1
|
Jiang H, Zhao Y, Jin M, Zhang Z, Wang L, Cheng X, Huang Y, Liu Z, Weng J, Xu S. Colchicine Inhibits Smooth Muscle Cell Phenotypic Switch and Aortic Dissection in Mice-Brief Report. Arterioscler Thromb Vasc Biol 2025; 45:979-984. [PMID: 40177774 DOI: 10.1161/atvbaha.124.322252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 03/20/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND Aortic dissection (AD) is a severe cardiovascular disorder characterized by intimal tearing and subsequent delamination of the aortic wall. The pathogenesis of AD primarily involves the phenotypic switch of vascular smooth muscle cells (VSMCs), degradation of the extracellular matrix, and chronic vascular inflammation. Colchicine, an alkaloid derived from Colchicum autumnale L, is a Food and Drug Administration-approved anti-inflammatory drug with established therapeutic applications in cardiovascular diseases. However, its potential role in modulating the development or progression of AD remains largely unexplored. METHODS To investigate the effects of colchicine on AD, a β-aminopropionitrile-induced AD mouse model was used. Colchicine was administered via oral gavage over a 3-week period to evaluate its impact on the incidence and mortality of AD in male C57BL/6J mice. Transcriptome sequencing was performed to identify genes and signaling pathways regulated by colchicine. Additionally, in vitro experiments using primary rat VSMCs were conducted to elucidate the mechanisms underlying colchicine-mediated regulation of VSMC phenotypic switch. RESULTS Colchicine demonstrated a protective effect against AD by attenuating vascular inflammation and suppressing VSMC phenotypic switch. Mechanistically, colchicine reverses VSMC phenotypic switch at least partially by modulating the expression of myocardin, a key regulator of VSMC contractile phenotype. Transcriptomic analysis further revealed specific genes and pathways influenced by colchicine, providing insights into its molecular mechanisms of action. CONCLUSIONS This study identifies colchicine as a potential therapeutic drug for AD, highlighting its ability to mitigate hallmark pathological processes such as vascular inflammation and VSMC phenotypic switch. These findings offer a foundation base for the repurposed clinical application of colchicine in AD, which warrants further clinical investigation.
Collapse
MESH Headings
- Animals
- Colchicine/pharmacology
- Aortic Dissection/pathology
- Aortic Dissection/prevention & control
- Aortic Dissection/chemically induced
- Aortic Dissection/metabolism
- Aortic Dissection/genetics
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Mice, Inbred C57BL
- Male
- Phenotype
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Disease Models, Animal
- Mice
- Cells, Cultured
- Trans-Activators/metabolism
- Trans-Activators/genetics
- Rats
- Nuclear Proteins/metabolism
- Nuclear Proteins/genetics
- Signal Transduction/drug effects
- Aortic Aneurysm/pathology
- Aortic Aneurysm/chemically induced
- Aortic Aneurysm/prevention & control
- Aortic Aneurysm/metabolism
- Aortic Aneurysm/genetics
- Aminopropionitrile
- Anti-Inflammatory Agents/pharmacology
Collapse
Affiliation(s)
- Hui Jiang
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei (H.J., Y.Z., Z.Z., J.W., S.X.)
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei (H.J.)
| | - Yaping Zhao
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei (H.J., Y.Z., Z.Z., J.W., S.X.)
- Department of Biomedical Sciences, City University of Hong Kong, China (Y.Z., L.W., Y.H.)
| | - Mei Jin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China (M.J., Z.L.)
| | - Zhidan Zhang
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei (H.J., Y.Z., Z.Z., J.W., S.X.)
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, China (Y.Z., L.W., Y.H.)
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (X.C.)
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, China (Y.Z., L.W., Y.H.)
| | - Zhiping Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China (M.J., Z.L.)
| | - Jianping Weng
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei (H.J., Y.Z., Z.Z., J.W., S.X.)
- Anhui Provincial Key Laboratory of Metabolic Health and Panvascular Diseases, Hefei, China (J.W., S.X.)
- Institute of Endocrine and Metabolic Diseases, University of Science and Technology of China, Hefei, Anhui (J.W., S.X.)
| | - Suowen Xu
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei (H.J., Y.Z., Z.Z., J.W., S.X.)
- Anhui Provincial Key Laboratory of Metabolic Health and Panvascular Diseases, Hefei, China (J.W., S.X.)
- Institute of Endocrine and Metabolic Diseases, University of Science and Technology of China, Hefei, Anhui (J.W., S.X.)
| |
Collapse
|
2
|
Bao Z, Li Q, Zhou Z, Yang J, Zhang L. Mitigating Uricase Immunogenicity through Zwitterionic Peptide Fusion for Enhanced Gout Therapy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:12664-12674. [PMID: 40374583 DOI: 10.1021/acs.langmuir.5c00829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2025]
Abstract
Uricase is a promising protein drug for treating gout, but its therapeutic efficacy is significantly limited by its high immunogenicity. In this study, we successfully mitigated the immunogenicity of uricase by fusing zwitterionic peptides (repeated VPKEG sequences) with varying lengths through synthetic biology. The results show that longer zwitterionic peptides can more efficiently improve the enzymatic activity, enhance the substrate affinity, prolong the blood circulation time, and reduce the antibody response. This is likely due to the longer zwitterionic peptides providing a more extensive protective hydration layer, which can shield the immunogenic sites of uricase. As a result, U-(VPKEG)60, the uricase with the longest zwitterionic peptide, significantly enhances therapeutic effects for rat gout models. Moreover, it also exhibits excellent biocompatibility with no hemolysis and negligible cytotoxicity to liver, heart, and kidney functions. Our findings provide new insights into the development of low-immunogenicity uricase through synthetic biology.
Collapse
Affiliation(s)
- Zhun Bao
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300072, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| | - Qingsi Li
- Department of Biological Engineering, Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), School of Synthetic Biology and Biomanufacturing, Tianjin University, Tianjin 300350, China
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300072, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| | - Zhaoyu Zhou
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300072, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| | - Jing Yang
- Department of Biological Engineering, Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), School of Synthetic Biology and Biomanufacturing, Tianjin University, Tianjin 300350, China
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300072, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| | - Lei Zhang
- Department of Biological Engineering, Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), School of Synthetic Biology and Biomanufacturing, Tianjin University, Tianjin 300350, China
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300072, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| |
Collapse
|
3
|
Chang T, He Y, Ling Y, Zhou Z, Lu H, Qiao L, Ruan X, Li Z, Liu J, Liu Y, Chen J. Relationship Between the Aggregate Index of Systemic Inflammation and the prognosis in Patients With CAD and CKD. Angiology 2025:33197251338428. [PMID: 40413562 DOI: 10.1177/00033197251338428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
Chronic kidney disease (CKD) is a complex disease that may exacerbate the inflammatory state in patients with coronary artery disease (CAD). The predictive value of the Aggregate Index of Systemic Inflammation (AISI) for individuals with CAD and chronic kidney disease (CKD) remains unclear. The data for this study were obtained from the Cardiorenal Improvement II (CIN-II) cohort. Patients (n = 15,133) with CAD combined with CKD were included. The AISI was calculated using the formula: neutrophil*platelet*monocyte/lymphocyte counts. During a median follow-up of 4.26 years, 3979 deaths occurred, including 2239 cardiovascular (CV) deaths. With adjustment for potential confounding factors, the level of AISI was associated with a higher risk of all-cause mortality (Q [quartile]2, Q3, Q4: hazard ratio [HR] [95 CI%] = 1.20 [1.09-1.32], 1.34 [1.22-1.48], and 1.59 [1.44-1.75], respectively; ptrend < 0.001) and CV mortality (Q2, Q3, Q4: HR [95 CI%] = 1.27 [1.11-1.45], 1.50 [1.32-1.71], 1.78 [1.55-2.03], respectively; ptrend < 0.001). Elevated AISI level was independently associated with increased risks of all-cause and CV-specific mortality in patients with CAD and CKD.
Collapse
Affiliation(s)
- Tian Chang
- School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yibo He
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yihang Ling
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ziyou Zhou
- School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Hongyu Lu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Linfang Qiao
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xianlin Ruan
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zeliang Li
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jin Liu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yong Liu
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jiyan Chen
- School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
4
|
Wang X, Chen L, Wei J, Zheng H, Zhou N, Xu X, Deng X, Liu T, Zou Y. The immune system in cardiovascular diseases: from basic mechanisms to therapeutic implications. Signal Transduct Target Ther 2025; 10:166. [PMID: 40404619 PMCID: PMC12098830 DOI: 10.1038/s41392-025-02220-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/22/2024] [Accepted: 03/20/2025] [Indexed: 05/24/2025] Open
Abstract
Immune system plays a crucial role in the physiological and pathological regulation of the cardiovascular system. The exploration history and milestones of immune system in cardiovascular diseases (CVDs) have evolved from the initial discovery of chronic inflammation in atherosclerosis to large-scale clinical studies confirming the importance of anti-inflammatory therapy in treating CVDs. This progress has been facilitated by advancements in various technological approaches, including multi-omics analysis (single-cell sequencing, spatial transcriptome et al.) and significant improvements in immunotherapy techniques such as chimeric antigen receptor (CAR)-T cell therapy. Both innate and adaptive immunity holds a pivotal role in CVDs, involving Toll-like receptor (TLR) signaling pathway, nucleotide-binding oligomerization domain-containing proteins 1 and 2 (NOD1/2) signaling pathway, inflammasome signaling pathway, RNA and DNA sensing signaling pathway, as well as antibody-mediated and complement-dependent systems. Meanwhile, immune responses are simultaneously regulated by multi-level regulations in CVDs, including epigenetics (DNA, RNA, protein) and other key signaling pathways in CVDs, interactions among immune cells, and interactions between immune and cardiac or vascular cells. Remarkably, based on the progress in basic research on immune responses in the cardiovascular system, significant advancements have also been made in pre-clinical and clinical studies of immunotherapy. This review provides an overview of the role of immune system in the cardiovascular system, providing in-depth insights into the physiological and pathological regulation of immune responses in various CVDs, highlighting the impact of multi-level regulation of immune responses in CVDs. Finally, we also discuss pre-clinical and clinical strategies targeting the immune system and translational implications in CVDs.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Liming Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianming Wei
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Hao Zheng
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Ning Zhou
- Department of Cardiovascular Medicine, Anzhen Hospital Affiliated to Capital Medical University, Beijing, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Deng
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tao Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China.
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Jiangsu, Nanjing, China.
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
- Institutes of Advanced Medical Sciences and Huaihe Hospital, Henan University, Kaifeng, Henan, China.
| |
Collapse
|
5
|
Trube J, Sabina M, Khanani A, Hernandez K, Khan Z, Bizanti A. Colchicine therapy in cardiovascular medicine: A literature review. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2025; 52:100525. [PMID: 40124716 PMCID: PMC11929935 DOI: 10.1016/j.ahjo.2025.100525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/14/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
Introduction Inflammation is a key risk factor in cardiovascular diseases, such as atherosclerosis, and has been linked to increased mortality following myocardial infarction. While inflammation promotes tissue repair, sustained inflammation can drive adverse cardiac remodeling, fibrosis, and impaired contractility, resulting in poorer outcomes. This maladaptive remodeling, compounded by oxidative stress, also predisposes patients to cardiovascular diseases. Colchicine has shown anti-inflammatory benefits in cardiovascular disease, but its role in individual diseases remains unclear. This literature review seeks to understand and evaluate the clinical trials evaluating colchicine in cardiovascular treatment. Methods A literature search identified randomized controlled trials (RCTs) evaluating colchicine in cardiovascular disease including coronary artery disease, post-myocardial infarction treatment, atrial fibrillation, heart failure, and stroke. Conclusions Colchicine has been studied across many cardiovascular conditions including atrial fibrillation (AF), coronary artery disease (CAD), post-myocardial infarction treatment, heart failure (HF) and stroke; however, evidence of its clinical effectiveness remains mixed. While colchicine has shown prfomise in reducing recurrent cardiovascular events in stable CAD, its impact in postoperative AF prevention, acute coronary syndrome (ACS), HF, and stroke prevention is limited.
Collapse
Affiliation(s)
- Jennifer Trube
- Lakeland Regional Health Medical Center, Lakeland, FL, USA
| | - Michael Sabina
- Lakeland Regional Health Medical Center, Lakeland, FL, USA
| | - Aqeel Khanani
- Lakeland Regional Health Medical Center, Lakeland, FL, USA
| | | | - Zoya Khan
- Lakeland Regional Health Medical Center, Lakeland, FL, USA
| | - Anas Bizanti
- Lakeland Regional Health Medical Center, Lakeland, FL, USA
| |
Collapse
|
6
|
Suryono S, Amien MI, Tohari AI, Saputra AD, Hidayat MRF, Ramadhan HF. In silico studies on quercetin, myricetin, and kaempferol in inhibiting TGF-β1 and galectin- 3 for cardiac fibrosis management. NARRA J 2025; 5:e1310. [PMID: 40352218 PMCID: PMC12059959 DOI: 10.52225/narra.v5i1.1310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 12/04/2024] [Indexed: 05/14/2025]
Abstract
Cardiac fibrosis remains as the leading cause of death worldwide and is often associated with elevated levels of transforming growth factor-β 1 (TGF-β1) and galectin-3, making them potential therapeutic targets. Recent studies revealed that quercetin, myricetin, and kaempferol have the biological effect for several cardiovascular diseases. However, the investigation into this topic through molecular models and analysis remain unexplored. The aim of this study was to evaluate the potential effect of quercetin, myricetin, and kaempferol which targeted TGF-β1 and galectin-3. In this study, quercetin, myricetin, and kaempferol roled as the tested ligands. Subsequently, colchicine and native ligand acted as control ligands that were screened through molecular docking against TGF-β1 and galectin-3 using AutoDock tools to identify the potential inhibitor. The stability of ligand- receptor complexes was assessed through molecular dynamic (MD) simulations using NMAD. Absorption, Distribution, Metabolism, Excretion and toxicity (ADMET) prediction were also performed using ADMETlab 2.0. Molecular docking analysis revealed that quercetin, myricetin, and kaempferol exhibited strong binding affinity which are -8.9 kcal/mol, -8.5 kcal/mol, -7.6 kcal/mol respectively with TGF-β1, and -7.5 kcal/mol, -7.0 kcal/mol, -5.7 kcal/mol respectively with galetcin-3; low inhibition constant (Ki); and stable interaction with the active sites of TGF-β1 and galectin-3. MD simulations confirmed the stability and compactness of the ligand-receptor complexes. ADMET analysis also showed high Plasma Protein Binding (PPB) values (quercetin: 95%, myricetin: 92%, and kaempferol: 97%) and moderate clearance values (quercetin: 8.284%, myricetin, and 7.716%, kaempferol: 6.868%) for the tested compounds. In conclusion, the in silico analyses suggested that quercetin, myricetin, and kaempferol are promising for cardiac fibrosis therapies by inhibiting TGF-β1 and galectin-3.
