1
|
Huang X, Sun T, Zhang B, Ma M, Chen Z, Zhao Z, Dong S, Zhou Y. Prognostic value of remnant-like particle cholesterol in ischemic heart failure patients following percutaneous coronary intervention. Ann Med 2025; 57:2458200. [PMID: 39898975 PMCID: PMC11792126 DOI: 10.1080/07853890.2025.2458200] [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: 08/23/2023] [Revised: 09/20/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND The relationship between remnant-like particle cholesterol (RLP-C) and cardiovascular disease risk and prognosis has been established, but its effect on the prognosis of ischemic heart failure (IHF) patients undergoing percutaneous coronary intervention (PCI) remains uncertain. METHOD In this study, 2036 patients with IHF who underwent PCI were included. Patients were categorized into tertiles based on their RLP-C levels. The primary outcome was major adverse cardiovascular events (MACE). Kaplan-Meier survival analysis was used to assess the incidence of MACE and other outcomes. Multivariate Cox regression models were employed to investigate the correlation between RLP-C and the studied outcomes. The nonlinear relationship between RLP-C and MACE was examined through the restricted cubic spline (RCS). Subgroup analyses were performed and interactions were assessed. RESULT The study results showed a clear association between higher RLP-C levels and an increased incidence of MACE in the participants. This association was validated by Kaplan-Meier analyses. The multivariate Cox regression demonstrated RLP-C was an independent risk factor for MACE, whether assessed as a continuous variable[hazard ratio (HR), 95% confidence interval (CI): 1.50, 1.15-1.98, p = 0.003] or categorized into tertiles[HR, 95% CI: 2.57, 2.03-3.26, p < 0.001, tertile 3 vs tertile 1]. A nonlinear relationship between RLP-C and MACE was observed, indicating that the risk of MACE increased with higher RLP-C levels(Nonlinear p < 0.001). This association remained consistent across various subgroups, as no significant interactions were found. CONCLUSION There was an independent and positive correlation between RLP-C and MACE in patients with IHF who underwent PCI.
Collapse
Affiliation(s)
- Xin Huang
- Department of Cardiology, Capital Medical University Affiliated Anzhen Hospital, Beijing, China
| | - Tienan Sun
- Department of Cardiology, Capital Medical University Affiliated Anzhen Hospital, Beijing, China
| | - Biyang Zhang
- Department of Cardiology, Capital Medical University Affiliated Anzhen Hospital, Beijing, China
| | - Meishi Ma
- Department of Cardiology, Capital Medical University Affiliated Anzhen Hospital, Beijing, China
| | - Zheng Chen
- Department of Cardiology, Capital Medical University Affiliated Anzhen Hospital, Beijing, China
| | - Zehao Zhao
- Department of Cardiology, Capital Medical University Affiliated Anzhen Hospital, Beijing, China
| | - Shutong Dong
- Department of Cardiology, Capital Medical University Affiliated Anzhen Hospital, Beijing, China
| | - Yujie Zhou
- Department of Cardiology, Capital Medical University Affiliated Anzhen Hospital, Beijing, 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
|
Xiong X, Lee HC, Lu T. Impact of Sorbs2 dysfunction on cardiovascular diseases. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167813. [PMID: 40139410 PMCID: PMC12037213 DOI: 10.1016/j.bbadis.2025.167813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
Despite significant advancements in prevention and treatment over the past decades, cardiovascular diseases (CVDs) remain the leading cause of death worldwide. CVDs involve multifactorial inheritance, but our understanding of the genetic impact on these diseases is still incomplete. Sorbin and SH3 domain-containing protein 2 (Sorbs2) is ubiquitously expressed in various tissues, including the cardiovascular system. Increasing evidence suggests that Sorbs2 malfunction contributes to CVDs. This manuscript will review our current understanding of the potential mechanisms underlying Sorbs2 dysregulation in the development of CVDs.
Collapse
Affiliation(s)
- Xiaowei Xiong
- The Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Hon-Chi Lee
- The Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Tong Lu
- The Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States of America.
| |
Collapse
|
4
|
Huntermann R, Peres de Oliveira J, Barbosa LM, Queiroz I, Nunes Cavalcante D, de Oliveira Fischer Bacca C. Colchicine in acute coronary syndromes: a systematic review and meta-analysis of randomised controlled trials. Heart 2025:heartjnl-2025-325826. [PMID: 40379469 DOI: 10.1136/heartjnl-2025-325826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/27/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND Acute coronary syndrome (ACS) is a global leading cause of morbidity, with residual inflammation contributing to recurrent events. Colchicine has been proposed as an adjunct therapy, but its efficacy remains uncertain. METHODS We performed a systematic review and meta-analysis. PubMed, Embase and Cochrane databases were searched for randomised controlled trials (RCTs) data comparing colchicine versus placebo in ACS. Risk ratio (RR) and mean difference with 95% CIs were computed for binary and continuous outcomes, respectively. Primary outcomes were adverse cardiovascular events (ACEs), mortality and safety. Random-effects models were used for pooled estimates. RESULTS Seventeen RCTs comprising 14 794 patients were included, of whom 7390 (50%) were randomised to colchicine. The mean patient age across the studies ranged from 54 to 63 years, in a follow-up period ranging from 5 days to 12 months. Colchicine reduced the incidence of recurrent ACS (RR 0.41, 95% CI 0.19 to 0.92; p=0.03; I²=55%) and unstable angina (RR 0.27, 95% CI 0.11 to 0.63; p<0.01; I²=0%). No meaningful differences were observed in all-cause mortality (RR 0.95, 95% CI 0.79 to 1.14; I²=12%), cardiovascular death (RR 1.03, 95% CI 0.82 to 1.30; I²=0%) or ACE (RR 0.77, 95% CI 0.59 to 1.01; p=0.05; I²=58%). Subgroup analyses suggested a dose-dependent effect, with 0.5 mg/day potentially reducing ACE (RR 0.63, 95% CI 0.45 to 0.88; I²=41%), but higher doses increasing gastrointestinal symptoms. CONCLUSION Low-dose colchicine may reduce recurrent ischaemic events in ACS, but evidence remains uncertain due to heterogeneity and limited long-term data. Safety and efficacy in women and optimal dosing require further investigation. TRIAL REGISTRATION NUMBER CRD42024627348.
Collapse
Affiliation(s)
- Ramon Huntermann
- Medical Sciences Research Center, University Center for the Development of Alto Vale, UNIDAVI, Rio do Sul, Brazil
| | - Juan Peres de Oliveira
- Medical Sciences Research Center, University Center for the Development of Alto Vale, UNIDAVI, Rio do Sul, Brazil
| | - Lucas M Barbosa
- Department of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Ivo Queiroz
- Radiology Department, University of Wisconsin-Madison, Madison, Winsconsin, USA
| | | | | |
Collapse
|
5
|
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
|
6
|
Xu J, Wang H, Han B, Zhang X. Mechanisms through which laparoscopic sleeve gastrectomy mitigates atherosclerosis risk: a focus on visceral adipose tissue. Eur J Med Res 2025; 30:370. [PMID: 40336107 PMCID: PMC12057030 DOI: 10.1186/s40001-025-02635-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: 11/28/2024] [Accepted: 04/23/2025] [Indexed: 05/09/2025] Open
Abstract
Bariatric surgery is currently considered the key treatment method for patients with obesity and related complications. Among the various surgeries, laparoscopic sleeve gastrectomy (LSG) is the most widely used. Obesity is a multifactor chronic disease characterized by the accumulation of visceral adipose tissue (VAT), leading to susceptibility to cardiac metabolic diseases. Many mechanisms, including abnormal lipid metabolism, insulin resistance, inflammation, endothelial dysfunction, adipocytokine imbalance and inflammasome activation, have been identified as the basis for the relationship between obesity and atherosclerosis. Bariatric surgery, such as LSG, reduces the risk of atherosclerosis in people living with obesity by reducing energy intake, disrupting energy balance and reducing the secretion of intestinal hormones to intervene in these risk factors. This review explores the current understanding of how LSG affects VAT and its impact on the risk of atherosclerosis.
Collapse
Affiliation(s)
- Juan Xu
- General Surgery Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| | - Heyue Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Bin Han
- Shanxi Provincial People's Hospital, Taiyuan, 030032, China
| | - Xiaomin Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| |
Collapse
|
7
|
Xing Y, Lin X. Challenges and advances in the management of inflammation in atherosclerosis. J Adv Res 2025; 71:317-335. [PMID: 38909884 DOI: 10.1016/j.jare.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 06/25/2024] Open
Abstract
INTRODUCTION Atherosclerosis, traditionally considered a lipid-related disease, is now understood as a chronic inflammatory condition with significant global health implications. OBJECTIVES This review aims to delve into the complex interactions among immune cells, cytokines, and the inflammatory cascade in atherosclerosis, shedding light on how these elements influence both the initiation and progression of the disease. METHODS This review draws on recent clinical research to elucidate the roles of key immune cells, macrophages, T cells, endothelial cells, and clonal hematopoiesis in atherosclerosis development. It focuses on how these cells and process contribute to disease initiation and progression, particularly through inflammation-driven processes that lead to plaque formation and stabilization. Macrophages ingest oxidized low-density lipoprotein (oxLDL), which partially converts to high-density lipoprotein (HDL) or accumulates as lipid droplets, forming foam cells crucial for plaque stability. Additionally, macrophages exhibit diverse phenotypes within plaques, with pro-inflammatory types predominating and others specializing in debris clearance at rupture sites. The involvement of CD4+ T and CD8+ T cells in these processes promotes inflammatory macrophage states, suppresses vascular smooth muscle cell proliferation, and enhances plaque instability. RESULTS The nuanced roles of macrophages, T cells, and the related immune cells within the atherosclerotic microenvironment are explored, revealing insights into the cellular and molecular pathways that fuel inflammation. This review also addresses recent advancements in imaging and biomarker technology that enhance our understanding of disease progression. Moreover, it points out the limitations of current treatment and highlights the potential of emerging anti-inflammatory strategies, including clinical trials for agents such as p38MAPK, tumor necrosis factor α (TNF-α), and IL-1β, their preliminary outcomes, and the promising effects of canakinumab, colchicine, and IL-6R antagonists. CONCLUSION This review explores cutting-edge anti-inflammatory interventions, their potential efficacy in preventing and alleviating atherosclerosis, and the role of nanotechnology in delivering drugs more effectively and safely.
Collapse
Affiliation(s)
- Yiming Xing
- Cardiology Department, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, 230022, China
| | - Xianhe Lin
- Cardiology Department, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, 230022, China.
| |
Collapse
|
8
|
Zheng Q, Wang T, Wang S, Chen Z, Jia X, Yang H, Chen H, Sun X, Wang K, Zhang L, Fu F. The anti-inflammatory effects of saponins from natural herbs. Pharmacol Ther 2025; 269:108827. [PMID: 40015518 DOI: 10.1016/j.pharmthera.2025.108827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 11/20/2024] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
Inflammation is a protective mechanism that also starts the healing process. However, inflammatory reaction may cause severe tissue damage. The increased influx of phagocytic leukocytes may produce excessive amount of reactive oxygen species, which leads to additional cell injury. Inflammatory response activates the leukocytes and thus induces tissue damage and prolongs inflammation. The inflammation-induced activation of the complement system may also contribute to cell injury. Non-steroidal anti-inflammatory drugs (NSAIDs) and glucocorticoids are chief agents for treating inflammation associated with the diseases. However, the unwanted side effects of NSAIDs (e.g., gastrointestinal disturbances, skin reactions, adverse renal effects, cardiovascular side effects) and glucocorticoids (e.g., suppression of immune system, Cushing's syndrome, osteoporosis, hyperglycemia) limit their use in patients. Natural herbs are important sources of anti-inflammatory drugs. The ingredients extracted from natural herbs display anti-inflammatory effects to work through multiple pathways with lower risk of adverse reaction. At present, the main anti-inflammatory natural agents include saponins, flavonoids, alkaloids, polysaccharides, and so on. The present article will review the anti-inflammatory effects of saponins including escin, ginsenosides, glycyrrhizin, astragaloside, Panax notoginseng saponins, saikosaponin, platycodin, timosaponin, ophiopogonin D, dioscin, senegenin.
Collapse
Affiliation(s)
- Qinpin Zheng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| | - Tian Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| | - Sensen Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| | - Zhuoxi Chen
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Xue Jia
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| | - Hui Yang
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Huijin Chen
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| | - Xin Sun
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Kejun Wang
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Leiming Zhang
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, China.
| | - Fenghua Fu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China.
| |
Collapse
|
9
|
Maguire C, Soloveichik E, Blinchevsky N, Miller J, Morrison R, Busch J, Michael Brode W, Wylie D, Rousseau J, Melamed E. Dissecting clinical features of COVID-19 in a cohort of 21,312 acute care patients. COMMUNICATIONS MEDICINE 2025; 5:138. [PMID: 40281203 PMCID: PMC12032146 DOI: 10.1038/s43856-025-00844-4] [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: 03/15/2024] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Although, COVID-19 has resulted in over 7 million deaths globally, many questions still remain about the risk factors for disease severity and the effects of variants and vaccinations over the course of the pandemic. To address this gap, we conducted a retrospective analysis of electronic health records from COVID-19 patients over 2.5 years of the COVID-19 pandemic to identify associated clinical features. METHODS We analyze a retrospective cohort of 21,312 acute-care patients over a 2.5 year period and define six clinical trajectory groups (TGs) associated with demographics, diagnoses, vitals, labs, imaging, consultations, and medications. RESULTS We show that the proportion of mild patients increased over time, particularly during Omicron waves. Additionally, while mild and fatal patients had differences in age, age did not distinguish patients with severe versus critical disease. Furthermore, we find that both male sex and Hispanic/Latino ethnicity are associated with more severe/critical TGs. More severe patients also have a higher rate of neuropsychiatric diagnoses and consultations, along with an immunological signature of high neutrophils and immature granulocytes, and low lymphocytes and monocytes. Interestingly, low albumin is one of the best lab predictors of COVID-19 severity in association with higher malnutrition in severe/critical patients, raising concern of nutritional insufficiency influencing COVID-19 outcomes. Despite this, only a small fraction of severe/critical patients had nutritional labs checked (e.g. Vitamin D, thiamine, B vitamins) or received vitamin supplementation. CONCLUSIONS Our findings expand on clinical risk factors in COVID-19, and highlight the interaction between severity, nutritional status, and neuropsychiatric complications in acute care patients to enable identification of patients at risk for severe disease.
Collapse
Affiliation(s)
- Cole Maguire
- Department of Neurology, The University of Texas at, Austin, Dell Medical School, Austin, TX, USA
| | - Elie Soloveichik
- Department of Neurology, The University of Texas at, Austin, Dell Medical School, Austin, TX, USA
| | - Netta Blinchevsky
- Department of Neurology, The University of Texas at, Austin, Dell Medical School, Austin, TX, USA
| | - Jaimie Miller
- Enterprise Data Intelligence, The University of Texas at Austin, Dell Medical School, Austin, TX, USA
| | - Robert Morrison
- Department of Internal Medicine, The University of Texas at Austin, Dell Medical School, Austin, TX, USA
| | - Johanna Busch
- Department of Internal Medicine, The University of Texas at Austin, Dell Medical School, Austin, TX, USA
| | - W Michael Brode
- Department of Internal Medicine, The University of Texas at Austin, Dell Medical School, Austin, TX, USA
| | - Dennis Wylie
- Center for Biomedical Support, The University of Texas at Austin, Austin, TX, USA
| | - Justin Rousseau
- Department of Neurology, The University of Texas at, Austin, Dell Medical School, Austin, TX, USA
- Biostatistics and Clinical Informatics Section, Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Esther Melamed
- Department of Neurology, The University of Texas at, Austin, Dell Medical School, Austin, TX, USA.
| |
Collapse
|
10
|
Grego A, Fernandes C, Fonseca I, Dias-Neto M, Costa R, Leite-Moreira A, Oliveira SM, Trindade F, Nogueira-Ferreira R. Endothelial dysfunction in cardiovascular diseases: mechanisms and in vitro models. Mol Cell Biochem 2025:10.1007/s11010-025-05289-w. [PMID: 40259179 DOI: 10.1007/s11010-025-05289-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 04/08/2025] [Indexed: 04/23/2025]
Abstract
Endothelial cells (ECs) are arranged side-by-side to create a semi-permeable monolayer, forming the inner lining of every blood vessel (micro and macrocirculation). Serving as the first barrier for circulating molecules and cells, ECs represent the main regulators of vascular homeostasis being able to respond to environmental changes, either physical or chemical signals, by producing several factors that regulate vascular tone and cellular adhesion. Healthy endothelium has anticoagulant properties that prevent the adhesion of leukocytes and platelets to the vessel walls, contributing to resistance to thrombus formation, and regulating inflammation, and vascular smooth muscle cell proliferation. Many risk factors of cardiovascular diseases (CVDs) promote the endothelial expression of chemokines, cytokines, and adhesion molecules. The resultant endothelial activation can lead to endothelial cell dysfunction (ECD). In vitro models of ECD allow the study of cellular and molecular mechanisms of disease and provide a research platform for screening potential therapeutic agents. Even though alternative models are available, such as animal models or ex vivo models, in vitro models offer higher experimental flexibility and reproducibility, making them a valuable tool for the understanding of pathophysiological mechanisms of several diseases, such as CVDs. Therefore, this review aims to synthesize the currently available in vitro models regarding ECD, emphasizing CVDs. This work will focus on 2D cell culture models (endothelial cell lines and primary ECs), 3D cell culture systems (scaffold-free and scaffold-based), and 3D cell culture models (such as organ-on-a-chip). We will dissect the role of external stimuli-chemical and mechanical-in triggering ECD.
