR 1.41) as significant predictors, with HDL-C offering a protective effect. Models incorporating lipid ratios (non-HDL-to-HDL-C or LDL-to-HDL-C) showed similar predictive power (AUROC 0.636). CONCLUSION Carotid plaque progression correlates with age, male sex, elevated BMI, hypertension, and adverse lipid profiles. Lipid ratios and age are consistent predictors, with HDL-C demonstrating protective effects. Comparable AUROC values across models underscore the value of lipid ratios for assessing atherosclerosis risk.
Collapse
Affiliation(s)
- Tzu-Wei Wu
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan, ROC.
| | - Chao-Liang Chou
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan, ROC; Department of Neurology, MacKay Memorial Hospital, New Taipei City, Taiwan, ROC.
| | - Chun-Chieh Liu
- Cardiovascular Center, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan, ROC.
| | - Chuen-Fei Chen
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan, ROC.
| | - Chun-Fang Cheng
- Tamsui Health Station, Department of Health, New Taipei City Government, New Taipei City, Taiwan, ROC.
| | - Li-Yu Wang
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan, ROC.
| |
Collapse
|
30
|
Ryu HE, Lee YJ, Park B, Jung DH. Comparisons of three novel markers for insulin resistance to predict incident cardiovascular disease: a Korean cohort study from three different regions. Eur J Med Res 2025; 30:188. [PMID: 40114229 PMCID: PMC11924704 DOI: 10.1186/s40001-025-02374-0] [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: 10/01/2023] [Accepted: 02/10/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Cardiovascular disease (CVD) is a crucial human health challenge. Previous studies have shown an association between CVD and the triglyceride-glucose (TyG) index, atherogenic index of plasma (AIP), and metabolic score for insulin resistance (METS-IR). However, a comparison of these novel markers for predicting CVD is not well known. Therefore, we aimed to assess the value of TyG, AIP, and METS-IR in predicting the incidence of CVD in three large cohorts of Korean adults. METHODS Data from 28 437 participants in the Korean Genome and Epidemiology Study (KoGES) and Korea Health Insurance Review and Assessment (HERAS-HIRA) were assessed. The participants were divided into four groups according to the quartiles of TyG index: ln ([triglyceride × fasting plasma glucose]/2), AIP calculated as log (triglyceride/high-density lipoprotein cholesterol), and METS-IR index: (ln ([2 × fasting plasma glucose] + triglyceride) × body mass index)/(ln [high-density lipoprotein cholesterol-cholesterol]). We prospectively assessed the hazard ratios (HRs) with 95% confidence intervals (CIs) for CVD using multivariate Cox proportional hazard regression models after adjusting for potential confounding variables. RESULTS During the follow-up period, 987 participants (3.5%) developed CVD. Compared with the referent first quartiles, the highest TyG index, AIP, and METS-IR quartiles, with HRs of 1.73 (95% CI 1.41-2.12), 1.47 (95% CI 1.19-1.80), and 2.61 (95% CI 1.83-3.72), respectively, significantly predicted future CVD, after adjusting for age, sex, and body mass index. When comparing the three biomarkers for insulin resistance, the TyG index and METS-IR showed similar predictive values, whereas AIP had a lower significance in predicting CVD. CONCLUSIONS Based on the current findings, novel surrogate markers of insulin resistance, particularly METS-IR and TyG index, may help predict the risk of CVD in Koreans.
Collapse
Affiliation(s)
- Ha Eun Ryu
- Department of Family Medicine, Yongin Severance Hospital, 363 Dongbaekjukjeondae-Ro, Giheung-Gu, Yongin-Si, Gyeonggi-Do, 16995, Republic of Korea
- Department of Family Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yong Jae Lee
- Department of Family Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- 3Department of Family Medicine, Gangnam Severance Hospital, Seoul, 06273, Republic of Korea
| | - Byoungjin Park
- Department of Family Medicine, Yongin Severance Hospital, 363 Dongbaekjukjeondae-Ro, Giheung-Gu, Yongin-Si, Gyeonggi-Do, 16995, Republic of Korea.
- Department of Family Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Dong Hyuk Jung
- Department of Family Medicine, Yongin Severance Hospital, 363 Dongbaekjukjeondae-Ro, Giheung-Gu, Yongin-Si, Gyeonggi-Do, 16995, Republic of Korea.
- Department of Family Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| |
Collapse
|
31
|
Yang L, Guan X, Cheng J, Ni L, Yao H, Gao Y, Zhu K, Shi X, Li B, Lin Y. VAMP8 as a biomarker and potential therapeutic target for endothelial cell dysfunction in atherosclerosis. Gene 2025; 942:149231. [PMID: 39800194 DOI: 10.1016/j.gene.2025.149231] [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: 05/30/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Endothelial cell dysfunction has a critical role in the pathophysiology of atherosclerosis. This study aims to uncover pivotal genes and pathways linked to endothelial cell dysfunction in atherosclerosis, as well as to ascertain the assumed causal effects and potential mechanisms. METHODS Datasets relevant to endothelial cell dysfunction in atherosclerosis were collected and divided into training and validation sets. Following differential analysis, we constructed a protein-protein interaction (PPI) network and a molecular interaction map of common-differentially expressed genes (co-DEGs) with proteins known to be involved in atherosclerotic endothelial cell dysfunction. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genome (KEGG), and Gene Set Enrichment Analysis (GSEA) were also conducted. Moreover, human umbilical vein endothelial cells (HUVECs) were cultured in circumstances characterized by elevated glucose levels to establish a cellular injury model simulating atherosclerotic conditions, and quantitative Polymerase Chain Reaction (qPCR) experiments were conducted to validate the differences of co-DEGs. Subsequently, the Summary-data-based Mendelian Randomization (SMR) method was employed. Additionally, we employed the Western Blot (WB) technique to validate the differential expression of VAMP8. Finally, we identified the differential expression of VAMP8 in the validation set and further validated its differential expression by collecting fresh blood samples from 20 patients with atherosclerosis and 20 healthy individuals. RESULTS 14 co-DEGs (FABP5, GULP1, COL4A5, VAMP8, FABP4, PFN2, ANGPT2, TFPI2, NUPR1, SULF1, FGF13, BASP1, EPB41L3, and PBK) were identified. SMR analysis confirmed 10 potential causal effect genes: PSRC1, VAMP8, FES, HNRNPUL1, CFDP1, SAP130, MDN1, OPRL1, UTP11, and HOXC4. The qPCR and WB experiments demonstrated that VAMP8 was significantly upregulated in the injured HUVECs group (p < 0.0001). Compared to the control group, VAMP8 was markedly increased in the blood samples of patients with atherosclerosis (p < 0.0001). CONCLUSIONS VAMP8 may potentially serve as a pathogenic gene in the process of endothelial dysfunction in atherosclerosis.