Collapse
Affiliation(s)
- Suryono Suryono
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Jember, Jember, Indonesia
- Department of Cardiology and Vascular Medicine, Dr. Soebandi General Hospital, Jember, Indonesia
| | | | | | | | | | | |
Collapse
|
7
|
Astiawati T, Rohman MS, Wihastuti T, Sujuti H, Endharti A, Sargowo D, Oceandy D, Lestari B, Triastuti E, Nugraha RA. The Emerging Role of Colchicine to Inhibit NOD-like Receptor Family, Pyrin Domain Containing 3 Inflammasome and Interleukin-1β Expression in In Vitro Models. Biomolecules 2025; 15:367. [PMID: 40149903 PMCID: PMC11940210 DOI: 10.3390/biom15030367] [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: 01/29/2025] [Revised: 02/22/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
While the beneficial effects of colchicine on inflammation and infarcted myocardium have been documented, its impact on cardiac fibroblast activation in the context of myocardial infarction (MI) remains unknown. This study aimed to investigate the effect of colchicine on the regulation of NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome activation and Interleukin-1β (IL-1β) expression in fibroblasts. 3T3 fibroblasts were exposed to 600 μM CoCl2 for 24 h to simulate hypoxia, with normoxic cells as controls. Colchicine (1 μM) was administered for 24 h. ASC-NLRP3 colocalization and IL-1β expression were evaluated using immunofluorescence and flow cytometry, respectively. Data were analyzed using t-tests and one-way ANOVA with post hoc tests. Hypoxia treatment significantly induced apoptosis-associated speck-like protein containing a CARD (ASC)-NLRP3 colocalization (p < 0.05). Colchicine treatment of hypoxic 3T3 cells reduced ASC-NLRP3 colocalization, although this reduction was not statistically significant. Additionally, IL-1β expression was significantly inhibited in colchicine-treated hypoxic 3T3 cells compared to those treated with placebo (p < 0.05). The findings of this study indicate that colchicine treatment inhibits the activation of the NLRP3 inflammasome by disrupting the colocalization of ASC and NLRP3, thereby reducing IL-1β expression in CoCl2-treated 3T3 cells.
Collapse
Affiliation(s)
- Tri Astiawati
- Doctoral Program of Medical Science, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
- Department of Cardiology, Doctor Iskak General Hospital, Tulungagung 62233, Indonesia
| | - Mohammad Saifur Rohman
- Department of Cardiology and Cardiovascular Medicine, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
- Cardiovascular Research Centre, Brawijaya University, Malang 65145, Indonesia
| | - Titin Wihastuti
- Department of Biomedical, Nursing Science, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Hidayat Sujuti
- Department of Biochemistry, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Agustina Endharti
- Doctoral Program of Medical Science, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Djanggan Sargowo
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Delvac Oceandy
- Division of Cardiovascular Science, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK;
| | - Bayu Lestari
- Department of Pharmacology, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK;
| | - Efta Triastuti
- Department of Pharmacy, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Ricardo Adrian Nugraha
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Airlangga—Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia;
| |
Collapse
|
8
|
Seecheran N, Grimaldos K, McCallum P, Ramcharan P, Kawall J, Katwaroo A, Grimaldos G, Seecheran V, Jagdeo CL, Rafeeq S, Seecheran R, Leyva Quert A, Ali N, Peram L, Motilal S, Ramtahal R, Bhagwandass N, Giddings S, Ramlackhansingh A, Sandy S. The Effect of Colchicine on Platelet Function Profiles in Patients with Stable Coronary Artery Disease: The ECLIPSE Pilot Study. Cardiol Ther 2025; 14:87-100. [PMID: 39826082 PMCID: PMC11893921 DOI: 10.1007/s40119-024-00393-2] [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: 10/09/2024] [Accepted: 12/04/2024] [Indexed: 01/20/2025] Open
Abstract
INTRODUCTION This prospective, single-arm pharmacodynamic study assessed the effect of colchicine (COLC) [Strides Pharma UK Ltd, Watford, Hertfordshire, England] 0.5 mg administered orally once daily for 14 days on platelet reactivity with respect to aspirin reaction units (ARUs) and P2Y12 reaction units (PRUs). METHODS Twenty-two patients with stable coronary artery disease (CAD) on dual antiplatelet therapy (DAPT) with daily maintenance aspirin and clopidogrel were recruited. Baseline platelet function was evaluated with the VerifyNow™ ARU and PRU assays (Werfen, Bedford, MA, USA) and assessed post-completion of COLC 0.5 mg once daily for 14 days. RESULTS In this study, the median ARU baseline score was 463, and post-COLC it was 436, which was not statistically significant (p = 0.485). The mean difference in scores was -18.31 (95% confidence interval [CI] -74.34 to 37.71, p = 0.504). At baseline, 27.3% of the patients had "aspirin resistance" or were non-responders, compared to 13.6% post-COLC (p = 0.423). The median baseline PRU score was 210, and post-COLC it was 199, which was also not statistically significant (p = 0.581). The mean difference in scores was -7.31 (95% CI -31.1 to 16.5, p = 0.530). At baseline, 50% of the patients had "clopidogrel resistance" or were non-responders, compared to 45.5% post-COLC (p = 0.999). Two patients experienced mild gastrointestinal upset during the trial without interruption of COLC, and there were no serious adverse events or treatment-emergent adverse events. CONCLUSIONS There were no significant differences in ARUs and PRUs post-COLC trial in patients with stable CAD. This pilot pharmacodynamic study could be clinically informative in patients on DAPT. Further studies are required to confirm these exploratory findings. TRIAL REGISTRATION ClinicalTrials.gov identifier, NCT06567678, prospectively registered 20/8/2024.
Collapse
Affiliation(s)
- Naveen Seecheran
- Adult Medicine, Department of Clinical Medical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago.
| | - Kathryn Grimaldos
- Department of Medicine, North Central Regional Health Authority, 2nd Floor, Building #67, Eric Williams Medical Sciences Complex, Mt. Hope, Trinidad and Tobago
| | - Penelope McCallum
- Department of Medicine, North Central Regional Health Authority, 2nd Floor, Building #67, Eric Williams Medical Sciences Complex, Mt. Hope, Trinidad and Tobago
| | - Priya Ramcharan
- Department of Medicine, North Central Regional Health Authority, 2nd Floor, Building #67, Eric Williams Medical Sciences Complex, Mt. Hope, Trinidad and Tobago
| | - Jessica Kawall
- Department of Medicine, Trinidad Institute of Medical Technology, St. Augustine, Trinidad and Tobago
| | - Arun Katwaroo
- Department of Medicine, Trinidad Institute of Medical Technology, St. Augustine, Trinidad and Tobago
| | - Gabriella Grimaldos
- Department of Medicine, North Central Regional Health Authority, 2nd Floor, Building #67, Eric Williams Medical Sciences Complex, Mt. Hope, Trinidad and Tobago
| | - Valmiki Seecheran
- Department of Medicine, North Central Regional Health Authority, 2nd Floor, Building #67, Eric Williams Medical Sciences Complex, Mt. Hope, Trinidad and Tobago
| | - Cathy-Lee Jagdeo
- Department of Medicine, North Central Regional Health Authority, 2nd Floor, Building #67, Eric Williams Medical Sciences Complex, Mt. Hope, Trinidad and Tobago
| | - Salma Rafeeq
- Department of Medicine, North Central Regional Health Authority, 2nd Floor, Building #67, Eric Williams Medical Sciences Complex, Mt. Hope, Trinidad and Tobago
| | - Rajeev Seecheran
- Department of Medicine, The University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Abel Leyva Quert
- Department of Medicine, North Central Regional Health Authority, 2nd Floor, Building #67, Eric Williams Medical Sciences Complex, Mt. Hope, Trinidad and Tobago
| | - Nafeesah Ali
- Department of Medicine, North Central Regional Health Authority, 2nd Floor, Building #67, Eric Williams Medical Sciences Complex, Mt. Hope, Trinidad and Tobago
| | - Lakshmipathi Peram
- Department of Medicine, North Central Regional Health Authority, 2nd Floor, Building #67, Eric Williams Medical Sciences Complex, Mt. Hope, Trinidad and Tobago
| | - Shastri Motilal
- Adult Medicine, Department of Clinical Medical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Rishi Ramtahal
- Adult Medicine, Department of Clinical Medical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Neal Bhagwandass
- Adult Medicine, Department of Clinical Medical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Stanley Giddings
- Adult Medicine, Department of Clinical Medical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Anil Ramlackhansingh
- Adult Medicine, Department of Clinical Medical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Sherry Sandy
- Adult Medicine, Department of Clinical Medical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| |
Collapse
|
9
|
Astiawati T, Rohman MS, Wihastuti T, Sujuti H, Endharti AT, Sargowo D, Oceandy D, Lestari B, Triastuti E, Nugraha RA. Efficacy of Colchicine in Reducing NT-proBNP, Caspase-1, TGF-β, and Galectin-3 Expression and Improving Echocardiography Parameters in Acute Myocardial Infarction: A Multi-Center, Randomized, Placebo-Controlled, Double-Blinded Clinical Trial. J Clin Med 2025; 14:1347. [PMID: 40004876 PMCID: PMC11856086 DOI: 10.3390/jcm14041347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/07/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Caspase-1 (reflects NOD-like receptor protein 3 inflammasome activity), transforming growth factor-β (TGF-β), and Galectin-3 play significant roles in post-AMI fibrosis and inflammation. Recently, colchicine was shown to dampen inflammation after AMI; however, its direct benefit remains controversial. Objectives: This study aimed to analyze the benefit of colchicine in reducing NT-proBNP, Caspase-1, TGF-β,and Galectin-3 expression and improving systolic-diastolic echocardiography parameters among AMI patients. Methods: A double-blinded, placebo-controlled, randomized, multicenter clinical trial was conducted at three hospitals in East Java, Indonesia: Dr. Saiful Anwar Hospital Malang, Dr. Soebandi Hospital Jember, and Dr. Iskak Hospital Tulungagung, between 1 June and 31 December 2023. A total of 161 eligible AMI subjects were randomly allocated 1:1 to colchicine (0.5 mg daily) or standard treatment for 30 days. Caspase-1, TGF-β, and Galectin-3 were tested on day 1 and day 5 by ELISA, while NT-proBNP was tested on days 5 and 30. Transthoracic echocardiography was also performed on day 5 and day 30. Results: By day 30, no significant improvements in systolic-diastolic echocardiography parameters had been shown in the colchicine group. However, colchicine reduced the level of NT-proBNP on day 30 more than placebo (ΔNT-proBNP: -73.74 ± 87.53 vs. -75.75 ± 12.44 pg/mL; p < 0.001). Moreover, colchicine lowered the level of Caspase-1 expression on day 5 and the levels of TGF-β and Galectin-3 expression on day 1. Conclusions: Colchicine can reduce NT-proBNP, Caspase-1, TGF-β, and Galectin-3 expression significantly among AMI patients. Colchicine administration was capable of reducing post-AMI inflammation, ventricular dysfunction, and heart failure but did not improve systolic-diastolic echocardiography parameters (ClinicalTrials.gov identifier: NCT06426537).
Collapse
Affiliation(s)
- Tri Astiawati
- Doctoral Program of Medical Science, Brawijaya University, Malang 65145, Indonesia
- Department of Cardiology and Vascular Medicine, Dr. Iskak General Hospital, Tulungagung 66223, Indonesia
| | - Mohammad Saifur Rohman
- Department of Cardiology and Cardiovascular Medicine, Faculty of Medicine, Brawijaya University, Dr. Saiful Anwar General Hospital, Malang 65145, Indonesia;
- Cardiovascular Research Centre, Universitas Brawijaya, Malang 65145, Indonesia
| | - Titin Wihastuti
- Department of Nursing Science, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Hidayat Sujuti
- Department of Biochemistry, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Agustina Tri Endharti
- Department of Parasitology, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Djanggan Sargowo
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Brawijaya University, Dr. Saiful Anwar General Hospital, Malang 65145, Indonesia;
| | - Delvac Oceandy
- Division of Cardiovascular Science, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK;
| | - Bayu Lestari
- Department of Pharmacology, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Efta Triastuti
- Department of Pharmacy, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Ricardo Adrian Nugraha
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo General Hospital, Surabaya 60286, Indonesia;
| |
Collapse
|
10
|
Corbali O, Nahab F. Colchicine in stroke prevention: potential benefits and limitations of an anti-inflammatory therapy. Front Neurol 2025; 16:1529458. [PMID: 40017533 PMCID: PMC11864930 DOI: 10.3389/fneur.2025.1529458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 01/29/2025] [Indexed: 03/01/2025] Open
Affiliation(s)
| | - Fadi Nahab
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, United States
| |
Collapse
|
11
|
Escalera E, Saver JL. Magnitude of effect of low dose colchicine, a newly food and drug administration approved treatment for stroke prevention. J Stroke Cerebrovasc Dis 2025; 34:108186. [PMID: 39667442 DOI: 10.1016/j.jstrokecerebrovasdis.2024.108186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 11/20/2024] [Accepted: 12/09/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND As the Food and Drug Administration in June 2023 approved low dose colchicine for primary prevention of stroke and other cardiovascular events, an updated meta-analysis of stroke outcomes in randomized trials would help inform clinical practice.**** METHODS: Systematic, study-level meta-analysis of randomized clinical trials of long-term colchicine in patients with established atherosclerotic cardiovascular disease (ASCVD, preponderantly primary prevention for stroke) or following non-cardioembolic ischemic stroke/transient ischemic attack (secondary prevention). Heterogeneity was assessed with the I2 statistic and Cochrane's Q and potential bias assessed with the Risk of Bias 2.0 scale. RESULTS Six randomized control trials met selection criteria, enrolling 14,987 patients (7495 colchicine, 7492 placebo), with median follow-up 26.3 months. Colchicine dosage in all trials was 0.5 mg once-daily. Across all trials, colchicine treatment produced a 28 % relative risk reduction in stroke (1.77 % vs 2.54 %, risk ratio (RR)=0.72, 95 %CI: 0.58-0.89; p = 0.003) and a comparable relative reduction on major adverse cardiovascular events. There was potential heterogeneity by subgroup (pinteraction = 0.06), with a stronger relative reduction for stroke in the five ASCVD trials (RR=0.48, 95 %CI:0.30-0.77; p = 0.003) than the non-cardioembolic ischemic stroke/TIA population (RR=0.80, 95 %CI:0.63-1.02; p = 0.07). Colchicine was associated with a small, non-significant increase in all-cause mortality (RR: 1.09; 95 %Cl: 0.85-1.40, p = 0.49) but not cardiovascular death (RR: 0.92; 95 %Cl: 0.65-1.29, p = 0.61). CONCLUSION Low-dose colchicine treatment decreases stroke and major adverse cardiovascular event risk in patients with ASCVD and potentially in patients following a non-cardioembolic ischemic stroke/TIA. Among every 1000 patients treated over 2 years, approximately 6.6 strokes and 24 major adverse cardiovascular events are avoided.
Collapse
Affiliation(s)
- Erica Escalera
- David Geffen School of Medicine, University of California, Los Angeles, CA USA.
| | - Jeffrey L Saver
- Comprehensive Stroke Center and Department of Neurology, Ronald Reagan - UCLA Medical Center, Los Angeles, CA USA.
| |
Collapse
|
12
|
Dong L, Dong C, Yu Y, Jiao X, Zhang X, Zhang X, Li Z. Transcriptomic analysis of Paraoxonase 1 expression in hepatocellular carcinoma and its potential impact on tumor immunity. Clin Transl Oncol 2025; 27:612-629. [PMID: 39031295 DOI: 10.1007/s12094-024-03598-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: 05/06/2024] [Accepted: 07/01/2024] [Indexed: 07/22/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is characterized by a complex pathogenesis that confers aggressive malignancy, leading to a lack of dependable biomarkers for predicting invasion and metastasis, which results in poor prognoses in patients with HCC. Glycogen storage disease (GSD) is an uncommon metabolic disorder marked by hepatomegaly and liver fibrosis. Notably, hepatic adenomas in GSD patients present a heightened risk of malignancy compared to those in individuals without the disorder. In this investigation, PON1 emerged as a potential pivotal gene for HCC through bioinformatics analysis. METHODS Transcriptomic profiling data of liver cancer were collected and integrated from TCGA and GEO databases. Bioinformatics analysis was conducted to identify mutated mRNAs associated with GSD, and the PON1 gene was selected as a key gene. Patients were grouped based on the expression levels of PON1, and differences in clinical characteristics, biological pathways, immune infiltration, and expression of immune checkpoints were compared. RESULTS The expression levels of the PON1 gene showed significant differences between the high-expression group and the low-expression group in HCC patients. Further analysis indicated that the PON1 gene at different expression levels might influence the clinical manifestations, biological processes, immune infiltration, and expression of immune checkpoints in HCC. Additionally, immunohistochemistry (IHC) results revealed high expression of PON1 in normal tissues and low expression in HCC tissues. These findings provide important clues and future research directions for the early diagnosis, prognosis, immunotherapy, and potential molecular interactions of HCC. CONCLUSION Our investigation underscores the noteworthy prognostic significance of PON1 in HCC, suggesting its potential pivotal role in modulating tumor progression and immune cell infiltration. These findings establish PON1 as a novel tumor biomarker with significant implications for the prognosis, targeted therapy, and immunotherapy of patients with HCC.