Collapse
Affiliation(s)
- Ana Grego
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Cristiana Fernandes
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Ivo Fonseca
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Marina Dias-Neto
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- Department of Angiology and Vascular Surgery, Unidade Local de Saúde de São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Raquel Costa
- Universidade Católica Portuguesa, CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005, Porto, Portugal
| | - Adelino Leite-Moreira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- Department of Cardiothoracic Surgery, Unidade Local de Saúde de São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Sandra Marisa Oliveira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Fábio Trindade
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Rita Nogueira-Ferreira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.
| |
Collapse
|
11
|
Borgas Y, Mohammad MA, Gisslander K, Rathmann J, Erlinge D, Jayne D, Mohammad AJ. Myocardial infarction in ANCA-associated vasculitis: a population-based cohort study. RMD Open 2025; 11:e005055. [PMID: 40250881 PMCID: PMC12007036 DOI: 10.1136/rmdopen-2024-005055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 03/24/2025] [Indexed: 04/20/2025] Open
Abstract
OBJECTIVES To determine the incidence rate (IR) and predictors of myocardial infarction (MI) in patients with antineutrophil cytoplasmic autoantibody (ANCA)-associated vasculitis (AAV) as well as to estimate the IR ratio (IRR) of MI in AAV versus the background population. METHODS 325 patients diagnosed with AAV 1997-2016 in Skåne, Sweden were included. Data were collected from the time of AAV diagnosis, and each patient was grouped with 10 age-matched and sex-matched reference subjects from the background population. MI after AAV diagnosis was identified using Swedish Web-System for Enhancement and Development of Evidence-based Care in Heart Disease Evaluated According to Recommended Therapies and the Skåne Healthcare Register, and IR of first MI calculated. The IRR was computed by dividing the IR for 282 AAV patients by the corresponding rate in the 2763 reference subjects. Predictors of MI were analysed using Cox regression. RESULTS 37 patients (11%) with AAV suffered an initial MI, yielding an IR of 1.6/100 person-years of follow-up (95% CI 1.2 to 2.2). The highest rate was recorded in the 3 months following AAV diagnosis, at 11.8/100 person-years (95% CI 6.2 to 22.7). The IRR of MI in AAV/reference was 1.9 (95% CI 1.3 to 2.8), highest in patients with myeloperoxidase-ANCA+disease (IRR 2.5, 95% CI 1.5 to 4.3) and those with high disease activity at diagnosis (2.1, 95% CI 1.3 to 3.3). Age at AAV diagnosis independently predicted MI. CONCLUSIONS The MI IR is greater in individuals diagnosed with AAV compared with background population, especially those with more severe disease, and highest in the 3 months following diagnosis. Age at diagnosis is the single independent predictor of MI in AAV in this study.
Collapse
Affiliation(s)
- Ylva Borgas
- Department of Rheumatology, Skåne University Hospital, Malmo, Sweden
- Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden
| | - Moman Aladdin Mohammad
- Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden
- Department of Cardiology, Skåne University Hospital, Lund, Sweden
| | - Karl Gisslander
- Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden
- Department of Rheumatology, Skåne University Hospital, Lund, Sweden
| | - Jens Rathmann
- Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden
- Department of Rheumatology, Skåne University Hospital, Lund, Sweden
| | - David Erlinge
- Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden
- Department of Cardiology, Skåne University Hospital, Lund, Sweden
| | - David Jayne
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Aladdin J Mohammad
- Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden
- Department of Rheumatology, Skåne University Hospital, Lund, Sweden
- Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
12
|
Georgescu CM, Butnariu I, Cojocea CR, Tiron AT, Anghel DN, Mitrică IAM, Lăptoiu VI, Bidea A, Antonescu-Ghelmez D, Tuță S, Antonescu F. Subacute Cardiomyopathy Due to Statin Treatment: Can It Be True?-Case Report and Literature Review. Life (Basel) 2025; 15:630. [PMID: 40283184 PMCID: PMC12028598 DOI: 10.3390/life15040630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Background and Clinical Significance: Statins are a widely used drug class associated with a plethora of muscular side effects ranging from the subclinical elevation of creatine kinase to fulminant rhabdomyolysis. Cardiac myopathy secondary to statin treatment is rare and was recently reported as a part of statin-induced necrotizing autoimmune myopathy (SINAM). Its occurrence outside of this context is still debated. Case Presentation: We present the case of a 60-year-old male who developed atorvastatin-induced rhabdomyolysis, without associated hydroxymethyl glutaryl coenzyme A reductase (HMGCR) antibodies, with clinical findings of cardiac failure and severe ECG anomalies. The symptoms slowly regressed with statin withdrawal, and the patient made a full recovery. We discuss the recently proposed statin-associated cardiomyopathy (SACM) and the possible mechanisms. We compare our case to the three other cases of statin-induced cardiac myositis found in the literature. Conclusions: We believe that in vulnerable patients, as was our case, statins can determine significant subacute cardiac toxicity. This would seem to occur in the context of severe skeletal muscle injury, probably due to higher metabolic resistance on the part of the myocardium. Also, the available evidence suggests myocardial involvement should be actively investigated in SINAM patients, preferably by cardiac MRI.
Collapse
Affiliation(s)
- Camelia Mihaela Georgescu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050471 Bucharest, Romania
- National Institute of Neurology and Neurovascular Diseases, “Carol Davila” University of Medicine and Pharmacy, 041902 Bucharest, Romania
| | - Ioana Butnariu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050471 Bucharest, Romania
- Department of Neurology, National Institute of Neurology and Neurovascular Diseases, “Carol Davila” University of Medicine and Pharmacy, 041902 Bucharest, Romania
| | - Cătălina Raluca Cojocea
- Department of Cardiology, “Sf. Ioan” Emergency Clinical Hospital, “Carol Davila” University of Medicine and Pharmacy, 041902 Bucharest, Romania
| | - Andreea Taisia Tiron
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050471 Bucharest, Romania
- Department of Cardiology, “Sf. Ioan” Emergency Clinical Hospital, “Carol Davila” University of Medicine and Pharmacy, 041902 Bucharest, Romania
| | - Daniela-Nicoleta Anghel
- Department of Neurology, National Institute of Neurology and Neurovascular Diseases, “Carol Davila” University of Medicine and Pharmacy, 041902 Bucharest, Romania
| | - Iulia Ana-Maria Mitrică
- Department of Neurology, National Institute of Neurology and Neurovascular Diseases, “Carol Davila” University of Medicine and Pharmacy, 041902 Bucharest, Romania
| | - Vlad-Iulian Lăptoiu
- Department of Neurology, National Institute of Neurology and Neurovascular Diseases, “Carol Davila” University of Medicine and Pharmacy, 041902 Bucharest, Romania
| | - Adriana Bidea
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050471 Bucharest, Romania
- Department of Neurology, National Institute of Neurology and Neurovascular Diseases, “Carol Davila” University of Medicine and Pharmacy, 041902 Bucharest, Romania
| | - Dana Antonescu-Ghelmez
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050471 Bucharest, Romania
- Department of Neurology, National Institute of Neurology and Neurovascular Diseases, “Carol Davila” University of Medicine and Pharmacy, 041902 Bucharest, Romania
| | - Sorin Tuță
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050471 Bucharest, Romania
- Department of Neurology, National Institute of Neurology and Neurovascular Diseases, “Carol Davila” University of Medicine and Pharmacy, 041902 Bucharest, Romania
| | - Florian Antonescu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050471 Bucharest, Romania
- Department of Neurology, National Institute of Neurology and Neurovascular Diseases, “Carol Davila” University of Medicine and Pharmacy, 041902 Bucharest, Romania
| |
Collapse
|
13
|
You SH, Cho SK, Kim JY, Song YJ, Jung SY, Sung YK. Risk of major adverse cardiovascular events following targeted therapy in patients with rheumatoid arthritis: a real-world analysis stratified by cardiovascular risk. Semin Arthritis Rheum 2025; 73:152721. [PMID: 40267667 DOI: 10.1016/j.semarthrit.2025.152721] [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: 10/11/2024] [Revised: 02/23/2025] [Accepted: 03/31/2025] [Indexed: 04/25/2025]
Abstract
OBJECTIVE To assess the risk of major adverse cardiovascular events (MACEs) associated with Janus kinase inhibitors (JAKi) compared to tumour necrosis factor inhibitors (TNFi) in rheumatoid arthritis (RA). METHODS Using the Korean nationwide claims database, we identified patients with RA prescribed JAKi or TNFi between 2015 and 2019. Patients were stratified into two groups based on cardiovascular (CV) risk and matched within each group using propensity score matching at a ratio of up to 1:4. Follow-up continued until MACE, death, or treatment discontinuation. Hazard ratios with 95 % confidence intervals were calculated using the Cox proportional hazards model. Additionally, MACE risk was analyzed separately in patients with and without a history of cardiovascular disease (Hx.CVD). RESULTS A total of 7575 patients prescribed either JAKi or TNFi were included. After propensity score matching, the hazard ratio for MACEs comparing JAKi to TNFi was 0.77 (95 % confidence interval 0.45-1.34) in the high CV risk group. No significant differences in MACEs were observed between JAKi and TNFi users across the low CV risk group, as well as Hx.CVD and non-Hx.CVD groups. Subgroup and sensitivity analyses showed no statistically significant differences in the risk of individual MACE components, such as myocardial infarction, stroke, or heart failure. CONCLUSION No significant differences in the risk of MACEs were observed between JAKi and TNFi users across various CV risk groups and in those with or without Hx.CVD. Subgroup and sensitivity analyses supported these findings, showing no elevated risks for individual MACE components.
Collapse
Affiliation(s)
- Seung-Hun You
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Soo-Kyung Cho
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, 222-1 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea; Hanyang University Institute for Rheumatology Research, 222-1 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Jeong-Yeon Kim
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Yeo-Jin Song
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, 222-1 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea; Hanyang University Institute for Rheumatology Research, 222-1 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Sun-Young Jung
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea.
| | - Yoon-Kyoung Sung
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, 222-1 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea; Hanyang University Institute for Rheumatology Research, 222-1 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea.
| |
Collapse
|
14
|
Xu S, Chuang CY, Hawkins CL, Hägglund P, Davies MJ. Quantitative analysis of the proteome and protein oxidative modifications in primary human coronary artery endothelial cells and associated extracellular matrix. Redox Biol 2025; 81:103524. [PMID: 39954365 PMCID: PMC11875191 DOI: 10.1016/j.redox.2025.103524] [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: 12/05/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/17/2025] Open
Abstract
Vascular endothelial cells (ECs) play a key role in physiology by controlling arterial contraction and relaxation, and molecular transport. EC dysfunction is associated with multiple pathologies. Here, we characterize the cellular and extracellular matrix (ECM) proteomes of primary human coronary artery ECs, from multiple donors, and oxidation/nitration products formed on these during cell culture, using liquid chromatography-mass spectrometry. In total ∼9900 proteins were identified in cells from 3 donors, with ∼7000 proteins per donor. Of these ∼5300 were consistently identified, indicating some heterogeneity across the donors, with age a possible cause. Multiple endogenous oxidation products were detected on both ECM and cellular proteins (and particularly endoplasmic reticulum species). In contrast, nitration was mostly detected on cell proteins and particularly cytoskeletal proteins, consistent with intracellular generation of nitrating agents, possibly from endothelial nitric oxide synthase (eNOS) or peroxidase enzymes. The modifications are ascribed to both physiological enzymatic activity (hydroxylation at proline/lysine; predominantly on ECM proteins and especially collagens) and the formation of reactive species (oxidation at tryptophan/tyrosine/histidine; nitration at tryptophan/tyrosine). The identified sites are present on a limited number of peptides (104 oxidized; 23 nitrated) from a modest number of proteins. A small number of proteins were detected with multiple modifications, consistent with these being selective and specific targets. Several nitrated peptides were consistently detected across all donors, and also in human smooth muscle cells suggesting that these are major targets in the vascular proteome. These data provide a 'background' proteome dataset for studies of endothelial dysfunction in disease.
Collapse
Affiliation(s)
- Shuqi Xu
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark; Department of Cardiovascular Medicine, The Affiliated Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Christine Y Chuang
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark
| | - Clare L Hawkins
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark
| | - Per Hägglund
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark.
| | - Michael J Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark.
| |
Collapse
|
15
|
Baidoo I, Sarbadhikary P, Abrahamse H, George BP. Metal-based nanoplatforms for enhancing the biomedical applications of berberine: current progress and future directions. Nanomedicine (Lond) 2025; 20:851-868. [PMID: 40110809 PMCID: PMC11999359 DOI: 10.1080/17435889.2025.2480051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
The isoquinoline alkaloid berberine, a bioactive compound derived from various plants, has demonstrated extensive therapeutic potential. However, its clinical application is hindered by poor water solubility, low bioavailability, rapid metabolism, and insufficient targeting. Metal-based nanoplatforms offer promising solutions, enhancing drug stability, controlled release, and targeted delivery. This review comprehensively explores the synthesis, physicochemical properties, and biomedical applications of metal-based nanocarriers, including gold, silver, iron oxide, zinc oxide, selenium, and magnetic nanoparticles, for berberine delivery to improve berberine's therapeutic efficacy. Recent advancements in metal-based nanocarrier systems have significantly improved berberine delivery by enhancing cellular uptake, extending circulation time, and enabling site-specific targeting. However, metal-based nanoplatforms encounter several limitations of potential toxicity, limited large-scale productions, and regulatory constraints. Addressing these limitations necessitates extensive studies on biocompatibility, long-term safety, and clinical translation. By summarizing the latest innovations and clinical perspectives, this review aims to guide future research toward optimizing berberine-based nanomedicine for improved therapeutic efficacy.
Collapse
Affiliation(s)
- Isaac Baidoo
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Paromita Sarbadhikary
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Blassan P. George
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|
16
|
Hossain R, Kongchain A, Chatatikun M, Klangbud WK, Yupanqui CT, Majima HJ, Indo HP, Sompol P, Sekeroglu N, Phongphithakchai A, Tangpong J. Green Tea Pressurized Hot Water Extract in Atherosclerosis: A Multi-Approach Study on Cellular, Animal, and Molecular Mechanisms. Antioxidants (Basel) 2025; 14:404. [PMID: 40298660 PMCID: PMC12024429 DOI: 10.3390/antiox14040404] [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: 02/21/2025] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/30/2025] Open
Abstract
Atherosclerosis is a persistent inflammatory disorder influenced by oxidative stress and lipid imbalances, and it continues to be a major contributor to cardiovascular diseases. Rich in catechins and flavonoids, green tea pressurized hot water extract (GPHWE) demonstrated potent antioxidant activity through DPPH, ABTS, hydroxyl, and nitric oxide scavenging assays. In vitro, GPHWE protected RAW264.7 macrophages from oxidized LDL (Ox-LDL)-induced cytotoxicity and apoptosis by mitigating oxidative stress and enhancing cell survival. Animal studies using mice fed a high-fat diet (HFD) revealed notable improvements in lipid profiles, including decreases in total cholesterol, LDL, the atherosclerosis index (AI), the coronary risk index (CRI), and triglycerides, as well as lower levels of malondialdehyde (MDA), an indicator of oxidative stress. These results were comparable to those achieved with Simvastatin. Molecular docking studies indicated strong binding affinities of catechins to essential targets such as LOX-1, HMG-CoA reductase, caspase-3, and Nrf2, implying that the mechanisms of GPHWE involve antioxidant properties, regulation of lipids, and stabilization of plaques. The catechins of GPHWE, including epigallocatechin gallate (EGCG), epicatechin gallate (ECG), and epigallocatechin (EGC), were tentatively identified through qualitative analysis performed by UHPLC-QTOF-MS. This comprehensive approach positions GPHWE as a promising natural remedy for preventing atherosclerosis and reducing cardiovascular risk.
Collapse
Affiliation(s)
- Rahni Hossain
- College of Graduate Studies, Walailak University, Nakhon Si Thammarat 80160, Thailand;
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand; (A.K.); (M.C.); (H.J.M.)