Collapse
Affiliation(s)
- Luqun Yang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Xin Guan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Jiangwei Cheng
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Lin Ni
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Huijing Yao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yuping Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Kaiyi Zhu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Xiushan Shi
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Bingjie Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yuanyuan Lin
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| |
Collapse
|
32
|
Zhang H, Long J, Wang C, Liu X, Lu H, Xu W, Sun X, Dou P, Zhou D, Zhu L, Xu K, Meng Y. Development of a nomogram model for predicting acute stroke events based on dual-energy CTA analysis of carotid intraplaque and perivascular adipose tissue. Front Neurol 2025; 16:1566395. [PMID: 40134696 PMCID: PMC11932918 DOI: 10.3389/fneur.2025.1566395] [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: 01/24/2025] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
Objective To evaluate the predictive value of dual-energy CT angiography (DECTA) parameters of carotid intraplaque and perivascular adipose tissue (PVAT) in acute stroke events. Methods A retrospective analysis was conducted using clinical, laboratory, and imaging data from patients who underwent dual-energy carotid CTA and cranial MRI. Acute cerebral infarctions occurring in the ipsilateral anterior circulation were classified as the symptomatic group (STA group), while other cases were categorized as the asymptomatic group (ATA group). LASSO regression was employed to identify key predictors. These predictors were used to develop three models: the intraplaque model (IP_Model), the perivascular adipose tissue model (PA_Model), and the nomogram model (Nomo_Model). The predictive accuracy of the models was evaluated using receiver operating characteristic (ROC) analysis, calibration curves, and decision curve analysis. Statistical significance was defined as p < 0.05. Results Seventy-five patients (mean age: 68.7 ± 8.7 years) were analyzed. LASSO regression identified seven significant variables (IP_Zeff, IP_40KH, IP_K, PA_FF, PA_VNC, PA_Rho, PA_K) for model construction. The Nomo_Model demonstrated superior predictive performance compared to the IP_Model and PA_Model, achieving an area under the curve (AUC) of 0.962, with a sensitivity of 95.8%, specificity of 82.4%, precision of 82.6%, an F1 score of 0.809, and an accuracy of 88.0%. The clinical decision curve analysis further validated the Nomo_Model's significant clinical utility. Conclusion DECTA imaging parameters revealed significant differences in carotid intraplaque and PVAT characteristics between the STA and ATA groups. Integrating these parameters into the nomogram (Nomo_Model) resulted in a highly accurate and clinically relevant tool for predicting acute stroke risk.
Collapse
Affiliation(s)
- He Zhang
- Department of Radiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- College of Medical Imaging, Xuzhou Medical University, Xuzhou, China
- Jiangsu Medical Imaging and Digital Medical Engineering Research Center, Xuzhou Medical University, Xuzhou, China
- Department of Radiology, Jiawang District People's Hospital, Xuzhou, China
| | - Juan Long
- Department of Radiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- College of Medical Imaging, Xuzhou Medical University, Xuzhou, China
- Jiangsu Medical Imaging and Digital Medical Engineering Research Center, Xuzhou Medical University, Xuzhou, China
| | - Chenzi Wang
- Department of Radiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- College of Medical Imaging, Xuzhou Medical University, Xuzhou, China
- Jiangsu Medical Imaging and Digital Medical Engineering Research Center, Xuzhou Medical University, Xuzhou, China
| | - Xiaohan Liu
- Department of Radiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- College of Medical Imaging, Xuzhou Medical University, Xuzhou, China
- Jiangsu Medical Imaging and Digital Medical Engineering Research Center, Xuzhou Medical University, Xuzhou, China
| | - He Lu
- Department of Radiology, Jiawang District People's Hospital, Xuzhou, China
| | - Wenbei Xu
- Department of Radiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- College of Medical Imaging, Xuzhou Medical University, Xuzhou, China
- Jiangsu Medical Imaging and Digital Medical Engineering Research Center, Xuzhou Medical University, Xuzhou, China
| | - Xiaonan Sun
- Department of Radiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- College of Medical Imaging, Xuzhou Medical University, Xuzhou, China
- Jiangsu Medical Imaging and Digital Medical Engineering Research Center, Xuzhou Medical University, Xuzhou, China
| | - Peipei Dou
- Department of Radiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- College of Medical Imaging, Xuzhou Medical University, Xuzhou, China
- Jiangsu Medical Imaging and Digital Medical Engineering Research Center, Xuzhou Medical University, Xuzhou, China
| | - Dexing Zhou
- Department of Radiology, Jiawang District People's Hospital, Xuzhou, China
| | - Lili Zhu
- Department of Medical Imaging, Xuzhou New Health Hospital, Xuzhou, China
| | - Kai Xu
- Department of Radiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- College of Medical Imaging, Xuzhou Medical University, Xuzhou, China
- Jiangsu Medical Imaging and Digital Medical Engineering Research Center, Xuzhou Medical University, Xuzhou, China
| | - Yankai Meng
- Department of Radiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- College of Medical Imaging, Xuzhou Medical University, Xuzhou, China
- Jiangsu Medical Imaging and Digital Medical Engineering Research Center, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
33
|
Xia B, Shaheen N, Chen H, Zhao J, Guo P, Zhao Y. RNA aptamer-mediated RNA nanotechnology for potential treatment of cardiopulmonary diseases. Pharmacol Res 2025; 213:107659. [PMID: 39978660 DOI: 10.1016/j.phrs.2025.107659] [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: 09/13/2024] [Revised: 01/14/2025] [Accepted: 02/14/2025] [Indexed: 02/22/2025]
Abstract
Ribonucleic acid (RNA) aptamers are single-stranded RNAs that bind to target proteins or other molecules with high specificity and affinity, modulating biological functions through distinct mechanisms. These aptamers can act n as antagonists to block pathological interactions, agonists to activate signaling pathways, or delivery vehicles for therapeutic cargos such as siRNAs and miRNAs. The advances in RNA nanotechnology further enhances the versatility of RNA aptamers, offering scalable platforms for engineering. In this review, we have summarized recent developments in RNA aptamer-mediated RNA nanotechnology and provide an overview of its potential in treating cardiovascular and respiratory disorders, including atherosclerosis, acute coronary syndromes, heart failure, lung cancer, pulmonary hypertension, asthma, chronic obstructive pulmonary disease (COPD), acute lung injury, viral respiratory infections, and pulmonary fibrosis. By integrating aptamer technologies with innovative delivery systems, RNA aptamers hold the potential to revolutionize the treatment landscape for cardiopulmonary diseases.