Collapse
Affiliation(s)
- Linhuan Dong
- Department of General surgery, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443000, China
| | - Changjun Dong
- Department of General surgery, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443000, China
| | - Yunlin Yu
- Department of General surgery, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443000, China
| | - Xin Jiao
- Department of General surgery, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443000, China
| | - Xiangwei Zhang
- Department of General surgery, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443000, China
| | - Xianlin Zhang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002, China.
| | - Zheng Li
- Department of General surgery, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443000, China.
| |
Collapse
|
13
|
Di Stolfo G, Mastroianno M, Pacilli MA, De Luca G, Coli CR, Bevere EML, Pacilli G, Potenza DR, Mastroianno S. Role of C-Reactive Protein as a Predictor of Early Revascularization and Mortality in Advanced Peripheral Arterial Disease. J Clin Med 2025; 14:815. [PMID: 39941486 PMCID: PMC11818854 DOI: 10.3390/jcm14030815] [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: 12/17/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Background: Elevated high-sensitivity C-reactive protein (hsCRP) levels are associated with poor cardiovascular outcomes, particularly in patients with advanced peripheral arterial disease (PAD). This study aimed to assess the impact of hsCRP on clinical characteristics and long-term outcomes in a cohort of PAD patients. Methods: A total of 346 patients with advanced PAD were enrolled and stratified into two groups based on their median hsCRP level (Group 1: <0.32 mg/dL, Group 2: >0.32 mg/dL). The patients were followed for a mean of 102.70 ± 44.13 months. Their clinical characteristics, comorbidities, and long-term cardiovascular events, including myocardial and/or peripheral revascularization, ischemia, and death, were analyzed. This study evaluated two composite endpoints: major adverse cardiovascular events (MACEs) and major adverse peripheral events (MAPEs). MACEs comprised fatal cardiovascular events, cerebral ischemia, cardiac infarction, myocardial revascularization, acute peripheral arterial occlusion, and peripheral reperfusion. MAPEs included carotid reperfusion, acute peripheral arterial occlusion, and lower limb revascularization. Results: The patients in Group 2 had a higher body mass index, waist circumference, and waist-hip ratio compared to those in Group 1 (all p < 0.05). Inflammatory markers, including fibrinogen and the erythrocyte sedimentation rate, were significantly elevated in Group 2 (both p < 0.01). While the overall incidence of peripheral revascularization was similar between groups, these interventions occurred significantly earlier in Group 2 (28.24 ± 38.87 months vs. 67.04 ± 49.97 months, p = 0.004; HR: 2.015, 95% CI: 1.134-3.580, p = 0.017). The MAPEs were comparable in number, but occurred earlier in Group 2 (36.60 ± 37.35 months vs. 66.19 ± 48.18 months, p < 0.01; HR: 1.99, 95% CI: 1.238-3.181, p = 0.004). Similarly, the MACEs had an earlier onset in Group 2 (40.31 ± 38.95 months vs. 55.89 ± 46.33 months, p = 0.04; HR: 1.62, 95% CI: 0.983-1.987, p = 0.062). A total of 169 deaths were recorded during the follow-up. Group 2 exhibited a significantly higher mortality rate (56% vs. 42%, p < 0.01) and an earlier trend in mortality (76.58 ± 43.53 months vs. 84.86 ± 5.18 months), although this difference did not reach statistical significance (p = 0.22). Conclusions: Elevated hsCRP levels (>0.32 mg/dL) are associated with a worse clinical profile and earlier adverse events in patients with advanced PAD. Group 2 experienced significantly earlier peripheral revascularization, MACEs, and MAPEs. The mortality rates were also significantly higher, highlighting the prognostic value of hsCRP in this population.
Collapse
Affiliation(s)
- Giuseppe Di Stolfo
- Cardiovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (M.A.P.); (G.D.L.); (C.R.C.); (E.M.L.B.); (D.R.P.); (S.M.)
| | - Mario Mastroianno
- Scientific Direction, Fondazione IRCCS Casa Sollievodella Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - Michele Antonio Pacilli
- Cardiovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (M.A.P.); (G.D.L.); (C.R.C.); (E.M.L.B.); (D.R.P.); (S.M.)
| | - Giovanni De Luca
- Cardiovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (M.A.P.); (G.D.L.); (C.R.C.); (E.M.L.B.); (D.R.P.); (S.M.)
| | - Carlo Rosario Coli
- Cardiovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (M.A.P.); (G.D.L.); (C.R.C.); (E.M.L.B.); (D.R.P.); (S.M.)
| | - Ester Maria Lucia Bevere
- Cardiovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (M.A.P.); (G.D.L.); (C.R.C.); (E.M.L.B.); (D.R.P.); (S.M.)
| | - Gabriella Pacilli
- Emergency Department, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - Domenico Rosario Potenza
- Cardiovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (M.A.P.); (G.D.L.); (C.R.C.); (E.M.L.B.); (D.R.P.); (S.M.)
| | - Sandra Mastroianno
- Cardiovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (M.A.P.); (G.D.L.); (C.R.C.); (E.M.L.B.); (D.R.P.); (S.M.)
| |
Collapse
|
14
|
Hamed EFA, Taha AA, Ghany SMA, Al-Attar ASR, Fawzy EM. Colchicine: A Dual Therapeutic Target for Trichinellosis. Acta Parasitol 2025; 70:42. [PMID: 39853509 PMCID: PMC11761976 DOI: 10.1007/s11686-024-00979-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/08/2024] [Indexed: 01/26/2025]
Abstract
PURPOSE Trichinellosis affects around 11 million people globally. Treatments for this medical condition are limited by adverse effects and resistance, emphasising the importance of effective and safe therapies. Consequentially, we sought to study colchicine's synergistic effects with atorvastatin or acetazolamide in the treatment of Trichinella spiralis (T. spiralis)-infected mice. METHODS Seventy mice were evenly divided into two groups (a and b) of 35 each. During the intestinal phase, group (a) began therapy on the second day post-infection (dpi) and lasted four days. Group (b) had treatment for four weeks during the muscle phase, beginning on the 12th dpi. While the other five infected groups received atorvastatin, colchicine, acetazolamide, a combination of acetazolamide and colchicine, or none, one group of infected mice received no treatment at all as a negative control. The efficacy was assessed by parasite count, histopathology and scanning electron microscopy. RESULTS Our data revealed that the combination treatment lowered T. spiralis adult worm and larvae counts in infected animals. Moreover, it restored the normal intestinal and muscular architecture, reduced edema, and alleviated inflammation, as demonstrated by reduced inflammatory infiltrate. Scanning electron microscopic examination of adults and larvae verified our findings. CONCLUSION Adjuvant treatment with colchicine as an antifibrotic can help treat muscle trichinellosis by reducing the production of fibrous tissue. This might help to enhance treatment results by enabling the admission of larvicidal medications and, as a result, reducing the number of larvae in the muscle, which together form the basis of pathology and can be debilitating for the patient.
Collapse
Affiliation(s)
- Enas Fakhry Abdel Hamed
- Department of Medical Parasitology, Faculty of Medicine, Zagazig University, El Kawmia Square, Zagazig, Sharkia Governorate, Egypt.
| | - Afaf A Taha
- Department of Medical Parasitology, Faculty of Medicine, Zagazig University, El Kawmia Square, Zagazig, Sharkia Governorate, Egypt
| | - Shereen M Abdel Ghany
- Department of Medical Parasitology, Faculty of Medicine, Zagazig University, El Kawmia Square, Zagazig, Sharkia Governorate, Egypt
| | - Al-Sayed R Al-Attar
- Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Sharkia, Egypt
| | - Eman M Fawzy
- Department of Medical Parasitology, Faculty of Medicine, Zagazig University, El Kawmia Square, Zagazig, Sharkia Governorate, Egypt
| |
Collapse
|
15
|
Yan M, Wang Z, Qiu Z, Cui Y, Xiang Q. Platelet signaling in immune landscape: comprehensive mechanism and clinical therapy. Biomark Res 2024; 12:164. [PMID: 39736771 DOI: 10.1186/s40364-024-00700-y] [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: 09/29/2024] [Accepted: 11/28/2024] [Indexed: 01/01/2025] Open
Abstract
Platelets are essential for blood clotting and maintaining normal hemostasis. In pathological conditions, platelets are increasingly recognized as crucial regulatory factors in various immune-mediated inflammatory diseases. Resting platelets are induced by various factors such as immune complexes through Fc receptors, platelet-targeting autoantibodies and other platelet-activating stimuli. Platelet activation in immunological processes involves the release of immune activation stimuli, antigen presentation and interaction with immune cells. Platelets participate in both the innate immune system (neutrophils, monocytes/macrophages, dendritic cells (DCs) and Natural Killer (NK) cells and the adaptive immune system (T and B cells). Clinical therapeutic strategies include targeting platelet activation, platelet-immune cell interaction and platelet-endothelial cell interaction, which display positive development prospects. Understanding the mechanisms of platelets in immunity is important, and developing targeted modulations of these mechanisms will pave the way for promising therapeutic strategies.
Collapse
Affiliation(s)
- Mengyao Yan
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China
| | - Zhe Wang
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China
| | - Zhiwei Qiu
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China
| | - Yimin Cui
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China.
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China.
| | - Qian Xiang
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China.
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China.
| |
Collapse
|
16
|
Zhang N, Zhao L, Li J, Li H, Chen Y. Harnessing Nanotechnology for Gout Therapy: Colchicine-Loaded Nanoparticles Regulate Macrophage Polarization and Reduce Inflammation. Biomater Res 2024; 28:0089. [PMID: 39665079 PMCID: PMC11632155 DOI: 10.34133/bmr.0089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 09/13/2024] [Accepted: 09/24/2024] [Indexed: 12/13/2024] Open
Abstract
Gout is a disease caused by hyperuricemia, characterized by inflammation reactions triggered by macrophage polarization. Colchicine is a commonly used drug for gout treatment, but its mechanism of action remains unclear. The aim of this study was to investigate the regulatory effect of colchicine on macrophage polarization to enhance the therapeutic effectiveness against gout inflammation. To accomplish this, a mouse model was established, and peripheral blood mononuclear cell samples were collected. Single-cell RNA sequencing was employed to reveal cellular heterogeneity and identify key genes. Molecular docking and experimental validation were performed to confirm the binding between the key genes and colchicine. Lentiviral intervention and biochemical indicator detection were conducted to assess the impact of key genes on gout mice. Additionally, the therapeutic effect of colchicine incorporated into neutrophil membrane-coated nanoparticles was investigated. The study found that macrophage polarization plays a critical role in gout, and AHNAK was identified as the key gene through which colchicine affects macrophage polarization. Lentiviral intervention to decrease AHNAK expression was shown to alleviate joint swelling in gout mice and regulate macrophage polarization. Colchicine encapsulated in R4F peptide-modified neutrophil membrane-coated Pluronic F127 nanoparticle (R4F-NM@F127) nanocarriers inhibited M1 macrophage polarization, induced M2 macrophage polarization, alleviated gout, and minimized toxicity to normal tissues. Colchicine suppressed M1 macrophage polarization and induced M2 macrophage polarization by binding to AHNAK protein, thereby alleviating gout. Colchicine incorporated into R4F-NM@F127 nanocarriers can serve as a targeted therapeutic drug to regulate macrophage polarization, alleviate gout, and reduce toxicity to normal tissues.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Rheumatology and Immunology,
Shengjing Hospital Affiliated to China Medical University, Shenyang 110000, China
| | - Lanqing Zhao
- Department of Sleep Medicine Center, The Shengjing Affiliated Hospital,
China Medical University, Shenyang 110000, Liaoning, China
| | - Jinwei Li
- Department of Neurology/Stroke Center, the First Affiliated Hospital ofChina Medical University, China Medical University, Shenyang 110000, Liaoning, China
| | - Hongxi Li
- Department of Pain Management,
Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Yu Chen
- Department of The Fourth Otolaryngology Head and Neck Surgery,
Shengjing Hospital of China Medical University, Shenyang 110000, China
| |
Collapse
|
17
|
Tabowei G, Faiza Rauf H, Dhungana M, Awais M, Blair K, Chaudhari SS, Habib I, Amin A. Efficacy of Colchicine for Secondary Prevention of Stroke: A Systematic Review and Meta-Analysis of Randomized Control Trials. Cureus 2024; 16:e75335. [PMID: 39776745 PMCID: PMC11706611 DOI: 10.7759/cureus.75335] [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] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Colchicine, a long-established anti-inflammatory medication, has emerged as a potential therapeutic agent for secondary prevention of stroke. This systematic review and meta-analysis aimed to evaluate the efficacy and safety of colchicine in preventing secondary stroke by comprehensively synthesizing available evidence. A systematic literature search was conducted across multiple electronic databases from inception to November 15, 2024, using comprehensive search strategies. Randomized controlled trials involving colchicine administration for stroke prevention were included. Two independent reviewers screened studies, extracted data, and assessed methodological quality using the Cochrane Risk of Bias tool. Meta-analysis was performed using Review Manager software, with risk ratios calculated for stroke incidence and all-cause mortality. The analysis encompassed seven studies involving 23,303 participants. The meta-analysis revealed a borderline significant 24% relative risk reduction in stroke incidence (risk ratio 0.76, 95% confidence interval 0.57-1.00, p = 0.05). Moderate heterogeneity was observed among studies (I² = 50%). Importantly, no significant difference was found in all-cause mortality between colchicine and control groups (risk ratio 1.03, 95% confidence interval 0.91-1.17, p = 0.66). While the findings suggest potential benefits of colchicine in stroke prevention, the results warrant cautious interpretation. The study emphasizes the need for larger, well-designed randomized controlled trials to definitively establish colchicine's role in comprehensive stroke prevention strategies.