- Research Excellence Center for Innovation and Health Products (RECIHP), School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Anawat Kongchain
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand; (A.K.); (M.C.); (H.J.M.)
| | - Moragot Chatatikun
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand; (A.K.); (M.C.); (H.J.M.)
- Research Excellence Center for Innovation and Health Products (RECIHP), School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Wiyada Kwanhian Klangbud
- Medical Technology Department, Faculty of Science, Nakhon Phanom University, Nakhon Phanom 48000, Thailand;
| | - Chutha Takahashi Yupanqui
- Center of Excellence in Functional Foods and Gastronomy, Faculty of Agro-Industry, Prince of Songkla University, Songkhla 90110, Thailand;
| | - Hideyuki J. Majima
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand; (A.K.); (M.C.); (H.J.M.)
- Research Excellence Center for Innovation and Health Products (RECIHP), School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Hiroko P. Indo
- Department of Oncology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima City 890-8544, Japan;
| | - Pradoldej Sompol
- Department of Pharmacology & Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536, USA;
| | - Nazim Sekeroglu
- Department of Biology, Faculty of Arts and Sciences, Gaziantep University, 27310 Gaziantep, Turkey;
| | - Atthaphong Phongphithakchai
- Nephrology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand;
| | - Jitbanjong Tangpong
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand; (A.K.); (M.C.); (H.J.M.)
- Research Excellence Center for Innovation and Health Products (RECIHP), School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
| |
Collapse
|
17
|
Ji H, Wang Y, Cao X, Liu Y, Xu M, Zhao X, Chen M. Neutrophil percentage to albumin ratio predicts cardiovascular and all-cause mortality in diabetes and pre diabetes patients. Sci Rep 2025; 15:10075. [PMID: 40128326 PMCID: PMC11933700 DOI: 10.1038/s41598-025-93558-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 03/07/2025] [Indexed: 03/26/2025] Open
Abstract
The association between Neutrophil-Percentage-to-Albumin Ratio (NPAR) and mortality in cardiovascular disease (CVD) patients with diabetes or pre-diabetes is not well understood. This study investigates the relationship between baseline NPAR levels and all-cause and cardiovascular mortality among American adults with CVD and diabetes or pre-diabetes. This study enrolled 6,080 patients with diabetes or prediabetes from the National Health and Nutrition Examination Survey (2001-2018). Mortality outcomes were determined by linkage to the National Death Index (NDI) records through December 31, 2019. Multivariate Cox proportional hazards models were used to explore associations between NPAR and mortality. Non-linear correlations were assessed with restricted cubic splines, and segmented Cox proportional hazards models were used to evaluate threshold effects. Receiver operating characteristic (ROC) curves were used to evaluate NPAR's predictive ability for all-cause mortality. Weighted Kaplan-Meier curves with log-rank tests assessed cumulative survival differences across NPAR levels. In this cohort study, with a total follow-up of 53,217 person-years, 1,378 deaths from all causes and 476 deaths from CVD were recorded. Restricted cubic spline analysis revealed a J-shaped association between NPAR and both all-cause and cardiovascular mortality. Threshold effect analysis identified inflection points for NPAR in relation to all-cause mortality at 15.1 and cardiovascular mortality at 14.2. When baseline NPAR exceeded these inflection points, a positive correlation was observed with all-cause mortality (HR: 1.55, 95% CI: 1.08-2.16) and cardiovascular mortality (HR: 1.25, 95% CI: 1.09-1.86). ROC curves for 3-year, 5-year, and 10-year survival rates for all-cause mortality had areas under the curve (AUC) of 0.83, 0.83, and 0.81, respectively. For cardiovascular mortality, the AUC values were 0.86, 0.87, and 0.84. Increased NPAR is significantly associated with increased all-cause and cardiovascular mortality in individuals with diabetes or prediabetes, suggesting its potential role as a prognostic marker.
Collapse
Affiliation(s)
- Hua Ji
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei City, Anhui Province, People's Republic of China
| | - Yongqi Wang
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei City, Anhui Province, People's Republic of China
| | - Xinyi Cao
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei City, Anhui Province, People's Republic of China
| | - Yichang Liu
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei City, Anhui Province, People's Republic of China
| | - Murong Xu
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei City, Anhui Province, People's Republic of China
| | - Xiaotong Zhao
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei City, Anhui Province, People's Republic of China.
| | - Mingwei Chen
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei City, Anhui Province, People's Republic of China.
| |
Collapse
|
18
|
Al-Kuraishy HM, Sulaiman GM, Mohammed HA, Dawood RA, Albuhadily AK, Al-Gareeb AI, Klionsky DJ, Abomughaid MM. Insight into the Mechanistic role of Colchicine in Atherosclerosis. Curr Atheroscler Rep 2025; 27:40. [PMID: 40111634 DOI: 10.1007/s11883-025-01291-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
PURPOSE OF REVIEW Globally, the prevalence of atherosclerosis (AS) is rising. Currently, there is no specific drug for AS. Therefore, this review aims to discuss the protective mechanisms of colchicine against the development and progression of atherosclerosis (AS). RECENT FINDINGS Many studies highlighted that the anti-inflammatory drug colchicine reduces the severity of AS, although the underlying mechanism for the beneficial effect of colchicine was not fully clarified. AS is a chronic progressive vascular disorder characterized by the formation of atherosclerotic plaques. Endothelial dysfunction is an initial stage in the pathogenesis of AS that is induced by oxidized low-density lipoprotein (oxLDL). Engulfment of oxLDL by macrophages triggers the development of inflammation due to the release of pro-inflammatory cytokines and growth factors. Inflammatory and adhesion molecules are involved in the pathogenesis of AS. Infiltration and accumulation of leukocytes provoke erosion, rupture, and thrombosis of the atherosclerotic plaque. Therefore, targeting inflammation and leukocyte infiltration by anti-inflammatory agents may reduce AS progression and complications. The anti-inflammatory drug colchicine reduces the severity of AS, although the underlying mechanism for the beneficial effect of colchicine was not fully elucidated. IN CONCLUSION colchicine through inhibition of vascular inflammation, oxidative stress, platelet aggregation and the modulation of autophagy reduces the development and progression of AS.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ghassan M Sulaiman
- Division of Biotechnology, Department of Applied Sciences, University of Technology, Baghdad, Iraq.
| | - Hamdoon A Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, 51452, Qassim, Saudi Arabia
| | - Retaj A Dawood
- Department of Biology, College of Science, Al-Mustaqbal University, Hilla, 51001, Iraq
| | - Ali K Albuhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Jabir Ibn Hayyan Medical University, Al-Ameer Qu, PO.Box13 Kufa, Najaf, Iraq
| | - Daniel J Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Mosleh M Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, 255, 67714, Bisha, Saudi Arabia
| |
Collapse
|
19
|
Gu X, Du L, Lin R, Ding Z, Guo Z, Wei J, Li Y. How Advanced Is Nanomedicine for Atherosclerosis? Int J Nanomedicine 2025; 20:3445-3470. [PMID: 40125442 PMCID: PMC11928726 DOI: 10.2147/ijn.s508757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
Advances in nanotechnology have opened new avenues for precision therapy, personalized medicine, and multifunctional theranostics in atherosclerosis (AS). This review provides a comprehensive overview of the role of nanoparticles (NPs) in precision medicine for AS, discussing their applications, challenges, and future prospects. The review first analyzes the current treatment landscape of AS and outlines potential biological targets for therapy. Various nanocarriers, including organic, inorganic, and hybrid systems, are evaluated for their therapeutic potential, with a focus on targeted drug delivery, anti-inflammatory therapy, vascular repair, plaque stabilization, and lipid clearance. Additionally, the review explores NP preparation methods, emphasizing strategies to enhance drug loading, stability, and controlled release. Finally, the translational challenges of NP-based therapies, including biocompatibility, large-scale production, regulatory hurdles, and clinical implementation, are critically analyzed. Future directions highlight the importance of interdisciplinary collaboration and technological innovation in advancing nanoparticle-based precision medicine for AS.
Collapse
Affiliation(s)
- Xiang Gu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Lixin Du
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ruifang Lin
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zehui Ding
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zhihua Guo
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Jiaming Wei
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ya Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| |
Collapse
|
20
|
Ratke I, Torsvik A, Bartz-Johannessen C, Fathian F, Joa I, Reitan SK, Løberg E, Rettenbacher M, Skrede S, Steen V, Johnsen E, Kroken R. Sex differences in the peripheral levels of cytokines during 12-month antipsychotic treatment in a drug-naïve schizophrenia spectrum cohort. Brain Behav Immun Health 2025; 44:100959. [PMID: 39990282 PMCID: PMC11846924 DOI: 10.1016/j.bbih.2025.100959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 02/02/2025] [Accepted: 02/02/2025] [Indexed: 02/25/2025] Open
Abstract
Background There are substantial sex differences in schizophrenia. However, research addressing sex differences regarding the antipsychotic effect on the immune system is lacking. The aim of our study was to compare changes in cytokine levels in men and women with schizophrenia spectrum disorder over 12 months of treatment with antipsychotics. Methods This study reports pre-planned secondary outcomes from the BeSt InTro Study - a pragmatic, semi-randomised, rater-blinded comparison of amisulpride, aripiprazole, and olanzapine. The groups were analysed collectively. Of the 144 enrolled patients with schizophrenia spectrum disorders and ongoing psychosis, 56 were antipsychotic-naïve at baseline (20 women and 36 men) and were included in this study. Blood samples from these 56 patients were drawn at baseline, prior to treatment with antipsychotics, and 1, 3, 6, 12, 26, 39, and 52 weeks after initiation of antipsychotic medication. Duration of treatment was 52 weeks. Serum cytokine levels were assessed with a multiplex immunoassay. Changes in the levels of IL-4, IL-6, TNF-α, IL-1β, IL-2, IL-10, IL-12p70, IL-17A, IFN-γ and CRP from baseline to the different follow-up times were analysed using linear mixed effects models separately for men and women, and then compared. Outcomes Cytokine levels were mainly stable in men during the study period. In women, IL-4 levels were lower at baseline compared with men (p = 0.048) and showed a consistent and significant increase at weeks 6 (p = 0.006), 26 (p < 0.001), 39 (p = 0.002), and 52 (p = 0.001). TNF-α increased in women at weeks 26 (p = 0.008) and 39 (p = 0.012). IL-6 had a transient increase in women at weeks 12 (p = 0.003) and 26 (p = 0.007). There were significant sex differences in progression of cytokine levels at weeks 3 (IL-6: p = 0.046), 6 (IL-4: p = 0.022, IL-6: p = 0.015), 12 (IL-6: p = 0.01), 26 (IL-4: p < 0.001, IL-6: p = 0.015, TNF-α: p = 0.026), 39 (IL-4: p = 0.003, TNF-α: p = 0.023) and 52 (IL-4: p < 0.001, TNF-α: p = 0.009). CRP levels did not differ between sexes at baseline or during the study period and did not change significantly during treatment with antipsychotics in either sex. Interpretation We found significant sex differences in serum cytokine changes in drug-naïve patients with schizophrenia during treatment with antipsychotics. Cytokine levels were mainly altered in women, with increased IL-4, IL-6, and TNF-α levels. Cytokine changes may dramatically affect mental as well as somatic health. Our findings add to already established sex differences in schizophrenia pathophysiology and might have a potential role for future treatment guidelines. Funding The Research Council of Norway, the Western Norway Regional Health Trust, and the participating hospitals and universities provided funding for this study.
Collapse
Affiliation(s)
- I. Ratke
- Haukeland University Hospital, Division of Psychiatry, Postboks, 1400, 5021, Bergen, Norway
- Haukeland University Hospital, Mohn Research Centre for Psychotic Disorders (MRCP), Postboks 1400, 5021, Bergen, Norway
| | - A. Torsvik
- University of Bergen, Dr. Einar Martens Research Group for Biological Psychiatry, Department of Clinical Science 2, Postboks 7804, 5020, Bergen, Norway
- Haukeland University Hospital, Department of Medical Genetics, Postboks 1400, 5021, Bergen, Norway
- Haukeland University Hospital, Mohn Research Centre for Psychotic Disorders (MRCP), Postboks 1400, 5021, Bergen, Norway
| | - C.A. Bartz-Johannessen
- Haukeland University Hospital, Division of Psychiatry, Postboks, 1400, 5021, Bergen, Norway
- Haukeland University Hospital, Mohn Research Centre for Psychotic Disorders (MRCP), Postboks 1400, 5021, Bergen, Norway
| | - F. Fathian
- Haukeland University Hospital, Division of Psychiatry, Postboks, 1400, 5021, Bergen, Norway
- Haukeland University Hospital, Mohn Research Centre for Psychotic Disorders (MRCP), Postboks 1400, 5021, Bergen, Norway
| | - I. Joa
- Stavanger University Hospital, TIPS – Network for Clinical Research in Psychosis, Postboks 8100, 4068, Stavanger, Norway
- University of Stavanger, Faculty of Health, Postboks 8600, 4036, Stavanger, Norway
| | - S.M. Klæbo Reitan
- Norwegian University of Science and Technology, Department of Mental Health, Postboks 8900, 7491, Trondheim, Norway
- St. Olavs Hospital, Nidelv DPS, Department of Mental Health, Postboks 3250, 7006, Trondheim, Norway
| | - E.M. Løberg
- Haukeland University Hospital, Division of Psychiatry, Postboks, 1400, 5021, Bergen, Norway
- University of Bergen, Department of Clinical Psychology, Postboks 7807, 5020, Bergen, Norway
- Haukeland University Hospital, Mohn Research Centre for Psychotic Disorders (MRCP), Postboks 1400, 5021, Bergen, Norway
| | - M. Rettenbacher
- Medical University Innsbruck, Department of Psychiatry, Psychotherapy and Psychosomatics, Anichstrasse 35, A-6020, Innsbruck, Austria
| | - S. Skrede
- University of Bergen, Dr. Einar Martens Research Group for Biological Psychiatry, Department of Clinical Science 2, Postboks 7804, 5020, Bergen, Norway
- Haukeland University Hospital, Department of Medical Biochemistry and Pharmacology, Postboks 1400, 5021, Bergen, Norway
- Haukeland University Hospital, Mohn Research Centre for Psychotic Disorders (MRCP), Postboks 1400, 5021, Bergen, Norway
| | - V.M. Steen
- University of Bergen, Dr. Einar Martens Research Group for Biological Psychiatry, Department of Clinical Science 2, Postboks 7804, 5020, Bergen, Norway
- Haukeland University Hospital, Department of Medical Genetics, Postboks 1400, 5021, Bergen, Norway
- Haukeland University Hospital, Mohn Research Centre for Psychotic Disorders (MRCP), Postboks 1400, 5021, Bergen, Norway
| | - E. Johnsen
- Haukeland University Hospital, Division of Psychiatry, Postboks, 1400, 5021, Bergen, Norway
- University of Bergen, Department of Clinical Medicine, Postboks 7804, 5020, Bergen, Norway
- Haukeland University Hospital, Mohn Research Centre for Psychotic Disorders (MRCP), Postboks 1400, 5021, Bergen, Norway
| | - R.A. Kroken
- Haukeland University Hospital, Division of Psychiatry, Postboks, 1400, 5021, Bergen, Norway
- University of Bergen, Department of Clinical Medicine, Postboks 7804, 5020, Bergen, Norway
- Haukeland University Hospital, Mohn Research Centre for Psychotic Disorders (MRCP), Postboks 1400, 5021, Bergen, Norway
| |
Collapse
|
21
|
Kardassis D, Vindis C, Stancu CS, Toma L, Gafencu AV, Georgescu A, Alexandru-Moise N, Molica F, Kwak BR, Burlacu A, Hall IF, Butoi E, Magni P, Wu J, Novella S, Gamon LF, Davies MJ, Caporali A, de la Cuesta F, Mitić T. Unravelling molecular mechanisms in atherosclerosis using cellular models and omics technologies. Vascul Pharmacol 2025; 158:107452. [PMID: 39667548 DOI: 10.1016/j.vph.2024.107452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/31/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024]
Abstract
Despite the discovery and prevalent clinical use of potent lipid-lowering therapies, including statins and PCSK9 inhibitors, cardiovascular diseases (CVD) caused by atherosclerosis remain a large unmet clinical need, accounting for frequent deaths worldwide. The pathogenesis of atherosclerosis is a complex process underlying the presence of modifiable and non-modifiable risk factors affecting several cell types including endothelial cells (ECs), monocytes/macrophages, smooth muscle cells (SMCs) and T cells. Heterogeneous composition of the plaque and its morphology could lead to rupture or erosion causing thrombosis, even a sudden death. To decipher this complexity, various cell model systems have been developed. With recent advances in systems biology approaches and single or multi-omics methods researchers can elucidate specific cell types, molecules and signalling pathways contributing to certain stages of disease progression. Compared with animals, in vitro models are economical, easily adjusted for high-throughput work, offering mechanistic insights. Hereby, we review the latest work performed employing the cellular models of atherosclerosis to generate a variety of omics data. We summarize their outputs and the impact they had in the field. Challenges in the translatability of the omics data obtained from the cell models will be discussed along with future perspectives.