Collapse
Affiliation(s)
- Boyu Xia
- Department of Physiology and Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Nargis Shaheen
- Department of Physiology and Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Huilong Chen
- Department of Physiology and Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jing Zhao
- Department of Physiology and Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Peixuan Guo
- Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Yutong Zhao
- Department of Physiology and Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
34
|
Assmann AK, Buschmann J, Reimers S, Karakas A, Weber E, Aubin H, Lichtenberg A, Assmann A. Development of an atherosclerosis rabbit model to evaluate the hemodynamic impact of extracorporeal circulation. Animal Model Exp Med 2025; 8:523-533. [PMID: 39909868 PMCID: PMC11904102 DOI: 10.1002/ame2.12556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/30/2024] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Aortic atherosclerosis increases the risk of embolic events under extracorporeal circulation (ECC). To evaluate the hemodynamic impact of ECC on atheromatous plaques, an atherosclerosis animal model, which is also eligible for ECC, is required. METHODS Twenty-nine New Zealand White rabbits received a pro-atherosclerotic diet (group diet, n = 10), a pro-atherosclerotic diet and additional intraaortic balloon insufflation injury (group BI, n = 9), or served as controls (n = 10). After 3 or 6 months, aortic explants were analyzed by (immuno-)histology and RT-PCR. RESULTS Blood serum analyses revealed increased cholesterol-levels in groups diet and BI compared to controls (3 months: p = 0.03 each, 6 months: p < 0.0001 each). Aortic inflammatory infiltration was significantly enhanced in groups diet (CD3 at 3 months: p < 0.0001, 6 months: p = 0.02; CD68 at 3 months: p = 0.01) and BI (CD3 at 3 months: p < 0.0001, 6 months: p = 0.03; CD68 at 3 months: p = 0.04, 6 months: p = 0.02). Increased intima hyperplasia occurred in both groups (p < 0.0001 each). Macroscopic analyses after 3 and 6 months showed ubiquitous lumen-narrowing aortic plaques. Calcification of the intima and media was increased in groups diet (intima: p < 0.0001 at 3 and 6 months; media at 3 months: p < 0.0001, 6 months: p = 0.01) and BI (intima: p < 0.0001 at 3 and 6 months; media at 3 months: p < 0.0001, 6 months: p = 0.02). Extensive lipid accumulation was found in the intima in both treatment groups (p < 0.0001 each). CONCLUSIONS A rabbit model with high aortic calcific plaque burden-diet-induced with no implicit need of an additional intimal injury by an intraaortic balloon insufflation due to comparable outcome-exhibiting multiple pathophysiological aspects of human atherosclerosis has been designed and thoroughly characterized. It is suitable for use in future studies on the interaction between atherosclerotic plaques and the arterial blood flow under ECC.
Collapse
Affiliation(s)
- Anna Kathrin Assmann
- Department of Cardiac Surgery and CURE 3D Lab, Medical FacultyHeinrich Heine University, University Hospital DüsseldorfDüsseldorfGermany
| | - Jan Buschmann
- CURE 3D Lab, Medical FacultyHeinrich Heine UniversityDüsseldorfGermany
| | - Sinje Reimers
- Department of Cardiac Surgery and CURE 3D Lab, Medical FacultyHeinrich Heine University, University Hospital DüsseldorfDüsseldorfGermany
| | - Aleyna Karakas
- CURE 3D Lab, Medical FacultyHeinrich Heine UniversityDüsseldorfGermany
| | - Elvira Weber
- CURE 3D Lab, Medical FacultyHeinrich Heine UniversityDüsseldorfGermany
| | - Hug Aubin
- Department of Cardiac Surgery and CURE 3D Lab, Medical FacultyHeinrich Heine University, University Hospital DüsseldorfDüsseldorfGermany
- Cardiovascular Research Institute Duesseldorf (CARID), Medical FacultyHeinrich‐Heine UniversityDüsseldorfGermany
| | - Artur Lichtenberg
- Department of Cardiac Surgery and CURE 3D Lab, Medical FacultyHeinrich Heine University, University Hospital DüsseldorfDüsseldorfGermany
- Cardiovascular Research Institute Duesseldorf (CARID), Medical FacultyHeinrich‐Heine UniversityDüsseldorfGermany
| | - Alexander Assmann
- Department of Cardiac Surgery and CURE 3D Lab, Medical FacultyHeinrich Heine University, University Hospital DüsseldorfDüsseldorfGermany
| |
Collapse
|
35
|
Puig N, Camps-Renom P, Garcia E, Benítez-Amaro A, Aguilera-Simón A, Fernández-León A, Quesada JLS, Llorente-Cortés V, Benitez S. sICAM-1 concentrations are associated with inflammation in contralateral carotid plaque in patients with ischemic stroke. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2025; 37:100729. [PMID: 39095307 DOI: 10.1016/j.arteri.2024.07.002] [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: 05/07/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Atherosclerotic plaques in the internal carotid artery are responsible for more than 15% of ischemic strokes. Carotid 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET) detects plaque inflammation. Plasma ICAM-1 and LRP1 concentrations have been associated with inflammation in ipsilateral carotid plaque. The aim of the present study was to test the association between the soluble (s) form of these biomarkers and contralateral carotid plaques. METHODS Prospective study conducted in 53 patients with a recent ischemic stroke and at least one atherosclerotic plaque in both carotid arteries. All of the patients underwent an early carotid 18F-FDG PET, and a blood sample was obtained at 7±1 days. Several plasma inflammatory markers were evaluated by Multiplex and sLRP1 levels were measured by commercial ELISA. Bivariate and multivariable linear regression was used to assess the association between inflammatory markers and the clinical variables, including contralateral maximum standardized uptake value (SUVmax) and mean SUVmax (mean of contralateral and ipsilateral SUVmax) of 18F-FDG uptake. Hazard ratio (HR) was estimated with Cox models adjusted for potential confounding factors to evaluate recurrence. RESULTS Multivariable linear regression analysis showed an independent association between sICAM-1 and sVCAM-1 and mean SUVmax (CI=-0.064-0.325, p=0.004; CI=0.079-0.554, p=0.010). In addition, in bivariate regression analysis, sICAM-1 was associated with contralateral SUVmax (CI=0.049-0.382, p=0.012). Cox regression showed that mean SUVmax was associated with stroke recurrence (HR=5.604, p=0.044). CONCLUSIONS sICAM-1 was independently associated with mean carotid plaque inflammation and with inflammation in contralateral plaque. sICAM-1 could be an indicator of plaque inflammation even in asymptomatic plaques.