Collapse
Affiliation(s)
- Godfrey Tabowei
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Odessa, USA
| | | | - Milan Dhungana
- Internal Medicine, Universal College of Medical Sciences, Bhairahawa, NPL
| | - Muhammad Awais
- Internal Medicine, Allama Iqbal Medical College, Lahore, PAK
| | - Keron Blair
- Medicine, American International School of Medicine, Georgetown, GUY
| | - Sandipkumar S Chaudhari
- Cardiothoracic Surgery, University of Alabama at Birmingham, Birmingham, USA
- Family Medicine, University of North Dakota School of Medicine and Health Sciences, Fargo, USA
| | - Ihtisham Habib
- Internal Medicine, Medical Teaching Institute, Lady Reading Hospital Peshawar, Peshawar, PAK
| | - Adil Amin
- Cardiology, Pakistan Navy Station (PNS) Shifa, Karachi, PAK
| |
Collapse
|
18
|
R Muralitharan R, Marques FZ, O'Donnell JA. Recent advancements in targeting the immune system to treat hypertension. Eur J Pharmacol 2024; 983:177008. [PMID: 39304109 DOI: 10.1016/j.ejphar.2024.177008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/10/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Hypertension is the key leading risk factor for death globally, affecting ∼1.3 billion adults, particularly in low- and middle-income countries. Most people living with hypertension have uncontrolled high blood pressure, increasing their likelihood of cardiovascular events. Significant issues preventing blood pressure control include lack of diagnosis, treatment, and response to existing therapy. For example, monotherapy and combination therapy are often unable to lower blood pressure to target levels. New therapies are urgently required to tackle this issue, particularly those that target the mechanisms behind hypertension instead of treating its symptoms. Acting via an increase in systemic and tissue-specific inflammation, the immune system is a critical contributor to blood pressure regulation and is considered an early mechanism leading to hypertension development. Here, we review the immune system's role in hypertension, evaluate clinical trials that target inflammation, and discuss knowledge gaps in pre-clinical and clinical data. We examine the effects of anti-inflammatory drugs colchicine and methotrexate on hypertension and evaluate the blockade of pro-inflammatory cytokines IL-1β and TNF-α on blood pressure in clinical trials. Lastly, we highlight how we can move forward to target specific components of the immune system to lower blood pressure. This includes targeting isolevuglandins, which accumulate in dendritic cells to promote T cell activation and cytokine production in salt-induced hypertension. We discuss the potential of the dietary fibre-derived metabolites short-chain fatty acids, which have anti-inflammatory and blood pressure-lowering effects via the gut microbiome. This would limit adverse events, leading to improved medication adherence and better blood pressure control.
Collapse
Affiliation(s)
- Rikeish R Muralitharan
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, VIC, Australia; Victorian Heart Institute, Monash University, Clayton, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, VIC, Australia; Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Victorian Heart Institute, Monash University, Clayton, Australia
| | - Joanne A O'Donnell
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
19
|
Lusta KA, Churov AV, Beloyartsev DF, Golovyuk AL, Lee AA, Sukhorukov VN, Orekhov AN. The two coin sides of bacterial extracellular membrane nanovesicles: atherosclerosis trigger or remedy. DISCOVER NANO 2024; 19:179. [PMID: 39532781 PMCID: PMC11557815 DOI: 10.1186/s11671-024-04149-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Among the numerous driving forces that cause the atherosclerotic cardiovascular disease (ASCVD), pathogenic bacterial extracellular membrane nanovesicles (BEMNs) containing toxins and virulence factors appear to be the key trigger of inflammation and atherogenesis, the major processes involved in the pathogenesis of ASCVD. Since BEMNs are the carriers of nanosized biomolecules to distant sites, they are now being considered as a novel drug delivery system. Nowadays, many therapeutic strategies are used to treat ASCVD. However, the conventional anti-atherosclerotic therapies are not effective enough. This primarily due to the inefficiency of non-targeted drug delivery systems to tissue affected areas, which, in turn, leads to numerous side effects, as well as faulty pharmacokinetics. In this regard, nanomedicine methods using nanoparticles (NPs) as targeted drug delivery vehicles proved to be extremely useful. Bioengineered BEMNs equipped with disease-specific ligand moieties and loaded with corresponding drugs represent a promising tool in nanomedicine, which can be used as a novel drug delivery system for a successful therapy of ASCVD. In this review, we outline the involvement of pathogenic BEMNs in the triggering of ASCVD, the conventional therapeutic strategies for the treatment of ASCVD, and the recent trends in nanomedicine using BEMNs and NPs as a vehicle for targeted drug delivery.
Collapse
Affiliation(s)
- Konstantin A Lusta
- Institute for Atherosclerosis Research, Ltd, Osennyaya Street 4-1-207, Moscow, Russia, 121609.
| | - Alexey V Churov
- Institute on Aging Research, Russian Gerontology Clinical Research Center, Pirogov Russian National Research Medical University, Moscow, Russia, 129226
- Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow, Russia, 125315
| | - Dmitry F Beloyartsev
- Vascular Surgery Department, A.V. Vishnevsky National Medical Research Center of Surgery, 27 Bolshaya Serpukhovskaya Street, Moscow, Russia, 117997
| | - Alexander L Golovyuk
- Vascular Surgery Department, A.V. Vishnevsky National Medical Research Center of Surgery, 27 Bolshaya Serpukhovskaya Street, Moscow, Russia, 117997
| | - Arthur A Lee
- Insitute of Human Morphology, Petrovsky Russian National Center of Surgery, 2 Abrikosovsky Lane, Moscow, Russia, 119991
| | - Vasily N Sukhorukov
- Insitute of Human Morphology, Petrovsky Russian National Center of Surgery, 2 Abrikosovsky Lane, Moscow, Russia, 119991
- Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow, Russia, 125315
| | - Alexander N Orekhov
- Insitute of Human Morphology, Petrovsky Russian National Center of Surgery, 2 Abrikosovsky Lane, Moscow, Russia, 119991
- Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow, Russia, 125315
| |
Collapse
|
20
|
Kumar R, Krishnaperumal G, Vellapandian C. Innovative mRNA Vaccine Approaches in Targeting Atherosclerosis: A New Era in Cardiovascular Therapy. Cureus 2024; 16:e74141. [PMID: 39712846 PMCID: PMC11662511 DOI: 10.7759/cureus.74141] [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: 10/19/2024] [Accepted: 11/21/2024] [Indexed: 12/24/2024] Open
Abstract
Atherosclerosis, a major cause of cardiovascular disease (CVD), involves plaque buildup in arteries driven by inflammation, endothelial dysfunction, and lipid metabolism disturbances. Current therapies aim to reduce cholesterol through statins and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, prevent blood clots with antiplatelet drugs like aspirin, and control inflammation, alongside lifestyle modifications. However, these approaches often fall short due to patient non-compliance and residual risks. This review explores emerging mRNA vaccine strategies targeting the complex mechanisms of atherosclerosis. These vaccines could produce therapeutic proteins to modulate inflammation by encoding sequences that inhibit pro-inflammatory cytokines such as interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α), stabilizing plaques. Key targets include interleukin-10 (IL-10) for plaque stability, PCSK9 for cholesterol regulation, and vascular endothelial growth factor (VEGF) for endothelial repair. Addressing these unmet needs, mRNA-based approaches offer the potential for more effective and personalized treatments for atherosclerosis. However, challenges remain, including difficulty replicating human atherosclerosis in preclinical models, regulatory concerns about long-term safety, and ensuring accessibility in low-resource settings. In addition, large and diverse clinical trials are needed to confirm the efficacy of these vaccines in reducing cardiovascular events.
Collapse
Affiliation(s)
- Rahul Kumar
- Pharmacy/Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology (SRMIST), Chengalpattu, IND
| | - Gowri Krishnaperumal
- Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology (SRMIST), Chengalpattu, IND
| | - Chitra Vellapandian
- Pharmacy/Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology (SRMIST), Chengalpattu, IND
| |
Collapse
|
21
|
Zarenezhad E, Hadi AT, Nournia E, Rostamnia S, Ghasemian A. A Comprehensive Review on Potential In Silico Screened Herbal Bioactive Compounds and Host Targets in the Cardiovascular Disease Therapy. BIOMED RESEARCH INTERNATIONAL 2024; 2024:2023620. [PMID: 39502274 PMCID: PMC11537750 DOI: 10.1155/2024/2023620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 05/15/2024] [Accepted: 09/28/2024] [Indexed: 11/08/2024]
Abstract
Herbal medicines (HMs) have deciphered indispensable therapeutic effects against cardiovascular disease (CVD) (the predominant cause of death worldwide). The conventional CVD therapy approaches have not been efficient and need alternative medicines. The objective of this study was a review of herbal bioactive compound efficacy for CVD therapy based on computational and in silico studies. HM bioactive compounds with potential anti-CVD traits include campesterol, naringenin, quercetin, stigmasterol, tanshinaldehyde, Bryophyllin A, Bryophyllin B, beta-sitosterol, punicalagin, butein, eriodyctiol, butin, luteolin, and kaempferol discovered using computational studies. Some of the bioactive compounds have exhibited therapeutic effects, as followed by in vitro (tanshinaldehyde, punicalagin, butein, eriodyctiol, and butin), in vivo (gallogen, luteolin, chebulic acid, butein, eriodyctiol, and butin), and clinical trials (quercetin, campesterol, and naringenin). The main mechanisms of action of bioactive compounds for CVD healing include cell signaling and inhibition of inflammation and oxidative stress, decrease of lipid accumulation, and regulation of metabolism and immune cells. Further experimental studies are required to verify the anti-CVD effects of herbal bioactive compounds and their pharmacokinetic/pharmacodynamic features.
Collapse
Affiliation(s)
- Elham Zarenezhad
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Ali Tareq Hadi
- Womens Obstetrics & Gynecology Hospital, Ministry of Health, Al Samawah, Iraq
| | - Ensieh Nournia
- Cardiology Department, Hamadan University of Medical Sciences, Hamedan, Iran
| | - Sadegh Rostamnia
- Organic and Nano Group, Department of Chemistry, Iran University of Science and Technology, PO Box 16846-13114, Tehran, Iran
| | - Abdolmajid Ghasemian
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
22
|
Kang M, Jia H, Feng M, Ren H, Gao J, Liu Y, Zhang L, Zhou MS. Cardiac macrophages in maintaining heart homeostasis and regulating ventricular remodeling of heart diseases. Front Immunol 2024; 15:1467089. [PMID: 39372400 PMCID: PMC11449765 DOI: 10.3389/fimmu.2024.1467089] [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: 07/19/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024] Open
Abstract
Macrophages are most important immune cell population in the heart. Cardiac macrophages have broad-spectrum and heterogeneity, with two extreme polarization phenotypes: M1 pro-inflammatory macrophages (CCR2-ly6Chi) and M2 anti-inflammatory macrophages (CCR2-ly6Clo). Cardiac macrophages can reshape their polarization states or phenotypes to adapt to their surrounding microenvironment by altering metabolic reprogramming. The phenotypes and polarization states of cardiac macrophages can be defined by specific signature markers on the cell surface, including tumor necrosis factor α, interleukin (IL)-1β, inducible nitric oxide synthase (iNOS), C-C chemokine receptor type (CCR)2, IL-4 and arginase (Arg)1, among them, CCR2+/- is one of most important markers which is used to distinguish between resident and non-resident cardiac macrophage as well as macrophage polarization states. Dedicated balance between M1 and M2 cardiac macrophages are crucial for maintaining heart development and cardiac functional and electric homeostasis, and imbalance between macrophage phenotypes may result in heart ventricular remodeling and various heart diseases. The therapy aiming at specific target on macrophage phenotype is a promising strategy for treatment of heart diseases. In this article, we comprehensively review cardiac macrophage phenotype, metabolic reprogramming, and their role in maintaining heart health and mediating ventricular remodeling and potential therapeutic strategy in heart diseases.
Collapse
Affiliation(s)
- Mengjie Kang
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
| | - Hui Jia
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
- School of Traditional Chinese Medicine, Shenyang Medical College, Shenyang, China
| | - Mei Feng
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
| | - Haolin Ren
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Junjia Gao
- Department of Cardiology, Second Affiliated Hospital, Shenyang Medical College, Shenyang, China
| | - Yueyang Liu
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
- School of Pharmacy, Shenyang Medical College, Shenyang, China
| | - Lu Zhang
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
| | - Ming-Sheng Zhou
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
| |
Collapse
|
23
|
Zhang Y, Zheng BY, Zhang QF, Zhao YN, Yu QM, Liu X, Ding SY, Qian SS, Wu H, Wu QY, Zhang YH, Zheng L, Zhang XH, Zhang HF, Hao YM, Lu JC, Wang L, Wen JK, Zheng B. Nanoparticles targeting OPN loaded with BY1 inhibits vascular restenosis by inducing FTH1-dependent ferroptosis in vascular smooth muscle cells. Biomaterials 2024; 309:122613. [PMID: 38759485 DOI: 10.1016/j.biomaterials.2024.122613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
Vascular restenosis following angioplasty continues to pose a significant challenge. The heterocyclic trioxirane compound [1, 3, 5-tris((oxiran-2-yl)methyl)-1, 3, 5-triazinane-2, 4, 6-trione (TGIC)], known for its anticancer activity, was utilized as the parent ring to conjugate with a non-steroidal anti-inflammatory drug, resulting in the creation of the spliced conjugated compound BY1. We found that BY1 induced ferroptosis in VSMCs as well as in neointima hyperplasia. Furthermore, ferroptosis inducers amplified BY1-induced cell death, while inhibitors mitigated it, indicating the contribution of ferroptosis to BY1-induced cell death. Additionally, we established that ferritin heavy chain1 (FTH1) played a pivotal role in BY1-induced ferroptosis, as evidenced by the fact that FTH1 overexpression abrogated BY1-induced ferroptosis, while FTH1 knockdown exacerbated it. Further study found that BY1 induced ferroptosis by enhancing the NCOA4-FTH1 interaction and increasing the amount of intracellular ferrous. We compared the effectiveness of various administration routes for BY1, including BY1-coated balloons, hydrogel-based BY1 delivery, and nanoparticles targeting OPN loaded with BY1 (TOP@MPDA@BY1) for targeting proliferated VSMCs, for prevention and treatment of the restenosis. Our results indicated that TOP@MPDA@BY1 was the most effective among the three administration routes, positioning BY1 as a highly promising candidate for the development of drug-eluting stents or treatments for restenosis.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Bo-Yang Zheng
- Department of tumor biotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Qian-Fan Zhang
- Chongqing School, University of Chinese Academy of Sciences, Chongqing, 400714, China
| | - Ya-Nan Zhao
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Qi-Ming Yu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xin Liu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Si-Ying Ding
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Shuang-Shuang Qian
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Han Wu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Qian-Yu Wu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yu-Han Zhang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Lei Zheng
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xin-Hua Zhang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China; Institution of Chinese Integrative Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, China
| | - Hao-Feng Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Hebei Province Key Laboratory of Innovative Drug Research and Evaluation, Shijiazhuang, 050017, China
| | - Yi-Ming Hao
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jing-Chao Lu
- Department of Cardiovascular Medicine, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Lei Wang
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Hebei Province Key Laboratory of Innovative Drug Research and Evaluation, Shijiazhuang, 050017, China.
| | - Jin-Kun Wen
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China.
| | - Bin Zheng
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China.
| |
Collapse
|
24
|
Mastana S, Halai KC, Akam L, Hunter DJ, Singh P. Genetic Polymorphisms and Genetic Risk Scores Contribute to the Risk of Coronary Artery Disease (CAD) in a North Indian Population. Int J Mol Sci 2024; 25:8552. [PMID: 39126122 PMCID: PMC11313018 DOI: 10.3390/ijms25158552] [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: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
Coronary artery disease (CAD) is the leading cause of death in India. Many genetic polymorphisms play a role in regulating oxidative stress, blood pressure and lipid metabolism, contributing to the pathophysiology of CAD. This study examined the association between ten polymorphisms and CAD in the Jat Sikh population from Northern India, also considering polygenic risk scores. This study included 177 CAD cases and 175 healthy controls. The genetic information of GSTM1 (rs366631), GSTT1 (rs17856199), ACE (rs4646994), AGT M235T (rs699), AGT T174M (rs4762), AGTR1 A1166C (rs5186), APOA5 (rs3135506), APOC3 (rs5128), APOE (rs7412) and APOE (rs429358) and clinical information was collated. Statistical analyses were performed using SPSS version 27.0 and SNPstats. Significant independent associations were found for GST*M1, GST*T1, ACE, AGT M235T, AGT T174M, AGTR1 A1166C and APOA5 polymorphisms and CAD risk (all p < 0.05). The AGT CT haplotype was significantly associated with a higher CAD risk, even after controlling for covariates (adjusted OR = 3.93, 95% CI [2.39-6.48], p < 0.0001). The APOA5/C3 CC haplotype was also significantly associated with CAD (adjusted OR = 1.86, 95% CI [1.14-3.03], p < 0.05). A higher polygenic risk score was associated with increased CAD risk (adjusted OR = 1.98, 95% CI [1.68-2.34], p < 0.001). Seven polymorphisms were independently associated with an increase in the risk of CAD in this North Indian population. A considerable risk association of AGT, APOA5/C3 haplotypes and higher genetic risk scores is documented, which may have implications for clinical and public health applications.