Collapse
Affiliation(s)
- Dimitris Kardassis
- University of Crete Medical School and Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology of Hellas, Heraklion, Greece.
| | - Cécile Vindis
- CARDIOMET, Center for Clinical Investigation 1436 (CIC1436)/INSERM, Toulouse, France
| | - Camelia Sorina Stancu
- Lipidomics Department, Institute of Cellular Biology and Pathology Nicolae Simionescu, Bucharest, Romania
| | - Laura Toma
- Lipidomics Department, Institute of Cellular Biology and Pathology Nicolae Simionescu, Bucharest, Romania
| | - Anca Violeta Gafencu
- Gene Regulation and Molecular Therapies Department, Institute of Cellular Biology and Pathology Nicolae Simionescu, Bucharest, Romania
| | - Adriana Georgescu
- Pathophysiology and Cellular Pharmacology Department, Institute of Cellular Biology and Pathology Nicolae Simionescu, Bucharest, Romania
| | - Nicoleta Alexandru-Moise
- Pathophysiology and Cellular Pharmacology Department, Institute of Cellular Biology and Pathology Nicolae Simionescu, Bucharest, Romania
| | - Filippo Molica
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Brenda R Kwak
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Alexandrina Burlacu
- Department of Stem Cell Biology, Institute of Cellular Biology and Pathology Nicolae Simionescu, Bucharest, Romania
| | - Ignacio Fernando Hall
- Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Elena Butoi
- Department of Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology Nicolae Simionescu, Bucharest, Romania
| | - Paolo Magni
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milano, Italy; IRCCS MultiMedica, Milan, Italy
| | - Junxi Wu
- University of Strathclyde, Glasgow, United Kingdom
| | - Susana Novella
- Department of Physiology, University of Valencia - INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Luke F Gamon
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael J Davies
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Caporali
- Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Fernando de la Cuesta
- Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Tijana Mitić
- Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
22
|
Baude JK, Mottaghy FM, Derlin T, Fischer A, Heinzel A, Bucerius J. PSMA - Targeted Clinical Molecular Imaging of Atherosclerosis: Correlation with Cardiovascular Risk Factors. Nuklearmedizin 2025; 64:13-21. [PMID: 39638325 DOI: 10.1055/a-2390-2829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
The early diagnosis of atherosclerotic changes to prevent ischemic events represents a clinical challenge.Prostate-specific membrane antigen (PSMA) as an established diagnostic in the field of prostate cancer also appears to detect neovascularization and inflammation in other diseases. We hypothesized that it might be also suited for detection of inflammation in atherosclerosis.We analyzed data of 78 prostate cancer patients who received a PSMA ligand PET/CT for re-staging. The cardiovascular risk factors (CVRF) of each patient were documented. Target-to-background-ratios (TBR) were calculated from the individual uptake values for three different sections of thoracic aorta [ascending (AA) and descending aorta (AD), aortic arch (AoAC)]. Statistical analyses included a linear regression analysis with the PSMA ligand uptake values of the different arterial segments versus different CVRF as independent variables.The meanTBRmax was measured highest in the AoAC (1.66 ± 0.33) compared to both other vessel sections (AA: 1.46 ± 0.21, p=0.001; AD: 1.59 ± 0.41, p=0.371). There was a correlation between the PSMA ligand uptake in all measured segments of the aorta and BMI, but only a significant correlation in the ascending aorta (r=0.347, p=0.001). This was confirmed in a subgroup analysis, which showed significantly higher uptake values in preadiposity (BMI >25) and obesity (BMI >30) patients in the ascending aorta (p=0.048).PSMA ligand uptake in the ascending aorta was linked to BMI. PET detection of vascular PSMA ligand uptake may be indicative of vessel wall inflammation to some extent. However, PSMA ligands appear to be less suitable than other tracers for this purpose, given their absent correlation with most established CVRFs.
Collapse
Affiliation(s)
- Julia K Baude
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, the Netherlands
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Alexander Fischer
- Philips GmbH Innovative Technologies, Research Laboratories, Aachen, Germany
| | - Alexander Heinzel
- Department of Nuclear Medicine, University Hospital, Martin-Luther-Universität, Halle-Wittenberg, Halle, Germany
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Jan Bucerius
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, the Netherlands
- Department of Nuclear Medicine, University Medicine Göttingen, Georg-August-University Göttingen, Göttingen, Germany
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC+), Maastricht, the Netherlands
| |
Collapse
|
23
|
Pogorielova OS, Korniienko VV, Chumachenko YD, Obukhova OA, Stroy YA, Harbuzova VY. Lack of Association of Serum MMP-9 Concentration and rs17576 Single Nucleotide Variant MMP-9 Gene With the Degree of Coronary Atherosclerosis and Other Risk Factors in Ukrainian Patients With Coronary Artery Disease. Cardiol Res Pract 2025; 2025:6610742. [PMID: 40224343 PMCID: PMC11986194 DOI: 10.1155/crp/6610742] [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: 10/24/2023] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 04/15/2025] Open
Abstract
This study probes the relationship between serum matrix metalloproteinase-9 (MMP-9) levels, the genetic variant rs17576 in the MMP-9 gene, and the extent of coronary atherosclerosis among Ukrainian patients diagnosed with coronary artery disease (CAD). A cohort of 128 patients was assessed, comprising 25 with angiographically intact (normal) coronary arteries, 40 with acute coronary syndrome (ACS), and 63 with chronic coronary syndrome (CCS). Utilizing clinical, anthropometric, and biochemical analyses, alongside ELISA immunoassays, genotyping, electrocardiography, and coronary angiography, we conducted a comprehensive evaluation. Our findings indicate that MMP-9 levels peaked in ACS patients, particularly those with single and triple-vessel coronary lesions, while the lowest levels were observed in individuals with unaltered coronary arteries. Notably, the glomerular filtration rate (GFR) was highest in patients with angiographically normal coronary arteries, averaging 79.91 ± 27.8 mL/min. In the context of ACS, individuals carrying the GG allele exhibited the highest GFR, whereas AA allele carriers had the lowest. Conversely, in the CCS cohort, GG carriers demonstrated the lowest GFR and heterozygotes the highest, although these differences did not reach statistical significance. A significant disparity in serum MMP-9 levels was observed between ACS patients, CCS patients, and individuals with unimpaired coronary arteries. Moreover, a substantial correlation was established between the degree of coronary artery lesions and GFR in the CCS group, providing a predictive measure for GFR in patients with triple-vessel involvement. However, no significant association was detected between serum MMP-9 levels, the rs17576 genetic variant in the MMP-9 gene, and the number of affected vessels or GFR in both ACS and CCS patients.
Collapse
Affiliation(s)
| | | | | | - Olha A. Obukhova
- Academic and Research Medical Institute, Sumy State University, Sumy, Ukraine
| | - Yelizaveta A. Stroy
- Academic and Research Medical Institute, Sumy State University, Sumy, Ukraine
| | | |
Collapse
|
24
|
Sapała AM, Staśkiewicz-Bartecka W, Kiciak A, Kardas M. Assessment of Nutritional Knowledge, Dietary Habits and Nutritional Status of Cardiology Patients, Considering Differences Between Individuals with Hypertension and Atherosclerosis and Those Without These Conditions. Nutrients 2025; 17:754. [PMID: 40077624 PMCID: PMC11901795 DOI: 10.3390/nu17050754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/13/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES Proper nutrition and correct habits and behaviours are crucial elements in the treatment and prevention of hypertension or atherosclerosis. This study aims to assess the nutritional knowledge, dietary habits, and nutritional status of cardiology patients, particularly those with hypertension and atherosclerosis. METHODS The study was conducted at St. Barbara Regional Specialized Hospital No. 5 in Sosnowiec from January to June 2021. It included 301 patients, 51.2% women (N = 154) and 48.8% men (N = 147), who were assessed for Body Mass Index, Nutritional Risk Score, and morphological and biochemical test results. Their knowledge and dietary habits were examined using a questionnaire and rating scale designed by the authors. While the study encompassed all cardiology patients, a subgroup analysis specifically examined individuals diagnosed with hypertension and/or atherosclerosis. RESULTS 80% of the respondents showed above-normal body weight, while there were no significant differences in the risk of malnutrition according to the Nutritional Risk Score. The diet analysis revealed insufficient intake of fruits, vegetables, legumes, dairy, whole grains, and fish, while the consumption of salty snacks, sweetened beverages, energy drinks, and alcohol was low. Analysis of morphology and biochemistry results showed no significant differences between patients with atherosclerosis, hypertension, and others. CONCLUSIONS The study reveals insufficient nutritional knowledge and poor dietary habits among cardiology patients, highlighting the need for targeted education to improve dietary behaviours and reduce cardiovascular risks. Implementing nutrition-focused interventions in cardiology care could enhance patient outcomes. Future research should explore the long-term effects of dietary education and identify effective strategies for behavioural change in this population.
Collapse
Affiliation(s)
- Anna-Maria Sapała
- Department of Dietetics and Food Science, Faculty of Science, Natural and Technical Sciences, Jan Długosz University in Częstochowa, al. Armii Krajowej 13/15, 42-200 Częstochowa, Poland;
| | - Wiktoria Staśkiewicz-Bartecka
- Department of Food Technology and Quality Assessment, School of Public Health in Bytom, Medical University of Silesia in Katowice, ul. Jordana 19, 41-808 Zabrze, Poland; (W.S.-B.); (M.K.)
| | - Agata Kiciak
- Department of Food Technology and Quality Assessment, School of Public Health in Bytom, Medical University of Silesia in Katowice, ul. Jordana 19, 41-808 Zabrze, Poland; (W.S.-B.); (M.K.)
| | - Marek Kardas
- Department of Food Technology and Quality Assessment, School of Public Health in Bytom, Medical University of Silesia in Katowice, ul. Jordana 19, 41-808 Zabrze, Poland; (W.S.-B.); (M.K.)
| |
Collapse
|
25
|
Shi Y, Gu W, Wei Y, Li S, Zhang S, Jiang Y, Chen C, Liu T, Shuai L, Zhou X, Tang F. Deficiency of Calpain-1 attenuates atherosclerotic plaque and calcification and improves vasomotor dysfunction in Apolipoprotein E knockout mice through inhibiting inflammation. Biochem Biophys Res Commun 2025; 749:151369. [PMID: 39892965 DOI: 10.1016/j.bbrc.2025.151369] [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: 11/08/2024] [Revised: 01/18/2025] [Accepted: 01/19/2025] [Indexed: 02/04/2025]
Abstract
PURPOSE Atherosclerosis (AS) and atherosclerotic calcification (AC) are closely related to the cardiovascular diseases, largely due to its induction of vasomotor dysfunction. We previously reported that Calpain-1 inhibitor attenuated AS and AC. The present study was designed to investigate the effects and potential mechanisms of Calpain-1 knockout (Calpain-1 KO) in Apolipoprotein E KO (ApoE KO) mice on AS, AC, aortic vasomotor function as well as the liver dysfunction. METHODS We hybridized ApoE KO mice with Calpain-1 KO mice to obtain ApoE/Calpain-1 double KO (A × C DKO) mice. The formation of AS and AC was evaluated and liver function was determined. Aortic vasomotor function was assessed. Contents of TNF-α, IL-6, IL-18, IL-1β and NO and the activity of AST, ALT, ALP and eNOS in serum were quantified. The mRNA expression of CD68, SR-A, CD36, PPAR-γ, LXR-α, ABCA1, BMP-2, OPN, ALP and Runx2 in aorta and/or liver were measured. RESULTS The results showed that in comparison to C57 mice, ApoE KO mice demonstrated the significant increases in the areas of AS and AC, the increases in the mRNA expression of CD68 in the aorta, the increases in the AST, ALT and ALP activity in serum. ApoE KO mice also showed the dysfunction of the aortic contraction induced by phenylephrine and of the relaxation induced by acetylcholine. However, compared with ApoE KO mice, A × C DKO mice exhibited the significant attenuation of AS and AC and the downregulation of mRNA expression of CD68 in aorta. A × C DKO mice revealed the reduction of AST, ALT and ALP activity in serum, the improvements in aortic contraction and relaxation as well as the increases in eNOS activity and NO content in serum. A × C DKO mice also showed the decreases in the contents of TNF-α, IL-6, IL-18 and IL-1β in serum. The mRNA expression of CD68 in aorta, SR-A and CD36 in both aorta and liver of A × C DKO mice was downregulated, while that of PPAR-γ, LXR-α, and ABCA1 was upregulated in comparison of ApoE KO mice. In addition, the mRNA expression of BMP-2, OPN, ALP and Runx2 in aorta of A × C DKO mice was downregulated in comparison of ApoE KO mice. CONCLUSION The results suggested that deficiency of Calpain-1 attenuated the formation of AS and AC and improved vasomotor and liver dysfunction in ApoE KO mice through anti-inflammation, and modulation of the mRNA expression of genes related to AS and AC.
Collapse
Affiliation(s)
- Youming Shi
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China; Department of Cardiovascular Disease, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China
| | - Wenjiao Gu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China; Department of Cardiovascular Disease, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China
| | - Yujie Wei
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China; Gansu Province Key Laboratory of Environmental Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China
| | - Shuangyi Li
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China; Department of Cardiovascular Disease, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China
| | - Shining Zhang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China; School of Nuclear and Technology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Ying Jiang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China; Department of Cardiovascular Disease, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China
| | - Chuanjian Chen
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China; Department of Urology Disease, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China
| | - Ting Liu
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Limei Shuai
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Xiaochun Zhou
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China; Department of Cardiovascular Disease, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China; Department of Nephrology, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China.
| | - Futian Tang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China; Department of Cardiovascular Disease, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China; Gansu Province Key Laboratory of Environmental Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China.
| |
Collapse
|
26
|
Martí-Carvajal AJ, Gemmato-Valecillos MA, Monge Martín D, De Sanctis JB, Martí-Amarista CE, Hidalgo R, Alegría-Barrero E, Riera Lizardo RJ, Correa-Pérez A. Colchicine for the primary prevention of cardiovascular events. Cochrane Database Syst Rev 2025; 2:CD015003. [PMID: 39927511 PMCID: PMC11808834 DOI: 10.1002/14651858.cd015003.pub2] [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] [Indexed: 02/11/2025]
Abstract
BACKGROUND Atherosclerotic cardiovascular diseases (ACVDs), a condition characterised by lipid accumulation in arterial walls, which is often exacerbated by chronic inflammation disorders, is the major cause of mortality and morbidity worldwide. Colchicine, with its first medicinal use in ancient Egypt, is an inexpensive drug with anti-inflammatory properties. However, its role in primary prevention of ACVDs in the general population remains unknown. OBJECTIVES To assess the clinical benefits and harms of colchicine as primary prevention of cardiovascular outcomes in the general population. SEARCH METHODS We searched the Cochrane Heart Specialised Register, the Cochrane Central Register of Controlled Trials (CENTRAL), Ovid MEDLINE (including In-Process & Other Non-Indexed Citations), Ovid Embase, Web of Science, and LILACS. We searched ClinicalTrials.gov and WHO ICTRP for ongoing and unpublished studies. We also scanned the reference lists of relevant included studies, reviews, meta-analyses, and health technology reports to identify additional studies. There were no limitations on language, date of publication, or study setting. The search results were updated on 31 May 2023. SELECTION CRITERIA Randomised controlled trials (RCTs) in any setting, recruiting adults without pre-existing cardiovascular disease. We included trials that compared colchicine versus placebo, non-steroidal anti-inflammatory drugs, corticosteroids, immunomodulating drugs, or usual care. Our primary outcomes were all-cause mortality, non-fatal myocardial infarction, stroke, and adverse events. DATA COLLECTION AND ANALYSIS Two or more review authors independently selected studies, extracted data, and performed risk of bias and GRADE assessments. MAIN RESULTS We identified 15 RCTs (1721 participants randomised; 1412 participants analysed) with follow-up periods ranging from 4 to 728 weeks. The intervention was oral colchicine compared with placebo, immunomodulating drugs, or usual care or no treatment. Due to biases and imprecision, the evidence was very uncertain for all outcomes. All trials but one had a high risk of bias. Five out of seven meta-analyses included fewer than six trials (71.4%). The objectives of the review were to assess cardiovascular outcomes in the general population, but many of the included trials focused on liver disease. Colchicine compared to placebo Colchicine may reduce all-cause mortality compared to placebo in primary prevention, but the evidence is very uncertain (risk ratio (RR) 0.68, 95% confidence interval (CI) 0.51 to 0.91; 6 studies, 463 participants; very low-certainty evidence; number needed to treat for an additional beneficial outcome (NNTB) 11, 95% CI 6 to 67). Colchicine may result in little to no difference in non-fatal myocardial infarction, but the evidence is very uncertain (RR 0.87, 95% CI 0.41 to 1.82; 1 study, 100 participants; very low-certainty evidence). Colchicine may not reduce the incidence of stroke, but the evidence is very uncertain (RR 2.43, 95% CI 0.67 to 8.86; 1 study, 100 participants; very low-certainty evidence). Regarding adverse events, colchicine may increase the incidence of diarrhoea (RR 3.99, 95% CI 1.44 to 11.06; 8 studies, 605 participants; very low-certainty evidence; number needed to treat for an additional harmful outcome (NNTH) 10, 95% CI 6 to 17), and may have little to no effect on neurological outcomes such as seizure or mental confusion (RR 0.72, 95% CI 0.31 to 1.66; 2 studies, 155 participants; very low-certainty evidence), but the evidence is very uncertain. The effect of colchicine on cardiovascular mortality is also very uncertain (RR 1.27, 95% CI 0.03 to 62.43; 2 studies, 160 participants; very low-certainty evidence). Colchicine may not reduce post-cardiac procedure atrial fibrillation, but the evidence is very uncertain (RR 0.74, 95% CI 0.25 to 2.19; 1 study, 100 participants). We found no trials reporting on pericardial effusion, peripheral artery disease, heart failure, or unstable angina. Colchicine compared to methotrexate (immunomodulating drug) Colchicine may result in little to no difference in all-cause mortality compared to methotrexate, but the evidence is very uncertain (RR 0.42, 95% CI 0.12 to 1.51; 1 study, 85 participants; very low-certainty evidence). We found no trials reporting other cardiovascular outcomes or adverse events for this comparison. Colchicine compared to usual care or no treatment The evidence is very uncertain about the effect of colchicine compared with usual care on all-cause mortality in primary prevention (RR 1.07, 95% CI 0.90 to 1.27; 2 studies, 729 participants; very low-certainty evidence). Regarding adverse events, colchicine may increase the incidence of diarrhoea compared to usual care, but the evidence is very uncertain (RR 3.32, 95% CI 1.56 to 7.03; 2 studies, 729 participants; very low-certainty evidence; NNTH 18, 95% CI 12 to 42). No trials reported other cardiovascular outcomes for this comparison. AUTHORS' CONCLUSIONS This Cochrane review evaluated the clinical benefits and harms of using colchicine for the primary prevention of cardiovascular events in the general population. Comparisons were made against placebo, immunomodulating medications, or usual care or no treatment. However, the certainty of the evidence for the predefined outcomes was very low, highlighting the pressing need for high-quality, rigorous studies to ascertain colchicine's clinical impact definitively. We identified numerous biases and inaccuracies in the included studies, limiting their generalisability and precluding a conclusive determination of colchicine's efficacy in preventing cardiovascular events. The existing evidence regarding colchicine's potential cardiovascular benefits or harms for primary prevention is inconclusive owing to the limitations inherent in the current studies. More robust clinical trials are needed to bridge this evidence gap effectively.