Collapse
Affiliation(s)
- Núria Puig
- Cardiovascular Biochemistry, Cardiovascular Biochemistry, Research Institute Sant Pau (Institut de Recerca Sant Pau, IR Sant Pau), Barcelona, Spain
| | - Pol Camps-Renom
- Stroke Unit, Department of Neurology, Hospital de La Santa Creu I Sant Pau, IR Sant Pau, Barcelona, Spain
| | - Eduardo Garcia
- Lipids and Cardiovascular Pathology, Institut d'Investigacions Biomèdiques de Barcelona (IIBB-CSIC), IR Sant Pau, Barcelona, Spain
| | - Aleyda Benítez-Amaro
- Lipids and Cardiovascular Pathology, Institut d'Investigacions Biomèdiques de Barcelona (IIBB-CSIC), IR Sant Pau, Barcelona, Spain
| | - Ana Aguilera-Simón
- Stroke Unit, Department of Neurology, Hospital de La Santa Creu I Sant Pau, IR Sant Pau, Barcelona, Spain
| | - Alejandro Fernández-León
- Department of Nuclear Medicine, Hospital de la Santa Creu i SantPau, IR SantPau, Barcelona, Spain
| | - Jose Luis Sanchez Quesada
- Cardiovascular Biochemistry, Cardiovascular Biochemistry, Research Institute Sant Pau (Institut de Recerca Sant Pau, IR Sant Pau), Barcelona, Spain; CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain
| | - Vicenta Llorente-Cortés
- Lipids and Cardiovascular Pathology, Institut d'Investigacions Biomèdiques de Barcelona (IIBB-CSIC), IR Sant Pau, Barcelona, Spain; CIBER of Cardiovascular (CIBERCV), Madrid, Spain.
| | - Sonia Benitez
- Cardiovascular Biochemistry, Cardiovascular Biochemistry, Research Institute Sant Pau (Institut de Recerca Sant Pau, IR Sant Pau), Barcelona, Spain; CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain.
| |
Collapse
|
36
|
Muheeb G, Yusuf J, Mehta V, Faizuddin M, Kurian S, M P G, Gupta MD, Safal S, Gautam A, Chauhan NK. Systemic immune inflammatory response index (SIIRI) in acute myocardial infarction. Coron Artery Dis 2025; 36:139-150. [PMID: 39501911 DOI: 10.1097/mca.0000000000001454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
BACKGROUND Different treatment approaches exist for non-ST elevation acute coronary syndrome (ACS) patients. This study assessed the systemic immune inflammatory response index (SIIRI) for its prognostic value and incremental clinical utility in determining optimal timing for percutaneous coronary intervention (PCI) in non-ST elevation myocardial infarction (NSTEMI) patients, particularly when troponin levels are initially negative. METHODS This study included 1270 ACS patients: 437 STEMI, 422 NSTEMI, and 411 unstable angina. Patients were stratified by SIIRI levels measured at admission, and coronary artery disease severity was evaluated using the SYNTAX score. The primary endpoint was major adverse cardiovascular events (MACE), including cardiovascular death, non-fatal myocardial infarction, stroke, and revascularization. Secondary endpoints encompassed individual MACE components and heart failure hospitalisations. RESULTS The mean age was 54.93 years (83% male). SIIRI levels were significantly higher in STEMI patients (6.83 ± 6.43 × 10 5 ) compared to NSTEMI (4.5 ± 5.39 × 10 5 ) and unstable angina (3.48 ± 2.83 × 10 5 ) ( P < 0.001). Area under the curve for SIIRI distinguished NSTEMI and unstable angina from STEMI (0.81 and 0.80), with optimal cut-off points of 4.80 × 10 5 and 4.25 × 10 5 . In NSTEMI, 24.6% presented within 2 h of symptom onset, were troponin-negative, yet had elevated SIIRI. Post-PCI, SIIRI > 4.93 × 10 5 correlated with increased MACE at 1 year (17.2% vs 5%). CONCLUSION NSTEMI and unstable angina patients with SIIRI values >4.80 × 10 5 and 4.25 × 10 5 respectively, may require urgent intervention (<2 h). SIIRI can be of significant utility in patients of NSTEMI who present earlier with negative troponins. SIIRI can also aid in identifying high-risk individuals post-PCI, providing a valuable tool for early and accurate assessment.
Collapse
Affiliation(s)
- Ghazi Muheeb
- Department of Cardiology, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research, Delhi, India
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Wong A, Sun Q, Latif II, Karwi QG. Macrophage energy metabolism in cardiometabolic disease. Mol Cell Biochem 2025; 480:1763-1783. [PMID: 39198360 PMCID: PMC11842501 DOI: 10.1007/s11010-024-05099-6] [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: 04/27/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
In a rapidly expanding body of literature, the major role of energy metabolism in determining the response and polarization status of macrophages has been examined, and it is currently a very active area of research. The metabolic flux through different metabolic pathways in the macrophage is interconnected and complex and could influence the polarization of macrophages. Earlier studies suggested glucose flux through cytosolic glycolysis is a prerequisite to trigger the pro-inflammatory phenotypes of macrophages while proposing that fatty acid oxidation is essential to support anti-inflammatory responses by macrophages. However, recent studies have shown that this understanding is oversimplified and that the metabolic control of macrophage polarization is highly complex and not fully defined yet. In this review, we systematically reviewed and summarized the literature regarding the role of energy metabolism in controlling macrophage activity and how that might be altered in cardiometabolic diseases, namely heart failure, obesity, and diabetes. We critically appraised the experimental studies and methodologies in the published studies. We also highlighted the challenging concepts in macrophage metabolism and identified several research questions yet to be addressed in future investigations.
Collapse
Affiliation(s)
- Angela Wong
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, A1B 3V6, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Qiuyu Sun
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, A1B 3V6, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Ismail I Latif
- Department of Microbiology, College of Medicine, University of Diyala, Baqubaa, Diyala, Iraq
| | - Qutuba G Karwi
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, A1B 3V6, Canada.
| |
Collapse
|
38
|
Ren J, Yao G, Ren L, Wang Y, Gao J, Zhang Y. Exploring the Associations Between Non-Traditional Lipid Parameters and Epicardial Adipose Tissue Volume. Angiology 2025; 76:239-248. [PMID: 37843829 DOI: 10.1177/00033197231207264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
The aim of this retrospective study was to determine the relationship between non-traditional lipid parameters and epicardial adipose tissue (EAT). A total of 770 patients with coronary computed tomography angiography examinations were included. The non-traditional lipid parameters included the atherogenic index of plasma (AIP), the atherogenic coefficient (AC), monocyte to high-density lipoprotein cholesterol (HDL-C) ratio (MHR), and lipoprotein combined index (LCI). To investigate the association between non-conventional lipid markers and the EAT-volume (EAT-v), a univariate and multivariate analyses were conducted. The receiver operating characteristic (ROC) analysis was used to compare the predictive ability among the four non-traditional lipid parameters. In the univariate analysis, we identified factors that might have effects on EAT-v (all P<.05) and adjusted for these in the multivariate analysis. We found that except for MHR, other non-traditional lipid parameters were still associated with high EAT-v after adjustment (all P<.05). In the ROC analysis, the area under the curve (AUC) of AIP was greater than that of other non-traditional lipid parameters and lipid profiles. There was an association between both non-traditional lipid parameters and EAT-v. After adjustment, the AIP remained an independent predictor of EAT-v and it outperformed other non-traditional lipid parameters.