Collapse
Affiliation(s)
- Sarabjit Mastana
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK (L.A.); (D.J.H.)
| | - Kushni Charisma Halai
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK (L.A.); (D.J.H.)
| | - Liz Akam
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK (L.A.); (D.J.H.)
| | - David John Hunter
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK (L.A.); (D.J.H.)
| | - Puneetpal Singh
- Department of Human Genetics, Punjabi University, Patiala 147002, India;
| |
Collapse
|
25
|
Zhang Y, Li B, Cai H, Fu Y, Zheng Y. Associations of iron metabolism and inflammation with all-cause and cardiovascular mortality in a large NHANES community sample: Moderating and mediating effects. Nutr Metab Cardiovasc Dis 2024; 34:1854-1863. [PMID: 38658228 DOI: 10.1016/j.numecd.2024.03.026] [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: 01/08/2024] [Revised: 03/01/2024] [Accepted: 03/21/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND AND AIMS This study aimed to assess the associations between serum iron concentration, C-reactive protein (CRP) concentration and the risk of all-cause mortality and cardiovascular mortality in the general population and to explore potential mediating and moderating effects. METHODS AND RESULTS This study analyzed data from the National Health and Nutrition Examination Survey spanning the years 1999-2010, encompassing 23,634 participants. Cox proportional hazards regression models were employed to investigate the independent associations of serum iron and CRP with all-cause and cardiovascular mortality. Moderation and mediation analyses explored the moderating effect of CRP on the association between the serum iron concentration and all-cause and cardiovascular mortality, and the mediating role of the serum iron concentration in the association between the CRP concentration and all-cause and cardiovascular mortality. After multivariate adjustments in the Cox model, serum iron and CRP levels were independently correlated with both all-cause and cardiovascular mortality risk. Moderation analyses revealed a more pronounced correlation between the serum iron concentration and both all-cause and cardiovascular mortality in participants with higher CRP levels. Mediation analysis indicated that the serum iron concentration partly mediated the impact of CRP on the risk of all-cause mortality (13.79%) and cardiovascular mortality (24.12%). CONCLUSION Serum iron and CRP are independently associated with all-cause and cardiovascular mortality. Moreover, the associations between serum iron concentrations and both all-cause and cardiovascular mortality are more pronounced in individuals with elevated CRP. Serum iron partially mediates the effect of CRP on all-cause and cardiovascular mortality.
Collapse
Affiliation(s)
- Yaoting Zhang
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Bing Li
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun, 130021, China.
| | - He Cai
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Yu Fu
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Yang Zheng
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
26
|
Guan J, Abudouaini H, Lin K, Yang K. Emerging insights into the role of IL-1 inhibitors and colchicine for inflammation control in type 2 diabetes. Diabetol Metab Syndr 2024; 16:140. [PMID: 38918878 PMCID: PMC11197348 DOI: 10.1186/s13098-024-01369-x] [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: 03/26/2024] [Accepted: 05/30/2024] [Indexed: 06/27/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM), a prevalent chronic metabolic disorder, is closely linked to persistent low-grade inflammation, significantly contributing to its development and progression. This review provides a comprehensive examination of the inflammatory mechanisms underlying T2DM, focusing on the role of the NLRP3 inflammasome and interleukin-1β (IL-1β) in mediating inflammatory responses. We discuss the therapeutic potential of IL-1 inhibitors and colchicine, highlighting their mechanisms in inhibiting the NLRP3 inflammasome and reducing IL-1β production. Recent studies indicate that these agents could effectively mitigate inflammation, offering promising avenues for the prevention and management of T2DM. By exploring the intricate connections between metabolic disturbances and chronic inflammation, this review underscores the need for novel anti-inflammatory strategies to address T2DM and its complications.
Collapse
Affiliation(s)
- Jianbin Guan
- Honghui-Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Haimiti Abudouaini
- Honghui-Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Kaiyuan Lin
- Honghui-Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.
| | - Kaitan Yang
- Honghui-Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.
- Truma Rehabilitation Department, Honghui-Hospital,Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.
| |
Collapse
|
27
|
Welt FGP, Batchelor W, Spears JR, Penna C, Pagliaro P, Ibanez B, Drakos SG, Dangas G, Kapur NK. Reperfusion Injury in Patients With Acute Myocardial Infarction: JACC Scientific Statement. J Am Coll Cardiol 2024; 83:2196-2213. [PMID: 38811097 DOI: 10.1016/j.jacc.2024.02.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/15/2024] [Accepted: 02/26/2024] [Indexed: 05/31/2024]
Abstract
Despite impressive improvements in the care of patients with ST-segment elevation myocardial infarction, mortality remains high. Reperfusion is necessary for myocardial salvage, but the abrupt return of flow sets off a cascade of injurious processes that can lead to further necrosis. This has been termed myocardial ischemia-reperfusion injury and is the subject of this review. The pathologic and molecular bases for myocardial ischemia-reperfusion injury are increasingly understood and include injury from reactive oxygen species, inflammation, calcium overload, endothelial dysfunction, and impaired microvascular flow. A variety of pharmacologic strategies have been developed that have worked well in preclinical models and some have shown promise in the clinical setting. In addition, there are newer mechanical approaches including mechanical unloading of the heart prior to reperfusion that are in current clinical trials.
Collapse
Affiliation(s)
- Frederick G P Welt
- Department of Medicine, Division of Cardiovascular Medicine, University of Utah Hospital, Salt Lake City, Utah, USA.
| | | | - J Richard Spears
- Department of Cardiovascular Medicine, Beaumont Systems, Royal Oak, Michigan, USA
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain; CIBER de Enfermedades Cardiovasculares, Madrid, Spain; Department of Cardiology, Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - Stavros G Drakos
- Department of Medicine, Division of Cardiovascular Medicine, University of Utah Hospital, Salt Lake City, Utah, USA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, USA
| | - George Dangas
- Division of Cardiology, Mount Sinai Health System, New York, New York, USA
| | - Navin K Kapur
- The CardioVascular Center and Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
28
|
Wang C, Wang X, Zhang Y, Mi Y, Han Y, Zhi Y, Zhao R, Cui N, Ma Q, Zhang H, Xue D, Qiao R, Han J, Yu Y, Li J, Shaiea M, Liu D, Gu G, Wang C. Inducible Fgf13 ablation alleviates cardiac fibrosis via regulation of microtubule stability. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1802-1812. [PMID: 38818580 PMCID: PMC11659771 DOI: 10.3724/abbs.2024075] [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: 01/26/2024] [Accepted: 04/11/2024] [Indexed: 06/01/2024] Open
Abstract
Fibroblast growth factor (FGF) isoform 13, a distinct type of FGF, boasts significant potential for therapeutic intervention in cardiovascular dysfunctions. However, its impact on regulating fibrosis remains unexplored. This study aims to elucidate the role and mechanism of FGF13 on cardiac fibrosis. Here, we show that following transverse aortic constriction (TAC) surgery, interstitial fibrosis and collagen content increase in mice, along with reduced ejection fraction and fractional shortening, augmented heart mass. However, following Fgf13 deletion, interstitial fibrosis is decreased, ejection fraction and fractional shortening are increased, and heart mass is decreased, compared with those in the TAC group. Mechanistically, incubation of cardiac fibroblasts with transforming growth factor β (TGFβ) increases the expressions of types I and III collagen proteins, as well as α-smooth muscle actin (α-SMA) proteins, and enhances fibroblast proliferation and migration. In the absence of Fgf13, the expressions of these proteins are decreased, and fibroblast proliferation and migration are suppressed, compared with those in the TGFβ-stimulated group. Overexpression of FGF13, but not FGF13 mutants defective in microtubule binding and stabilization, rescues the decrease in collagen and α-SMA protein and weakens the proliferation and migration function of the Fgf13 knockdown group. Furthermore, Fgf13 knockdown decreases ROCK protein expression via microtubule disruption. Collectively, cardiac Fgf13 knockdown protects the heart from fibrosis in response to haemodynamic stress by modulating microtubule stabilization and ROCK signaling pathway.
Collapse
Affiliation(s)
- Cong Wang
- Department of Pharmacologythe Key Laboratory of Neural and Vascular BiologyMinistry of Educationthe Key Laboratory of New Drug Pharmacology and Toxicologythe Hebei Collaboration Innovation Center for MechanismDiagnosis and Treatment of Neurological and Psychiatric DiseaseHebei Medical UniversityShijiazhuang050017China
| | - Xiangchong Wang
- Department of PharmacologyHebei International Cooperation Center for Ion Channel Function and Innovative Traditional Chinese MedicineHebei Higher Education Institute Applied Technology Research Center on TCM Formula PreparationHebei University of Chinese MedicineShijiazhuang050091China
| | - Yiyi Zhang
- Department of Pharmacologythe Key Laboratory of Neural and Vascular BiologyMinistry of Educationthe Key Laboratory of New Drug Pharmacology and Toxicologythe Hebei Collaboration Innovation Center for MechanismDiagnosis and Treatment of Neurological and Psychiatric DiseaseHebei Medical UniversityShijiazhuang050017China
| | - Yuan Mi
- Department of Emergencythe Fourth Hospital of Hebei Medical UniversityShijiazhuang050011China
| | - Yanxue Han
- Department of Pharmacologythe Key Laboratory of Neural and Vascular BiologyMinistry of Educationthe Key Laboratory of New Drug Pharmacology and Toxicologythe Hebei Collaboration Innovation Center for MechanismDiagnosis and Treatment of Neurological and Psychiatric DiseaseHebei Medical UniversityShijiazhuang050017China
| | - Yaxin Zhi
- Department of Cardiologythe Second Hospital of Hebei Medical UniversityShijiazhuang050000China
| | - Ran Zhao
- Department of Pharmacologythe Key Laboratory of Neural and Vascular BiologyMinistry of Educationthe Key Laboratory of New Drug Pharmacology and Toxicologythe Hebei Collaboration Innovation Center for MechanismDiagnosis and Treatment of Neurological and Psychiatric DiseaseHebei Medical UniversityShijiazhuang050017China
| | - Nanqi Cui
- Department of Vascular Surgerythe Second Hospital of Hebei Medical UniversityShijiazhuang050000China
| | - Qianli Ma
- of Cardiac Surgerythe Second Hospital of Hebei Medical UniversityShijiazhuang050000China
| | - Huaxing Zhang
- Core Facilities and CentersHebei Medical UniversityShijiazhuang050017China
| | - Dazhong Xue
- Department of Pharmacologythe Key Laboratory of Neural and Vascular BiologyMinistry of Educationthe Key Laboratory of New Drug Pharmacology and Toxicologythe Hebei Collaboration Innovation Center for MechanismDiagnosis and Treatment of Neurological and Psychiatric DiseaseHebei Medical UniversityShijiazhuang050017China
| | - Ruoyang Qiao
- College of Basic MedicineHebei Medical UniversityShijiazhuang050017China
| | - Jiabing Han
- Department of Pharmacologythe Key Laboratory of Neural and Vascular BiologyMinistry of Educationthe Key Laboratory of New Drug Pharmacology and Toxicologythe Hebei Collaboration Innovation Center for MechanismDiagnosis and Treatment of Neurological and Psychiatric DiseaseHebei Medical UniversityShijiazhuang050017China
| | - Yulou Yu
- Department of Pharmacologythe Key Laboratory of Neural and Vascular BiologyMinistry of Educationthe Key Laboratory of New Drug Pharmacology and Toxicologythe Hebei Collaboration Innovation Center for MechanismDiagnosis and Treatment of Neurological and Psychiatric DiseaseHebei Medical UniversityShijiazhuang050017China
| | - Jiaxuan Li
- SchoolHebei Medical UniversityShijiazhuang050017China
| | - Mohammed Shaiea
- Department of Pharmacologythe Key Laboratory of Neural and Vascular BiologyMinistry of Educationthe Key Laboratory of New Drug Pharmacology and Toxicologythe Hebei Collaboration Innovation Center for MechanismDiagnosis and Treatment of Neurological and Psychiatric DiseaseHebei Medical UniversityShijiazhuang050017China
| | - Demin Liu
- Department of Cardiologythe Second Hospital of Hebei Medical UniversityShijiazhuang050000China
| | - Guoqiang Gu
- Department of Cardiologythe Second Hospital of Hebei Medical UniversityShijiazhuang050000China
| | - Chuan Wang
- Department of Pharmacologythe Key Laboratory of Neural and Vascular BiologyMinistry of Educationthe Key Laboratory of New Drug Pharmacology and Toxicologythe Hebei Collaboration Innovation Center for MechanismDiagnosis and Treatment of Neurological and Psychiatric DiseaseHebei Medical UniversityShijiazhuang050017China
| |
Collapse
|
29
|
Sun H, Huang C, Li L, Zhu W, Li J, Sun P, A G, Fonarow GC, Yang Q, Zhou X. Time to benefit of colchicine in patients with cardiovascular disease: A pooled analysis of randomized controlled trials. Heliyon 2024; 10:e30408. [PMID: 38726146 PMCID: PMC11079085 DOI: 10.1016/j.heliyon.2024.e30408] [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: 04/15/2024] [Accepted: 04/25/2024] [Indexed: 05/12/2024] Open
Abstract
Background Low-dose colchicine has been shown to lower major adverse cardiovascular events (MACE) among those with cardiovascular disease (CVD). It remains unclear how long a CVD patient needs to live to potentially benefit from colchicine. Our study aimed to determine the time to benefit (TTB) of colchicine in individuals with CVD. Methods Literature searches were performed in PubMed for the cardiovascular outcome trial of colchicine in patients with CVD until October 12, 2023. The primary outcome measured was MACE. Reconstructed individual participant data (IPD) and the stratified Cox proportional hazards model were used to calculate the hazard ratio (HR) and 95 % confidence interval (CI) to estimate the efficacy of colchicine, and Weibull survival curves were fitted to estimate TTB for specific absolute risk reduction (ARR) thresholds (0.002, 0.005, and 0.01). Results Four trials randomizing 11,594 adults aged between 59.8 and 66.5 years were included (follow-up duration: 12-28.6 months). Compared with placebo, colchicine reduced the risk of MACE (HR 0.68, 95 % CI: 0.60 to 0.78) but had no impact on cardiovascular and all-cause mortality. A TTB of 11.0 months (95 % CI: 0.59 to 21.3) was estimated to be needed to prevent 1 MACE in 100-colchicine-treated patients. The TTB for acute coronary syndrome was similar compared to stable coronary artery disease (10.7 vs. 11.2 months for ARR = 0.010). Conclusions By using reconstructed IPD, this pooled analysis demonstrated that colchicine was associated with reduced nonfatal MACE, and the TTB was approximately 11.0 months to prevent 1 MACE per 100 patients.