Collapse
Affiliation(s)
- Arturo J Martí-Carvajal
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador, Universidad UTE, Quito, Ecuador
- Faculty of Medicine, Universidad Francisco de Vitoria, Madrid, Spain
- Cátedra Rectoral de Medicina Basada en la Evidencia, Universidad de Carabobo, Valencia, Venezuela
| | - Mario A Gemmato-Valecillos
- Department of Medicine, NYC Health + Hospitals / Elmhurst, Icahn School of Medicine at Mount Sinai, New York, USA
| | | | - Juan Bautista De Sanctis
- The Institute of Molecular and Translational Medicine, Palacky University Olomouc, Faculty of Medicine and Dentistry, Olomouc, Czech Republic
| | | | - Ricardo Hidalgo
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador., Universidad UTE, Quito, Ecuador
| | | | - Ricardo J Riera Lizardo
- Cátedra Rectoral de Medicina Basada en la Evidencia, Universidad de Carabobo, Valencia, Venezuela
- Medicine Department, Rheumatology Unit, Universidad de Carabobo, Valencia, Venezuela
| | - Andrea Correa-Pérez
- Faculty of Medicine, Universidad Francisco de Vitoria, Madrid, Spain
- Department of Hospital Pharmacy and Medical Devices, Hospital Central de la Defensa Gómez Ulla, Madrid, Spain
| |
Collapse
|
27
|
Li X, Zhu D, Zhao B, Li Q, Jin P. Alternative splicing: Therapeutic target for vasculopathy in diabetic complications. Life Sci 2025; 362:123331. [PMID: 39734014 DOI: 10.1016/j.lfs.2024.123331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/03/2024] [Accepted: 12/19/2024] [Indexed: 12/31/2024]
Abstract
It is becoming increasingly evident that diabetic vascular complications seriously threaten human health. The most prevalent microvascular complications include kidney disease, retinal disease, cardiovascular diseases and amputation. Conventional treatments can only relieve the progression of the diseases, and is no longer appropriate for the long-term management of diabetic patients. Exploring a novel therapeutic regimens and improvements in management of Diabetic Complications is required. Alternative splicing has been found to play a crucial role in the occurrence and treatment of diseases, including the destruction and generation of blood vessels in diabetes. Alternative splicing is an important factor in the high complexity of multicellular eukaryotic transcriptome, and angiogenesis, which is an important process controlled by alternative splicing mechanism. This review mainly introduces the current understanding of alternative splicing and the role that alternative splicing plays in the diabetic complications, with a special focus on vascular system. In this study, we summarized alternative splicing in relation to diabetes complications and the pathogenesis of diabetic vasculopathy. It discussed potential treatment strategies for correcting aberrant splicing and suggested novel approaches for addressing diabetes complications.
Collapse
Affiliation(s)
- Xiaoyue Li
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Xuzhou Medical University, Xuzhou, China
| | - Dong Zhu
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Xuzhou Medical University, Xuzhou, China
| | - Bingkun Zhao
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| | - Qiang Li
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| | - Peisheng Jin
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
28
|
Zhang H, Lu W, Tang H, Chen A, Gao X, Zhu C, Zhang J. Novel Insight of N6-Methyladenosine in Cardiovascular System. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:222. [PMID: 40005339 PMCID: PMC11857502 DOI: 10.3390/medicina61020222] [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] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/19/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025]
Abstract
N6-methyladenosine (m6A) is the most common and abundant internal co-transcriptional modification in eukaryotic RNAs. This modification is catalyzed by m6A methyltransferases, known as "writers", including METTL3/14 and WTAP, and removed by demethylases, or "erasers", such as FTO and ALKBH5. It is recognized by m6A-binding proteins, or "readers", such as YTHDF1/2/3, YTHDC1/2, IGF2BP1/2/3, and HNRNPA2B1. Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide. Recent studies indicate that m6A RNA modification plays a critical role in both the physiological and pathological processes involved in the initiation and progression of CVDs. In this review, we will explore how m6A RNA methylation impacts both the normal and disease states of the cardiovascular system. Our focus will be on recent advancements in understanding the biological functions, molecular mechanisms, and regulatory factors of m6A RNA methylation, along with its downstream target genes in various CVDs, such as atherosclerosis, ischemic diseases, metabolic disorders, and heart failure. We propose that the m6A RNA methylation pathway holds promise as a potential therapeutic target in cardiovascular disease.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (H.Z.); (W.L.); (H.T.); (A.C.); (X.G.)
| | - Wei Lu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (H.Z.); (W.L.); (H.T.); (A.C.); (X.G.)
| | - Haoyue Tang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (H.Z.); (W.L.); (H.T.); (A.C.); (X.G.)
| | - Aiqun Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (H.Z.); (W.L.); (H.T.); (A.C.); (X.G.)
| | - Xiaofei Gao
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (H.Z.); (W.L.); (H.T.); (A.C.); (X.G.)
| | - Congfei Zhu
- Department of Cardiology, Lianshui County People’s Hospital, Affiliated Hospital of Kangda College, Nanjing Medical University, Huaian 223400, China
| | - Junjie Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (H.Z.); (W.L.); (H.T.); (A.C.); (X.G.)
| |
Collapse
|
29
|
Sun L, Liu J, Cheng Y, Wu Y, He T, Zhang Y, Bai X, Zhou Z, Xu X, Yao Y, Tan Y, Qiu Q, Liu C. Metabolomics with gut microbiota analysis of podophyllotoxin-mediated cardiotoxicity in mice based on the toxicological evidence chain (TEC) concept. Chem Biol Interact 2025; 406:111360. [PMID: 39706312 DOI: 10.1016/j.cbi.2024.111360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
Podophyllotoxin (PPT) is a lignan isolated from the traditional Chinese medicine Dysosma Versipellis, with significant anti-tumor activity. However, its cardiotoxicity restricts its clinical application. This study aims to investigate the cardiotoxicity of PPT in mice and its underlying mechanisms based on the concept of toxicological evidence chain (TEC). In this study, alterations in body weight, behavior, and the levels of myocardial enzymes and histopathology in mice were observed. Additionally, microbiome and metabolome were integrated to identify potential microorganisms, metabolic markers and major pathways with correlation analysis. The results indicated that PPT induced pathological changes in mice, including weight loss, diarrhea, alopecia and dehydration accompanied by increased levels of serum myocardial enzymes. The results of microbiome showed that PPT altered the gut microbiota composition, changing the abundance of microbial community. The results of metabolome studies indicated total of 55 differential metabolites were involved in glycine, serine, and threonine metabolism, alanine, glutamate, and aspartate metabolism, purine, pyrimidine metabolism, and steroid hormone metabolism. Integrating the results of microbiome and metabolome, it was concluded that PPT remodeled the gut microbiota composition, which in turn modified the gut microbiota metabolism, affecting amino acid metabolisms, nucleotide metabolism, and steroid hormone metabolism in the heart, potentially leading to energy metabolism disorders, apoptosis, and oxidative stress, ultimately inducing cardiotoxicity.
Collapse
Affiliation(s)
- Lu Sun
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Jiaojiao Liu
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Yangyang Cheng
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Yikang Wu
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Tao He
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Yingyue Zhang
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Xiaorui Bai
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Zixin Zhou
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Xiayu Xu
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Yuxin Yao
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Yafei Tan
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Qiang Qiu
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Chuanxin Liu
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China.
| |
Collapse
|
30
|
Lorentzen LG, Yeung K, Zitkeviciute A, Yang-Jensen KC, Eldrup N, Eiberg JP, Davies MJ. N-Terminal Proteomics Reveals Distinct Protein Degradation Patterns in Different Types of Human Atherosclerotic Plaques. J Proteome Res 2025; 24:144-157. [PMID: 39665830 DOI: 10.1021/acs.jproteome.4c00548] [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: 12/13/2024]
Abstract
Atherosclerotic plaque rupture is a major cause of cardiovascular events. Plaque destabilization is associated with extracellular matrix (ECM) modification involving proteases which generate protein fragments with new N-termini. We hypothesized that rupture-prone plaques would contain elevated fragment levels, and their sequences would allow identification of active proteases and target proteins. Plaques from 21 patients who underwent surgery for symptomatic carotid artery stenosis were examined in an observational/cross-sectional study. Plaques were analyzed by liquid chromatography-mass spectrometry for the presence of N-terminal fragments. 33920 peptides were identified, with 17814 being N-terminal species. 5735 distinct N-terminal peptides were quantified and subjected to multidimensional scaling analysis and consensus clustering. These analyses indicated three clusters, which correlate with gross macroscopic plaque morphology (soft/mixed/hard), ultrasound classification (echolucent/echogenic), and the presence of hemorrhage/ulceration. Differences in the fragment complements are consistent with plaque-type-dependent turnover and degradation pathways. Identified peptides include signal and pro-peptides from synthesis and those from protein fragmentation. Sequence analysis indicates that targeted proteins include ECM species and responsible proteases (meprins, cathepsins, matrix metalloproteinases, elastase, and kallikreins). This study provides a large data set of peptide fragments and proteases present in plaques of differing stability. These species may have potential as biomarkers for improved atherosclerosis risk profiling.
Collapse
Affiliation(s)
- Lasse G Lorentzen
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen 2200, Denmark
- Department of Vascular Surgery, Heart Centre, University Hospital Copenhagen─Rigshospitalet, Copenhagen 2100, Denmark
| | - Karin Yeung
- Department of Vascular Surgery, Heart Centre, University Hospital Copenhagen─Rigshospitalet, Copenhagen 2100, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Auguste Zitkeviciute
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen 2200, Denmark
| | - Karen C Yang-Jensen
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen 2200, Denmark
| | - Nikolaj Eldrup
- Department of Vascular Surgery, Heart Centre, University Hospital Copenhagen─Rigshospitalet, Copenhagen 2100, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Jonas P Eiberg
- Department of Vascular Surgery, Heart Centre, University Hospital Copenhagen─Rigshospitalet, Copenhagen 2100, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
- Copenhagen Academy for Medical Education and Simulation, Capital Region of Denmark, Copenhagen 2100, Denmark
| | - Michael J Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen 2200, Denmark
| |
Collapse
|
31
|
Abdulrahim AO, Doddapaneni NSP, Salman N, Giridharan A, Thomas J, Sharma K, Abboud E, Rochill K, Shreelakshmi B, Gupta V, Lakkimsetti M, Mowo-Wale A, Ali N. The gut-heart axis: a review of gut microbiota, dysbiosis, and cardiovascular disease development. Ann Med Surg (Lond) 2025; 87:177-191. [PMID: 40109640 PMCID: PMC11918638 DOI: 10.1097/ms9.0000000000002789] [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: 06/25/2024] [Accepted: 11/20/2024] [Indexed: 03/22/2025] Open
Abstract
Background Cardiovascular diseases (CVDs) are a major cause of morbidity and mortality worldwide and there are strong links existing between gut health and cardiovascular health. Gut microbial diversity determines gut health. Dysbiosis, described as altered gut microbiota, causes bacterial translocations and abnormal gut byproducts resulting in systemic inflammation. Objective To review the current literature on the relationships between gut microbiota, dysbiosis, and CVD development, and explore therapeutic methods to prevent dysbiosis and support cardiovascular health. Summary Dysbiosis increases levels of pro-inflammatory substances while reducing those of anti-inflammatory substances. This accumulative inflammatory effect negatively modulates the immune system and promotes vascular dysfunction and atherosclerosis. High Firmicutes to Bacteroidetes ratios, high trimethylamine-n-oxide to short-chain fatty acid ratios, high indole sulfate levels, low cardiac output, and polypharmacy are all associated with worse cardiovascular outcomes. Supplementation with prebiotics and probiotics potentially alleviates some CVD risk. Blood and stool samples may be used in clinical practice to quantify and qualify gut bacterial ratios and byproducts, assess patients' risk for adverse cardiovascular outcomes, and track their gut health progress. Further research is required to set population-based cutoffs for normal and abnormal gut microbiota and byproduct ratios.
Collapse
Affiliation(s)
| | | | - Nadhra Salman
- Department of Internal Medicine, Baqai Medical University, Karachi, Pakistan
| | | | | | - Kavya Sharma
- Maharishi Markandeshwar Medical College and Hospital, Himachal Pradesh, India
| | - Elias Abboud
- Faculty of Medicine, University of Saint Joseph, Beirut, Lebanon
| | | | - B Shreelakshmi
- Navodaya Medical College Hospital & Research Centre, Karnataka, India
| | | | | | | | - Noor Ali
- Dubai Medical College, Dubai, United Arab Emirates
| |
Collapse
|
32
|
Sarlak Z, Naderi N, Amidi B, Ghorbanzadeh V. Sodium Butyrate, A Gut Microbiota Derived Metabolite in Type 2 Diabetes Mellitus and Cardiovascular Disease: A Review. Cardiovasc Hematol Agents Med Chem 2025; 23:1-10. [PMID: 39206487 DOI: 10.2174/0118715257307380240820052940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/13/2024] [Accepted: 07/03/2024] [Indexed: 09/04/2024]
Abstract
Type 2 diabetes is characterized by elevated blood glucose levels, leading to an increased risk of cardiovascular diseases. Sodium butyrate, the sodium salt of the short-chain fatty acid butyric acid produced by gut microbiota fermentation, has shown promising effects on metabolic diseases, including type 2 diabetes and cardiovascular diseases. Sodium butyrate demonstrates anti-inflammatory, anti-oxidative, and lipid-lowering properties and can improve insulin sensitivity and reduce hepatic steatosis. In this review, we investigate how sodium butyrate influences cardiovascular complications of type 2 diabetes, including atherosclerosis (AS), heart failure (HF), hypertension, and angiogenesis. Moreover, we explore the pathophysiology of cardiovascular disease in type 2 diabetes, focusing on hyperglycemia, oxidative stress, inflammation, and genetic factors playing crucial roles. The review suggests that sodium butyrate can be a potential preventive and therapeutic agent for cardiovascular complications in individuals with type 2 diabetes.