Collapse
Affiliation(s)
- Junli Ren
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guang Yao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lichen Ren
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiran Wang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianbo Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yonggao Zhang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
39
|
Guo Y, Wang F, Ma S, Mao Z, Zhao S, Sui L, Jiao C, Lu R, Zhu X, Pan X. Relationship between atherogenic index of plasma and length of stay in critically ill patients with atherosclerotic cardiovascular disease: a retrospective cohort study and predictive modeling based on machine learning. Cardiovasc Diabetol 2025; 24:95. [PMID: 40022165 PMCID: PMC11871731 DOI: 10.1186/s12933-025-02654-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 02/18/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND The atherogenic index of plasma (AIP) is considered an important marker of atherosclerosis and cardiovascular risk. However, its potential role in predicting length of stay (LOS), especially in patients with atherosclerotic cardiovascular disease (ASCVD), remains to be explored. We investigated the effect of AIP on hospital LOS in critically ill ASCVD patients and explored the risk factors affecting LOS in conjunction with machine learning. METHODS Using data from the Medical Information Mart for Intensive Care (MIMIC)-IV. AIP was calculated as the logarithmic ratio of TG to HDL-C, and patients were stratified into four groups based on AIP values. We investigated the association between AIP and two key clinical outcomes: ICU LOS and total hospital LOS. Multivariate logistic regression models were used to evaluate these associations, while restricted cubic spline (RCS) regressions assessed potential nonlinear relationships. Additionally, machine learning (ML) techniques, including logistic regression (LR), decision tree (DT), random forest (RF), extreme gradient boosting (XGB), and light gradient boosting machine (LGB), were applied, with the Shapley additive explanation (SHAP) method used to determine feature importance. RESULTS The study enrolled a total of 2423 patients with critically ill ASCVD, predominantly male (54.91%), and revealed that higher AIP values were independently associated with longer ICU and hospital stays. Specifically, for each unit increase in AIP, the odds of prolonged ICU and hospital stays were significantly higher, with adjusted odds ratios (OR) of 1.42 (95% CI, 1.11-1.81; P = 0.006) and 1.73 (95% CI, 1.34-2.24; P < 0.001), respectively. The RCS regression demonstrated a linear relationship between increasing AIP and both ICU LOS and hospital LOS. ML models, specifically LGB (ROC:0.740) and LR (ROC:0.832) demonstrated superior predictive accuracy for these endpoints, identifying AIP as a vital component of hospitalization duration. CONCLUSION AIP is a significant predictor of ICU and hospital LOS in patients with critically ill ASCVD. AIP could serve as an early prognostic tool for guiding clinical decision-making and managing patient outcomes.
Collapse
Affiliation(s)
- Yu Guo
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fuxu Wang
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shiyin Ma
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhi Mao
- Department of Critical Care Medicine, The First Medical Center of PLA General Hospital, Beijing, China
| | - Shuangmei Zhao
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Liutao Sui
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chucheng Jiao
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ruogu Lu
- Medical Innovation Research Department, Chinese PLA General Hospital, Beijing, China.
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
40
|
Le Goff W, Bourron O, Materne C, Galier S, Phan F, Tan-Chen S, Guillas I, Hartemann A, Salem JE, Redheuil A, Foufelle F, Le Stunff H, Hajduch E, Guerin M. Inverse relationship between circulating sphingosine-1-phosphate and precursor species and coronary artery calcification score in type 2 diabetes. Cardiovasc Diabetol 2025; 24:85. [PMID: 39984928 PMCID: PMC11846453 DOI: 10.1186/s12933-025-02624-9] [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: 11/27/2024] [Accepted: 01/31/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Sphingosine 1-phosphate (S1P) is a key mediator of lipid signaling with strong immunomodulatory and anti-inflammatory effects. Circulating S1P levels including S1P in high-density lipoproteins (HDL) were demonstrated to be inversely associated with cardiovascular diseases (CVD). However, no studies are available regarding a potential implication of S1P on the risk of CVD in type 2 diabetes (T2D). The objective of this study is to determine if the increased CVD risk in T2D may involve an alteration of circulating S1P species as well as their precursors. METHODS A total of 168 and 31 patients with T2D (154 men and 45 women) with available Coronary artery calcification (CAC) score from the DIACART and CERABIAB cohorts, respectively, were included in the study. Quantification of S1P species and their precursors was carried out by LC-MS/MS in plasma and isolated HDL. CAC score was modeled as a binary variable (0/1 below or equal/above 100) using CAC < 100 for reference. S1P species or precursors were modeled as binary variables dichotomized at the median (0/1: below or equal/above the median). The relationships between S1P species and CAC score modeled as a binary variable (below or equal/above 100) was evaluated by linear regression analyses. In vitro experiments were conducted to evaluate the contribution of HDL-S1P content on anti-inflammatory properties of HDL particles. RESULTS Multivariate analysis revealed that plasma S1P levels, especially d18:1-S1P, and sphingosine in HDL were inversely associated with the high risk of CVD (CAC > 100) in patients with T2D. Clustering of HDL according to their concentration in S1P species and their precursors revealed that S1P-impoverished HDL is a major feature of patients with a CAC > 100. In vitro analysis of monocyte adhesion and inflammation in human umbilical vein endothelial cells as well as inflammatory phenotype of human macrophages demonstrated that low HDL-S1P exhibited impaired anti-inflammatory properties in comparison to high HDL-S1P. CONCLUSION This study unraveled that circulating S1P and their precursors are biomarkers of coronary atherosclerosis in T2D, which may underlie the lower abundance of S1P and anti-inflammatory activities of HDL. Trial registration ClinicalTrials.gov number, NCT02431234.