Collapse
Affiliation(s)
- Haonan Sun
- Department of Cardiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Chuanyi Huang
- Department of Cardiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Linjie Li
- Department of Cardiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Wenjun Zhu
- Department of Cardiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Jingge Li
- Department of Cardiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Pengfei Sun
- Department of Cardiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Geru A
- Department of Cardiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Gregg C. Fonarow
- Division of Cardiology, David Geffen School of Medicine at University of California, Geffen Hall 885 Tiverton Drive Los Angeles, CA 90095, USA
| | - Qing Yang
- Department of Cardiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Xin Zhou
- Department of Cardiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| |
Collapse
|
30
|
Abstract
An intense, stereotyped inflammatory response occurs in response to ischaemic and non-ischaemic injury to the myocardium. The NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome is a finely regulated macromolecular protein complex that senses the injury and triggers and amplifies the inflammatory response by activation of caspase 1; cleavage of pro-inflammatory cytokines, such as pro-IL-1β and pro-IL-18, to their mature forms; and induction of inflammatory cell death (pyroptosis). Inhibitors of the NLRP3 inflammasome and blockers of IL-1β and IL-18 activity have been shown to reduce injury to the myocardium and pericardium, favour resolution of the inflammation and preserve cardiac function. In this Review, we discuss the components of the NLRP3 inflammasome and how it is formed and activated in various ischaemic and non-ischaemic cardiac pathologies (acute myocardial infarction, cardiac dysfunction and remodelling, atherothrombosis, myocarditis and pericarditis, cardiotoxicity and cardiac sarcoidosis). We also summarize current preclinical and clinical evidence from studies of agents that target the NLRP3 inflammasome and related cytokines.
Collapse
Affiliation(s)
- Stefano Toldo
- Robert M. Berne Cardiovascular Research Center and Division of Cardiology, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center and Division of Cardiology, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
31
|
Sadik O, Tahir S, Sahibzada A, Iguh C, Ezenwa V, Bhavanam S. Colchicine Therapy for Glenohumeral Osteoarthritis: A Case Report. Cureus 2024; 16:e59181. [PMID: 38807809 PMCID: PMC11130563 DOI: 10.7759/cureus.59181] [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] [Accepted: 04/27/2024] [Indexed: 05/30/2024] Open
Abstract
Osteoarthritis management primarily focuses on targeting pain. Conventional modalities for pain management include acetaminophen, non-steroidal anti-inflammatory drugs (NSAIDs), and intra-articular corticosteroid injections. However, these approaches may provide minimal pain relief and can be contraindicated for some patients, highlighting the ongoing need for alternative pain management. Colchicine, commonly used in the management of gout, has emerged as a potential option for pain management in osteoarthritis. There are implications of colchicine use for knee and hand osteoarthritis but remains inconclusive. In this context, we present a case of a 68-year-old diabetic woman with glenohumeral osteoarthritis and associated right shoulder pain. Due to minimal pain relief from previous treatments, the patient was given a combination trial of colchicine and acetaminophen for three months. After completion of this treatment, the patient experienced significant pain relief and improved functionality. The aim of this case is to highlight the efficacy of colchicine as a possible treatment option for managing shoulder pain in osteoarthritis.
Collapse
Affiliation(s)
- Oman Sadik
- Family Medicine, Jackson Park Hospital, Chicago, USA
| | - Sophia Tahir
- Internal Medicine, Windsor University School of Medicine, Cayon, KNA
| | - Anum Sahibzada
- Internal Medicine, Saint James School of Medicine, The Quarter, AIA
| | - Chinenye Iguh
- Medicine, Windsor University School of Medicine, Cayon, KNA
| | - Virginia Ezenwa
- Internal Medicine, Windsor University School of Medicine, Cayon, KNA
| | | |
Collapse
|
32
|
Zhu Y, Fang Y, Wang Y, Han D, Liu J, Tian L, Xu M, Wang Y, Cao F. Cluster of Differentiation-44-Targeting Prussian Blue Nanoparticles Onloaded with Colchicine for Atherosclerotic Plaque Regression in a Mice Model. ACS Biomater Sci Eng 2024; 10:1530-1543. [PMID: 38372216 DOI: 10.1021/acsbiomaterials.3c01518] [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: 02/20/2024]
Abstract
Atherosclerosis management heavily relies on the suppression of the inflammatory response of macrophages. Colchicine's potent anti-inflammatory properties make it a promising candidate for secondary prevention against cardiovascular disease. However, its high toxicity and numerous adverse effects limit its clinical use. To address this, there is an urgent need for specific drug delivery systems to boost the level of accumulation of colchicine within atherosclerotic plaques. In this study, the cluster of differentiation-44 receptor was verified to be overexpressed in inflammatory macrophages within plaques both in vitro and in vivo. Subsequently, a Prussian blue-based nanomedical loading system with hyaluronic acid (HA) coating was constructed, and its effects were observed on the atherosclerosis regression. Colchicine and Cy5.5 were encapsulated within Prussian blue nanoparticles through self-assembly, followed by conjugation with hyaluronic acid to create col@PBNP@HA. The formulated col@PBNP@HA displayed a cubic shape and scattered distribution. Importantly, col@PBNP@HA demonstrated specific cellular uptake into lipopolysaccharide-stimulated macrophages. In vitro experiments showed that col@PBNP@HA more effectively inhibited expression of inflammatory factors and scavenged reactive oxygen species compared with the control group, which were treated with colchicine. Furthermore, col@PBNP@HA exhibited its specific and higher accumulation in aortic plaque analysis via fluorescence imaging of aortas. After 4 weeks, administration of col@PBNP@HA resulted in significant atherosclerosis regression in the mice model, with therapeutic effects superior to those of free colchicine. Similar to colchicine, col@PBNP@HA inhibited the secretion of inflammation factors and scavenged ROS through the regulation of the toll-like receptor 4 (TLR4)/myeloid differentiation factor 88 (Myd88)/nuclear factor kappa-B (NF-κB) and peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α) signaling pathway. In summary, col@PBNP@HA demonstrated specific targeting ability to inflammatory plaques and exerted beneficial effects on atherosclerosis regression through TLR4/Myd88/NF-κB and PGC-1α modulation.
Collapse
Affiliation(s)
- Yan Zhu
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of Cardiology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Second Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Yan Fang
- Department of Cardiology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Second Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Yujia Wang
- Department of Cardiology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Second Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Dong Han
- Department of Cardiology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Second Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Jing Liu
- Department of Cardiology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Second Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Lei Tian
- Department of Cardiology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Second Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Mengqi Xu
- Department of Cardiology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Second Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Yabin Wang
- Department of Cardiology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Second Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Feng Cao
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of Cardiology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Second Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
33
|
Luo Z, Yin F, Wang X, Kong L. Progress in approved drugs from natural product resources. Chin J Nat Med 2024; 22:195-211. [PMID: 38553188 DOI: 10.1016/s1875-5364(24)60582-0] [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: 12/03/2023] [Indexed: 04/02/2024]
Abstract
Natural products (NPs) have consistently played a pivotal role in pharmaceutical research, exerting profound impacts on the treatment of human diseases. A significant proportion of approved molecular entity drugs are either directly derived from NPs or indirectly through modifications of NPs. This review presents an overview of NP drugs recently approved in China, the United States, and other countries, spanning various disease categories, including cancers, cardiovascular and cerebrovascular diseases, central nervous system disorders, and infectious diseases. The article provides a succinct introduction to the origin, activity, development process, approval details, and mechanism of action of these NP drugs.
Collapse
Affiliation(s)
- Zhongwen Luo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Fucheng Yin
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaobing Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
34
|
Fu J, Liang Y, Yu D, Wang Y, Lu F, Liu S. Radix Saposhnikoviae enhancing Huangqi Chifeng Decoction improves lipid metabolism in AS mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 320:117479. [PMID: 37992882 DOI: 10.1016/j.jep.2023.117479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/24/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huangqi Chifeng decoction (HQCF) combined with parsnips is a classic Chinese traditional medicine formula that has certain advantages in the clinical treatment of cardiovascular and cerebrovascular diseases. At present, there is an absence of research on the regulatory effect and mechanism of this formula on atherosclerosis (AS). The synergistic effect of Radix Saposhnikoviae (RS) in HQCF is also unclear. AIM OF THE STUDY This study was designed to investigate the role of RS, which is designed as a guide drug for HQCF, in improving the lipid metabolism of AS. MATERIALS AND METHODS In this study, we studied the effect of HQCF on ApoE-/- mice before and after RS compatibility. Hematoxylin and eosin (HE) staining and oil red staining were used to evaluate atherosclerotic lesions and lipid accumulation in the aorta and liver, respectively. The expression of adenosine monophosphate-activated protein kinase (AMPK) and pAMPK in the aorta was measured by immunofluorescence, and AMPK and sterol regulatory element binding protein-1 (SREBP-1),fatty acid synthase (FAS) and acetyl-CoA carboxylase (ACC) in liver tissue were measured by Western blot analysis. Metabolomics was used to compare the changes in serum and liver metabolites of ApoE-/- mice before and after RS combination. RESULTS Compared with the control group, the serum lipid levels of ApoE-/- mice increased, the aortic intima thickened with plaque formation, and liver tissue pathological changes and lipid deposition occurred. Both (HQCFT without RS)HQCS and HQCF can improve the pathological condition of tissue and regulate the blood lipid level. It was noted that HQCF could promote the phosphorylation of AMPK to activate it, inhibit the expression of SREBP-1c and FAS, reduce lipid synthesis, and inhibit ACC to promote the oxidative decomposition of fatty acids. Serum and liver metabolome results showed that HQCS and HQCF treated AS mainly by regulating glycerophospholipid metabolism, sphingolipid metabolism and the arachidonic acid metabolism pathway. Importantly, HQCF showed better efficacy in regulating lipid metabolism than the HQCS group. CONCLUSION HQCF decoction reduces atherosclerotic lesions in the aorta and lipid accumulation in the liver, which may regulate lipid transport and metabolic function by activating the AMPK pathway. These effects can be attributed to the guidance and synergism of RS.
Collapse
Affiliation(s)
- Jiaqi Fu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yuqin Liang
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Donghua Yu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yu Wang
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Fang Lu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Shumin Liu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| |
Collapse
|
35
|
Yu M, Cheng X. Editorial Commentary: Top Five Stories of the Cellular Landscape and Therapies of Atherosclerosis: Current Knowledge and Future Perspectives. Curr Med Sci 2024; 44:241-243. [PMID: 38277018 DOI: 10.1007/s11596-023-2825-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Affiliation(s)
- Miao Yu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
36
|
Maidana D, Arroyo-Álvarez A, Arenas-Loriente A, Barreres-Martín G, Muñoz-Alfonso C, Bompart Berroteran D, Esteve Claramunt F, Blanco del Burgo R, Cepas-Guillén P, Garcia-Blas S, Bonanad C. Inflammation as a New Therapeutic Target among Older Patients with Ischemic Heart Disease. J Clin Med 2024; 13:363. [PMID: 38256497 PMCID: PMC10816645 DOI: 10.3390/jcm13020363] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Cardiovascular (CV) diseases remain a global health challenge, with ischemic heart disease (IHD) being the primary cause of both morbidity and mortality. Despite optimal pharmacological therapy, older patients with IHD exhibit an increased susceptibility to recurrent ischemic events, significantly impacting their prognosis. Inflammation is intricately linked with the aging process and plays a pivotal role in the evolution of atherosclerosis. Emerging anti-inflammatory therapies have shown promise in reducing ischemic events among high-risk populations. This review aims to explore the potential of targeted anti-inflammatory interventions in improving clinical outcomes and the quality of life for older patients with IHD.
Collapse
Affiliation(s)
- Daniela Maidana
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | - Andrea Arroyo-Álvarez
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | | | - Guillermo Barreres-Martín
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | - Carles Muñoz-Alfonso
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | - Daznia Bompart Berroteran
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | - Francisca Esteve Claramunt
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | - Regina Blanco del Burgo
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | | | - Sergio Garcia-Blas
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
- Cardiology Department, Clinic University Hospital of Valencia, 46026 Valencia, Spain
| | - Clara Bonanad
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
- Cardiology Department, Clinic University Hospital of Valencia, 46026 Valencia, Spain
| |
Collapse
|
37
|
Natsheh IY, Alsaleh MM, Alkhawaldeh AK, Albadawi DK, Darwish MM, Shammout MJA. The dark side of drug repurposing. From clinical trial challenges to antimicrobial resistance: analysis based on three major fields. Drug Target Insights 2024; 18:8-19. [PMID: 38751378 PMCID: PMC11094707 DOI: 10.33393/dti.2024.3019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024] Open
Abstract
Drug repurposing is a strategic endeavor that entails the identification of novel therapeutic applications for pharmaceuticals that are already available in the market. Despite the advantageous nature of implementing this particular strategy owing to its cost-effectiveness and efficiency in reducing the time required for the drug discovery process, it is essential to bear in mind that there are various factors that must be meticulously considered and taken into account. Up to this point, there has been a noticeable absence of comprehensive analyses that shed light on the limitations of repurposing drugs. The primary aim of this review is to conduct a thorough illustration of the various challenges that arise when contemplating drug repurposing from a clinical perspective in three major fields-cardiovascular, cancer, and diabetes-and to further underscore the potential risks associated with the emergence of antimicrobial resistance (AMR) when employing repurposed antibiotics for the treatment of noninfectious and infectious diseases. The process of developing repurposed medications necessitates the application of creativity and innovation in designing the development program, as the body of evidence may differ for each specific case. In order to effectively repurpose drugs, it is crucial to consider the clinical implications and potential drawbacks that may arise during this process. By comprehensively analyzing these challenges, we can attain a deeper comprehension of the intricacies involved in drug repurposing, which will ultimately lead to the development of more efficacious and safe therapeutic approaches.