Collapse
Affiliation(s)
- Zeynab Sarlak
- Department of Biology, Khorramabad branch, Islamic Azad University, Khorramabad, Iran
| | - Narges Naderi
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Bardia Amidi
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Vajihe Ghorbanzadeh
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
33
|
Woxholt S, Ueland T, Aukrust P, Anstensrud AK, Broch K, Tøllefsen IM, Seljeflot I, Halvorsen B, Dahl TB, Huse C, Andersen GØ, Gullestad L, Wiseth R, Damås JK, Kleveland O. Effect of tocilizumab on endothelial and platelet-derived CXC-chemokines and their association with inflammation and myocardial injury in STEMI patients undergoing primary PCI. Int J Cardiol 2025; 418:132613. [PMID: 39374793 DOI: 10.1016/j.ijcard.2024.132613] [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: 05/30/2024] [Revised: 08/29/2024] [Accepted: 10/02/2024] [Indexed: 10/09/2024]
Abstract
BACKGROUND Tocilizumab improves myocardial salvage in ST-elevation myocardial infarction (STEMI) patients when administered before percutaneous coronary intervention (PCI). The mechanisms underlying ischemia-reperfusion injury remain unclear. In this sub-study, we investigated whether endothelial and platelet-derived CXC chemokines are involved, as they represent inflammatory mediators from two cell types relevant to myocardial infarction. Associations between these chemokines and neutrophils, C-reactive protein (CRP), troponin T (TnT), myocardial salvage index (MSI), microvascular obstruction (MVO), and infarct size. METHODS This is a sub-study of the ASSAIL-MI trial, a double-blind clinical trial that randomized 199 STEMI patients to receive either 280 mg tocilizumab (n = 101) or placebo (n = 98) intravenously before PCI. Blood samples were collected prior to infusion, at day 1-2, 3-7, and at 3 and 6 months. Heparin was administered before baseline in 150 patients, while 49 received it after. We measured CXC-chemokines CXCL4, CXCL5, CXCL6, CXCL7, and CXCL12 using immunoassays. Cardiac MRI was performed in the first week and at 6 months. RESULTS Tocilizumab did not significantly affect CXC-chemokines levels. Although some correlations were observed between chemokine levels and neutrophil counts and CRP, none of the CXC chemokines were associated with infarct size, MSI, MVO, or TnT levels. Notably, CXCL 12 levels increased in patients who received heparin before baseline, while other CXC-chemokines decreased significantly. CONCLUSION This study suggests that the beneficial effects of tocilizumab in STEMI patients are not due to changes in circulating endothelial or platelet-derived CXC-chemokines, compared to placebo. However, heparin significantly influences the levels of these chemokines.
Collapse
Affiliation(s)
- Sindre Woxholt
- Clinic of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Thrombosis Research and Expertise Center (TREC), The Arctic University of Norway, Tromsø, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Section of Clinical Immunology and Infectious Disease, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Kaspar Broch
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway; K. G. Jebsen Cardiac Research Centre and Centre for Heart Failure Research, University of Oslo, Oslo, Norway
| | | | - Ingebjørg Seljeflot
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Cardiology, Oslo Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Tuva B Dahl
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Camilla Huse
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Geir Øystein Andersen
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway; Department of Cardiology, Oslo Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Lars Gullestad
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway; K. G. Jebsen Cardiac Research Centre and Centre for Heart Failure Research, University of Oslo, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Rune Wiseth
- Clinic of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Jan Kristian Damås
- Department of Infectious Disease, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway; Department of clinical and Molecular medicine, Centre of Molecular Inflammation Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ola Kleveland
- Clinic of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
34
|
Fawzy El‐Sayed K, Mahlandt E, Schlicht K, Enthammer K, Tölle J, Wagner J, Hartmann K, Ebeling PR, Graetz C, Laudes M, Dörfer CE, Schulte DM. Effects of oxidized LDL versus IL-1ß/TNF-ɑ/INFɣ on human gingival mesenchymal stem cells properties. J Periodontal Res 2025; 60:77-89. [PMID: 38952262 PMCID: PMC11840472 DOI: 10.1111/jre.13319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/12/2024] [Accepted: 06/19/2024] [Indexed: 07/03/2024]
Abstract
AIMS Oxidized low-density lipoprotein (oxLDL) is an important player in the course of metabolic inflammatory diseases. oxLDL was identified in the gingival crevicular fluid, denoting possible associations between oxLDL-induced inflammation and periodontal disease. The current investigation compared for the first-time direct effects of oxLDL to a cytokine cocktail of IL-1ß/TNF-ɑ/INF-γ on gingival mesenchymal stem cells' (G-MSCs) attributes. METHODS Human third passage G-MSCs, isolated from connective tissue biopsies (n = 5) and characterized, were stimulated in three groups over 7 days: control group, cytokine group (IL-1β[1 ng/mL], TNF-α[10 ng/mL], IFN-γ[100 ng/mL]), or oxLDL group (oxLDL [50 μg/mL]). Next Generation Sequencing and KEGG pathway enrichment analysis, stemness gene expression (NANOG/SOX2/OCT4A), cellular proliferation, colony-formation, multilinear potential, and altered intracellular pathways were investigated via histochemistry, next-generation sequencing, and RT-qPCR. RESULTS G-MSCs exhibited all mesenchymal stem cells' characteristics. oxLDL group and cytokine group displayed no disparities in their stemness markers (p > .05). Next-generation-sequencing revealed altered expression of the TXNIP gene in response to oxLDL treatment compared with controls (p = .04). Following an initial boosting for up to 5 days by inflammatory stimuli, over 14 day, cellular counts [median count ×10-5 (Q25/Q75)] were utmost in control - [2.6607 (2.0804/4.5357)], followed by cytokine - [0.0433 (0.0026/1.4215)] and significantly lowered in the oxLDL group [0.0274 (0.0023/0.7290); p = .0047]. Osteogenic differentiation [median relative Ca2+ content(Q25/Q75)] was significantly lower in cytokine - [0.0066 (0.0052/0.0105)] compared to oxLDL - [0.0144 (0.0108/0.0216)] (p = .0133), with no differences notable for chondrogenic and adipogenic differentiation (p > .05). CONCLUSIONS Within the current investigation's limitations, in contrast to cytokine-mediated inflammation, G-MSCs appear to be minimally responsive to oxLDL-mediated metabolic inflammation, with little negative effect on their differentiation attributes and significantly reduced cellular proliferation.
Collapse
Affiliation(s)
- Karim Fawzy El‐Sayed
- Clinic for Conservative Dentistry and PeriodontologyUniversity Hospital of Schleswig‐HolsteinKielGermany
- Oral Medicine and Periodontology Department, Faculty of DentistryCairo UniversityCairoEgypt
- Stem Cells and Tissue Engineering Unit, Faculty of DentistryCairo UniversityCairoEgypt
| | - Elena Mahlandt
- Institute of Diabetes and Clinical Metabolic ResearchUniversity Hospital of Schleswig‐HolsteinKielGermany
| | - Kristina Schlicht
- Institute of Diabetes and Clinical Metabolic ResearchUniversity Hospital of Schleswig‐HolsteinKielGermany
| | - Kim Enthammer
- Institute of Diabetes and Clinical Metabolic ResearchUniversity Hospital of Schleswig‐HolsteinKielGermany
| | - Johannes Tölle
- Clinic for Conservative Dentistry and PeriodontologyUniversity Hospital of Schleswig‐HolsteinKielGermany
- Department of DermatologyUniversity Hospital Schleswig‐HolsteinKielGermany
- Institute of ImmunologyUniversity Hospital Schleswig‐HolsteinKielGermany
| | - Juliane Wagner
- Department of Oral and Maxillofacial SurgeryUniversity Hospital of Schleswig‐HolsteinKielGermany
| | - Katharina Hartmann
- Institute of Diabetes and Clinical Metabolic ResearchUniversity Hospital of Schleswig‐HolsteinKielGermany
| | - Peter R. Ebeling
- Department of Medicine, School of Clinical Sciences at Monash HealthMonash UniversityMelbourneVictoriaAustralia
| | - Christian Graetz
- Clinic for Conservative Dentistry and PeriodontologyUniversity Hospital of Schleswig‐HolsteinKielGermany
| | - Mathias Laudes
- Institute of Diabetes and Clinical Metabolic ResearchUniversity Hospital of Schleswig‐HolsteinKielGermany
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Internal Medicine IUniversity Hospital Schleswig‐HolsteinKielGermany
| | - Christof E. Dörfer
- Clinic for Conservative Dentistry and PeriodontologyUniversity Hospital of Schleswig‐HolsteinKielGermany
| | - Dominik M. Schulte
- Institute of Diabetes and Clinical Metabolic ResearchUniversity Hospital of Schleswig‐HolsteinKielGermany
- Department of Medicine, School of Clinical Sciences at Monash HealthMonash UniversityMelbourneVictoriaAustralia
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Internal Medicine IUniversity Hospital Schleswig‐HolsteinKielGermany
| |
Collapse
|
35
|
Wang Y, Fang X, Liu J, Lv X, Lu K, Lu Y, Jiang Y. PCSK9 in T-cell function and the immune response. Biomark Res 2024; 12:163. [PMID: 39736777 DOI: 10.1186/s40364-024-00712-8] [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: 08/16/2024] [Accepted: 12/19/2024] [Indexed: 01/01/2025] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) was first reported in 2003 and confirmed to be strongly associated with familial hypercholesterolemia. Small-molecule inhibitors targeting PCSK9 provide an effective and safe method for managing hypercholesterolemia and reducing the cardiovascular risk. In recent years, increasing evidence has indicated other important roles for PCSK9 in inflammation, tumors, and even immune regulation. PCSK9 might be an attractive regulator of T-cell activation and expansion. It might mediate inflammation and regulate other types of immune cells. In this review, we summarize the current advances in the field of PCSK9 and provide a narrative of the biological processes associated with PCSK9. The relationships between PCSK9 and different T cells were investigated in depth. Finally, the signaling pathways associated with PCSK9 and the immune response are also summarized in this review.
Collapse
Affiliation(s)
- Yuying Wang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Xiaosheng Fang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Jiarui Liu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Xiao Lv
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Kang Lu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Yingxue Lu
- Department of Nephrology, Shandong Second Provincial General Hospital, Jinan , Shandong, 250021, China
| | - Yujie Jiang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China.
| |
Collapse
|
36
|
Zhang Z, Zheng Z, Luo W, Li J, Liao J, Chen F, Wang D, Lin Y. Identifying immune cell infiltration and effective diagnostic biomarkers for ischemic stroke using bioinformatics analysis. PLoS One 2024; 19:e0310108. [PMID: 39636891 PMCID: PMC11620413 DOI: 10.1371/journal.pone.0310108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 08/25/2024] [Indexed: 12/07/2024] Open
Abstract
Ischemic stroke (IS) is a leading cause of death and disability worldwide. Screening for marker genes in IS is crucial for its early diagnosis and improvement in clinical outcomes. In the study, the gene expression profiles in the GSE22255 and GSE37587 datasets were extracted from the public database Gene Expression Omnibus. Weighted gene co‑expression network analysis (WGCNA) was used to investigate the gene sets that were related to ubiquitination. A total of 33 ubiquitination-related differentially expressed genes (DEGs) were identified using "limma (version 3.50.0)". Gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) analysis enriched multiple pathways that were closely related to IS. The correlations between the HALLMARK signaling pathways and DGEs were analyzed. Receiver operating characteristic analysis was used to validate the diagnostic value of the key genes. Among them, 16 genes were identified as hub genes. Single-sample GSEA was performed to evaluate the infiltration status of immune cells in IS. To understand the potential molecular mechanisms of the hub genes in IS, we constructed RBP-mRNA and mRNA-miRNA-lncRNA interaction networks. Additionally, we used the GeneMANIA database to create a PPI network for the signature genes to investigate their functions. As a result, there was a significant difference in the overall infiltration of immune cells between the IS and control groups. Among the 28 types of immune cells, the degree of infiltration of seven types was significantly different between the two groups (p<0.05). The expression of four types of immune cells, namely type 1 T helper cell, type 17 T helper cell, eosinophil, and mast cell, in the IS group were significantly higher than that in the control group. The expressions of DHFR2 (R = -0.575; p<0.001) and DNAAF2 (R = -0.562; p<0.001) were significantly negatively correlated with eosinophil infiltration. The PPI network demonstrated that the 16 hub genes interacted with each other. In conclusion, we identified DEGs, WGCNA modules, hub genes, enriched pathways, and infiltrating immune cells that may be closely involved in IS. Further studies are required to explore the functions of these genes.
Collapse
Affiliation(s)
- Zongyong Zhang
- Department of Neurosurgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Zongqing Zheng
- Department of Neurosurgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Wenwei Luo
- Department of Neurosurgery, Nanping First Hospital Affiliated to Fujian Medical University, Fuzhou, China
| | - Jiebo Li
- Department of Neurosurgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jiushan Liao
- Department of Neurosurgery, Luoyuan County Hospital, Fuzhou, China
| | - Fuxiang Chen
- Department of Neurosurgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Dengliang Wang
- Department of Neurosurgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yuanxiang Lin
- Department of Neurosurgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
37
|
Lee YN, Wu YJ, Su CH, Wang BJ, Yang SH, Lee HI, Chou YH, Tien TY, Lin CF, Chan WH, Chung CH, Wang SW, Yeh HI. Fluorescent gold nanoclusters possess multiple actions against atherosclerosis. Redox Biol 2024; 78:103427. [PMID: 39566163 PMCID: PMC11612375 DOI: 10.1016/j.redox.2024.103427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024] Open
Abstract
Atherosclerosis caused major morbidity and mortality worldwide. Molecules possessing lipid-lowering and/or anti-inflammatory properties are potential druggable targets against atherosclerosis. We examined the anti-atherosclerotic effects of fluorescent gold nanoclusters (FANC), which were dihydrolipoic acid (DHLA)-capped 2-nm gold nanoparticles. We evaluated the 8-week effects of FANC in Western-type diet-fed ApoE-deficient mice by either continuous intraperitoneal delivery (20 μM, 50 μl weekly) or via drinking water (300 nM). FANC reduced aortic atheroma burden, serum total cholesterol, and oxidative stress markers malondialdehyde and 4-hydroxynonenal levels. FANC attenuated hepatic lipid deposit, with changed expression of lipid homeostasis-related genes HMGCR, SREBP, PCSK9, and LDLR in a pattern similar to mice treated with ezetimibe. FANC also inhibited intestinal cholesterol absorption, resembling the action of ezetimibe. The lipid-lowering and anti-atherosclerotic effects of FANC reappeared in Western-type diet-fed LDLr-deficient mice. FANC bound insulin receptor β (IRβ) via DHLA, leading to AKT activation. However, unlike insulin, which also bound IRβ to activate AKT to induce HO-1, activation of AKT by FANC was independent of HO-1 expression in human aortic endothelial cells (HAECs). Alternatively, FANC up-regulated NRF2, interfered the binding of KEAP1 to NRF2, and promoted KEAP1 degradation to free NRF2 for nuclear entry to induce HO-1 that suppressed the expression of ICAM-1 and VCAM-1. Consistently, FANC suppressed ox-LDL-induced enhanced attachment of THP-derived macrophages onto HAECs. In macrophages, FANC up-regulated ABCA1, and reversed ox-LDL-induced suppression of cholesterol efflux. FANC effected in vitro at nano moles. In conclusion, our findings showed novel actions and multiple mechanisms of FANC worked coherently against atherosclerosis.
Collapse
Affiliation(s)
- Yi-Nan Lee
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan
| | - Yih-Jer Wu
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Cheng-Huang Su
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan
| | - Bo-Jeng Wang
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan
| | - Sheng-Hsun Yang
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan
| | - Hsin-I Lee
- Department of Medicine, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Yen-Hung Chou
- Department of Medicine, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Ting-Yi Tien
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan
| | - Chao-Feng Lin
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Wen-Hsiung Chan
- Department of Bioscience Technology and Center for Nanotechnology, Chung Yuan Christian University, Zhongbei Road, Zhongli District, Taoyuan City, 32023, Taiwan
| | - Ching-Hu Chung
- Department of Medicine, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Shin-Wei Wang
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Hung-I Yeh
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan.
| |
Collapse
|
38
|
Killick R, Stranks L, Hoyos CM. Sleep Deficiency and Cardiometabolic Disease. Sleep Med Clin 2024; 19:653-670. [PMID: 39455184 DOI: 10.1016/j.jsmc.2024.07.011] [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: 10/28/2024]
Abstract
Epidemiologic studies have demonstrated that short sleep duration is associated with an increased risk of cardio-metabolic health outcomes including cardiovascular disease mortality, coronary heart disease, type 2 diabetes mellitus, hypertension, and metabolic syndrome. Experimental sleep restriction studies have sought to explain these findings. This review describes the main evidence of these associations and possible mechanisms explaining them. Whether sleep extension reverses these now widely acknowledged adverse health effects and the feasibility of implementing such strategies on a public health level is discussed.