Collapse
Affiliation(s)
- Wilfried Le Goff
- INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), UMR_S1166, Sorbonne Université, 75013, Paris, France
| | - Olivier Bourron
- INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), UMR_S1166, Sorbonne Université, 75013, Paris, France
- Diabetology Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Paris, France
| | - Clément Materne
- INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), UMR_S1166, Sorbonne Université, 75013, Paris, France
| | - Sophie Galier
- INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), UMR_S1166, Sorbonne Université, 75013, Paris, France
| | - Franck Phan
- INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), UMR_S1166, Sorbonne Université, 75013, Paris, France
- Diabetology Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Paris, France
| | - Sophie Tan-Chen
- Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Paris, France
| | - Isabelle Guillas
- INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), UMR_S1166, Sorbonne Université, 75013, Paris, France
| | - Agnès Hartemann
- Diabetology Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Paris, France
| | - Joe-Elie Salem
- AP-HP, INSERM, CIC-1901, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Alban Redheuil
- Laboratoire d'Imagerie Biomédicale INSERM_1146, CNRS_7371, ICT Cardiovascular and Thoracic Imaging Unit, Assistance Publique‑Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Paris, France
| | - Fabienne Foufelle
- INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), UMR_S1166, Sorbonne Université, 75013, Paris, France
| | - Hervé Le Stunff
- CNRS UMR 9197, Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, Saclay, France.
| | - Eric Hajduch
- INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), UMR_S1166, Sorbonne Université, 75013, Paris, France.
| | - Maryse Guerin
- INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), UMR_S1166, Sorbonne Université, 75013, Paris, France.
| |
Collapse
|
41
|
Ma Y, Gong H, Cheng L, Zhang D. Discoid Domain Receptors Signaling in Macrophages-Mediated Diseases. Int J Gen Med 2025; 18:907-926. [PMID: 39990299 PMCID: PMC11847422 DOI: 10.2147/ijgm.s487093] [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: 07/15/2024] [Accepted: 01/18/2025] [Indexed: 02/25/2025] Open
Abstract
Macrophages, as a crucial component of the body's immune system, play a vital role in the onset, progression, and outcome of diseases. Discoidin domain receptors (DDRs), important members of the novel receptor tyrosine kinase superfamily, exhibit unique functions in macrophage physiology. Through interactions with the extracellular matrix, DDRs activate signaling pathways such as p38 MAPK and NF-κB, regulating macrophage adhesion, migration, and secretory functions, thereby influencing their behavior in diseases. Recent studies have indicated a direct correlation between DDRs and the progression of various diseases, including inflammation, cancer, and fibrosis. However, there remain numerous knowledge gaps regarding the specific mechanisms by which DDRs function in macrophage-mediated diseases. This article provides an in-depth summary of the regulatory mechanisms of DDRs on macrophages, detailing their modulatory roles in various diseases through macrophages and their underlying mechanisms. The aim is to offer new insights into biomedical therapies targeting DDRs and the development of novel drugs.
Collapse
Affiliation(s)
- Yaohui Ma
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| | - Hang Gong
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| | - Long Cheng
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| | - Dekui Zhang
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| |
Collapse
|
42
|
Li K, Feng J, Li M, Han L, Wu Y. Systematic Review of Interleukin-35 in Endothelial Dysfunction: A New Target for Therapeutic Intervention. Mediators Inflamm 2025; 2025:2003124. [PMID: 39974277 PMCID: PMC11839265 DOI: 10.1155/mi/2003124] [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: 06/19/2024] [Accepted: 01/21/2025] [Indexed: 02/21/2025] Open
Abstract
Endothelial dysfunction is a significant factor in the pathogenesis of various diseases. In pathological states, endothelial cells (ECs) undergo activation, resulting in dysfunction characterized by the stimulation of inflammatory responses, oxidative stress, cell proliferation, blood coagulation, and vascular adhesions. Interleukin-35 (IL-35), a novel member of the IL-12 family, is primarily secreted by regulatory T cells (Tregs) and regulatory B cells (Bregs). The role of IL-35 in immunomodulation, antioxidative stress, resistance to apoptosis, control of EC activation, adhesion, and angiogenesis in ECs remains incompletely understood, as the specific mechanisms of IL-35 action and its regulation have yet to be fully elucidated. Therefore, this systematic review aims to comprehensively investigate the impact of IL-35 on ECs and their physiological roles in a range of conditions, including cardiovascular diseases, tumors, sepsis, and rheumatoid arthritis (RA), with the objective of elucidating the potential of IL-35 as a therapeutic target for these ailments.
Collapse
Affiliation(s)
- Kai Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Jie Feng
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Meng Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Leilei Han
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| |
Collapse
|
43
|
Gong S, Li Y, Yan K, Shi Z, Leng J, Bao Y, Ning K. The Crosstalk Between Endothelial Cells, Smooth Muscle Cells, and Macrophages in Atherosclerosis. Int J Mol Sci 2025; 26:1457. [PMID: 40003923 PMCID: PMC11855868 DOI: 10.3390/ijms26041457] [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: 12/31/2024] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory vascular disease closely tied to cellular metabolism. Recent genome-wide association study data have suggested the significant roles of endothelial cells, smooth muscle cells, and macrophages in the regression and exacerbation of AS. However, the impact of cellular crosstalk and cellular metabolic derangements on disease progression in AS is vaguely understood. In this review, we analyze the roles of the three cell types in AS. We also summarize the crosstalk between the two of them, and the associated molecules and consequences involved. In addition, we emphasize potential therapeutic targets and highlight the importance of the three-cell co-culture model and extracellular vesicles in AS-related research, providing ideas for future studies.
Collapse
Affiliation(s)
- Sihe Gong
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Yanni Li
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Kaijie Yan
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Zhonghong Shi
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Jing Leng
- Preclinical Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China;
| | - Yimin Bao
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Ke Ning
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| |
Collapse
|
44
|
Liang L, Peng F, Wang L, Lin C, Chi H. Effects of exercise on high-density lipoprotein levels in middle-aged and older individuals: A systematic review and meta-analysis. Medicine (Baltimore) 2025; 104:e41493. [PMID: 39928779 PMCID: PMC11813029 DOI: 10.1097/md.0000000000041493] [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: 02/06/2024] [Revised: 11/28/2024] [Accepted: 01/23/2025] [Indexed: 02/12/2025] Open
Abstract
BACKGROUND This study aimed to perform a systematic review and meta-analysis of randomized controlled trials investigating the effect of exercise on high-density lipoprotein (HDL) levels in middle-aged and older individuals. METHODS A systematic search was conducted in PubMed, Web of Science, and EMBASE, including meta-analyses of randomized controlled trials that investigated the effects of exercise on HDL-C levels in middle-aged and older individuals. RESULTS Forty-two trials were analyzed. We found that exercise had a significant effect on HDL cholesterol levels (standard mean difference [95% confidence interval] = 0.22 [0.09-0.35], Z = 3.28, P = .001, I2 = 54%), and the heterogeneity across the studies was significant and high. After subgroup analysis, we found that aerobic exercise had the most significant effect on the levels of HDLs in middle-aged and older individuals, which was better than that of other types of exercise. CONCLUSIONS Aerobic exercise significantly improves HDL levels in middle-aged and older individuals, but resistance and stretching exercises had no significant effect on HDL levels.