Collapse
Affiliation(s)
- Iyad Y. Natsheh
- Department of Medical Applied Sciences, Zarqa University College, Al-Balqa Applied University, Salt - Jordan
| | - Majd M. Alsaleh
- Department of Medical Applied Sciences, Zarqa University College, Al-Balqa Applied University, Salt - Jordan
- Department of Biology, School of Science, University of Jordan, Amman - Jordan
| | - Ahmad K. Alkhawaldeh
- Department of Medical Applied Sciences, Zarqa University College, Al-Balqa Applied University, Salt - Jordan
| | - Duaa K. Albadawi
- Department of Medical Applied Sciences, Zarqa University College, Al-Balqa Applied University, Salt - Jordan
| | - Maisa’ M. Darwish
- Department of Biology, School of Science, University of Jordan, Amman - Jordan
- National Agricultural Research Center, Amman - Jordan
| | | |
Collapse
|
38
|
Zou LF, Liu DF, Yang H, Zhou CH, Deng SB, Xu NS, He XM, Liu YQ, Shao M, Yu LZ, Liu JS. Salvianolic acids from Salvia miltiorrhiza Bunge and their anti-inflammatory effects through the activation of α7nAchR signaling. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116743. [PMID: 37331452 DOI: 10.1016/j.jep.2023.116743] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/05/2023] [Accepted: 06/05/2023] [Indexed: 06/20/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cardiovascular disease (CVD) is a serious disease with a high incidence rate and mortality. Inflammation is closely related to the occurrence of CVDs. As an essential medicine of promoting blood circulation and removing blood stasis in China, Salvia miltiorrhiza Bunge (Danshen) is widely used to treat CVDs due to its anti-inflammatory and cardiovascular protective effects. Salvianolic acids are the most abundant component in the water extract of S. miltiorrhiza, which has a significant effect on the treatment of CVDs. However, due to the complex composition of salvianolic acids, the active molecules and their underlying mechanisms have not been fully explored. AIM OF THIS STUDY The present study aims to isolate and identify salvianolic acids from Danshen with anti-inflammatory activity and explore the potential mechanisms of isolates. METHODS The structures of isolated salvianolic acids were elucidated by UV, IR, NMR, MS and electronic circular dichroism (ECD) calculations. Then anti-inflammatory activities of isolates were screened out by the zebrafish inflammation models. The most active compound was further used to explore the anti-inflammatory mechanisms on LPS-stimulated RAW 264.7 cells. The key inflammatory cytokines IL-6 and TNF-α were measured by enzyme-linked immunosorbent assay (ELISA). The protein expression levels of STAT3, p-STAT3 (Tyr705), NF-κB p65, IκBα, p-IκBα (Ser32) and α7nAchR were determined by Western blotting. The nuclear translocation of p-STAT3 (Tyr705) and NF-κB p65 was evaluated by immunofluorescence assays. Finally, the in vivo anti-inflammatory mechanisms were investigated by observation of neutrophil migration, H&E staining, survival analysis and quantitative PCR (Q-PCR) in LPS-microinjected zebrafish. RESULTS Two new and four known compounds were isolated from Danshen. Among them, isosalvianolic acid A-1 (C1) and ethyl lithospermate (C5) inhibited neutrophil migrations in three zebrafish inflammation models and C1 with the best activities decreased the secretion of IL-6 and TNF-α and inhibited the expression level of p-IκBα (Ser32) in LPS stimulated RAW 264.7 cells. In addition, C1 also reduced the nuclear translocation of NF-κB p65 and p-STAT3 (Tyr705). Moreover, C1 significantly upregulated the protein expression of α7nAchR, and the knockdown of α7nAchR counteracted the effects of C1 on the production of IL-6 and TNF-α and the expression levels of p-STAT3 (Tyr705), NF-κB p65 and p-IκBα (Ser32). In vivo experiments, C1 decreased the migration and infiltration of inflammatory cells, increased the survival ratio and inhibited the mRNA level of IL-6, TNF-α, STAT3, NF-κB and IκBα in LPS-microinjected zebrafish. CONCLUSION Two new and four known compounds were isolated from Danshen. Among them, C1 exerted anti-inflammatory activities by activating α7nAchR signaling and subsequently inhibiting STAT3 and NF-κB pathways. This study provided evidence for the clinical application of Danshen and contributed to the development of C1 as a novel in the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Li-Fang Zou
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China
| | - Di-Fa Liu
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, School of Pharmacy, Jinan University, Guangzhou, 510632, PR China; State Key Laboratory of Innovative Natural Medicine and TCM Injections, Jiangxi Qingfeng Pharmaceutical Co. Ltd., Ganzhou, 341000, PR China
| | - Hua Yang
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China
| | - Chun-Hong Zhou
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China
| | - Shuang-Bing Deng
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Jiangxi Qingfeng Pharmaceutical Co. Ltd., Ganzhou, 341000, PR China
| | - Ni-Shan Xu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China
| | - Xue-Mei He
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China
| | - Yao-Qi Liu
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Jiangxi Qingfeng Pharmaceutical Co. Ltd., Ganzhou, 341000, PR China
| | - Meng Shao
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China.
| | - Lin-Zhong Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China; Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, PR China.
| | - Jun-Shan Liu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China; Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, PR China.
| |
Collapse
|
39
|
Jouabadi SM, Ataabadi EA, Golshiri K, Bos D, Stricker BHC, Danser AHJ, Mattace-Raso F, Roks AJM. Clinical Impact and Mechanisms of Nonatherosclerotic Vascular Aging: The New Kid to Be Blocked. Can J Cardiol 2023; 39:1839-1858. [PMID: 37495207 DOI: 10.1016/j.cjca.2023.07.022] [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: 05/05/2023] [Revised: 07/07/2023] [Accepted: 07/20/2023] [Indexed: 07/28/2023] Open
Abstract
Ischemic cardiovascular disease and stroke remain the leading cause of global morbidity and mortality. During aging, protective mechanisms in the body gradually deteriorate, resulting in functional, structural, and morphologic changes that affect the vascular system. Because atherosclerotic plaques are not always present along with these alterations, we refer to this kind of vascular aging as nonatherosclerotic vascular aging (NAVA). To maintain proper vascular function during NAVA, it is important to preserve intracellular signalling, prevent inflammation, and block the development of senescent cells. Pharmacologic interventions targeting these components are potential therapeutic approaches for NAVA, with a particular emphasis on inflammation and senescence. This review provides an overview of the pathophysiology of vascular aging and explores potential pharmacotherapies that can improve the function of aged vasculature, focusing on NAVA.
Collapse
Affiliation(s)
- Soroush Mohammadi Jouabadi
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ehsan Ataei Ataabadi
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Keivan Golshiri
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Daniel Bos
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Bruno H C Stricker
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Francesco Mattace-Raso
- Division of Geriatric Medicine, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Anton J M Roks
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
40
|
Tang J, Li T, Xiong X, Yang Q, Su Z, Zheng M, Chen Q. Colchicine delivered by a novel nanoparticle platform alleviates atherosclerosis by targeted inhibition of NF-κB/NLRP3 pathways in inflammatory endothelial cells. J Nanobiotechnology 2023; 21:460. [PMID: 38037046 PMCID: PMC10690998 DOI: 10.1186/s12951-023-02228-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023] Open
Abstract
Atherosclerosis, a chronic inflammatory disease characterized by arterial plaque formation, is one of the most prominent causes of cardiovascular diseases. However, the current treatments often do not adequately compromise the chronic inflammation-mediated plaque accumulation and the disease progression. Therefore, a new and effective strategy that blocks atherosclerosis-associated inflammation is urgently needed to further reduce the risk. Colchicine, a potent anti-inflammatory medication, has shown great potential in the treatment of atherosclerosis, but its adverse effects have hampered its clinical application. Herein, we developed a novel delivery nanosystem encapsulated with colchicine (VHPK-PLGA@COL), which exhibited improved biosafety and sustained drug release along with the gradual degradation of PLGA and PEG as confirmed both in vitro and in vivo. Surface modification of the nanoparticles with the VHPK peptide ensured its capability to specifically target inflammatory endothelial cells and alleviate atherosclerotic plaque accumulation. In the ApoE - / - atherosclerotic mouse model, both colchicine and VHPK-PLGA@COL treatment significantly decreased the plaque area and enhanced plaque stability by blocking the NF-κB/NLRP3 pathways, while VHPK-PLGA@COL exhibited enhanced therapeutic effects due to its unique ability to target inflammatory endothelial cells without obvious long-term safety concerns. In summary, VHPK-PLGA@COL has the potential to overcome the key translational barriers of colchicine and open new avenues to repurpose this drug for anti-atherosclerotic therapy.
Collapse
Affiliation(s)
- Juan Tang
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Department of Endocrinology, The First People's Hospital of Ziyang, Sichuan, 641300, China
| | - Tao Li
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Department of Ophthalmology, The First People's Hospital of Ziyang, Sichuan, 641300, China
| | - Xiaojing Xiong
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Qiaoyun Yang
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Zedazhong Su
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Minming Zheng
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| | - Qingwei Chen
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
41
|
Gao H, Kuang Y, Liu Y, Zhang Y, Wang P, Ma Q. Changes of plasma Rap1A levels in patients with in-stent restenosis after percutaneous coronary intervention and the underlying mechanisms. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2023; 48:1650-1658. [PMID: 38432855 PMCID: PMC10929945 DOI: 10.11817/j.issn.1672-7347.2023.230285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Indexed: 03/05/2024]
Abstract
OBJECTIVES Percutaneous coronary intervention (PCI) is one of the most important treatments for coronary artery disease (CAD). However, in-stent restenosis (ISR) after PCI is a serious complication without effective measures for prevention and treatment. This study aims to investigate the Ras-related protein 1A (Rap1A) level in ISR patients and in the tumor necrosis factor-α (TNF-α)-induced inflammatory injury model of human umbilical vein endothelial cells (HUVECs), to explore the role of Rap1A in regulating TNF-α-induced inflammation in HUVECs and to provide a new potential target for ISR prevention and treatment. METHODS A total of 60 CAD patients, who underwent PCI between December 2020 and July 2022 from the Department of Cardiovascular Medicine of Xiangya Hospital, Central South University, and re-examined coronary angiography (CAG) 1 year after the operation, were included. After admission, 27 patients were diagnosed with ISR and 33 patients were diagnosed with non-in-stent restenosis (non-ISR) according to the CAG. Clinical data were collected, and the plasma Rap1A level was determined by enzyme linked immunosorbent assay (ELISA). In cell experiments, an inflammatory injury model was established with TNF-α treatment (10 ng/mL, 24 h) in HUVECs. The mRNA and protein expression levels of Rap1A, interlukin-6 (IL-6), and vascular cell adhesion molecule-1 (VCAM-1) were measured by real-time reverse transcription PCR and Western blotting. Small interfering RNA (siRNA) was used to explore the role of Rap1A in regulating TNF-α-induced inflammation in HUVECs. RESULTS Compared with the non-ISR patients, a higher proportion of ISR patients had a history of smoking (P=0.005) and diabetes (P=0.028), and higher levels of glycosylated hemoglobin (HbA1c) (P=0.012), low-density lipoprotein cholesterol (LDL-c) (P=0.014), and hypersensitive C-reactive protein (hs-CRP) (P=0.027). The remaining projects did not show significant differences (all P>0.05). The plasma level of Rap1A in the ISR group was significantly higher than that in the non-ISR group [942.14 (873.28 to 1 133.81) μg/mL vs 886.93 (812.61 to 930.98) μg/mL; P=0.004]. Diabetes, LDL-c, and Rap1A were risk factors for ISR by univariate logistic regression analysis (all P<0.05). The mRNA and protein expression levels of inflammatory factors IL-6 and VCAM-1 were increased in HUVECs after 10 ng/mL TNF-α treatment for 24 h compared with the control group (all P<0.05), while the mRNA and protein levels of Rap1A were increased (both P<0.05). After inhibition of Rap1A in HUVECs, the mRNA and protein expression levels of IL-6 and VCAM-1 were significantly decreased (all P<0.05). CONCLUSIONS The plasma Rap1A level was significantly elevated in patients with ISR, suggesting that Rap1A may be a potential biomarker for predicting ISR. In the TNF-α- induced HUVECs inflammatory injury model, the expression level of Rap1A was increased. The level of TNF-α-induced endothelial cell inflammation was decreased after inhibition of Rap1A expression, suggesting that Rap1A may be a potential target for the treatment of endothelial cell inflammation in ISR.
Collapse
Affiliation(s)
- Haodong Gao
- Department of Cardiology, Xiangya Hospital, Central South University; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008.
| | - Yuanyuan Kuang
- Department of Cardiology, Xiangya Hospital, Central South University; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008
| | - Yubo Liu
- Department of Cardiology, Xiangya Hospital, Central South University; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008
| | - Yinzhuang Zhang
- Department of Cardiology, First Hospital of Changsha, Changsha 410005, China
| | - Ping Wang
- Department of Cardiology, Xiangya Hospital, Central South University; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008
| | - Qilin Ma
- Department of Cardiology, Xiangya Hospital, Central South University; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008.
| |
Collapse
|
42
|
Cheng DCY, Climie RE, Shu M, Grieve SM, Kozor R, Figtree GA. Vascular aging and cardiovascular disease: pathophysiology and measurement in the coronary arteries. Front Cardiovasc Med 2023; 10:1206156. [PMID: 38089775 PMCID: PMC10715672 DOI: 10.3389/fcvm.2023.1206156] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 11/13/2023] [Indexed: 11/29/2024] Open
Abstract
Age is a key risk factor for cardiovascular disease, including atherosclerosis. However, pathophysiological disease processes in the arteries are not an inevitable feature of aging. Large cohort studies with arterial phenotyping along with clinical and demographic data are essential to better understand factors related to the susceptibility or resilience to age-related vascular pathophysiology in humans. This review explores the mechanisms by which vascular structure and function alters with age, and how these changes relate to cardiovascular pathophysiology and disease. Features of vascular aging in the coronary arteries have historically been difficult to quantify pre-mortem due to their size and location. However, non-invasive imaging modalities including CT Coronary Angiogram are now being used to assess coronary vascular age, and further advances in imaging analysis such as the CT Fat Attenuation Index will help provide further measurement of features associated with coronary vascular aging. Currently, markers of vascular aging are not used as therapeutic targets in routine clinical practice, but non-pharmacological interventions including aerobic exercise and low salt diet, as well as anti-hypertensives have been demonstrated to reduce arterial stiffness. Advances in imaging technology, both in acquisition and advanced analysis, as well as harmonisation of measurements for researchers across the globe will be invaluable in understanding what constitutes healthy vascular aging and in identifying features of vascular aging that are associated with coronary artery disease and its adverse outcomes. Assessing such images in large cohorts can facilitate improved definitions of resilient and susceptible phenotypes to vascular aging in the coronary arteries. This is a critical step in identifying further risk factors and biomarkers within these groups and driving forward the development of novel therapies aimed at slowing or stopping age-related vascular changes in the coronary arteries.
Collapse
Affiliation(s)
- Daniel C. Y. Cheng
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Rachel E. Climie
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Matthew Shu
- Northern Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Stuart M. Grieve
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
- Imaging and Phenotyping Laboratory, Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Rebecca Kozor
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
- Department of Cardiology, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Gemma A. Figtree
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
- Imaging and Phenotyping Laboratory, Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Department of Cardiology, Royal North Shore Hospital, Sydney, NSW, Australia
| |
Collapse
|
43
|
Loescher CM, Freundt JK, Unger A, Hessel AL, Kühn M, Koser F, Linke WA. Titin governs myocardial passive stiffness with major support from microtubules and actin and the extracellular matrix. NATURE CARDIOVASCULAR RESEARCH 2023; 2:991-1002. [PMID: 39196092 PMCID: PMC11358001 DOI: 10.1038/s44161-023-00348-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 09/19/2023] [Indexed: 08/29/2024]
Abstract
Myocardial passive stiffness is crucial for the heart's pump function and is determined by mechanical elements, including the extracellular matrix and cytoskeletal filaments; however, their individual contributions are controversially discussed and difficult to quantify. In this study, we targeted the cytoskeletal filaments in a mouse model, which enables the specific, acute and complete cleavage of the sarcomeric titin springs. We show in vitro that each cytoskeletal filament's stiffness contribution varies depending on whether the elastic or the viscous forces are considered and on strain level. Titin governs myocardial elastic forces, with the largest contribution provided at both low and high strain. Viscous force contributions are more uniformly distributed among the microtubules, titin and actin. The extracellular matrix contributes at high strain. The remaining forces after total target element disruption are likely derived from desmin filaments. Our findings answer longstanding questions about cardiac mechanical architecture and allow better targeting of passive myocardial stiffness in heart failure.