Collapse
Affiliation(s)
- Roo Killick
- Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, University of Sydney, Sydney, Australia
| | - Lachlan Stranks
- Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, University of Sydney, Sydney, Australia; The University of Adelaide, Faculty of Health and Medical Sciences, Adelaide, Australia
| | - Camilla M Hoyos
- Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, University of Sydney, Sydney, Australia; The University of Sydney, Faculty of Science, School of Psychology and Brain and Mind Centre, Sydney, Australia.
| |
Collapse
|
39
|
Qian P, Cao X, Zhang Q, Gao M, Liu X, Yan L. Circ_0004872 deficiency attenuates ox-LDL-induced vascular smooth muscle cell dysfunction by miR-424-5p-dependent regulation of FRS2. Mol Cell Biochem 2024; 479:3425-3435. [PMID: 38376663 DOI: 10.1007/s11010-024-04929-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 01/05/2024] [Indexed: 02/21/2024]
Abstract
Atherosclerosis (AS) is a pivotal pathological basis of cardiovascular and cerebrovascular diseases, and circular RNAs (circRNAs) has been disclosed to exert a vital part in the progression of AS. However, the functions of circ_0004872 in the progression of AS is indistinct. In this context, we aimed to elucidate the role of circ_0004872 and the potential mechanism in AS. The level of circ_0004872, miR-424-5p and fibroblast growth factor receptor substrate 2 (FRS2) was detected using quantitative real-time polymerase chain reaction (qRT-PCR). Cell proliferation was monitored by Cell Counting Kit-8 and 5-ethynyl-2'-deoxyuridine (EDU) assays. The invasion and migration capabilities of VSMCs were tested by transwell assays and wound-healing assay, respectively. Western blot was adopted to check the protein levels of CyclinD1, Vimentin and FRS2. Dual-luciferase reporter and RNA immunoprecipitation assay were executed to manifest the interaction between miR-424-5p and circ_0004872 or FRS2. The level of circ_0004872 was increased in the serum samples of AS patients and ox-LDL-exposed VSMCs. Ox-LDL exposure triggered cell proliferation, invasion and migration ability of VSMCs. depletion of circ_0004872 partly weakened ox-LDL-mediated effects in VSMCs. Mechanistically, circ_0004872 functioned as a sponge of miR-424-5p, and miR-424-5p inhibition partly alleviated circ_0004872 deficiency-mediated influences in VSMCs. Additionally, miR-424-5p interacted with FRS2, and miR-424-5p constrained dysfunction in ox-LDL-stimulated VSMCs via reducing FRS2 level. Notably, circ_0004872 functioned as a sponge of miR-424-5p to elevate FRS2 expression. Circ_0004872 accelerated ox-LDL-induced damage via mediating miR-424-5p/FRS2 axis.
Collapse
MESH Headings
- MicroRNAs/genetics
- MicroRNAs/metabolism
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Humans
- Lipoproteins, LDL/metabolism
- Membrane Proteins/metabolism
- Membrane Proteins/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Adaptor Proteins, Signal Transducing/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/genetics
- Cell Proliferation
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Cell Movement
- Male
Collapse
Affiliation(s)
- Peng Qian
- Department of Geriatric Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, 450003, China
| | - Xuanchao Cao
- Department of Geriatric Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, 450003, China
| | - Qian Zhang
- Department of Geriatric Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, 450003, China
| | - Meihua Gao
- Department of Geriatric Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, 450003, China
| | - Xin Liu
- Department of Geriatric Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, 450003, China
| | - Lijie Yan
- Heart Center of Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital, Fuwai Central China Hospital of Zhengzhou University, No. 1 Fuwai Street, Zhengdong New District, Zhengzhou, 451464, China.
| |
Collapse
|
40
|
Tang MY, Xie H, Tao JT, Zhang C, Luo YH, Zhang C, Peng SQ, Xie LX, Lv WB, Zhang C, Huang L. Pathophysiological relevance and therapeutic outlook of GPR43 in atherosclerosis. Biochem Cell Biol 2024; 102:418-429. [PMID: 39013204 DOI: 10.1139/bcb-2024-0053] [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: 07/18/2024] Open
Abstract
Atherosclerosis (AS) is an inflammatory arterial disorder that occurs due to the deposition of the excessive lipoprotein under the artery intima, mainly including low-density lipoprotein and other apolipoprotein B-containing lipoproteins. G protein-coupled receptors (GPCRs) play a crucial role in transmitting signals in physiological and pathophysiological conditions. GPCRs recognize inflammatory mediators, thereby serving as important players during chronic inflammatory processes. It has been demonstrated that free fatty acids can function as ligands for various GPCRs, such as free fatty acid receptor (FFAR)1/GPR40, FFAR2/GPR43, FFAR3/GPR41, FFAR4/GPR120, and the lipid metabolite binding glucose-dependent insulinotropic receptor (GPR119). This review discusses GPR43 and its ligands in the pathogenesis of AS, especially focusing on its distinct role in regulating chronic vascular inflammation, inhibiting oxidative stress, ameliorating endothelial dysfunction and improving dyslipidemia. It is hoped that this review may provide guidance for further studies aimed at GPR43 as a promising target for drug development in the prevention and therapy of AS.
Collapse
Affiliation(s)
- Mu-Yao Tang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Hao Xie
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Jin-Tao Tao
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Chun Zhang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Yao-Hua Luo
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Cong Zhang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Si-Qin Peng
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Lin-Xi Xie
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Wen-Bo Lv
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Chi Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Liang Huang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| |
Collapse
|
41
|
Najmi EA, Mirzaasgari Z, Motamed MR, Aslani A. Carotid intima-media thickness and risk of atherosclerosis in multiple sclerosis: A cross-sectional study. PLoS One 2024; 19:e0314031. [PMID: 39561151 PMCID: PMC11575821 DOI: 10.1371/journal.pone.0314031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is characterized by inflammation and demyelination in the central nervous system. Recent studies have suggested a potential association between MS and an increased risk of atherosclerosis, a systemic vascular disease involving arterial wall thickening. Understanding this relationship is crucial, given the heightened cardiovascular risk observed in MS patients. OBJECTIVE To investigate factors influencing the development of atherosclerosis in patients with multiple sclerosis (pwMS), focusing on carotid intima-media thickness (CIMT) as a marker. METHODS 114 pwMS (82 females and 32 males) and 127 control subjects (57 females and 70 males) were included. The mean CIMT between the two groups was compared. Additionally, the effects of annual relapse rate, EDSS (Expanded Disability Status Scale), MS duration, treatment duration, and type of Disease-modifying treatment (DMT) on CIMT were evaluated. RESULTS This study included 241 participants with a mean (SD) age of 41.13 years (10.93). CIMT was significantly higher in pwMS compared to controls (p < 0.001). Even after adjusting for age, sex, and BMI (Body Mass Index), CIMT remained significantly higher in the pwMS group (p < 0.001). Spearman's correlation analysis revealed significant associations between CIMT and age, BMI, EDSS score, and disease duration (all p < 0.05). Additionally, the SPMS (Secondary Progressive Multiple Sclerosis) disease course was significantly associated with higher CIMT (p < 0.001). Linear regression analysis identified age as the most significant predictor of increased CIMT in pwMS (p < 0.001), followed by BMI (p = 0.054). CONCLUSION This study demonstrates a significant association between MS and increased CIMT. Additionally, age emerged as the most prominent predictor of high CIMT in pwMS, followed by BMI. These findings suggest a potential link between MS and increased cardiovascular risk. Further research is warranted to explore the underlying mechanisms and investigate the long-term cardiovascular outcomes in this population.
Collapse
Affiliation(s)
- Elyar Alizadeh Najmi
- Firouzgar Clinical Research and Development Center (FCRDC), Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Neurology, Firouzgar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Mirzaasgari
- Firouzgar Clinical Research and Development Center (FCRDC), Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Neurology, Firouzgar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- School of Advanced Technologies in Medicine, Iran University of Medical, Tehran, Iran
| | - Mohammad Reza Motamed
- Firouzgar Clinical Research and Development Center (FCRDC), Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Neurology, Firouzgar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Armin Aslani
- Department of Community Medicine, Social Determinants of Health Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
42
|
Pan Z, Lv J, Zhao L, Xing K, Ye R, Zhang Y, Chen S, Yang P, Yu H, Lin Y, Li R, Wang D, Fang J, Dong Y, Sheng J, Wang X, Shan G, Zhang S, Cheng H, Xu Q, Guo X. CircARCN1 aggravates atherosclerosis by regulating HuR-mediated USP31 mRNA in macrophages. Cardiovasc Res 2024; 120:1531-1549. [PMID: 39028686 DOI: 10.1093/cvr/cvae148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 03/08/2024] [Accepted: 05/05/2024] [Indexed: 07/21/2024] Open
Abstract
AIMS Circular RNAs (circRNAs) are considered important regulators of biological processes, but their impact on atherosclerosis development, a key factor in coronary artery disease (CAD), has not been fully elucidated. We aimed to investigate their potential use in patients with CAD and the pathogenesis of atherosclerosis. METHODS AND RESULTS Patients with stable angina (SA) or acute coronary syndrome (ACS) and controls were selected for transcriptomic screening and quantification of circRNAs in blood cells. We stained carotid plaque samples for circRNAs and performed gain- and loss-of-function studies in vitro. Western blots, protein interaction analysis, and molecular approaches were used to perform the mechanistic study. ApoE-/- mouse models were employed in functional studies with adeno-associated virus-mediated genetic intervention. We demonstrated elevated circARCN1 expression in peripheral blood mononuclear cells from patients with SA or ACS, especially in those with ACS. Furthermore, higher circARCN1 levels were associated with a higher risk of developing SA and ACS. We also observed elevated expression of circARCN1 in carotid artery plaques. Further analysis indicated that circARCN1 was mainly expressed in monocytes and macrophages, which was also confirmed in atherosclerotic plaques. Our in vitro studies provided evidence that circARCN1 affected the interaction between HuR and ubiquitin-specific peptidase 31 (USP31) mRNA, resulting in attenuated USP31-mediated NF-κB activation. Interestingly, macrophage accumulation and inflammation in atherosclerotic plaques were markedly decreased when circARCN1 was knocked down with adeno-associated virus in macrophages of ApoE-/- mice, while circARCN1 overexpression in the model exacerbated atherosclerotic lesions. CONCLUSIONS Our findings provide solid evidence macrophagic-expressed circARCN1 plays a role in atherosclerosis development by regulating HuR-mediated USP31 mRNA stability and NF-κB activation, suggesting that circARCN1 may serve as a factor for atherosclerotic lesion formation.
Collapse
MESH Headings
- Aged
- Animals
- Female
- Humans
- Male
- Mice
- Middle Aged
- Acute Coronary Syndrome/genetics
- Acute Coronary Syndrome/pathology
- Acute Coronary Syndrome/metabolism
- Angina, Stable/genetics
- Angina, Stable/metabolism
- Angina, Stable/pathology
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Carotid Artery Diseases/genetics
- Carotid Artery Diseases/metabolism
- Carotid Artery Diseases/pathology
- Case-Control Studies
- Coronary Artery Disease/genetics
- Coronary Artery Disease/pathology
- Coronary Artery Disease/metabolism
- Disease Models, Animal
- ELAV-Like Protein 1/metabolism
- ELAV-Like Protein 1/genetics
- Gene Expression Regulation
- Macrophages/metabolism
- Macrophages/pathology
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- NF-kappa B/metabolism
- Plaque, Atherosclerotic
- RAW 264.7 Cells
- RNA, Circular/genetics
- RNA, Circular/metabolism
- RNA, Messenger/metabolism
- RNA, Messenger/genetics
- Signal Transduction
- THP-1 Cells
- Ubiquitin Thiolesterase/genetics
- Ubiquitin Thiolesterase/metabolism
- Ubiquitin-Specific Proteases/genetics
- Ubiquitin-Specific Proteases/metabolism
Collapse
Affiliation(s)
- Zhicheng Pan
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jialan Lv
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liding Zhao
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kaidi Xing
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Runze Ye
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuesheng Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Siyuan Chen
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Yang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hailong Yu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yangkai Lin
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruobing Li
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongfei Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Juan Fang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yang Dong
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianpeng Sheng
- Zhejiang Province Key Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaolin Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, The RNA Institute, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China (UTSC), Hefei, China
| | - Ge Shan
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, The RNA Institute, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China (UTSC), Hefei, China
| | - Shan Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongqiang Cheng
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, China
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingbo Xu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
43
|
Abudukeyimu Z, Luo J, Liu F, Ma Y, Li J, Wang J, Li X. Early growth response factor 3 may regulate coronary atherosclerosis through the NF-κB signaling pathway and VEGF expression. Am J Med Sci 2024; 368:476-484. [PMID: 38992750 DOI: 10.1016/j.amjms.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 07/13/2024]
Abstract
AIM The present study was conducted to measure the expression of early growth response factor 3 (Egr3), inflammatory cytokines (IL-1β, IL-6), vascular endothelial growth factor (VEGF) and NF-κB in patients with coronary artery disease (CAD) to investigate the relationships of these molecules and Egr3 gene expression. METHODS We recruited 132 CAD patients and 63 healthy individuals. The expression levels of Egr3, VEGF, p50 and p65 were measured by reverse transcription quantitative polymerase chain reaction and the levels of Egr3, IL-1β and IL-6 in patients serum and in human coronary artery endothelial cells (HCAECs) were measured by enzyme-linked immunosorbent assay (ELISAs) in CAD patients. HCAECs were treated with ox-LDL to establish an in vitro atherosclerosis model. An oil red O staining assay was used to assess the lipid droplet formation. A colloidal external lumen formed by Matrigel was used to test the migration of HCAECs. The expression of Egr3, VEGF and NF-κB was determined by Western blotting. RESULTS The levels of serum Egr3 and IL-6 in the severe stenosis group were greater than those in the mild stenosis group and controls (p < 0.05). The level of serum IL-1β in the severe stenosis group was greater than that in the control group (p < 0.05). Moreover, Egr3 expression was positively associated with IL-6 levels (r = 0.55, p < 0.001), IL-1β levels (r = 0.21, p = 0.004) and the Gensini score (r = 0.20, p = 0.02). We also found that Egr3 expression was significantly greater in CAD patients than that in controls. And its expression was highest in the mild patients. The expression of VEGF, P50 and P65 was also greater in CAD patients. In the in vitro experiment, we found that the inhibition of Egr3 expression significantly reduced the expression levels of p50, p65, IL-6 and CRP. Moreover, the inhibition of Egr3 expression significantly reduced the lipid droplet formation and decreased capability of lumen formation. CONCLUSIONS In the pathogenesis of atherosclerosis, Egr3 gene expression may induce the expression of inflammatory factors and lipid droplet formation and lumen formation, which could promote the atherosclerosis development.
Collapse
Affiliation(s)
- Zumureti Abudukeyimu
- Department of General Practice, the Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, China
| | - Junyi Luo
- Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, China
| | - Fang Liu
- Department of General Practice, the Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, China
| | - Yanling Ma
- Department of General Practice, the Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, China
| | - Jiao Li
- Department of General Practice, the Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, China
| | - Juan Wang
- Department of Cardiology, the Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, China.
| | - Xia Li
- Department of General Practice, the Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, China.
| |
Collapse
|
44
|
Yu D, Guo G, Wan F, Hu B. The association between C-reactive protein to albumin ratio and adverse outcomes in acute ischemic stroke patients: A study in the Korean population. Heliyon 2024; 10:e39212. [PMID: 39497966 PMCID: PMC11533568 DOI: 10.1016/j.heliyon.2024.e39212] [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: 07/02/2024] [Revised: 09/19/2024] [Accepted: 10/09/2024] [Indexed: 11/07/2024] Open
Abstract
Introduction Our study aimed to explore the relationship between the C-reactive protein to albumin ratio (CAR) and adverse outcomes at 3 months in Korean patients with acute ischemic cerebral infarction. Methods This study focused on a sample of 1654 individuals with acute ischemic stroke (AIS), who received medical treatment at a reputable hospital in South Korea between January 2010 and December 2016. We used multivariate logistic regression models to show the effect of CAR admission on 3-month adverse outcomes in patients with AIS, as well as generalized additive modeling (GAM) and two-stage linear regression modeling to explore whether there was a linear relationship. Results A total of 1654 patients with acute ischemic cerebral infarction were enrolled in the study, and the CAR was determined to be associated with poor results for patients with AIS after 3 months. with correction for potential confounders (odds ratio [OR], 1.23; 95 % confidence interval [CI], 1.08-1; p < 0.001). A nonlinear relationship was found between the CAR and adverse outcomes at 3 months for patients with AIS, with a threshold of approximately 0.6. The effect sizes, CIs, and p-values above and below the threshold were 1.39 (1.25-1.55, p < 0.001) and 1.08 (0.81-1.43, p = 0.60). Conclusions The adverse outcomes of patients with AIS at three months were independently correlated with the CAR. In addition, there was a nonlinear relationship between adverse outcomes and the CAR, with the CAR increasing the risk of adverse outcomes at 3 months for patients with AIS when the CAR was less than 0.6.