Collapse
Affiliation(s)
- Lunmin Liang
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Fenglin Peng
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Linyuan Wang
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Chen Lin
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Hong Chi
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| |
Collapse
|
45
|
Liu Z, Huang N, Liu C, Wu C, Zhou L, Liu X, Lei H. Mitochondrial DNA in atherosclerosis research progress: a mini review. Front Immunol 2025; 16:1526390. [PMID: 39991161 PMCID: PMC11842404 DOI: 10.3389/fimmu.2025.1526390] [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: 11/11/2024] [Accepted: 01/23/2025] [Indexed: 02/25/2025] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease that primarily affects large and medium-sized arteries and is one of the leading causes of death worldwide. This article reviews the multifaceted role of mitochondrial DNA (mtDNA) in AS, including its structure, function, release, and relationship with inflammation. Damage and release of mtDNA are considered central drivers in the development of AS, as they participate in the progression of AS by activating inflammatory pathways and affecting lipid metabolism. Therefore, therapeutic strategies targeting mtDNA and its downstream effects may provide new avenues to address this global health challenge.
Collapse
Affiliation(s)
- Zheng Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China
| | - Nan Huang
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China
| | - Chan Liu
- Department of Clinical Pharmacy, Liuyang People’s Hospital, Liuyang, China
| | - Can Wu
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China
| | - Ling Zhou
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China
| | - Xiang Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China
| | - Haibo Lei
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China
| |
Collapse
|
46
|
Zeng C, Peng Z, Huang S, Xu Z, Peng Z, Wu Z, Lei J, Zhang X, Qin J, Ye K, Li B, Zhao Z, Pan Y, Yin M, Lu X. Metal-organic framework-based nanoplatforms for synergistic anti-atherosclerosis therapy by regulating the PI3K/AKT/MSR1 pathway in macrophages. NANOSCALE 2025; 17:3071-3085. [PMID: 39704073 DOI: 10.1039/d4nr04058a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Atherosclerosis is the main pathogenic factor of various cardiovascular diseases. During the pathogenesis of atherosclerosis, macrophages play a major role, mainly by secreting pro-inflammatory cytokines and taking in lipids to form foam cells. Thiamine pyrophosphate (TPP) is an antagonist of the P2Y6 receptor, which is overexpressed on macrophages during atherosclerosis and facilitates the lipid phagocytosis of macrophages. However, the excessive accumulation of TPP may interfere with some vital metabolic processes like the tricarboxylic acid cycle, oxidative phosphorylation and the pentose phosphate pathway. Herein, we designed and constructed a nanoparticle ZIF-8@TPP for the treatment of atherosclerosis. The as-established ZIF-8@TPP nanoplatform exhibited specific cytotoxicity towards macrophages in vitro. Meanwhile, histological analysis confirmed the excellent therapeutic efficacy of ZIF-8@TPP in vivo. Mechanistic studies indicated that ZIF-8@TPP potentially lowered lipid phagocytosis and lipid metabolism of macrophages via the PI3K/AKT/MSR1 pathway. This study also demonstrated that the anti-atherosclerotic effect of TPP was enhanced after combination with a prototypical metal-organic framework (MOF), ZIF-8. This synergistic controlled-release drug delivery system may provide a novel idea for anti-atherosclerosis therapy by combining reagents that can inhibit lipid phagocytosis of macrophages with MOFs.
Collapse
Affiliation(s)
- Chenlin Zeng
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Vascular Center of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Zhiyou Peng
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Vascular Center of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Sida Huang
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
| | - Zhijue Xu
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhaoxi Peng
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Vascular Center of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Zhaoyu Wu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Vascular Center of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Jiahao Lei
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Vascular Center of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Xing Zhang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Vascular Center of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Jinbao Qin
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Vascular Center of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Kaichuang Ye
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Vascular Center of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Bo Li
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Vascular Center of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Zhen Zhao
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Vascular Center of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Ying Pan
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
| | - Minyi Yin
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Vascular Center of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Xinwu Lu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Vascular Center of Shanghai Jiao Tong University, Shanghai, 200011, China
| |
Collapse
|
47
|
Liu S, Miao X, Hu M, Song Z, Xie X, Sun Y, Li M, Tang G, Leng S. Monocyte to High-Density Lipoprotein Ratio Is Associated With Carotid Plaque: A Retrospective Cohort Study. J Am Heart Assoc 2025; 14:e037210. [PMID: 39846278 PMCID: PMC12074780 DOI: 10.1161/jaha.124.037210] [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: 06/18/2024] [Accepted: 11/27/2024] [Indexed: 01/24/2025]
Abstract
BACKGROUND The level of monocyte to high-density lipoprotein ratio (MHR) is associated with cardiovascular diseases. Carotid plaque (CP) is an independent risk factor for cardiovascular diseases. However, evidence for association of MHR with risk of CP is scarce. METHODS AND RESULTS This study involved 5260 participants aged >18 years old from the Dalian health management cohort in 2014 to 2022. The subjects were stratified into 4 groups based on the quartile of the MHR at baseline. Multivariable-adjusted Cox regression models were used to calculate the MHR-associated risk of incident CP. The mean age of the population was 46.14 years and 58.8% (n=3093) of the participants were male. Seven hundred fifty-nine (14.4%) of participants developed new-onset CP. During the follow-up of 9725 person-years, the MHR at quartile 4 group experienced a significantly higher incidence of CP than the MHR at quartile 1 group (56.9 versus 101.5 per 1000 person-years; log-rank P <0.001). Compared with the MHR at quartile 1 group, the MHR at quartile 4 group had the highest CP risk (hazard ratio. 1.389 [95% CI, 1.059-1.823]) and 10-year cardiovascular risk (China-PAR Project score: odds ratio, 1.975 [95% CI, 1441-2.708 in men]; odds ratio, 6.015 [95% CI, 1.949-18.564 in women) (P <0.001). Meanwhile, similar results were observed in multiple sensitivity analyses. CONCLUSIONS Elevated MHR was associated with the risk of CP. The assessment and management of MHR is helpful for the early detection of patients with CP and the primary prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Shuang Liu
- Health Management Center of the Second Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
- School of Public HealthDalian Medical UniversityDalianLiaoningChina
| | - Xinlei Miao
- Health Management Center of the Second Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Manling Hu
- Department of Gastroenterology of the Second Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Ziping Song
- Department of Gastroenterology of the Second Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Xiaoling Xie
- Health Management Center of the Second Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
- School of Public HealthDalian Medical UniversityDalianLiaoningChina
| | - Yuting Sun
- Department of Gastroenterology of the Second Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Meng Li
- Health Management Center of the Second Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
- School of Public HealthDalian Medical UniversityDalianLiaoningChina
| | - Guimin Tang
- Health Management Center of the Second Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
- School of Public HealthDalian Medical UniversityDalianLiaoningChina
| | - Song Leng
- Health Management Center of the Second Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
- School of Public HealthDalian Medical UniversityDalianLiaoningChina
| |
Collapse
|
48
|
Kilic M, Karakoy Z, Halici H, Cadirci E, Halici Z. Novel therapeutic approaches targeting 5-HT7 receptors outside the central nervous system. J Recept Signal Transduct Res 2025; 45:55-60. [PMID: 39754608 DOI: 10.1080/10799893.2024.2446401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/06/2025]
Abstract
Serotonin (5-HT) is a neurotransmitter found throughout the human body that regulates many physiological events arising from the brain and central nervous system (CNS), such as sleep and appetite. However, it has many other functions in systems outside. In addition to the routine expression of 5-HT7 receptors in CNS regions, such as the pituitary gland, spinal cord, and hippocampus, many studies have reported the expression of these receptors in pathological conditions outside. The role of 5-HT7 receptors outside the CNS has been attracting increased attention in recent years. This review highlights the fact that 5-HT7 receptors are associated with diseases and systems beyond the CNS increasing or decreasing in response to cellular changes. Clinical, basic, in vivo and in vitro studies to date are described, but more research is needed to better understand the role of 5-HT7 receptors outside the CNS.