Collapse
Affiliation(s)
| | - Johanna K Freundt
- Institute of Physiology II, University of Muenster, Muenster, Germany
| | - Andreas Unger
- Institute of Physiology II, University of Muenster, Muenster, Germany
| | - Anthony L Hessel
- Institute of Physiology II, University of Muenster, Muenster, Germany
| | - Michel Kühn
- Institute of Physiology II, University of Muenster, Muenster, Germany
| | - Franziska Koser
- Institute of Physiology II, University of Muenster, Muenster, Germany
| | - Wolfgang A Linke
- Institute of Physiology II, University of Muenster, Muenster, Germany.
| |
Collapse
|
44
|
Lin JJ, Lin CL, Chen CC, Lin YH, Cho DY, Chen X, Chen DC, Chen HY. Unlocking Colchicine's Untapped Potential: A Paradigm Shift in Hepatocellular Carcinoma Prevention. Cancers (Basel) 2023; 15:5031. [PMID: 37894398 PMCID: PMC10605746 DOI: 10.3390/cancers15205031] [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: 09/07/2023] [Revised: 10/05/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Background: Liver cancer and notably hepatocellular carcinoma (HCC), results in significantly high mortality rates worldwide. Chronic hepatitis and fatty liver, recognized precursors, underscore the imperative need for effective preventive strategies. This study explores colchicine, traditionally acknowledged for its anti-inflammatory properties and investigates its potential in liver cancer prevention. Methods: Utilizing the iHi Data Platform of China Medical University Hospital, Taiwan, this study analyzed two decades of medical data, incorporating 10,353 patients each in the Colchicine and Non-Colchicine cohorts, to investigate the association between colchicine use and liver cancer risk. Results: The study identified that colchicine users exhibited a 19% reduction in liver cancer risk, with a multivariable-adjusted odds ratio of 0.81 after accounting for confounding variables. Additionally, the influence of gender and comorbidities like diabetes mellitus on liver cancer risk was identified, corroborating the existing literature. A notable finding was that the prolonged use of colchicine was associated with improved outcomes, indicating a potential dose-response relationship. Conclusions: This study proposes a potential new role for colchicine in liver cancer prevention, extending beyond its established anti-inflammatory applications. While the findings are promising, further research is essential to validate these results. This research may serve as a foundation for future studies, aiming to further explore colchicine's role via clinical trials and in-depth investigations, potentially impacting preventive strategies for liver cancer.
Collapse
Affiliation(s)
- Jung-Ju Lin
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan;
| | - Cheng-Li Lin
- Management Office for Health Data, China Medical University Hospital, Taichung 404, Taiwan;
| | - Chun-Chung Chen
- Department of Neurosurgery, China Medical University Hospital, Taichung 404, Taiwan; (C.-C.C.); (Y.-H.L.); (D.-Y.C.); (X.C.)
- Graduate Institute of Integrated Medicine, China Medical University, Taichung 404, Taiwan
- Graduate Institute of Acupuncture Science, China Medical University, Taichung 404, Taiwan
- Neuroscience and Brain Disease Center, China Medical University, Taichung 404, Taiwan
| | - Yu-Hsiang Lin
- Department of Neurosurgery, China Medical University Hospital, Taichung 404, Taiwan; (C.-C.C.); (Y.-H.L.); (D.-Y.C.); (X.C.)
| | - Der-Yang Cho
- Department of Neurosurgery, China Medical University Hospital, Taichung 404, Taiwan; (C.-C.C.); (Y.-H.L.); (D.-Y.C.); (X.C.)
| | - XianXiu Chen
- Department of Neurosurgery, China Medical University Hospital, Taichung 404, Taiwan; (C.-C.C.); (Y.-H.L.); (D.-Y.C.); (X.C.)
- Neuroscience and Brain Disease Center, China Medical University, Taichung 404, Taiwan
| | - Der-Cherng Chen
- Department of Neurosurgery, China Medical University Hospital, Taichung 404, Taiwan; (C.-C.C.); (Y.-H.L.); (D.-Y.C.); (X.C.)
| | - Hung-Yao Chen
- School of Medicine, China Medical University, Taichung 404, Taiwan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan
| |
Collapse
|
45
|
Zhao Y, Shen QR, Chen YX, Shi Y, Wu WB, Li Q, Li DJ, Shen FM, Fu H. Colchicine protects against the development of experimental abdominal aortic aneurysm. Clin Sci (Lond) 2023; 137:1533-1545. [PMID: 37748024 PMCID: PMC10550771 DOI: 10.1042/cs20230499] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 09/27/2023]
Abstract
Abdominal aortic aneurysm (AAA) is characterized by at least 1.5-fold enlargement of the infrarenal aorta, a ruptured AAA is life-threatening. Colchicine is a medicine used to treat gout and familial Mediterranean fever, and recently, it was approved to reduce the risk of cardiovascular events in adult patients with established atherosclerotic disease. With an AAA mice model created by treatment with porcine pancreatic elastase (PPE) and β-aminopropionitrile (BAPN), this work was designed to explore whether colchicine could protect against the development of AAA. Here, we showed that colchicine could limit AAA formation, as evidenced by the decreased total aortic weight per body weight, AAA incidence, maximal abdominal aortic diameter and collagen deposition. We also found that colchicine could prevent the phenotypic switching of vascular smooth muscle cells from a contractile to synthetic state during AAA. In addition, it was demonstrated that colchicine was able to reduce vascular inflammation, oxidative stress, cell pyroptosis and immune cells infiltration to the aortic wall in the AAA mice model. Finally, it was proved that the protective action of colchicine against AAA formation was mainly mediated by preventing immune cells infiltration to the aortic wall. In summary, our findings demonstrated that colchicine could protect against the development of experimental AAA, providing a potential therapeutic strategy for AAA intervention in the clinic.
Collapse
Affiliation(s)
- Yi Zhao
- Department of Pharmacy, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qi-Rui Shen
- Department of Pharmacy, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu-Xin Chen
- Department of Pharmacy, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu Shi
- Department of Pharmacy, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen-Bing Wu
- Department of Pharmacology, School of Pharmacy, Second Military Medical University/ Naval Medical University, Shanghai, China
| | - Qiao Li
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Dong-Jie Li
- Department of Pharmacy, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fu-Ming Shen
- Department of Pharmacy, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hui Fu
- Department of Pharmacy, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
46
|
Gracheva IA, Schmalz HG, Svirshchevskaya EV, Shchegravina ES, Fedorov AY. Design of an aryne-platform for the synthesis of non-racemic heterocyclic allocolchicinoids. Org Biomol Chem 2023; 21:6141-6150. [PMID: 37458676 DOI: 10.1039/d3ob00827d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
A four-step semisynthetic approach towards a highly versatile allocolchicine-related chiral aryne intermediate starting from naturally occurring colchicine was developed, and some of its synthetic transformations were studied. The in situ generated benzyne intermediate afforded a number of non-racemic heterocyclic allocolchicinoids, which were shown to exhibit potent cytotoxicity towards COLO 357, OSA and Raji cells. The proposed methodology is attractive for the synthesis of libraries of new cytotoxic tubulin inhibitors.
Collapse
Affiliation(s)
- Iuliia A Gracheva
- Department of Organic Chemistry, National Research Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Aenue, 603950 Nizhny Novgorod, Russian Federation.
| | - Hans-Günther Schmalz
- Department of Chemistry, University of Cologne, Greinstrasse 4, 50939 Cologne, Germany
| | - Elena V Svirshchevskaya
- Department of Organic Chemistry, National Research Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Aenue, 603950 Nizhny Novgorod, Russian Federation.
- Laboratory of Cell Interactions, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 16/10 Miklukho-Maklaya Street, 117997 Moscow, Russian Federation
| | - Ekaterina S Shchegravina
- Department of Organic Chemistry, National Research Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Aenue, 603950 Nizhny Novgorod, Russian Federation.
| | - Alexey Yu Fedorov
- Department of Organic Chemistry, National Research Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Aenue, 603950 Nizhny Novgorod, Russian Federation.
| |
Collapse
|
47
|
Ma W, Zhu J, Bai L, Zhao P, Li F, Zhang S. The role of neutrophil extracellular traps and proinflammatory damage-associated molecular patterns in idiopathic inflammatory myopathies. Clin Exp Immunol 2023; 213:202-208. [PMID: 37289984 PMCID: PMC10361739 DOI: 10.1093/cei/uxad059] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/13/2023] [Accepted: 05/25/2023] [Indexed: 06/10/2023] Open
Abstract
Idiopathic inflammatory myopathies (IIMs) are a group of systemic autoimmune diseases characterized by immune-mediated muscle injury. Abnormal neutrophil extracellular traps (NETs) can be used as a biomarker of IIM disease activity, but the mechanism of NET involvement in IIMs needs to be elucidated. Important components of NETs, including high-mobility group box 1, DNA, histones, extracellular matrix, serum amyloid A, and S100A8/A9, act as damage-associated molecular patterns (DAMPs) to promote inflammation in IIMs. NETs can act on different cells to release large amounts of cytokines and activate the inflammasome, which can subsequently aggravate the inflammatory response. Based on the idea that NETs may be proinflammatory DAMPs of IIMs, we describe the role of NETs, DAMPs, and their interaction in the pathogenesis of IIMs and discuss the possible targeted treatment strategies in IIMs.
Collapse
Affiliation(s)
- Wenlan Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jiarui Zhu
- Department of Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Ling Bai
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Peipei Zhao
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Feifei Li
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Sigong Zhang
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
48
|
Yu Y, Zhou M, Long X, Yin S, Hu G, Yang X, Jian W, Yu R. Study on the mechanism of action of colchicine in the treatment of coronary artery disease based on network pharmacology and molecular docking technology. Front Pharmacol 2023; 14:1147360. [PMID: 37405052 PMCID: PMC10315633 DOI: 10.3389/fphar.2023.1147360] [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: 01/18/2023] [Accepted: 06/07/2023] [Indexed: 07/06/2023] Open
Abstract
Objective: This is the first study to explore the mechanism of colchicine in treating coronary artery disease using network pharmacology and molecular docking technology, aiming to predict the key targets and main approaches of colchicine in treating coronary artery disease. It is expected to provide new ideas for research on disease mechanism and drug development. Methods: Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP), Swiss Target Prediction and PharmMapper databases were used to obtain drug targets. GeneCards, Online Mendelian Inheritance in Man (OMIM), Therapeutic Target Database (TTD), DrugBank and DisGeNET databases were utilized to gain disease targets. The intersection of the two was taken to access the intersection targets of colchicine for the treatment of coronary artery disease. The Sting database was employed to analyze the protein-protein interaction network. Gene Ontology (GO) functional enrichment analysis was performed using Webgestalt database. Reactom database was applied for Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Molecular docking was simulated using AutoDock 4.2.6 and PyMOL2.4 software. Results: A total of 70 intersecting targets of colchicine for the treatment of coronary artery disease were obtained, and there were interactions among 50 targets. GO functional enrichment analysis yielded 13 biological processes, 18 cellular components and 16 molecular functions. 549 signaling pathways were obtained by KEGG enrichment analysis. The molecular docking results of key targets were generally good. Conclusion: Colchicine may treat coronary artery disease through targets such as Cytochrome c (CYCS), Myeloperoxidase (MPO) and Histone deacetylase 1 (HDAC1). The mechanism of action may be related to the cellular response to chemical stimulus and p75NTR-mediated negative regulation of cell cycle by SC1, which is valuable for further research exploration. However, this research still needs to be verified by experiments. Future research will explore new drugs for treating coronary artery disease from these targets.
Collapse
Affiliation(s)
- Yunfeng Yu
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Manli Zhou
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xi Long
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Shuang Yin
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Gang Hu
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xinyu Yang
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Weixiong Jian
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Chinese Medicine Diagnostics in Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Rong Yu
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
49
|
Immanuel J, Yun S. Vascular Inflammatory Diseases and Endothelial Phenotypes. Cells 2023; 12:1640. [PMID: 37371110 PMCID: PMC10297687 DOI: 10.3390/cells12121640] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/06/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
The physiological functions of endothelial cells control vascular tone, permeability, inflammation, and angiogenesis, which significantly help to maintain a healthy vascular system. Several cardiovascular diseases are characterized by endothelial cell activation or dysfunction triggered by external stimuli such as disturbed flow, hypoxia, growth factors, and cytokines in response to high levels of low-density lipoprotein and cholesterol, hypertension, diabetes, aging, drugs, and smoking. Increasing evidence suggests that uncontrolled proinflammatory signaling and further alteration in endothelial cell phenotypes such as barrier disruption, increased permeability, endothelial to mesenchymal transition (EndMT), and metabolic reprogramming further induce vascular diseases, and multiple studies are focusing on finding the pathways and mechanisms involved in it. This review highlights the main proinflammatory stimuli and their effects on endothelial cell function. In order to provide a rational direction for future research, we also compiled the most recent data regarding the impact of endothelial cell dysfunction on vascular diseases and potential targets that impede the pathogenic process.
Collapse
Affiliation(s)
| | - Sanguk Yun
- Department of Biotechnology, Inje University, Gimhae-si 50834, Republic of Korea;
| |
Collapse
|
50
|
Work M, Scudder C, Bergum Hjellegjerde K, Dunning M, Gajanayake I, Kent A, Tintle L, Sparks T, Allerton F. A survey on Shar Pei autoinflammatory disease in the United Kingdom. J Small Anim Pract 2023; 64:401-408. [PMID: 36978210 DOI: 10.1111/jsap.13602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/03/2022] [Accepted: 01/16/2023] [Indexed: 03/30/2023]
Abstract
OBJECTIVES To characterise the fever episodes attributed to Shar Pei autoinflammatory disease and to identify common diagnostic and management strategies in the United Kingdom. A secondary objective was to determine risk factors associated with Shar Pei autoinflammatory disease fever episodes. METHODS A retrospective survey was performed to characterise episodes of Shar Pei autoinflammatory disease fever and to identify commonly used treatments in affected dogs. Clinical data were collected from owners and veterinarians. Frequencies of previously proposed risk factors (skin thickness and folding, muzzle conformation) and comorbid conditions were compared between dogs that had exhibited fever episodes consistent with Shar Pei autoinflammatory disease and those who had not. RESULTS At least one episode of fever attributed to Shar Pei autoinflammatory disease was reported in 52 of 106 (49%) Shar Pei. Nine other dogs had fever episodes consistent with Shar Pei autoinflammatory disease reported by their owners but not by veterinarians. Median rectal temperature at presentation for Shar Pei autoinflammatory disease fever was 40.1°C [104.2°F] (39.9 to 41.3°C [103.8 to 106.3°F]) and owners reported associated hyporexia (n=33, 63%) and vomiting (n=8, 15%) more frequently than veterinary records (n=22, 42% and n=0, 0%, respectively). The median number of veterinary appointments for Shar Pei autoinflammatory disease was two per dog (1 to 15) while owners reported a median of four episodes per dog per year. None of the assessed phenotypic variants or comorbidities were significantly associated with exhibiting Shar Pei autoinflammatory disease fever episodes. CLINICAL SIGNIFICANCE Episodes of Shar Pei autoinflammatory disease fever were reported approximately twice as frequently by owners compared to veterinary records, suggesting the burden of this condition may be underestimated by veterinarians. Specific risk factors for Shar Pei autoinflammatory disease fever were not identified.
Collapse
Affiliation(s)
- M Work
- Willows Veterinary Centre and Referral Service, Solihull, UK
| | - C Scudder
- Southfields Veterinary Specialists, Essex, UK
- Royal Veterinary College, Potters Bar, Hertfordshire, UK
| | | | - M Dunning
- Willows Veterinary Centre and Referral Service, Solihull, UK
| | - I Gajanayake
- Willows Veterinary Centre and Referral Service, Solihull, UK
| | - A Kent
- Willows Veterinary Centre and Referral Service, Solihull, UK
| | - L Tintle
- Wurtsboro Veterinary Clinic, Wurtsboro, New York, USA
| | - T Sparks
- Waltham Petcare Science Institute, Leicestershire, UK
| | - F Allerton
- Willows Veterinary Centre and Referral Service, Solihull, UK
| |
Collapse
|