Collapse
Affiliation(s)
- Dandan Yu
- Department of Electrocardiogram, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, China
| | - Guixiang Guo
- Department of Neurology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, China
| | - Fangchao Wan
- Department of Neurology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, China
| | - Bohong Hu
- Department of Neurology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, China
| |
Collapse
|
45
|
Grzeczka A, Graczyk S, Kordowitzki P. Pleiotropic Effects of Resveratrol on Aging-Related Cardiovascular Diseases-What Can We Learn from Research in Dogs? Cells 2024; 13:1732. [PMID: 39451250 PMCID: PMC11505706 DOI: 10.3390/cells13201732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024] Open
Abstract
Resveratrol (RES) is a polyphenol with natural anti-inflammatory and antioxidant properties. It is found in abundance in plants, i.e., grapes and mulberry fruit. In addition, synthetic forms of RES exist. Since the discovery of its specific biological properties, RES has emerged as a candidate substance not only with modeling effects on the immune response but also as an important factor in preventing the onset and progression of cardiovascular disease (CVD). Previous research provided strong evidence of the effects of RES on platelets, mitochondria, cardiomyocytes, and vascular endothelial function. In addition, RES positively affects the coagulation system and vasodilatory function and improves blood flow. Not only in humans but also in veterinary medicine, cardiovascular diseases have one of the highest incidence rates. Canine and human species co-evolved and share recent evolutionary selection processes, and interestingly, numerous pathologies of companion dogs have a human counterpart. Knowledge of the impact of RES on the cardiovascular system of dogs is becoming clearer in the literature. Dogs have long been recognized as valuable animal models for the study of various human diseases as they share many physiological and genetic similarities with humans. In this review, we aim to shed light on the pleiotropic effects of resveratrol on cardiovascular health in dogs as a translational model for human cardiovascular diseases.
Collapse
Affiliation(s)
| | | | - Pawel Kordowitzki
- Department for Basic and Preclinical Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Toruń, Poland; (A.G.)
| |
Collapse
|
46
|
Wang L, Luo W, Zhang S, Zhang J, He L, Shi Y, Gao L, Wu B, Nie X, Hu C, Han X, He C, Xu B, Liang G. Macrophage-derived FGFR1 drives atherosclerosis through PLCγ-mediated activation of NF-κB inflammatory signalling pathway. Cardiovasc Res 2024; 120:1385-1399. [PMID: 38842387 DOI: 10.1093/cvr/cvae131] [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: 01/16/2024] [Revised: 04/22/2024] [Accepted: 05/04/2024] [Indexed: 06/07/2024] Open
Abstract
AIMS Atherosclerosis (AS) is a leading cause of cardiovascular morbidity and mortality. Atherosclerotic lesions show increased levels of proteins associated with the fibroblast growth factor receptor (FGFR) pathway. However, the functional significance and mechanisms governed by FGFR signalling in AS are not known. In the present study, we investigated fibroblast growth factor receptor 1 (FGFR1) signalling in AS development and progression. METHODS AND RESULTS Examination of human atherosclerotic lesions and aortas of Apoe-/- mice fed a high-fat diet (HFD) showed increased levels of FGFR1 in macrophages. We deleted myeloid-expressed Fgfr1 in Apoe-/- mice and showed that Fgfr1 deficiency reduces atherosclerotic lesions and lipid accumulations in both male and female mice upon HFD feeding. These protective effects of myeloid Fgfr1 deficiency were also observed when mice with intact FGFR1 were treated with FGFR inhibitor AZD4547. To understand the mechanistic basis of this protection, we harvested macrophages from mice and show that FGFR1 is required for macrophage inflammatory responses and uptake of oxidized LDL. RNA sequencing showed that FGFR1 activity is mediated through phospholipase-C-gamma (PLCγ) and the activation of nuclear factor-κB (NF-κB) but is independent of FGFR substrate 2. CONCLUSION Our study provides evidence of a new FGFR1-PLCγ-NF-κB axis in macrophages in inflammatory AS, supporting FGFR1 as a potentially therapeutic target for AS-related diseases.
Collapse
MESH Headings
- Animals
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/genetics
- Phospholipase C gamma/metabolism
- Phospholipase C gamma/genetics
- NF-kappa B/metabolism
- Signal Transduction
- Macrophages/metabolism
- Male
- Female
- Disease Models, Animal
- Aortic Diseases/pathology
- Aortic Diseases/metabolism
- Aortic Diseases/genetics
- Aortic Diseases/prevention & control
- Aortic Diseases/immunology
- Humans
- Plaque, Atherosclerotic
- Mice, Knockout, ApoE
- Mice, Inbred C57BL
- Lipoproteins, LDL/metabolism
- Diet, High-Fat
- Pyrazoles/pharmacology
- Inflammation Mediators/metabolism
- Benzamides/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Piperazines
Collapse
Affiliation(s)
- Lintao Wang
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Shangtang Road 158, Hangzhou, Zhejiang 310014, China
- Department of Cardiology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu 210008, China
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Longmian Avenue 639, Nanjing, Jiangsu 210009, China
| | - Wu Luo
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Shangtang Road 158, Hangzhou, Zhejiang 310014, China
- Department of Cardiology, The Affiliated First Hospital of Wenzhou Medical University, Nanbaixiang Street, Wenzhou, Zhejiang 325035, China
| | - Suya Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Longmian Avenue 639, Nanjing, Jiangsu 210009, China
| | - Junsheng Zhang
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, China
- Department of Pathology, Anhui Public Health Clinical Center, Hefei, Anhui 230032, China
| | - Lu He
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Longmian Avenue 639, Nanjing, Jiangsu 210009, China
| | - Yifan Shi
- Department of Cardiology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu 210008, China
| | - Li Gao
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Longmian Avenue 639, Nanjing, Jiangsu 210009, China
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Baochuan Wu
- Department of Cardiology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu 210008, China
| | - Xiaoyan Nie
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Longmian Avenue 639, Nanjing, Jiangsu 210009, China
| | - Chenghong Hu
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Shangtang Road 158, Hangzhou, Zhejiang 310014, China
- Department of Cardiology, The Affiliated First Hospital of Wenzhou Medical University, Nanbaixiang Street, Wenzhou, Zhejiang 325035, China
| | - Xue Han
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Shangtang Road 158, Hangzhou, Zhejiang 310014, China
| | - Chaoyong He
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Longmian Avenue 639, Nanjing, Jiangsu 210009, China
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu 210008, China
| | - Guang Liang
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Shangtang Road 158, Hangzhou, Zhejiang 310014, China
- Department of Cardiology, The Affiliated First Hospital of Wenzhou Medical University, Nanbaixiang Street, Wenzhou, Zhejiang 325035, China
| |
Collapse
|
47
|
Galhom RA, Ali SNS, El-Fark MMO, Ali MHM, Hussein HH. Assessment of therapeutic efficacy of adipose tissue-derived mesenchymal stem cells administration in hyperlipidemia-induced aortic atherosclerosis in adult male albino rats. Tissue Cell 2024; 90:102498. [PMID: 39079452 DOI: 10.1016/j.tice.2024.102498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/21/2024] [Accepted: 07/24/2024] [Indexed: 09/03/2024]
Abstract
Atherosclerosis (AS) is a common disease seriously detrimental to human health. AS is a chronic progressive disease related to inflammatory reactions. The present study aimed to characterize and evaluate the effects of adipose tissue stem cells (ADSCs) in high-fat diet-induced atherosclerosis in a rat model. The present study comprises thirty-six rats and they were divided into three groups: the control group, the high-fat diet (HFD) group; which received a high-fat diet, and the high-fat diet + stem cells (HFD+SC) group; which was fed with a high-fat diet along with the administration of intravenous ADSCs. Food was given to the animals for 20 weeks to establish dyslipidemia models. After 20 weeks, animals were sacrificed by cervical dislocation; blood was collected to measure total cholesterol (TC), triglycerides (TG), low-density lipoprotein (LDL), and high-density lipoprotein (HDL); aortae were collected to detect morphologic changes. Rats of the HFD group showed a significant increase in body weight (B.Wt), altered lipid profile increased expression of inducible nitric oxide synthase (iNOS), and decreased expression of endothelial nitric oxide synthase (eNOS). However, in HFD+SC there was a significant decrease in body weight gain and an improvement in lipid profile. Histopathological and ultrastructural variations observed in the aorta of the HFD group when treated with ADSCs showed preserved normal histological architecture and reduced atherosclerosis compared with the HFD group. This was evidenced by laboratory, histological, immunohistochemical, and morphometric studies. Thus, ADSCs reduced TC, TG, and LDL, reduced the expression of iNOS, and increased the expression of eNOS. The high-fat diet was likely to cause damage to the wall of blood vessels. Systemically transplanted ADSCs could home to the aorta, and further protect the aorta from HFD-induced damage.
Collapse
Affiliation(s)
- Rania A Galhom
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt; Department of Human Anatomy and Embryology, Faculty of Medicine, Badr University in Cairo (BUC), Egypt.
| | - Saleh Nasser Saleh Ali
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt; Department of Human Anatomy and Embryology, Faculty of Medicine and Health Sciences, Thamar University, Thamar, Yemen.
| | - Magdy Mohamed Omar El-Fark
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | - Mona Hassan Mohammed Ali
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | - Hoda Hassan Hussein
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| |
Collapse
|
48
|
Liu Y, Qiu Z, Shen G, Sun Y, Mei J, Liu Z, Wang L, Li J. Associations between neutrophil-percentage-to-albumin ratio level and all-cause mortality and cardiovascular disease-cause mortality in general population: evidence from NHANES 1999-2010. Front Cardiovasc Med 2024; 11:1393513. [PMID: 39386385 PMCID: PMC11461234 DOI: 10.3389/fcvm.2024.1393513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction Chronic inflammation is a recognized independent risk factor for cardiovascular disease (CVD), highlighting the need for reliable inflammatory indicator to predict CVDs. As an inflammatory indicator which has been proved to have predictive value for prognosis of CVDs, neutrophil percentage-to-albumin ratio (NPAR) has obtained increasing attention, but further research is needed to confirm the relationship with mortality in the general population. Method This prospective cohort study included 21,317 individuals who participated in the National Health and Nutrition Examination Survey (NHANES) from 1999 to 2010, where baseline characteristics and NPAR level were extracted. Data for CVD and all-cause mortality were acquired by linking the cohort database with the National Death Index through December 31, 2019. We employed restricted cubic spline analyses to examine the nonlinear association. Weighted Kaplan-Meier curves with log-rank tests were conducted to access cumulative survival differences across different NPAR results. Multivariable Cox proportional hazards regression models were used to compute hazard ratios and 95% CIs. Receiver Operating Characteristic (ROC) curves were used to compare predictive value of NPAR with systemic immune inflammation index (SII) and neutrophils percent. Results In this cohort study, during 270,014 person-years of follow-up, 4,074 all-cause deaths and 1,116 CVD-cause deaths were documented. NPAR levels exhibited significant nonlinear associations with both CVD-cause (P = 0.018 for nonlinearity) and all-cause mortality (P < 0.001 for nonlinearity). Participants in the highest NPAR tertile had a significantly increased risk of all-cause mortality (HR: 1.46, 95% CI: 1.33-1.61) and CVD-cause mortality (HR: 1.54, 95% CI: 1.32-1.80) compared to those in the lowest tertile in the fully adjusted model, while no association was detected for individuals in the middle tertile. Further ROC analysis confirmed that NPAR had higher predictive value than neutrophil percent segment and SII. Conclusions Elevated NPAR level was significantly associated with an increased risk of all-cause and CVD-cause mortality in general population. The high predictive value of NPAR, combined with the easy-to-calculate property, suggests that its potential as a novel inflammatory indicator is worthy of further investigation.
Collapse
Affiliation(s)
- Yuting Liu
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Zifeng Qiu
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Geng Shen
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - YangYang Sun
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Jiarong Mei
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Zhihao Liu
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Leyi Wang
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Jianping Li
- Department of Cardiology, Peking University First Hospital, Beijing, China
- Institute of Cardiovascular Disease, Peking University First Hospital, Beijing, China
| |
Collapse
|
49
|
Samara I, Moula AI, Moulas AN, Katsouras CS. The Effect of Retinoids in Vascular Smooth Muscle Cells: From Phenotyping Switching to Proliferation and Migration. Int J Mol Sci 2024; 25:10303. [PMID: 39408632 PMCID: PMC11477379 DOI: 10.3390/ijms251910303] [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: 09/05/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Atherosclerosis, a term derived from the Greek "athero" (atheroma) and "sclerosis" (hardening), is a long-standing process that leads to the formation of atheromatous plaques in the arterial wall, contributing to the development of atherosclerotic cardiovascular disease. The proliferation and migration of vascular smooth muscle cells (VSMCs) and the switching of their phenotype play a crucial role in the whole process. Retinoic acid (RA), a natural derivative of vitamin A, has been used in the treatment of various inflammatory diseases and cell proliferation disorders. Numerous studies have demonstrated that RA has an important inhibitory effect on the proliferation, migration, and dedifferentiation of vascular smooth muscle cells, leading to a significant reduction in atherosclerotic lesions. In this review article, we explore the effects of RA on the pathogenesis of atherosclerosis, focusing on its regulatory action in VSMCs and its role in the phenotypic switching, proliferation, and migration of VSMCs. Despite the potential impact that RA may have on the process of atherosclerosis, further studies are required to examine its safety and efficacy in clinical practice.
Collapse
Affiliation(s)
- Ioanna Samara
- Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
| | - Amalia I. Moula
- Department of Surgery, “Achillopouleion” General Hospital, 38222 Volos, Greece;
| | | | - Christos S. Katsouras
- Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
| |
Collapse
|
50
|
Liu Y, Wang Z, Fang L, Xu Y, Zhao B, Kang X, Zhao Y, Han J, Zhang Y, Dong E, Wang N. Deficiency of 5-HT 2B receptors alleviates atherosclerosis by regulating macrophage phenotype through inhibiting interferon signalling. Br J Pharmacol 2024. [PMID: 39232850 DOI: 10.1111/bph.17315] [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: 12/30/2023] [Revised: 06/09/2024] [Accepted: 07/10/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND AND PURPOSE Elevated levels of 5-HT have been correlated with coronary artery disease and cardiac events, suggesting 5-HT is a potential novel factor in the development of atherosclerotic cardiovascular disease. However, the underlying pathological mechanisms of the 5-HT system in atherosclerosis remain unclear. The 5-HT2B receptor (5-HT2BR), which establishes a positive feedback loop with 5-HT, has been identified as a contributor to pathophysiological processes in various vascular disorders. In this study, we investigated the immunological impact of 5-HT2BR in atherosclerosis-prone apolipoprotein E-deficient (ApoE-/-) mice. EXPERIMENTAL APPROACH Plasma levels of 5-HT were measured in mice using an ELISA kit. Atherosclerotic plaque formation, macrophage infiltration and inflammatory signalling were assessed in ApoE-/- mice by employing both pharmacological inhibition and genetic deficiency of 5-HT2BR. Inflammasome activation was elucidated using peritoneal macrophages isolated from 5-HT2BR-deficient mice. KEY RESULTS An upregulation of 5-HT2BR expression was observed in the aortas of ApoE-/- mice, exhibiting a strong correlation with the presence of macrophages in plaques. Atherosclerosis was attenuated in mice through pharmacological inhibition and genetic deficiency of 5-HT2BR. Additionally, a significant reduction in atherosclerotic plaque size was achieved through bone marrow reconstitution with 5-HT2BR-deficient cells. 5-HT2BR-deficient macrophages showed attenuated interferon (IFN) signalling, NLRP3 inflammasome activation, and interleukin-1β release. Moreover, macrophages primed with 5-HT2BR deficiency displayed an anti-inflammatory phenotype. CONCLUSION AND IMPLICATIONS These findings support the hypothesis that 5-HT2BR in macrophages plays a causal role in the development of atherosclerosis, revealing a novel perspective for potential therapeutic strategies in atherosclerosis-related diseases.
Collapse
Affiliation(s)
- Yahan Liu
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Zhipeng Wang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Li Fang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yaohua Xu
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Beilei Zhao
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Xuya Kang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yanqing Zhao
- Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, China
| | - Jintao Han
- Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, China
| | - Yan Zhang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- Institute of Cardiovascular Diseases, The first affiliated Hospital of Dalian Medical University, Dalian, China
| | - Erdan Dong
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital); School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Nanping Wang
- Wuhu Hospital, East China Normal University (ECNU), Wuhu, China
- East China Normal University Health Science Center, Shanghai, China
| |
Collapse
|