Collapse
Affiliation(s)
- Melis Kilic
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Zeynep Karakoy
- Department of Pharmacology, Faculty of Pharmacy, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Hamza Halici
- Department of Hınıs Vocational Training School, Ataturk University, Erzurum, Turkey
| | - Elif Cadirci
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Zekai Halici
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| |
Collapse
|
49
|
Zhao Y, Yang M, Liu Y, Wan Z, Chen M, He Q, Liao Y, Shuai P, Shi J, Guo S. Pathogenesis of cardiovascular diseases: effects of mitochondrial CF6 on endothelial cell function. Mol Cell Biochem 2025; 480:841-853. [PMID: 38985252 DOI: 10.1007/s11010-024-05065-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 06/29/2024] [Indexed: 07/11/2024]
Abstract
Cardiovascular disease (CVD) stands as a predominant global cause of morbidity and mortality, necessitating effective and cost-efficient therapies for cardiovascular risk reduction. Mitochondrial coupling factor 6 (CF6), identified as a novel proatherogenic peptide, emerges as a significant risk factor in endothelial dysfunction development, correlating with CVD severity. CF6 expression can be heightened by CVD risk factors like mechanical force, hypoxia, or high glucose stimuli through the NF-κB pathway. Many studies have explored the CF6-CVD relationship, revealing elevated plasma CF6 levels in essential hypertension, atherosclerotic cardiovascular disease (ASCVD), stroke, and preeclampsia patients. CF6 acts as a vasoactive and proatherogenic peptide in CVD, inducing intracellular acidosis in vascular endothelial cells, inhibiting nitric oxide (NO) and prostacyclin generation, increasing blood pressure, and producing proatherogenic molecules, significantly contributing to CVD development. CF6 induces an imbalance in endothelium-dependent factors, including NO, prostacyclin, and asymmetric dimethylarginine (ADMA), promoting vasoconstriction, vascular remodeling, thrombosis, and insulin resistance, possibly via C-src Ca2+ and PRMT-1/DDAH-2-ADMA-NO pathways. This review offers a comprehensive exploration of CF6 in the context of CVD, providing mechanistic insights into its role in processes impacting CVD, with a focus on CF6 functions, intracellular signaling, and regulatory mechanisms in vascular endothelial cells.
Collapse
Affiliation(s)
- Yingying Zhao
- Department of Geriatric Medicine, School of Medicine and Life Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ming Yang
- The Lab of Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Youren Liu
- Department of Health Management Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Zhengwei Wan
- Department of Health Management Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Mengchun Chen
- Department of Geriatric Medicine, School of Medicine and Life Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiumei He
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yang Liao
- Department of Geriatric Medicine, School of Medicine and Life Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ping Shuai
- Department of Health Management Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Shujin Guo
- Department of Health Management Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
50
|
Zhang N, Zhang T, Feng J, Shang J, Zhang B, Dong Q, Zhang Z, Sun C. Application of therapeutical nanoparticles with neutrophil membrane camouflaging for inflammatory plaques targeting against atherosclerosis. Mater Today Bio 2025; 30:101397. [PMID: 39802828 PMCID: PMC11722182 DOI: 10.1016/j.mtbio.2024.101397] [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: 08/22/2024] [Revised: 11/30/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
Atherosclerosis is the leading cause of cardiovascular disease and myocardial infarction. Precise and effective plaque targeting is a major objective for therapeutic outcomes throughout various stages of atherosclerosis. Inspired by the natural recruitment of neutrophils in atherosclerotic plaques, we fabricated a simvastatin (ST)-loaded and neutrophil membrane-cloaked nanoplatform (NNPST) for enhancing localized payload delivery and atherosclerosis management. The resulting NNPST mimicked neutrophil function and significantly decreased macrophage-mediated phagocytosis to prolong its own circulation time in the blood. Compared to pristine nanoparticles (NPST) without a membrane coating, NNPST achieved better plaque targeting in ApoE-/- mice, as indicated by neutrophils actively recruited in atherosclerotic lesions. The higher plaque homing with NNPST was monitored by dynamic fluorescence/magnetic resonance (MR) dual-modality imaging. The results further showed that NNPST efficiently prevented atherosclerosis development mainly by suppressing local inflammatory macrophages, and the percentage of plaques in the entire aortic area was reduced to 4.75 ± 1.48 % following NNPST treatment. A biosafety assessment indicated that the biomimetic NNPST induced no noticeable toxicity in the body. This approach of neutrophil membrane-camouflaged nanoparticles offers new opportunities to various therapeutic agents for on-demand delivery in neutrophil-involved inflammatory diseases.
Collapse
Affiliation(s)
- Ningnannan Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Tianzhu Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Jintang Feng
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Jian Shang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Beibei Zhang
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450002, PR China
| | - Qingyang Dong
- Department of Environmental Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, PR China
| | - Zhang Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Chunyang Sun
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| |
Collapse
|